1
|
Agrawal R, Sengupta S. p53 regulates DREAM complex-mediated repression in a p21-independent manner. EMBO J 2025; 44:2279-2297. [PMID: 40038454 PMCID: PMC12000331 DOI: 10.1038/s44318-025-00402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 01/18/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The DREAM repressor complex regulates genes involved in the cell cycle and DNA repair, vital for maintaining genome stability. Although it mediates p53-driven repression through the canonical p53-p21-Rb axis, the potential for p53 to directly regulate DREAM targets independently of its transcriptional activity has not been explored. Here, we demonstrate that in asynchronously growing cells, p53 loss leads to greater de-repression of DREAM targets compared to p21 loss alone. Both wild-type and transactivation-deficient p53 mutants are capable of repressing DREAM targets, suggesting a transactivation-independent "non-canonical" repression mechanism. These p53 variants bind p130/p107, irrespective of their phosphorylation status, while cancer-associated p53 mutants disrupt DREAM complex function by sequestering E2F4. Re-ChIP analysis shows co-recruitment of p53 and E2F4 to known and newly identified DREAM target promoters, indicating direct repression of these targets by p53. These findings reveal a novel, transactivation-independent mechanism of p53-mediated repression, expanding our understanding of p53's tumor-suppressive functions and suggesting DREAM complex targeting as potential future avenues in cancer therapy.
Collapse
Affiliation(s)
- Ritu Agrawal
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sagar Sengupta
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India.
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), PO: NSS, Kalyani, 741251, India.
| |
Collapse
|
2
|
Wang Y, Lv Y, Wen Y, Wang J, Hu P, Wu K, Chai B, Gan S, Liu J, Wu Y, Zhu L, Dong N, Tan Y, Wu H, Zhang G, Zhu L, Ren D, Zhang Q, Wang Y, Qian Q, Hu J. GS2 cooperates with IPA1 to control panicle architecture. THE NEW PHYTOLOGIST 2025; 245:2726-2743. [PMID: 39887382 PMCID: PMC11840411 DOI: 10.1111/nph.20412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
Panicle size and grain number are important agronomic traits that determine grain yield in rice. However, the underlying mechanism regulating panicle size and grain number remains largely unknown. Here, we report that GS2 plays an important role in regulating panicle architecture. The RNAi of GS2™ (target site mutation, TM) produced erect and dense panicle with increased primary and secondary branches and grain number per panicle, whereas the overexpression of GS2™ showed longer panicles and fewer grains than wild-type. GS2 directly binds to the GCCA motif and significantly enhances the transcriptional activation ability through the interaction with IPA1. DEP1 is a common target gene of GS2 and IPA1 in regulating branch number and grain number per panicle. The pyramiding of GS2™ and IPA1™1 (Target site mutation1, TM1) on hybrid rice can significantly increase rice yield. Our findings reveal the novel function of GS2 and the molecular mechanism of GS2/IPA1-DEP1 module in controlling panicle architecture.
Collapse
Affiliation(s)
- Yueying Wang
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Yang Lv
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Yi Wen
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Junge Wang
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Peng Hu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Kaixiong Wu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Bingze Chai
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Shuxian Gan
- Institute of Agricultural Sciences, Xishuangbanna PrefectureJinghongYunnan Province666100China
| | - Jialong Liu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Yue Wu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Lixin Zhu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Nannan Dong
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Yiqing Tan
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Hao Wu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Guangheng Zhang
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
- Academician Workstation, National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural SciencesSanya572024China
| | - Li Zhu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Deyong Ren
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Qiang Zhang
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
- Academician Workstation, National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural SciencesSanya572024China
| | - Yuexing Wang
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
| | - Qian Qian
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
- Academician Workstation, National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural SciencesSanya572024China
| | - Jiang Hu
- State Key Laboratory of Rice Biology and BreedingChina National Rice Research InstituteHangzhou311401China
- Academician Workstation, National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural SciencesSanya572024China
- Hainan Seed Industry LaboratorySanya572024China
| |
Collapse
|
3
|
Zhang J, Lin Q, Wang X, Shao J, Ren Y, Liu X, Feng M, Li S, Sun Q, Luo S, Liu B, Xing X, Chang Y, Cheng Z, Wan J. The DENSE AND ERECT PANICLE1-GRAIN NUMBER ASSOCIATED module enhances rice yield by repressing CYTOKININ OXIDASE 2 expression. THE PLANT CELL 2024; 37:koae309. [PMID: 39660553 DOI: 10.1093/plcell/koae309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024]
Abstract
The phytohormone cytokinin (CK) positively regulates the activity of the inflorescence meristem (IM). Cytokinin oxidase 2/Grain number 1a (OsCKX2/Gn1a)-mediated degradation of CK in rice (Oryza sativa L.) negatively regulates panicle grain number, whereas DENSE AND ERECT PANICLE 1 (DEP1) positively regulates grain number per panicle (GNP). However, the detailed regulatory mechanism between DEP1 and OsCKX2 remains elusive. Here, we report the GRAS (GIBBERELLIN ACID INSENSITIVE, REPRESSOR OF GA1, and SCARECROW) transcription factor GRAIN NUMBER ASSOCIATED (GNA), previously thought to be involved in the Brassinosteroids (BRs) signaling pathway, directly inhibits OsCKX2 expression in the IM through a DEP1-GNA regulatory module. Overexpressing GNA leads to increased CK levels and consequently higher branch number, GNP, and yield. Both DEP1 and dep1 enhance the inhibitory effect of GNA on OsCKX2 expression through interacting with GNA. GNA promotes the translocation of DEP1 to the nucleus, while the gain-of-function mutant dep1 translocates into the nucleus in the absence of GNA. Our findings provide insight into the regulatory mechanism underlying OsCKX2 and a strategy to improve rice yield.
Collapse
Affiliation(s)
- Jinhui Zhang
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Qibing Lin
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Xin Wang
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Jiale Shao
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Yulong Ren
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Xin Liu
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Miao Feng
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Shuai Li
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Qi Sun
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Sheng Luo
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Bojuan Liu
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Xinxin Xing
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Yanqi Chang
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
| | - Zhijun Cheng
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
- Chinese Academy of Agricultural Sciences, Nanfan Research Institute, Sanya 572025, China
| | - Jianmin Wan
- State Key Laboratory of Crop Gene Resources and Breeding, Chinese Academy of Agricultural Sciences, Institute of Crop Sciences, Beijing 100081, China
- State Key Laboratory for Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China
- Zhongshan Biological Breeding Laboratory, Nanjing 210014, China
| |
Collapse
|
4
|
Pradhan S, Bush K, Zhang N, Pandita RK, Tsai CL, Smith C, Pandlebury DF, Gaikwad S, Leonard F, Nie L, Tao A, Russell W, Yuan S, Choudhary S, Ramos KS, Elferink C, Wairkar YP, Tainer JA, Thompson LM, Pandita TK, Sarkar PS. Chromatin remodeler BRG1 recruits huntingtin to repair DNA double-strand breaks in neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613927. [PMID: 39345557 PMCID: PMC11429940 DOI: 10.1101/2024.09.19.613927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Persistent DNA double-strand breaks (DSBs) are enigmatically implicated in neurodegenerative diseases including Huntington's disease (HD), the inherited late-onset disorder caused by CAG repeat elongations in Huntingtin (HTT). Here we combine biochemistry, computation and molecular cell biology to unveil a mechanism whereby HTT coordinates a Transcription-Coupled Non-Homologous End-Joining (TC-NHEJ) complex. HTT joins TC-NHEJ proteins PNKP, Ku70/80, and XRCC4 with chromatin remodeler Brahma-related Gene 1 (BRG1) to resolve transcription-associated DSBs in brain. HTT recruitment to DSBs in transcriptionally active gene- rich regions is BRG1-dependent while efficient TC-NHEJ protein recruitment is HTT-dependent. Notably, mHTT compromises TC-NHEJ interactions and repair activity, promoting DSB accumulation in HD tissues. Importantly, HTT or PNKP overexpression restores TC-NHEJ in a Drosophila HD model dramatically improving genome integrity, motor defects, and lifespan. Collective results uncover HTT stimulation of DSB repair by organizing a TC-NHEJ complex that is impaired by mHTT thereby implicating dysregulation of transcription-coupled DSB repair in mHTT pathophysiology. Highlights BRG1 recruits HTT and NHEJ components to transcriptionally active DSBs.HTT joins BRG1 and PNKP to efficiently repair transcription related DSBs in brain.Mutant HTT impairs the functional integrity of TC-NHEJ complex for DSB repair.HTT expression improves DSB repair, genome integrity and phenotypes in HD flies.
Collapse
|
5
|
Xiong S, Zhou J, Tan TK, Chung TH, Tan TZ, Toh SHM, Tang NXN, Jia Y, See YX, Fullwood MJ, Sanda T, Chng WJ. Super enhancer acquisition drives expression of oncogenic PPP1R15B that regulates protein homeostasis in multiple myeloma. Nat Commun 2024; 15:6810. [PMID: 39122682 PMCID: PMC11316114 DOI: 10.1038/s41467-024-50910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Multiple myeloma is a hematological malignancy arising from immunoglobulin-secreting plasma cells. It remains poorly understood how chromatin rewiring of regulatory elements contributes to tumorigenesis and therapy resistance in myeloma. Here we generate a high-resolution contact map of myeloma-associated super-enhancers by integrating H3K27ac ChIP-seq and HiChIP from myeloma cell lines, patient-derived myeloma cells and normal plasma cells. Our comprehensive transcriptomic and phenomic analyses prioritize candidate genes with biological and clinical implications in myeloma. We show that myeloma cells frequently acquire SE that transcriptionally activate an oncogene PPP1R15B, which encodes a regulatory subunit of the holophosphatase complex that dephosphorylates translation initiation factor eIF2α. Epigenetic silencing or knockdown of PPP1R15B activates pro-apoptotic eIF2α-ATF4-CHOP pathway, while inhibiting protein synthesis and immunoglobulin production. Pharmacological inhibition of PPP1R15B using Raphin1 potentiates the anti-myeloma effect of bortezomib. Our study reveals that myeloma cells are vulnerable to perturbation of PPP1R15B-dependent protein homeostasis, highlighting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Sinan Xiong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jianbiao Zhou
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Tae-Hoon Chung
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Sabrina Hui-Min Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Nicole Xin Ning Tang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yunlu Jia
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yi Xiang See
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Melissa Jane Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Takaomi Sanda
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wee-Joo Chng
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), National University Health System (NUHS), Singapore, Singapore.
| |
Collapse
|
6
|
Sun H, Ge Y, Liu J, Li Z, Li H, Zhao T, Wang X, Feng Y, Wang H, Gao S, Shi L, Yang S, Sun P, Chang A, Hao J, Huang C. Tumor-derived interleukin 35 mediates the dissemination of gemcitabine resistance in pancreatic adenocarcinoma. Oncogene 2024; 43:776-788. [PMID: 38243080 DOI: 10.1038/s41388-024-02938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
Rapid development of drug resistance after chemotherapy is a major cause of treatment failure in individuals with pancreatic ductal adenocarcinoma (PDAC). In this study, we illustrate that tumor-derived interleukin 35 (IL-35) mediates the accelerated resistance of PDAC to gemcitabine (GEM). We observe that GEM resistance can spread from GEM-resistant PDAC cells to GEM-sensitive cells, and that IL-35 is responsible for the propagation of chemoresistance, which is supported by sequencing and experimental data. Additionally, we discover that GEM-resistant cells have significantly higher levels of IL-35 expression. Mechanistically, aberrantly expressed IL-35 triggers transcriptional activation of SOD2 expression via GP130-STAT1 signaling, scavenging reactive oxygen species (ROS) and leading to GEM resistance. Furthermore, GEM treatment stimulates IL-35 expression through activation of the NF-κB pathway, resulting in acquired chemoresistance. In the mouse model, a neutralizing antibody against IL-35 enhances the tumor suppressive effect of GEM. Collectively, our data suggests that IL-35 is critical in mediating GEM resistance in pancreatic cancer, and therefore could be a valuable therapeutic target in overcoming PDAC chemoresistance.
Collapse
Affiliation(s)
- Huizhi Sun
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yi Ge
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jing Liu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zengxun Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hui Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Tiansuo Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiuchao Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hongwei Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Song Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shengyu Yang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Antao Chang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Chongbiao Huang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
7
|
Seneviratne JA, Ho WWH, Glancy E, Eckersley-Maslin MA. A low-input high resolution sequential chromatin immunoprecipitation method captures genome-wide dynamics of bivalent chromatin. Epigenetics Chromatin 2024; 17:3. [PMID: 38336688 PMCID: PMC10858499 DOI: 10.1186/s13072-024-00527-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/11/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Bivalent chromatin is an exemplar of epigenetic plasticity. This co-occurrence of active-associated H3K4me3 and inactive-associated H3K27me3 histone modifications on opposite tails of the same nucleosome occurs predominantly at promoters that are poised for future transcriptional upregulation or terminal silencing. We know little of the dynamics, resolution, and regulation of this chromatin state outside of embryonic stem cells where it was first described. This is partly due to the technical challenges distinguishing bone-fide bivalent chromatin, where both marks are on the same nucleosome, from allelic or sample heterogeneity where there is a mix of H3K4me3-only and H3K27me3-only mononucleosomes. RESULTS Here, we present a robust and sensitive method to accurately map bivalent chromatin genome-wide, along with controls, from as little as 2 million cells. We optimized and refined the sequential ChIP protocol which uses two sequential overnight immunoprecipitation reactions to robustly purify nucleosomes that are truly bivalent and contain both H3K4me3 and H3K27me3 modifications. Our method generates high quality genome-wide maps with strong peak enrichment and low background, which can be analyzed using standard bioinformatic packages. Using this method, we detect 8,789 bivalent regions in mouse embryonic stem cells corresponding to 3,918 predominantly CpG rich and developmentally regulated gene promoters. Furthermore, profiling Dppa2/4 knockout mouse embryonic stem cells, which lose both H3K4me3 and H3K27me3 at approximately 10% of bivalent promoters, demonstrated the ability of our method to capture bivalent chromatin dynamics. CONCLUSIONS Our optimized sequential reChIP method enables high-resolution genome-wide assessment of bivalent chromatin together with all required controls in as little as 2 million cells. We share a detailed protocol and guidelines that will enable bivalent chromatin landscapes to be generated in a range of cellular contexts, greatly enhancing our understanding of bivalent chromatin and epigenetic plasticity beyond embryonic stem cells.
Collapse
Affiliation(s)
- Janith A Seneviratne
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, 3010, Australia
| | - William W H Ho
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Eleanor Glancy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, 3010, Australia
| | - Melanie A Eckersley-Maslin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, 3010, Australia.
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
8
|
Liu C, Shen A, Song J, Cheng L, Zhang M, Wang Y, Liu X. LncRNA-CCAT5-mediated crosstalk between Wnt/β-Catenin and STAT3 signaling suggests novel therapeutic approaches for metastatic gastric cancer with high Wnt activity. Cancer Commun (Lond) 2024; 44:76-100. [PMID: 38010289 PMCID: PMC10794011 DOI: 10.1002/cac2.12507] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Although the constitutively activated Wnt/β-catenin signaling pathway plays vital roles in gastric cancer (GC) progression, few Wnt inhibitors are approved for clinical use. Additionally, the clinical significance of long non-coding RNAs (lncRNAs) in GC intraperitoneal dissemination (IPD) remains elusive. Here, we investigated the function and therapeutic potential of Wnt-transactivated lncRNA, colon cancer-associated transcript 5 (CCAT5), in GC metastasis. METHODS LncRNA-sequencing assay was performed to document abundance changes of lncRNAs induced by Wnt family member 3A (Wnt3a) and degradation-resistant β-catenin (S33Y mutated) in ascites-derived GC cells with low Wnt activity. Luciferase reporter, Chromatin immunoprecipitation (ChIP)-re-ChIP assays were performed to determine how CCAT5 was transcribed. The clinical significance of CCAT5 was examined in 2 cohorts of GC patients. The biological function of CCAT5 was investigated through gain- and loss-of-function studies. The molecular mechanism was explored through RNA-sequencing, mass spectrometry, and CRISPR/Cas9-knocknout system. The therapeutic potential of CCAT5 was examined through RNAi-based cell xenograft model and patient-derived xenograft (PDX) model of IPD. RESULTS We identified a novel Wnt-regulated lncRNA, CCAT5, which was transactivated by the β-catenin/transcription factor 3 (TCF3) complex. CCAT5 was significantly upregulated in GC and predicted poor prognosis. Functional studies confirmed the promotive role of CCAT5 in GC growth and metastasis. Mechanistically, CCAT5 bound to the C-end domain of signal transducer and activator of transcription 3 (STAT3) and blocks Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1)-mediated STAT3Y705 dephosphorylation, leading to STAT3 nuclear entry and transactivation, thus accelerating GC progression. Furthermore, we demonstrated that both Wnt3a and β-catenin acted as activator of STAT3 signaling pathway, and the interplay between CCAT5 and STAT3 was functionally essential for Wnt-drived STAT3 signaling and tumor evolution. Finally, we revealed in vivo si-CCAT5 selectively attenuated growth and metastasis of Wnthigh GC, but not Wntlow GC. The combination of si-CCAT5 and oxaliplatin displayed obvious synergistic therapeutic effects on Wnthigh PDX mice. CONCLUSIONS We identified a novel Wnt-transactivated lncRNA, CCAT5. Our study revealed a mechanism of STAT3 signaling regulation via canonical Wnt signaling and the functional significance of CCAT5 as critical mediator. We provided conceptual advance that lncRNAs serve as therapeutic targets reversing GC progression.
Collapse
Affiliation(s)
- Chenchen Liu
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiP. R. China
| | - Aiwen Shen
- Department of NephrologyShanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Junquan Song
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiP. R. China
| | - Lei Cheng
- Department of PulmonaryShanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiP. R. China
| | - Meng Zhang
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiP. R. China
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Yanong Wang
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiP. R. China
| | - Xiaowen Liu
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiP. R. China
| |
Collapse
|
9
|
Schinderle JD, Bochkis IM. Sequential ChIP-Seq. Methods Mol Biol 2024; 2846:109-121. [PMID: 39141232 DOI: 10.1007/978-1-0716-4071-5_7] [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: 08/15/2024]
Abstract
ChIP-Seq has been used extensively to profile genome-wide transcription factor binding and post-translational histone modifications. A sequential ChIP assay determines the in vivo co-localization of two proteins to the same genomic locus. In this chapter, we combine the two protocols in Sequential ChIP-Seq, a method for identifying genome-wide sites of in vivo protein co-occupancy.
Collapse
Affiliation(s)
- Joseph D Schinderle
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Irina M Bochkis
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Grunert M, Dorn C, Dopazo A, Sánchez-Cabo F, Vázquez J, Rickert-Sperling S, Lara-Pezzi E. Technologies to Study Genetics and Molecular Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:435-458. [PMID: 38884724 DOI: 10.1007/978-3-031-44087-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Over the last few decades, the study of congenital heart disease (CHD) has benefited from various model systems and the development of molecular biological techniques enabling the analysis of single gene as well as global effects. In this chapter, we first describe different models including CHD patients and their families, animal models ranging from invertebrates to mammals, and various cell culture systems. Moreover, techniques to experimentally manipulate these models are discussed. Second, we introduce cardiac phenotyping technologies comprising the analysis of mouse and cell culture models, live imaging of cardiogenesis, and histological methods for fixed hearts. Finally, the most important and latest molecular biotechniques are described. These include genotyping technologies, different applications of next-generation sequencing, and the analysis of transcriptome, epigenome, proteome, and metabolome. In summary, the models and technologies presented in this chapter are essential to study the function and development of the heart and to understand the molecular pathways underlying CHD.
Collapse
Affiliation(s)
- Marcel Grunert
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DiNAQOR AG, Schlieren, Switzerland
| | - Cornelia Dorn
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Jésus Vázquez
- Proteomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Enrique Lara-Pezzi
- Myocardial Homeostasis and Cardiac Injury Programme, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| |
Collapse
|
11
|
Wang Y, Zhu W, Jang Y, Sommers JA, Yi G, Puligilla C, Croteau DL, Yang Y, Kai M, Liu Y. The RNA-binding motif protein 14 regulates telomere integrity at the interface of TERRA and telomeric R-loops. Nucleic Acids Res 2023; 51:12242-12260. [PMID: 37930826 PMCID: PMC10711441 DOI: 10.1093/nar/gkad967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023] Open
Abstract
Telomeric repeat-containing RNA (TERRA) and its formation of RNA:DNA hybrids (or TERRA R-loops), influence telomere maintenance, particularly in human cancer cells that use homologous recombination-mediated alternative lengthening of telomeres. Here, we report that the RNA-binding motif protein 14 (RBM14) is associated with telomeres in human cancer cells. RBM14 negatively regulates TERRA expression. It also binds to TERRA and inhibits it from forming TERRA R-loops at telomeres. RBM14 depletion has several effects, including elevated TERRA levels, telomeric R-loops, telomere dysfunction-induced DNA damage foci formation, particularly in the presence of DNA replication stress, pRPA32 accumulation at telomeres and telomere signal-free ends. Thus, RBM14 protects telomere integrity via modulating TERRA levels and its R-loop formation at telomeres.
Collapse
Affiliation(s)
- Yajun Wang
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Wei Zhu
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| | - Yumi Jang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua A Sommers
- Translational Gerontology Branch, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| | - Gong Yi
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| | - Chandrakala Puligilla
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| | - Deborah L Croteau
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| | - Yibin Yang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Mihoko Kai
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yie Liu
- Laboratory of Genetics and Genomics, 251 Bayview Blvd, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
12
|
Ríos-López DG, Tecalco-Cruz AC, Martínez-Pastor D, Sosa-Garrocho M, Tapia-Urzúa G, Aranda-López Y, Ortega-Domínguez B, Recillas-Targa F, Vázquez-Victorio G, Macías-Silva M. TGF-β/SMAD canonical pathway induces the expression of transcriptional cofactor TAZ in liver cancer cells. Heliyon 2023; 9:e21519. [PMID: 38027697 PMCID: PMC10660035 DOI: 10.1016/j.heliyon.2023.e21519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
The TGF-β and Hippo pathways are critical for liver size control, regeneration, and cancer progression. The transcriptional cofactor TAZ, also named WWTR1, is a downstream effector of Hippo pathway and plays a key role in the maintenance of liver physiological functions. However, the up-regulation of TAZ expression has been associated with liver cancer progression. Recent evidence shows crosstalk of TGF-β and Hippo pathways, since TGF-β modulates TAZ expression through different mechanisms in a cellular context-dependent manner but supposedly independent of SMADs. Here, we evaluate the molecular interplay between TGF-β pathway and TAZ expression and observe that TGF-β induces TAZ expression through SMAD canonical pathway in liver cancer HepG2 cells. Therefore, TAZ cofactor is a primary target of TGF-β/SMAD-signaling, one of the pathways altered in liver cancer.
Collapse
Affiliation(s)
- Diana G. Ríos-López
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Angeles C. Tecalco-Cruz
- Programa en Ciencias Genómicas, Universidad Autónoma de La Ciudad de México, Ciudad de México 03100, Mexico
| | - David Martínez-Pastor
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Gustavo Tapia-Urzúa
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yuli Aranda-López
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Bibiana Ortega-Domínguez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Félix Recillas-Targa
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Genaro Vázquez-Victorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
13
|
Wang J, Chen Q, Wang X, Zhao S, Deng H, Guo B, Zhang C, Song X, Deng W, Zhang T, Ni H. TFIIB-related factor 1 is a nucleolar protein that promotes RNA polymerase I-directed transcription and tumour cell growth. Hum Mol Genet 2023; 32:104-121. [PMID: 35925837 DOI: 10.1093/hmg/ddac152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/26/2022] [Accepted: 07/01/2022] [Indexed: 01/25/2023] Open
Abstract
Eukaryotic RNA polymerase I (Pol I) products play fundamental roles in ribosomal assembly, protein synthesis, metabolism and cell growth. Abnormal expression of both Pol I transcription-related factors and Pol I products causes a range of diseases, including ribosomopathies and cancers. However, the factors and mechanisms governing Pol I-dependent transcription remain to be elucidated. Here, we report that transcription factor IIB-related factor 1 (BRF1), a subunit of transcription factor IIIB required for RNA polymerase III (Pol III)-mediated transcription, is a nucleolar protein and modulates Pol I-mediated transcription. We showed that BRF1 can be localized to the nucleolus in several human cell types. BRF1 expression correlates positively with Pol I product levels and tumour cell growth in vitro and in vivo. Pol III transcription inhibition assays confirmed that BRF1 modulates Pol I-directed transcription in an independent manner rather than through a Pol III product-to-45S pre-rRNA feedback mode. Mechanistically, BRF1 binds to the Pol I transcription machinery components and can be recruited to the rDNA promoter along with them. Additionally, alteration of BRF1 expression affects the recruitment of Pol I transcription machinery components to the rDNA promoter and the expression of TBP and TAF1A. These findings indicate that BRF1 modulates Pol I-directed transcription by controlling the expression of selective factor 1 subunits. In summary, we identified a novel role of BRF1 in Pol I-directed transcription, suggesting that BRF1 can independently regulate both Pol I- and Pol III-mediated transcription and act as a key coordinator of Pol I and Pol III.
Collapse
Affiliation(s)
- Juan Wang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China.,School of Materials and Metallurgy, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Qiyue Chen
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xin Wang
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PL, UK
| | - Shasha Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Baoqiang Guo
- School of Healthcare Science, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Cheng Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xiaoye Song
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Wensheng Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Tongcun Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Hongwei Ni
- School of Materials and Metallurgy, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
14
|
Chromatin remodeler CHD7 targets active enhancer region to regulate cell type-specific gene expression in human neural crest cells. Sci Rep 2022; 12:22648. [PMID: 36587182 PMCID: PMC9805427 DOI: 10.1038/s41598-022-27293-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023] Open
Abstract
A mutation in the chromatin remodeler chromodomain helicase DNA-binding 7 (CHD7) gene causes the multiple congenital anomaly CHARGE syndrome. The craniofacial anomalies observed in CHARGE syndrome are caused by dysfunctions of neural crest cells (NCCs), which originate from the neural tube. However, the mechanism by which CHD7 regulates the function of human NCCs (hNCCs) remains unclear. We aimed to characterize the cis-regulatory elements governed by CHD7 in hNCCs by analyzing genome-wide ChIP-Seq data and identifying hNCC-specific CHD7-binding profiles. We compared CHD7-binding regions among cell types, including human induced pluripotent stem cells and human neuroepithelial cells, to determine the comprehensive properties of CHD7-binding in hNCCs. Importantly, analysis of the hNCC-specific CHD7-bound region revealed transcription factor AP-2α as a potential co-factor facilitating the cell type-specific transcriptional program in hNCCs. CHD7 was strongly associated with active enhancer regions, permitting the expression of hNCC-specific genes to sustain the function of hNCCs. Our findings reveal the regulatory mechanisms of CHD7 in hNCCs, thus providing additional information regarding the transcriptional programs in hNCCs.
Collapse
|
15
|
Identification of PAX6 and NFAT4 as the Transcriptional Regulators of the Long Noncoding RNA Mrhl in Neuronal Progenitors. Mol Cell Biol 2022; 42:e0003622. [PMID: 36317923 PMCID: PMC9670966 DOI: 10.1128/mcb.00036-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The long noncoding RNA (lncRNA) Mrhl has been shown to be involved in coordinating meiotic commitment of mouse spermatogonial progenitors and differentiation events in mouse embryonic stem cells. Here, we characterized the interplay of Mrhl with lineage-specific transcription factors during mouse neuronal lineage development. Our results demonstrate that Mrhl is expressed in the neuronal progenitor populations in mouse embryonic brains and in retinoic acid-derived radial-glia-like neuronal progenitor cells. Depletion of Mrhl leads to early differentiation of neuronal progenitors to a more committed state. A master transcription factor, PAX6, directly binds to the Mrhl promoter at a major site in the distal promoter, located at 2.9 kb upstream of the transcription start site (TSS) of Mrhl. Furthermore, NFAT4 occupies the Mrhl-proximal promoter at two sites, at 437 base pairs (bp) and 143 bp upstream of the TSS. Independent knockdown studies for PAX6 and NFAT4 confirm that they regulate Mrhl expression in neuronal progenitors. We also show that PAX6 and NFAT4 associate with each other in the same chromatin complex. NFAT4 occupies the Mrhl promoter in PAX6-bound chromatin, implying possible coregulation of Mrhl. Our studies are crucial for understanding how lncRNAs are regulated by major lineage-specific transcription factors, in order to define specific development and differentiation events.
Collapse
|
16
|
Zimmerman SM, Nixon SJ, Chen PY, Raj L, Smith SR, Paolini RL, Lin PN, Souroullas GP. Ezh2 Y641F mutations co-operate with Stat3 to regulate MHC class I antigen processing and alter the tumor immune response in melanoma. Oncogene 2022; 41:4983-4993. [PMID: 36220978 PMCID: PMC9669177 DOI: 10.1038/s41388-022-02492-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022]
Abstract
Enhancer of Zeste Homolog 2 (EZH2) is the catalytic component of the Polycomb Repressive Complex 2, a chromatin modifying complex, which mediates methylation of lysine 27 on histone 3 (H3K27me3), a repressive chromatin mark. Genetic alterations in EZH2 in melanoma include amplifications and activating point mutations at tyrosine 641 (Y641) whose underlying oncogenic mechanisms remain largely unknown. Here, we found that expression of Ezh2Y641F causes upregulation of a subset of interferon-regulated genes in melanoma cells. Upregulation of these genes was not a direct effect of changes in H3K27me3, but via a non-canonical interaction between Ezh2 and Signal Transducer and Activator of Transcription 3 (Stat3). Ezh2 and Stat3 together function as transcriptional activators to mediate gene activation of numerous genes, including MHC Class 1b antigen processing genes. Furthermore, expression of Stat3 is required to maintain an anti-tumor immune response in Ezh2Y641F melanomas and to prevent melanoma progression and recurrence.
Collapse
Affiliation(s)
- Sarah M Zimmerman
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Samantha J Nixon
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Pei Yu Chen
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Leela Raj
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Sofia R Smith
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Rachel L Paolini
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Phyo Nay Lin
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - George P Souroullas
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA.
- Division of Oncology, Molecular Oncology Section, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA.
- Siteman Comprehensive Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
17
|
Patiyal S, Dhall A, Raghava GPS. A deep learning-based method for the prediction of DNA interacting residues in a protein. Brief Bioinform 2022; 23:6658239. [PMID: 35943134 DOI: 10.1093/bib/bbac322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/01/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
DNA-protein interaction is one of the most crucial interactions in the biological system, which decides the fate of many processes such as transcription, regulation and splicing of genes. In this study, we trained our models on a training dataset of 646 DNA-binding proteins having 15 636 DNA interacting and 298 503 non-interacting residues. Our trained models were evaluated on an independent dataset of 46 DNA-binding proteins having 965 DNA interacting and 9911 non-interacting residues. All proteins in the independent dataset have less than 30% of sequence similarity with proteins in the training dataset. A wide range of traditional machine learning and deep learning (1D-CNN) techniques-based models have been developed using binary, physicochemical properties and Position-Specific Scoring Matrix (PSSM)/evolutionary profiles. In the case of machine learning technique, eXtreme Gradient Boosting-based model achieved a maximum area under the receiver operating characteristics (AUROC) curve of 0.77 on the independent dataset using PSSM profile. Deep learning-based model achieved the highest AUROC of 0.79 on the independent dataset using a combination of all three profiles. We evaluated the performance of existing methods on the independent dataset and observed that our proposed method outperformed all the existing methods. In order to facilitate scientific community, we developed standalone software and web server, which are accessible from https://webs.iiitd.edu.in/raghava/dbpred.
Collapse
Affiliation(s)
- Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi-110020, India
| | - Anjali Dhall
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi-110020, India
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi-110020, India
| |
Collapse
|
18
|
Gong W, Das S, Sierra-Pagan JE, Skie E, Dsouza N, Larson TA, Garry MG, Luzete-Monteiro E, Zaret KS, Garry DJ. ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage. Nat Cell Biol 2022; 24:672-684. [PMID: 35550615 PMCID: PMC11827897 DOI: 10.1038/s41556-022-00901-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022]
Abstract
The vasculature is an essential organ for the delivery of blood and oxygen to all tissues of the body and is thus relevant to the treatment of ischaemic diseases, injury-induced regeneration and solid tumour growth. Previously, we demonstrated that ETV2 is an essential transcription factor for the development of cardiac, endothelial and haematopoietic lineages. Here we report that ETV2 functions as a pioneer factor that relaxes closed chromatin and regulates endothelial development. By comparing engineered embryonic stem cell differentiation and reprogramming models with multi-omics techniques, we demonstrated that ETV2 was able to bind nucleosomal DNA and recruit BRG1. BRG1 recruitment remodelled chromatin around endothelial genes and helped to maintain an open configuration, resulting in increased H3K27ac deposition. Collectively, these results will serve as a platform for the development of therapeutic initiatives directed towards cardiovascular diseases and solid tumours.
Collapse
Affiliation(s)
- Wuming Gong
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Satyabrata Das
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Javier E Sierra-Pagan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Erik Skie
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Nikita Dsouza
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Thijs A Larson
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Mary G Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, USA
| | - Edgar Luzete-Monteiro
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
19
|
Lin J, Liu H, Fukumoto T, Zundell J, Yan Q, Tang CHA, Wu S, Zhou W, Guo D, Karakashev S, Hu CCA, Sarma K, Kossenkov AV, Zhang R. Targeting the IRE1α/XBP1s pathway suppresses CARM1-expressing ovarian cancer. Nat Commun 2021; 12:5321. [PMID: 34493732 PMCID: PMC8423755 DOI: 10.1038/s41467-021-25684-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
CARM1 is often overexpressed in human cancers including in ovarian cancer. However, therapeutic approaches based on CARM1 expression remain to be an unmet need. Cancer cells exploit adaptive responses such as the endoplasmic reticulum (ER) stress response for their survival through activating pathways such as the IRE1α/XBP1s pathway. Here, we report that CARM1-expressing ovarian cancer cells are selectively sensitive to inhibition of the IRE1α/XBP1s pathway. CARM1 regulates XBP1s target gene expression and directly interacts with XBP1s during ER stress response. Inhibition of the IRE1α/XBP1s pathway was effective against ovarian cancer in a CARM1-dependent manner both in vitro and in vivo in orthotopic and patient-derived xenograft models. In addition, IRE1α inhibitor B-I09 synergizes with immune checkpoint blockade anti-PD1 antibody in an immunocompetent CARM1-expressing ovarian cancer model. Our data show that pharmacological inhibition of the IRE1α/XBP1s pathway alone or in combination with immune checkpoint blockade represents a therapeutic strategy for CARM1-expressing cancers.
Collapse
Affiliation(s)
- Jianhuang Lin
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Heng Liu
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Takeshi Fukumoto
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Joseph Zundell
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Qingqing Yan
- grid.251075.40000 0001 1956 6678Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA USA
| | - Chih-Hang Anthony Tang
- grid.63368.380000 0004 0445 0041Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX USA
| | - Shuai Wu
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Wei Zhou
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Dajiang Guo
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Sergey Karakashev
- grid.251075.40000 0001 1956 6678Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA USA
| | - Chih-Chi Andrew Hu
- grid.63368.380000 0004 0445 0041Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX USA
| | - Kavitha Sarma
- grid.251075.40000 0001 1956 6678Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA USA
| | - Andrew V. Kossenkov
- grid.251075.40000 0001 1956 6678Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA USA
| | - Rugang Zhang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Wang W, Li K, Yang Z, Hou Q, Zhao WW, Sun Q. RNase H1C collaborates with ssDNA binding proteins WHY1/3 and recombinase RecA1 to fulfill the DNA damage repair in Arabidopsis chloroplasts. Nucleic Acids Res 2021; 49:6771-6787. [PMID: 34133716 PMCID: PMC8266629 DOI: 10.1093/nar/gkab479] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/25/2021] [Accepted: 05/19/2021] [Indexed: 12/28/2022] Open
Abstract
Proper repair of damaged DNA is crucial for genetic integrity and organismal survival. As semi-autonomous organelles, plastids have their own genomes whose integrity must be preserved. Several factors have been shown to participate in plastid DNA damage repair; however, the underlying mechanism remains unclear. Here, we elucidate a mechanism of homologous recombination (HR) repair in chloroplasts that involves R-loops. We find that the recombinase RecA1 forms filaments in chloroplasts during HR repair, but aggregates as puncta when RNA:DNA hybrids accumulate. ssDNA-binding proteins WHY1/3 and chloroplast RNase H1 AtRNH1C are recruited to the same genomic sites to promote HR repair. Depletion of AtRNH1C or WHY1/3 significantly suppresses the binding of RNA polymerase to the damaged DNA, thus reducing HR repair and modulating microhomology-mediated double-strand break repair. Furthermore, we show that DNA polymerase IB works with AtRNH1C genetically to complete the DNA damage repair process. This study reveals the positive role of R-loops in facilitating the activities of WHY1/3 and RecA1, which in turn secures HR repair and organellar development.
Collapse
Affiliation(s)
- Wenjie Wang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Kuan Li
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Zhuo Yang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Quancan Hou
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei W Zhao
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Qianwen Sun
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
21
|
FOXC1 Downregulates Nanog Expression by Recruiting HDAC2 to Its Promoter in F9 Cells Treated by Retinoic Acid. Int J Mol Sci 2021; 22:ijms22052255. [PMID: 33668324 PMCID: PMC7956269 DOI: 10.3390/ijms22052255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
FOXC1, a transcription factor involved in cell differentiation and embryogenesis, is demonstrated to be a negative regulator of Nanog in this study. FOXC1 is up-regulated in retinoic acid-induced differentiation of F9 Embryonal Carcinoma (EC) cells; furthermore, FOXC1 specifically inhibits the core pluripotency factor Nanog by binding to the proximal promoter. Overexpression of FOXC1 in F9 or knockdown in 3T3 results in the down-regulation or up-regulation of Nanog mRNA and proteins, respectively. In order to explain the mechanism by which FOXC1 inhibits Nanog expression, we identified the co-repressor HDAC2 from the FOXC1 interactome. FOXC1 recruits HDAC2 to Nanog promoter to decrease H3K27ac enrichment, resulting in transcription inhibition of Nanog. To the best of our knowledge, this is the first report that FOXC1 is involved in the epigenetic regulation of gene expression.
Collapse
|
22
|
RNA-cDNA hybrids mediate transposition via different mechanisms. Sci Rep 2020; 10:16034. [PMID: 32994470 PMCID: PMC7524711 DOI: 10.1038/s41598-020-73018-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/04/2020] [Indexed: 11/21/2022] Open
Abstract
Retrotransposons can represent half of eukaryotic genomes. Retrotransposon dysregulation destabilizes genomes and has been linked to various human diseases. Emerging regulators of retromobility include RNA–DNA hybrid-containing structures known as R-loops. Accumulation of these structures at the transposons of yeast 1 (Ty1) elements has been shown to increase Ty1 retromobility through an unknown mechanism. Here, via a targeted genetic screen, we identified the rnh1Δ rad27Δ yeast mutant, which lacked both the Ty1 inhibitor Rad27 and the RNA–DNA hybrid suppressor Rnh1. The mutant exhibited elevated levels of Ty1 cDNA-associated RNA–DNA hybrids that promoted Ty1 mobility. Moreover, in this rnh1Δ rad27Δ mutant, but not in the double RNase H mutant rnh1Δ rnh201Δ, RNA–DNA hybrids preferentially existed as duplex nucleic acid structures and increased Ty1 mobility in a Rad52-dependent manner. The data indicate that in cells lacking RNA–DNA hybrid and Ty1 repressors, elevated levels of RNA-cDNA hybrids, which are associated with duplex nucleic acid structures, boost Ty1 mobility via a Rad52-dependent mechanism. In contrast, in cells lacking RNA–DNA hybrid repressors alone, elevated levels of RNA-cDNA hybrids, which are associated with triplex nucleic acid structures, boost Ty1 mobility via a Rad52-independent process. We propose that duplex and triplex RNA–DNA hybrids promote transposon mobility via Rad52-dependent or -independent mechanisms.
Collapse
|
23
|
Abstract
Chromatin immunoprecipitation, commonly referred to as ChIP, is a powerful technique for the evaluation of in vivo interactions of proteins with specific regions of genomic DNA. Formaldehyde is used in this technique to cross-link proteins to DNA in vivo, followed by the extraction of chromatin from cross-linked cells and tissues. Harvested chromatin is sheared and subsequently used in an immunoprecipitation incorporating antibodies specific to protein(s) of interest and thus coprecipitating and enriching the cross-linked, protein-associated DNA. The cross-linking process can be reversed, and protein-bound DNA fragments of optimal length ranging from 200 to 1000 base pairs (bp) can subsequently be purified and measured or sequenced by numerous analytical methods. In this protocol, two different fixation methods are described in detail. The first involves the standard fixation of cells and tissue by formaldehyde if the target antigen is highly abundant. The dual cross-linking procedure presented at the end includes an additional preformaldehyde cross-linking step and can be especially useful when the target protein is in low abundance or if it is indirectly associated with chromatin DNA through another protein.
Collapse
|
24
|
A parallelized, automated platform enabling individual or sequential ChIP of histone marks and transcription factors. Proc Natl Acad Sci U S A 2020; 117:13828-13838. [PMID: 32461370 DOI: 10.1073/pnas.1913261117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite its popularity, chromatin immunoprecipitation followed by sequencing (ChIP-seq) remains a tedious (>2 d), manually intensive, low-sensitivity and low-throughput approach. Here, we combine principles of microengineering, surface chemistry, and molecular biology to address the major limitations of standard ChIP-seq. The resulting technology, FloChIP, automates and miniaturizes ChIP in a beadless fashion while facilitating the downstream library preparation process through on-chip chromatin tagmentation. FloChIP is fast (<2 h), has a wide dynamic range (from 106 to 500 cells), is scalable and parallelized, and supports antibody- or sample-multiplexed ChIP on both histone marks and transcription factors. In addition, FloChIP's interconnected design allows for straightforward chromatin reimmunoprecipitation, which allows this technology to also act as a microfluidic sequential ChIP-seq system. Finally, we ran FloChIP for the transcription factor MEF2A in 32 distinct human lymphoblastoid cell lines, providing insights into the main factors driving collaborative DNA binding of MEF2A and into its role in B cell-specific gene regulation. Together, our results validate FloChIP as a flexible and reproducible automated solution for individual or sequential ChIP-seq.
Collapse
|
25
|
Dhadve AC, Hari K, Rekhi B, Jolly MK, De A, Ray P. Decoding molecular interplay between RUNX1 and FOXO3a underlying the pulsatile IGF1R expression during acquirement of chemoresistance. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165754. [PMID: 32142859 DOI: 10.1016/j.bbadis.2020.165754] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/11/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022]
Abstract
Hyperactive Insulin like growth factor-1-receptor (IGF1R) signalling is associated with development of therapy resistance in many cancers. We recently reported a pulsatile nature of IGF1R during acquirement of platinum-taxol resistance in Epithelial Ovarian Cancer (EOC) cells and a therapy induced upregulation in IGF1R expression in tumors of a small cohort of high grade serous EOC patients. Here, we report Runt-related transcription factor 1 (RUNX1) as a novel transcriptional regulator which along with another known regulator Forkhead Box O3 (FOXO3a), drives the dynamic modulation of IGF1R expression during platinum-taxol resistance development in EOC cells. RUNX1-FOXO3a cooperatively bind to IGF1R promoter and produce a transcriptional surge during onset of resistance and such co-operativity falls apart when cells attain maximal resistance resulting in decreased IGF1R expression. The intriguing descending trend in IGF1R and FOXO3a expressions is caused by a Protein Kinase B (AKT)-FOXO3a negative feedback loop exclusively present in the highly resistant cells eliciting the pulsatile behaviour of IGF1R and FOXO3a. In vivo molecular imaging revealed that RUNX1 inhibition causes significant attenuation of the IGF1R promoter activity, decreased tumorigenicity and enhanced drug sensitivity of tumors of early resistant cells. Altogether our findings delineate a dynamic interplay between several molecular regulators driving pulsatile IGF1R expression and identify a new avenue for targeting EOC through RUNX1-IGF1R axis during acquirement of chemoresistance.
Collapse
Affiliation(s)
- Ajit C Dhadve
- Imaging Cell Signaling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Kishore Hari
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Bharat Rekhi
- Tata Memorial Hospital, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Pritha Ray
- Imaging Cell Signaling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India.
| |
Collapse
|
26
|
Timp W, Timp G. Beyond mass spectrometry, the next step in proteomics. SCIENCE ADVANCES 2020; 6:eaax8978. [PMID: 31950079 PMCID: PMC6954058 DOI: 10.1126/sciadv.aax8978] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/19/2019] [Indexed: 05/08/2023]
Abstract
Proteins can be the root cause of a disease, and they can be used to cure it. The need to identify these critical actors was recognized early (1951) by Sanger; the first biopolymer sequenced was a peptide, insulin. With the advent of scalable, single-molecule DNA sequencing, genomics and transcriptomics have since propelled medicine through improved sensitivity and lower costs, but proteomics has lagged behind. Currently, proteomics relies mainly on mass spectrometry (MS), but instead of truly sequencing, it classifies a protein and typically requires about a billion copies of a protein to do it. Here, we offer a survey that illuminates a few alternatives with the brightest prospects for identifying whole proteins and displacing MS for sequencing them. These alternatives all boast sensitivity superior to MS and promise to be scalable and seem to be adaptable to bioinformatics tools for calling the sequence of amino acids that constitute a protein.
Collapse
Affiliation(s)
- Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gregory Timp
- Departments of Electrical Engineering and Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
27
|
Tributyrin Inhibits Ethanol-Induced Epigenetic Repression of CPT-1A and Attenuates Hepatic Steatosis and Injury. Cell Mol Gastroenterol Hepatol 2019; 9:569-585. [PMID: 31654770 PMCID: PMC7078548 DOI: 10.1016/j.jcmgh.2019.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 10/10/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
UNLABELLED Ethanol-mediated down-regulation of carnitine palmitoyltransferase-1 (CPT-1A) gene expression plays a major role in the development of hepatic steatosis; however, the underlying mechanisms are not completely elucidated. Tributyrin, a butyrate prodrug that can inhibit histone deacetylase (HDAC) activity, attenuates hepatic steatosis and injury. The present study examined the beneficial effect of tributyrin/butyrate in attenuating ethanol-induced pathogenic epigenetic mechanisms affecting CPT-1A promoter-histone modifications and gene expression and hepatic steatosis/injury. METHODS Mice were fed a liquid Lieber-DeCarli diet (Research Diet Inc, New Brunswick, NJ) with or without ethanol for 4 weeks. In a subset of mice, tributyrin (2 g/kg) was administered orally by gavage. Primary rat hepatocytes were treated with 50 mmol/L ethanol and/or 2 mmol/L butyrate. Gene expression and epigenetic modifications at the CPT-1A promoter were analyzed by chromatin immunoprecipitation analysis. RESULTS In vivo, ethanol induced hepatic CPT-1A promoter histone H3K9 deacetylation, which is indicative of a repressive chromatin state, and decreased CPT-1A gene expression. Our data identified HDAC1 as the predominant HDAC causing CPT-1A promoter histone H3K9 deacetylation and epigenetic down-regulation of gene expression. Significantly, Specificity Protein 1 (SP1) and Hepatocyte Nuclear Factor 4 Alpha (HNF4α) participated in the recruitment of HDAC1 to the proximal and distal regions of CPT-1A promoter, respectively, and mediated transcriptional repression. Importantly, butyrate, a dietary HDAC inhibitor, attenuated ethanol-induced recruitment of HDAC1 and facilitated p300-HAT binding by enabling SP1/p300 interaction at the proximal region and HNF4α/peroxisomal proliferator-activated receptor-γ coactivator-1α/p300 interactions at the distal region, leading to promoter histone acetylation and enhanced CPT-1A transcription. CONCLUSIONS This study identifies HDAC1-mediated repressive epigenetic mechanisms that underlie an ethanol-mediated decrease in CPT-1A expression. Importantly, tributyrin/butyrate inhibits HDAC1, rescues CPT-1A expression, and attenuates ethanol-mediated hepatic steatosis and injury, suggesting its potential use in therapeutic strategies for alcoholic liver disease.
Collapse
|
28
|
Palmer N, Talib SZA, Ratnacaram CK, Low D, Bisteau X, Lee JHS, Pfeiffenberger E, Wollmann H, Tan JHL, Wee S, Sobota R, Gunaratne J, Messerschmidt DM, Guccione E, Kaldis P. CDK2 regulates the NRF1/ Ehmt1 axis during meiotic prophase I. J Cell Biol 2019; 218:2896-2918. [PMID: 31350280 PMCID: PMC6719441 DOI: 10.1083/jcb.201903125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/10/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
Palmer et al. identify NRF1 as a novel CDK2 interactor and substrate. This interaction was found to be important for the DNA-binding activity of NRF1. Their findings demonstrate that the loss of CDK2 expression impairs the regulation of NRF1 transcriptional activity, leading to inappropriate transcription during meiotic division. Meiosis generates four genetically distinct haploid gametes over the course of two reductional cell divisions. Meiotic divisions are characterized by the coordinated deposition and removal of various epigenetic marks. Here we propose that nuclear respiratory factor 1 (NRF1) regulates transcription of euchromatic histone methyltransferase 1 (EHMT1) to ensure normal patterns of H3K9 methylation during meiotic prophase I. We demonstrate that cyclin-dependent kinase (CDK2) can bind to the promoters of a number of genes in male germ cells including that of Ehmt1 through interaction with the NRF1 transcription factor. Our data indicate that CDK2-mediated phosphorylation of NRF1 can occur at two distinct serine residues and negatively regulates NRF1 DNA binding activity in vitro. Furthermore, induced deletion of Cdk2 in spermatocytes results in increased expression of many NRF1 target genes including Ehmt1. We hypothesize that the regulation of NRF1 transcriptional activity by CDK2 may allow the modulation of Ehmt1 expression, therefore controlling the dynamic methylation of H3K9 during meiotic prophase.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Biochemistry, National University of Singapore, Singapore
| | - S Zakiah A Talib
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | | | - Diana Low
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Xavier Bisteau
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Joanna Hui Si Lee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | | | - Heike Wollmann
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Joel Heng Loong Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Biochemistry, National University of Singapore, Singapore
| | - Sheena Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Radoslaw Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Daniel M Messerschmidt
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Ernesto Guccione
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore .,Department of Biochemistry, National University of Singapore, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore .,Department of Biochemistry, National University of Singapore, Singapore
| |
Collapse
|
29
|
Cogoi S, Ferino A, Miglietta G, Pedersen EB, Xodo LE. The regulatory G4 motif of the Kirsten ras (KRAS) gene is sensitive to guanine oxidation: implications on transcription. Nucleic Acids Res 2019; 46:661-676. [PMID: 29165690 PMCID: PMC5778462 DOI: 10.1093/nar/gkx1142] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/31/2017] [Indexed: 01/10/2023] Open
Abstract
KRAS is one of the most mutated genes in human cancer. It is controlled by a G4 motif located upstream of the transcription start site. In this paper, we demonstrate that 8-oxoguanine (8-oxoG), being more abundant in G4 than in non-G4 regions, is a new player in the regulation of this oncogene. We designed oligonucleotides mimicking the KRAS G4-motif and found that 8-oxoG impacts folding and stability of the G-quadruplex. Dimethylsulphate-footprinting showed that the G-run carrying 8-oxoG is excluded from the G-tetrads and replaced by a redundant G-run in the KRAS G4-motif. Chromatin immunoprecipitation revealed that the base-excision repair protein OGG1 is recruited to the KRAS promoter when the level of 8-oxoG in the G4 region is raised by H2O2. Polyacrylamide gel electrophoresis evidenced that OGG1 removes 8-oxoG from the G4-motif in duplex, but when folded it binds to the G-quadruplex in a non-productive way. We also found that 8-oxoG enhances the recruitment to the KRAS promoter of MAZ and hnRNP A1, two nuclear factors essential for transcription. All this suggests that 8-oxoG in the promoter G4 region could have an epigenetic potential for the control of gene expression.
Collapse
Affiliation(s)
- Susanna Cogoi
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Annalisa Ferino
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | | | - Erik B Pedersen
- Nucleic Acid Center, Institute of Physics and Chemistry, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Luigi E Xodo
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
30
|
Emamjomeh A, Choobineh D, Hajieghrari B, MahdiNezhad N, Khodavirdipour A. DNA-protein interaction: identification, prediction and data analysis. Mol Biol Rep 2019; 46:3571-3596. [PMID: 30915687 DOI: 10.1007/s11033-019-04763-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/14/2019] [Indexed: 12/30/2022]
Abstract
Life in living organisms is dependent on specific and purposeful interaction between other molecules. Such purposeful interactions make the various processes inside the cells and the bodies of living organisms possible. DNA-protein interactions, among all the types of interactions between different molecules, are of considerable importance. Currently, with the development of numerous experimental techniques, diverse methods are convenient for recognition and investigating such interactions. While the traditional experimental techniques to identify DNA-protein complexes are time-consuming and are unsuitable for genome-scale studies, the current high throughput approaches are more efficient in determining such interaction at a large-scale, but they are clearly too costly to be practice for daily applications. Hence, according to the availability of much information related to different biological sequences and clearing different dimensions of conditions in which such interactions are formed, with the developments related to the computer, mathematics, and statistics motivate scientists to develop bioinformatics tools for prediction the interaction site(s). Until now, there has been much progress in this field. In this review, the factors and conditions governing the interaction and the laboratory techniques for examining such interactions are addressed. In addition, developed bioinformatics tools are introduced and compared for this reason and, in the end, several suggestions are offered for the promotion of such tools in prediction with much more precision.
Collapse
Affiliation(s)
- Abbasali Emamjomeh
- Laboratory of Computational Biotechnology and Bioinformatics (CBB), Department of Plant Breeding and Biotechnology (PBB), University of Zabol, Zabol, 98615-538, Iran.
| | - Darush Choobineh
- Agricultural Biotechnology, Department of Plant Breeding and Biotechnology (PBB), Faculty of Agriculture, University of Zabol, Zabol, Iran
| | - Behzad Hajieghrari
- Department of Agricultural Biotechnology, College of Agriculture, Jahrom University, Jahrom, 74135-111, Iran.
| | - Nafiseh MahdiNezhad
- Laboratory of Computational Biotechnology and Bioinformatics (CBB), Department of Plant Breeding and Biotechnology (PBB), University of Zabol, Zabol, 98615-538, Iran
| | - Amir Khodavirdipour
- Division of Human Genetics, Department of Anatomy, St. John's hospital, Bangalore, India
| |
Collapse
|
31
|
Mishra LN, Shalini V, Gupta N, Ghosh K, Suthar N, Bhaduri U, Rao MRS. Spermatid-specific linker histone HILS1 is a poor condenser of DNA and chromatin and preferentially associates with LINE-1 elements. Epigenetics Chromatin 2018; 11:43. [PMID: 30068355 PMCID: PMC6069787 DOI: 10.1186/s13072-018-0214-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/25/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Linker histones establish and maintain higher-order chromatin structure. Eleven linker histone subtypes have been reported in mammals. HILS1 is a spermatid-specific linker histone, and its expression overlaps with the histone-protamine exchange process during mammalian spermiogenesis. However, the role of HILS1 in spermatid chromatin remodeling is largely unknown. RESULTS In this study, we demonstrate using circular dichroism spectroscopy that HILS1 is a poor condenser of DNA and chromatin compared to somatic linker histone H1d. Genome-wide occupancy study in elongating/condensing spermatids revealed the preferential binding of HILS1 to the LINE-1 (L1) elements within the intergenic and intronic regions of rat spermatid genome. We observed specific enrichment of the histone PTMs like H3K9me3, H4K20me3 and H4 acetylation marks (H4K5ac and H4K12ac) in the HILS1-bound chromatin complex, whereas H3K4me3 and H3K27me3 marks were absent. CONCLUSIONS HILS1 possesses significantly lower α-helicity compared to other linker histones such as H1t and H1d. Interestingly, in contrast to the somatic histone variant H1d, HILS1 is a poor condenser of chromatin which demonstrate the idea that this particular linker histone variant may have distinct role in histone to protamine replacement. Based on HILS1 ChIP-seq analysis of elongating/condensing spermatids, we speculate that HILS1 may provide a platform for the structural transitions and forms the higher-order chromatin structures encompassing LINE-1 elements during spermiogenesis.
Collapse
Affiliation(s)
- Laxmi Narayan Mishra
- Chromatin Biology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India.,Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Vasantha Shalini
- Chromatin Biology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India
| | - Nikhil Gupta
- Chromatin Biology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India.,Epigenetics and Cell Fate, UMR7216, CNRS, University Paris Diderot, Sorbonne Paris Cite, 75013, Paris, France
| | - Krittika Ghosh
- InterpretOmics India Pvt. Ltd., #329, 7th Main, HAL II Stage 80 Feet Road, Indira Nagar, Bangalore, 560008, India
| | - Neeraj Suthar
- InterpretOmics India Pvt. Ltd., #329, 7th Main, HAL II Stage 80 Feet Road, Indira Nagar, Bangalore, 560008, India
| | - Utsa Bhaduri
- Chromatin Biology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India
| | - M R Satyanarayana Rao
- Chromatin Biology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India.
| |
Collapse
|
32
|
Abstract
Thoracic aortic aneurysm (TAA) has been associated with mutations affecting members of the TGF-β signaling pathway, or components and regulators of the vascular smooth muscle cell (VSMC) actomyosin cytoskeleton. Although both clinical groups present similar phenotypes, the existence of potential common mechanisms of pathogenesis remain obscure. Here we show that mutations affecting TGF-β signaling and VSMC cytoskeleton both lead to the formation of a ternary complex comprising the histone deacetylase HDAC9, the chromatin-remodeling enzyme BRG1, and the long noncoding RNA MALAT1. The HDAC9–MALAT1–BRG1 complex binds chromatin and represses contractile protein gene expression in association with gain of histone H3-lysine 27 trimethylation modifications. Disruption of Malat1 or Hdac9 restores contractile protein expression, improves aortic mural architecture, and inhibits experimental aneurysm growth. Thus, we highlight a shared epigenetic pathway responsible for VSMC dysfunction in both forms of TAA, with potential therapeutic implication for other known HDAC9-associated vascular diseases. Vascular smooth muscle cell (VSMC) dysfunction is a common feature of thoracic aortic aneurysms (TAAs). Here, Lino Cardenas and colleagues show that the formation of a HDAC9-MALAT1-BRG1 complex promotes VSMC dysfunction in TAA by epigenetically altering the expression of key components of the cytoskeleton in VSMCs.
Collapse
|
33
|
Vaughan CA, Pearsall I, Singh S, Windle B, Deb SP, Grossman SR, Yeudall WA, Deb S. Addiction of lung cancer cells to GOF p53 is promoted by up-regulation of epidermal growth factor receptor through multiple contacts with p53 transactivation domain and promoter. Oncotarget 2017; 7:12426-46. [PMID: 26820293 PMCID: PMC4914296 DOI: 10.18632/oncotarget.6998] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/16/2016] [Indexed: 12/20/2022] Open
Abstract
Human lung cancers harboring gain-of-function (GOF) p53 alleles express higher levels of the epidermal growth factor receptor (EGFR). We demonstrate that a number of GOF p53 alleles directly upregulate EGFR. Knock-down of p53 in lung cancer cells lowers EGFR expression and reduces tumorigenicity and other GOF p53 properties. However, addiction of lung cancer cells to GOF p53 can be compensated by overexpressing EGFR, suggesting that EGFR plays a critical role in addiction. Chromatin immunoprecipitation (ChIP) using lung cancer cells expressing GOF p53 alleles showed that GOF p53 localized to the EGFR promoter. The sequence where GOF p53 is found to interact by ChIP seq can act as a GOF p53 response element. The presence of GOF p53 on the EGFR promoter increased histone H3 acetylation, indicating a mechanism whereby GOF p53 enhances chromatin opening for improved access to transcription factors (TFs). ChIP and ChIP-re-ChIP with p53, Sp1 and CBP histone acetylase (HAT) antibodies revealed docking of GOF p53 on Sp1, leading to increased binding of Sp1 and CBP to the EGFR promoter. Up-regulation of EGFR can occur via GOF p53 contact at other novel sites in the EGFR promoter even when TAD-I is inactivated; these sites are used by both intact and TAD-I mutated GOF p53 and might reflect redundancy in GOF p53 mechanisms for EGFR transactivation. Thus, the oncogenic action of GOF p53 in lung cancer is highly dependent on transactivation of the EGFR promoter via a novel transcriptional mechanism involving coordinated interactions of TFs, HATs and GOF p53.
Collapse
Affiliation(s)
- Catherine A Vaughan
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Isabella Pearsall
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Shilpa Singh
- Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA
| | - Brad Windle
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Philips Institute, Virginia Commonwealth University, Richmond, VA, USA
| | - Swati P Deb
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven R Grossman
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | - W Andrew Yeudall
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Philips Institute, Virginia Commonwealth University, Richmond, VA, USA.,Department of Oral Biology, Augusta University, Augusta, GA, USA
| | - Sumitra Deb
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
34
|
Sciuto MR, Warnken U, Schnölzer M, Valvo C, Brunetto L, Boe A, Biffoni M, Krammer PH, De Maria R, Haas TL. Two-Step Coimmunoprecipitation (TIP) Enables Efficient and Highly Selective Isolation of Native Protein Complexes. Mol Cell Proteomics 2017; 17:993-1009. [PMID: 29217617 DOI: 10.1074/mcp.o116.065920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 11/22/2017] [Indexed: 11/06/2022] Open
Abstract
Coimmunoprecipitation (co-IP) is one of the most frequently used techniques to study protein-protein (PPIs) or protein-nucleic acid interactions (PNIs). However, the presence of coprecipitated contaminants is a well-recognized issue associated with single-step co-IPs. To overcome this limitation, we developed the two-step co-IP (TIP) strategy that enables sequential coimmunoprecipitations of endogenous protein complexes. TIP can be performed with a broad range of mono- and polyclonal antibodies targeting a single protein or different components of a given complex. TIP results in a highly selective enrichment of protein complexes and thus outperforms single-step co-IPs for downstream applications such as mass spectrometry for the identification of PPIs and quantitative PCR for the analysis of PNIs. We benchmarked TIP for the identification of CD95/FAS-interacting proteins in primary human CD4+ T cells, which recapitulated all major known interactors, but also enabled the proteomics discovery of PPM1G and IPO7 as new interaction partners. For its feasibility and high performance, we propose TIP as an advanced tool for the isolation of highly purified protein-protein and protein-nucleic acid complexes under native expression conditions.
Collapse
Affiliation(s)
- Maria Rita Sciuto
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy;
| | - Uwe Warnken
- §Functional Proteome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Martina Schnölzer
- §Functional Proteome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Cecilia Valvo
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.,¶Institute of General Pathology, Catholic University and Gemelli Polyclinic, Largo F. Vito 1, 00168, Rome, Italy
| | - Lidia Brunetto
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Boe
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Mauro Biffoni
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Peter H Krammer
- ‖Department of Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ruggero De Maria
- ¶Institute of General Pathology, Catholic University and Gemelli Polyclinic, Largo F. Vito 1, 00168, Rome, Italy
| | - Tobias L Haas
- From the ‡Department of Hematology and Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy; .,¶Institute of General Pathology, Catholic University and Gemelli Polyclinic, Largo F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
35
|
An C, Li L, Zhai Q, You Y, Deng L, Wu F, Chen R, Jiang H, Wang H, Chen Q, Li C. Mediator subunit MED25 links the jasmonate receptor to transcriptionally active chromatin. Proc Natl Acad Sci U S A 2017; 114:E8930-E8939. [PMID: 28973940 PMCID: PMC5651773 DOI: 10.1073/pnas.1710885114] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Jasmonoyl-isoleucine (JA-Ile), the active form of the plant hormone jasmonate (JA), is sensed by the F-box protein CORONATINE INSENSITIVE 1 (COI1), a component of a functional Skp-Cullin-F-box E3 ubiquitin ligase complex. Sensing of JA-Ile by COI1 rapidly triggers genome-wide transcriptional changes that are largely regulated by the basic helix-loop-helix transcription factor MYC2. However, it remains unclear how the JA-Ile receptor protein COI1 relays hormone-specific regulatory signals to the RNA polymerase II general transcriptional machinery. Here, we report that the plant transcriptional coactivator complex Mediator directly links COI1 to the promoters of MYC2 target genes. MED25, a subunit of the Mediator complex, brings COI1 to MYC2 target promoters and facilitates COI1-dependent degradation of jasmonate-ZIM domain (JAZ) transcriptional repressors. MED25 and COI1 influence each other's enrichment on MYC2 target promoters. Furthermore, MED25 physically and functionally interacts with HISTONE ACETYLTRANSFERASE1 (HAC1), which plays an important role in JA signaling by selectively regulating histone (H) 3 lysine (K) 9 (H3K9) acetylation of MYC2 target promoters. Moreover, the enrichment and function of HAC1 on MYC2 target promoters depend on COI1 and MED25. Therefore, the MED25 interface of Mediator links COI1 with HAC1-dependent H3K9 acetylation to activate MYC2-regulated transcription of JA-responsive genes. This study exemplifies how a single Mediator subunit integrates the actions of both genetic and epigenetic regulators into a concerted transcriptional program.
Collapse
Affiliation(s)
- Chunpeng An
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Li
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qingzhe Zhai
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China;
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yanrong You
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Deng
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fangming Wu
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rong Chen
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongling Jiang
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hang Wang
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Qian Chen
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chuanyou Li
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China;
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| |
Collapse
|
36
|
Yang F, Ma Q, Liu Z, Li W, Tan Y, Jin C, Ma W, Hu Y, Shen J, Ohgi KA, Telese F, Liu W, Rosenfeld MG. Glucocorticoid Receptor:MegaTrans Switching Mediates the Repression of an ERα-Regulated Transcriptional Program. Mol Cell 2017; 66:321-331.e6. [PMID: 28475868 PMCID: PMC5510478 DOI: 10.1016/j.molcel.2017.03.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/01/2016] [Accepted: 03/31/2017] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms underlying the opposing functions of glucocorticoid receptors (GRs) and estrogen receptor α (ERα) in breast cancer development remain poorly understood. Here we report that, in breast cancer cells, liganded GR represses a large ERα-activated transcriptional program by binding, in trans, to ERα-occupied enhancers. This abolishes effective activation of these enhancers and their cognate target genes, and it leads to the inhibition of ERα-dependent binding of components of the MegaTrans complex. Consistent with the effects of SUMOylation on other classes of nuclear receptors, dexamethasone (Dex)-induced trans-repression of the estrogen E2 program appears to depend on GR SUMOylation, which leads to stable trans-recruitment of the GR-N-CoR/SMRT-HDAC3 corepressor complex on these enhancers. Together, these results uncover a mechanism by which competitive recruitment of DNA-binding nuclear receptors/transcription factors in trans to hot spot enhancers serves as an effective biological strategy for trans-repression, with clear implications for breast cancer and other diseases.
Collapse
Affiliation(s)
- Feng Yang
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qi Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhijie Liu
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Yuliang Tan
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chunyu Jin
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wubin Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yiren Hu
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jia Shen
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kenneth A Ohgi
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Francesca Telese
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wen Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian 361102, China
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
37
|
Vaughan CA, Singh S, Grossman SR, Windle B, Deb SP, Deb S. Gain-of-function p53 activates multiple signaling pathways to induce oncogenicity in lung cancer cells. Mol Oncol 2017; 11:696-711. [PMID: 28423230 PMCID: PMC5467493 DOI: 10.1002/1878-0261.12068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/20/2017] [Accepted: 04/06/2017] [Indexed: 01/26/2023] Open
Abstract
Gain-of-function (GOF) mutants of p53 upregulate genes implicated in cell proliferation and oncogenesis. Here, we report that GOF p53 induces tumorigenicity through simultaneous activation of key oncogenic pathways including those controlling putative tumor-initiating cell functions. We determined that in cells expressing p53-R273H, GOF p53 simultaneously upregulates genes from multiple signaling pathways by recognizing promoters containing distinct transcription factor (TF) binding sites. Our analytical data support a model in which GOF p53 complexes with two TFs on the promoter-a mediator protein, Med17, and a histone acetyl transferase, activating histone acetylation-and enhances gene expression to signal cell proliferation and oncogenesis. Thus, therapeutic inhibition of one GOF p53-induced pathway would be insufficient to prevent tumor growth as the oncoprotein activates a multitude of parallel pathways. This discovery suggests enormous selection advantage for cancer cells with GOF p53 to induce oncogenic growth, highlighting the problems of cancer therapy.
Collapse
Affiliation(s)
- Catherine A Vaughan
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Shilpa Singh
- Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven R Grossman
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, Richmond, VA, USA
| | - Brad Windle
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.,Philips Institute, Virginia Commonwealth University, Richmond, VA, USA
| | - Swati Palit Deb
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Sumitra Deb
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA.,Integrated Life Sciences Program, Virginia Commonwealth University, Richmond, VA, USA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
38
|
Cruz-Molina S, Respuela P, Tebartz C, Kolovos P, Nikolic M, Fueyo R, van Ijcken WF, Grosveld F, Frommolt P, Bazzi H, Rada-Iglesias A. PRC2 Facilitates the Regulatory Topology Required for Poised Enhancer Function during Pluripotent Stem Cell Differentiation. Cell Stem Cell 2017; 20:689-705.e9. [DOI: 10.1016/j.stem.2017.02.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/19/2016] [Accepted: 02/07/2017] [Indexed: 01/28/2023]
|
39
|
Histone Post-Translational Modifications and Nucleosome Organisation in Transcriptional Regulation: Some Open Questions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28639249 DOI: 10.1007/5584_2017_58] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The organisation of chromatin is first discussed to conclude that nucleosomes play both structural and transcription-regulatory roles. The presence of nucleosomes makes difficult the access of transcriptional factors to their target sequences and the action of RNA polymerases. The histone post-translational modifications and nucleosome remodelling are first discussed, from a historical point of view, as mechanisms to remove the obstacles imposed by chromatin structure to transcription. Instead of reviewing the state of the art of the whole field, this review is centred on some open questions. First, some "non-classical" histone modifications, such as short-chain acylations other than acetylation, are considered to conclude that their relationship with the concentration of metabolic intermediaries might make of them a sensor of the physiological state of the cells. Then attention is paid to the interest of studying chromatin organisation and epigenetic marks at a single nucleosome level as a complement to genome-wide approaches. Finally, as a consequence of the above questions, the review focuses on the presence of multiple histone post-translational modifications on a single nucleosome. The methods to detect them and their meaning, with special emphasis on bivalent marks, are discussed.
Collapse
|
40
|
Han Y, He X. Integrating Epigenomics into the Understanding of Biomedical Insight. Bioinform Biol Insights 2016; 10:267-289. [PMID: 27980397 PMCID: PMC5138066 DOI: 10.4137/bbi.s38427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/01/2016] [Accepted: 11/06/2016] [Indexed: 12/13/2022] Open
Abstract
Epigenetics is one of the most rapidly expanding fields in biomedical research, and the popularity of the high-throughput next-generation sequencing (NGS) highlights the accelerating speed of epigenomics discovery over the past decade. Epigenetics studies the heritable phenotypes resulting from chromatin changes but without alteration on DNA sequence. Epigenetic factors and their interactive network regulate almost all of the fundamental biological procedures, and incorrect epigenetic information may lead to complex diseases. A comprehensive understanding of epigenetic mechanisms, their interactions, and alterations in health and diseases genome widely has become a priority in biological research. Bioinformatics is expected to make a remarkable contribution for this purpose, especially in processing and interpreting the large-scale NGS datasets. In this review, we introduce the epigenetics pioneering achievements in health status and complex diseases; next, we give a systematic review of the epigenomics data generation, summarize public resources and integrative analysis approaches, and finally outline the challenges and future directions in computational epigenomics.
Collapse
Affiliation(s)
- Yixing Han
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.; Present address: Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ximiao He
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.; Present address: Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
41
|
Tarradas A, Pinsach-Abuin ML, Mackintosh C, Llorà-Batlle O, Pérez-Serra A, Batlle M, Pérez-Villa F, Zimmer T, Garcia-Bassets I, Brugada R, Beltran-Alvarez P, Pagans S. Transcriptional regulation of the sodium channel gene (SCN5A) by GATA4 in human heart. J Mol Cell Cardiol 2016; 102:74-82. [PMID: 27894866 DOI: 10.1016/j.yjmcc.2016.10.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/07/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023]
Abstract
Aberrant expression of the sodium channel gene (SCN5A) has been proposed to disrupt cardiac action potential and cause human cardiac arrhythmias, but the mechanisms of SCN5A gene regulation and dysregulation still remain largely unexplored. To gain insight into the transcriptional regulatory networks of SCN5A, we surveyed the promoter and first intronic regions of the SCN5A gene, predicting the presence of several binding sites for GATA transcription factors (TFs). Consistent with this prediction, chromatin immunoprecipitation (ChIP) and sequential ChIP (Re-ChIP) assays show co-occupancy of cardiac GATA TFs GATA4 and GATA5 on promoter and intron 1 SCN5A regions in fresh-frozen human left ventricle samples. Gene reporter experiments show GATA4 and GATA5 synergism in the activation of the SCN5A promoter, and its dependence on predicted GATA binding sites. GATA4 and GATA6 mRNAs are robustly expressed in fresh-frozen human left ventricle samples as measured by highly sensitive droplet digital PCR (ddPCR). GATA5 mRNA is marginally but still clearly detected in the same samples. Importantly, GATA4 mRNA levels are strongly and positively correlated with SCN5A transcript levels in the human heart. Together, our findings uncover a novel mechanism of GATA TFs in the regulation of the SCN5A gene in human heart tissue. Our studies suggest that GATA5 but especially GATA4 are main contributors to SCN5A gene expression, thus providing a new paradigm of SCN5A expression regulation that may shed new light into the understanding of cardiac disease.
Collapse
Affiliation(s)
- Anna Tarradas
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain
| | - Mel Lina Pinsach-Abuin
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain; School of Medicine, University of California San Diego, La Jolla, CA 92093-0648, USA
| | - Carlos Mackintosh
- School of Medicine, University of California San Diego, La Jolla, CA 92093-0648, USA
| | - Oriol Llorà-Batlle
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain
| | - Alexandra Pérez-Serra
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain
| | - Montserrat Batlle
- Thorax Institute, Cardiology Department, Hospital Clínic, University of Barcelona, Institute of Biomedical Research August Pi i Sunyer, 08036 Barcelona, Spain
| | - Félix Pérez-Villa
- Thorax Institute, Cardiology Department, Hospital Clínic, University of Barcelona, Institute of Biomedical Research August Pi i Sunyer, 08036 Barcelona, Spain
| | - Thomas Zimmer
- Institute for Physiology II, University Hospital, 07743 Jena, Germany
| | - Ivan Garcia-Bassets
- School of Medicine, University of California San Diego, La Jolla, CA 92093-0648, USA
| | - Ramon Brugada
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain; Hospital Universitari Dr. Josep Trueta, 17001 Girona, Spain
| | - Pedro Beltran-Alvarez
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain; School of Biological, Biomedical, and Environmental Sciences, University of Hull, HU6 7RX, Hull, UK.
| | - Sara Pagans
- Medical Sciences Department, School of Medicine, University of Girona, 17071 Girona, Spain; Institut d'Investigació Biomèdica de Girona, 17190 Salt, Spain.
| |
Collapse
|
42
|
Papageorgiou DN, Karkoulia E, Amaral-Psarris A, Burda P, Kolodziej K, Demmers J, Bungert J, Stopka T, Strouboulis J. Distinct and overlapping DNMT1 interactions with multiple transcription factors in erythroid cells: Evidence for co-repressor functions. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1515-1526. [PMID: 27693117 DOI: 10.1016/j.bbagrm.2016.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/14/2016] [Accepted: 09/26/2016] [Indexed: 01/14/2023]
Abstract
DNMT1 is the maintenance DNA methyltransferase shown to be essential for embryonic development and cellular growth and differentiation in many somatic tissues in mammals. Increasing evidence has also suggested a role for DNMT1 in repressing gene expression through interactions with specific transcription factors. Previously, we identified DNMT1 as an interacting partner of the TR2/TR4 nuclear receptor heterodimer in erythroid cells, implicated in the developmental silencing of fetal β-type globin genes in the adult stage of human erythropoiesis. Here, we extended this work by using a biotinylation tagging approach to characterize DNMT1 protein complexes in mouse erythroleukemic cells. We identified novel DNMT1 interactions with several hematopoietic transcription factors with essential roles in erythroid differentiation, including GATA1, GFI-1b and FOG-1. We provide evidence for DNMT1 forming distinct protein subcomplexes with specific transcription factors and propose the existence of a "core" DNMT1 complex with the transcription factors ZBP-89 and ZNF143, which is also present in non-hematopoietic cells. Furthermore, we identified the short (17a.a.) PCNA Binding Domain (PBD) located near the N-terminus of DNMT1 as being necessary for mediating interactions with the transcription factors described herein. Lastly, we provide evidence for DNMT1 serving as a co-repressor of ZBP-89 and GATA1 acting through upstream regulatory elements of the PU.1 and GATA1 gene loci.
Collapse
Affiliation(s)
- Dimitris N Papageorgiou
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Elena Karkoulia
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Alexandra Amaral-Psarris
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Pavel Burda
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Katarzyna Kolodziej
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jeroen Demmers
- Proteomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jörg Bungert
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Tomas Stopka
- Biocev, 1st Medical Faculty, Charles University, Prague, Czech Republic
| | - John Strouboulis
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| |
Collapse
|
43
|
Kinkley S, Helmuth J, Polansky JK, Dunkel I, Gasparoni G, Fröhler S, Chen W, Walter J, Hamann A, Chung HR. reChIP-seq reveals widespread bivalency of H3K4me3 and H3K27me3 in CD4(+) memory T cells. Nat Commun 2016; 7:12514. [PMID: 27530917 PMCID: PMC4992058 DOI: 10.1038/ncomms12514] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/11/2016] [Indexed: 01/06/2023] Open
Abstract
The combinatorial action of co-localizing chromatin modifications and regulators determines chromatin structure and function. However, identifying co-localizing chromatin features in a high-throughput manner remains a technical challenge. Here we describe a novel reChIP-seq approach and tailored bioinformatic analysis tool, normR that allows for the sequential enrichment and detection of co-localizing DNA-associated proteins in an unbiased and genome-wide manner. We illustrate the utility of the reChIP-seq method and normR by identifying H3K4me3 or H3K27me3 bivalently modified nucleosomes in primary human CD4(+) memory T cells. We unravel widespread bivalency at hypomethylated CpG-islands coinciding with inactive promoters of developmental regulators. reChIP-seq additionally uncovered heterogeneous bivalency in the population, which was undetectable by intersecting H3K4me3 and H3K27me3 ChIP-seq tracks. Finally, we provide evidence that bivalency is established and stabilized by an interplay between the genome and epigenome. Our reChIP-seq approach augments conventional ChIP-seq and is broadly applicable to unravel combinatorial modes of action.
Collapse
Affiliation(s)
- Sarah Kinkley
- Otto-Warburg-Laboratory: Epigenomics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| | - Johannes Helmuth
- Otto-Warburg-Laboratory: Epigenomics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| | - Julia K Polansky
- Experimental Rheumatology, German Rheumatism Research Center Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ilona Dunkel
- Otto-Warburg-Laboratory: Epigenomics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| | - Gilles Gasparoni
- The Department of Genetics and Epigenetics, University of Saarland, Campus A2.4 66123 Saarbrücken, Germany
| | - Sebastian Fröhler
- The Laboratory of Functional Genomics and Systems Biology, Max Delbruck Centrum for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Wei Chen
- The Laboratory of Functional Genomics and Systems Biology, Max Delbruck Centrum for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jörn Walter
- The Department of Genetics and Epigenetics, University of Saarland, Campus A2.4 66123 Saarbrücken, Germany
| | - Alf Hamann
- Experimental Rheumatology, German Rheumatism Research Center Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ho-Ryun Chung
- Otto-Warburg-Laboratory: Epigenomics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| |
Collapse
|
44
|
Co-ChIP enables genome-wide mapping of histone mark co-occurrence at single-molecule resolution. Nat Biotechnol 2016; 34:953-61. [PMID: 27454738 DOI: 10.1038/nbt.3652] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/18/2016] [Indexed: 11/08/2022]
Abstract
Histone modifications play an important role in chromatin organization and transcriptional regulation, but despite the large amount of genome-wide histone modification data collected in different cells and tissues, little is known about co-occurrence of modifications on the same nucleosome. Here we present a genome-wide quantitative method for combinatorial indexed chromatin immunoprecipitation (co-ChIP) to characterize co-occurrence of histone modifications on nucleosomes. Using co-ChIP, we study the genome-wide co-occurrence of 14 chromatin marks (70 pairwise combinations), and find previously undescribed co-occurrence patterns, including the co-occurrence of H3K9me1 and H3K27ac in super-enhancers. Finally, we apply co-ChIP to measure the distribution of the bivalent H3K4me3-H3K27me3 domains in two distinct mouse embryonic stem cell (mESC) states and in four adult tissues. We observe dynamic changes in 5,786 regions and discover both loss and de novo gain of bivalency in key tissue-specific regulatory genes, suggesting a functional role for bivalent domains during different stages of development. These results show that co-ChIP can reveal the complex interactions between histone modifications.
Collapse
|
45
|
Insulin induction of SREBP-1c in rodent liver requires LXRα-C/EBPβ complex. Proc Natl Acad Sci U S A 2016; 113:8182-7. [PMID: 27382175 DOI: 10.1073/pnas.1608987113] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Insulin increases lipid synthesis in liver by activating transcription of the gene encoding sterol regulatory element-binding protein-1c (SREBP-1c). SREBP-1c activates the transcription of all genes necessary for fatty acid synthesis. Insulin induction of SREBP-1c requires LXRα, a nuclear receptor. Transcription of SREBP-1c also requires transcription factor C/EBPβ, but a connection between LXRα and C/EBPβ has not been made. Here we show that LXRα and C/EBPβ form a complex that can be immunoprecipitated from rat liver nuclei. Chromatin immunoprecipitation assays showed that the LXRα-C/EBPβ complex binds to the SREBP-1c promoter in a region that contains two binding sites for LXRα and is known to be required for insulin induction. Knockdown of C/EBPβ in fresh rat hepatocytes or mouse livers in vivo reduces the ability of insulin to increase SREBP-1c mRNA. The LXRα-C/EBPβ complex is bound to the SREBP-1c promoter in the absence or presence of insulin, indicating that insulin acts not by increasing the formation of this complex, but rather by activating it.
Collapse
|
46
|
Abstract
Hundreds of distinct chemical modifications to DNA and histone amino acids have been described. Regulation exerted by these so-called epigenetic marks is vital to normal development, stability of cell identity through mitosis, and nongenetic transmission of traits between generations through meiosis. Loss of this regulation contributes to many diseases. Evidence indicates epigenetic marks function in combinations, whereby a given modification has distinct effects on local genome control, depending on which additional modifications are locally present. This review summarizes emerging methods for assessing combinatorial epigenomic states, as well as challenges and opportunities for their refinement.
Collapse
Affiliation(s)
- Paul D. Soloway
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
47
|
Hass MR, Liow HH, Chen X, Sharma A, Inoue YU, Inoue T, Reeb A, Martens A, Fulbright M, Raju S, Stevens M, Boyle S, Park JS, Weirauch MT, Brent MR, Kopan R. SpDamID: Marking DNA Bound by Protein Complexes Identifies Notch-Dimer Responsive Enhancers. Mol Cell 2015; 59:685-97. [PMID: 26257285 DOI: 10.1016/j.molcel.2015.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/11/2015] [Accepted: 07/02/2015] [Indexed: 12/20/2022]
Abstract
We developed Split DamID (SpDamID), a protein complementation version of DamID, to mark genomic DNA bound in vivo by interacting or juxtapositioned transcription factors. Inactive halves of DAM (DNA adenine methyltransferase) were fused to protein pairs to be queried. Either direct interaction between proteins or proximity enabled DAM reconstitution and methylation of adenine in GATC. Inducible SpDamID was used to analyze Notch-mediated transcriptional activation. We demonstrate that Notch complexes label RBP sites broadly across the genome and show that a subset of these complexes that recruit MAML and p300 undergo changes in chromatin accessibility in response to Notch signaling. SpDamID differentiates between monomeric and dimeric binding, thereby allowing for identification of half-site motifs used by Notch dimers. Motif enrichment of Notch enhancers coupled with SpDamID reveals co-targeting of regulatory sequences by Notch and Runx1. SpDamID represents a sensitive and powerful tool that enables dynamic analysis of combinatorial protein-DNA transactions at a genome-wide level.
Collapse
Affiliation(s)
- Matthew R Hass
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Hien-Haw Liow
- Center for Genome Sciences and Systems Biology, Washington University, Saint Louis, MO 63108, USA
| | - Xiaoting Chen
- School of Electronic and Computing Systems, University of Cincinnati, Cincinnati, OH 45221, USA; Center for Autoimmune Genomics and Etiology (CAGE) and Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ankur Sharma
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Ashley Reeb
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Andrew Martens
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Mary Fulbright
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Saravanan Raju
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Michael Stevens
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Scott Boyle
- Department of Developmental Biology, Washington University, Saint Louis, MO 63110, USA
| | - Joo-Seop Park
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Pediatric Urology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Matthew T Weirauch
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Autoimmune Genomics and Etiology (CAGE) and Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael R Brent
- Center for Genome Sciences and Systems Biology, Washington University, Saint Louis, MO 63108, USA
| | - Raphael Kopan
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
48
|
Schmid M, Smith J, Burt DW, Aken BL, Antin PB, Archibald AL, Ashwell C, Blackshear PJ, Boschiero C, Brown CT, Burgess SC, Cheng HH, Chow W, Coble DJ, Cooksey A, Crooijmans RPMA, Damas J, Davis RVN, de Koning DJ, Delany ME, Derrien T, Desta TT, Dunn IC, Dunn M, Ellegren H, Eöry L, Erb I, Farré M, Fasold M, Fleming D, Flicek P, Fowler KE, Frésard L, Froman DP, Garceau V, Gardner PP, Gheyas AA, Griffin DK, Groenen MAM, Haaf T, Hanotte O, Hart A, Häsler J, Hedges SB, Hertel J, Howe K, Hubbard A, Hume DA, Kaiser P, Kedra D, Kemp SJ, Klopp C, Kniel KE, Kuo R, Lagarrigue S, Lamont SJ, Larkin DM, Lawal RA, Markland SM, McCarthy F, McCormack HA, McPherson MC, Motegi A, Muljo SA, Münsterberg A, Nag R, Nanda I, Neuberger M, Nitsche A, Notredame C, Noyes H, O'Connor R, O'Hare EA, Oler AJ, Ommeh SC, Pais H, Persia M, Pitel F, Preeyanon L, Prieto Barja P, Pritchett EM, Rhoads DD, Robinson CM, Romanov MN, Rothschild M, Roux PF, Schmidt CJ, Schneider AS, Schwartz MG, Searle SM, Skinner MA, Smith CA, Stadler PF, Steeves TE, Steinlein C, Sun L, Takata M, Ulitsky I, Wang Q, Wang Y, et alSchmid M, Smith J, Burt DW, Aken BL, Antin PB, Archibald AL, Ashwell C, Blackshear PJ, Boschiero C, Brown CT, Burgess SC, Cheng HH, Chow W, Coble DJ, Cooksey A, Crooijmans RPMA, Damas J, Davis RVN, de Koning DJ, Delany ME, Derrien T, Desta TT, Dunn IC, Dunn M, Ellegren H, Eöry L, Erb I, Farré M, Fasold M, Fleming D, Flicek P, Fowler KE, Frésard L, Froman DP, Garceau V, Gardner PP, Gheyas AA, Griffin DK, Groenen MAM, Haaf T, Hanotte O, Hart A, Häsler J, Hedges SB, Hertel J, Howe K, Hubbard A, Hume DA, Kaiser P, Kedra D, Kemp SJ, Klopp C, Kniel KE, Kuo R, Lagarrigue S, Lamont SJ, Larkin DM, Lawal RA, Markland SM, McCarthy F, McCormack HA, McPherson MC, Motegi A, Muljo SA, Münsterberg A, Nag R, Nanda I, Neuberger M, Nitsche A, Notredame C, Noyes H, O'Connor R, O'Hare EA, Oler AJ, Ommeh SC, Pais H, Persia M, Pitel F, Preeyanon L, Prieto Barja P, Pritchett EM, Rhoads DD, Robinson CM, Romanov MN, Rothschild M, Roux PF, Schmidt CJ, Schneider AS, Schwartz MG, Searle SM, Skinner MA, Smith CA, Stadler PF, Steeves TE, Steinlein C, Sun L, Takata M, Ulitsky I, Wang Q, Wang Y, Warren WC, Wood JMD, Wragg D, Zhou H. Third Report on Chicken Genes and Chromosomes 2015. Cytogenet Genome Res 2015; 145:78-179. [PMID: 26282327 PMCID: PMC5120589 DOI: 10.1159/000430927] [Show More Authors] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Michael Schmid
- Department of Human Genetics, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nasipak BT, Padilla-Benavides T, Green KM, Leszyk JD, Mao W, Konda S, Sif S, Shaffer SA, Ohkawa Y, Imbalzano AN. Opposing calcium-dependent signalling pathways control skeletal muscle differentiation by regulating a chromatin remodelling enzyme. Nat Commun 2015; 6:7441. [PMID: 26081415 PMCID: PMC4530624 DOI: 10.1038/ncomms8441] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 05/06/2015] [Indexed: 01/03/2023] Open
Abstract
Calcium signaling is important for differentiation-dependent gene expression, but is also involved in other cellular functions. Therefore mechanisms must exist to distinguish calcium signaling relevant to differentiation. Calcineurin is a calcium-regulated phosphatase that is required for myogenic gene expression and skeletal muscle differentiation. Here, we demonstrate that inhibition of calcineurin blocks chromatin remodeling and that the Brg1 ATPase of the SWI/SNF chromatin remodeling enzyme, which is required for the activation of myogenic gene expression, is a calcineurin substrate. Furthermore, we identify the calcium-regulated classical protein kinase C beta (PKCβ) as a repressor of myogenesis and as the enzyme that opposes calcineurin function. Replacement of endogenous Brg1 with a phosphomimetic mutant in primary myoblasts inhibits myogenesis, while replacement with a non-phosphorylatable mutant allows myogenesis despite inhibition of calcineurin signaling, demonstrating the functionality of calcineurin/PKC modified residues. Thus the Brg1 chromatin remodeling enzyme integrates two antagonistic calcium-dependent signaling pathways that control myogenic differentiation.
Collapse
Affiliation(s)
- Brian T Nasipak
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Teresita Padilla-Benavides
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Karin M Green
- Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - John D Leszyk
- Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Wenjie Mao
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Silvana Konda
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Saïd Sif
- Department of Internal Medicine, College of Medicine, Ohio State University, Columbus, Ohio 43210, USA.,Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, PO Box 2713, Doha, Qatar
| | - Scott A Shaffer
- Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Yasuyuki Ohkawa
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA.,Department Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, 3-1-1 Maidashi Fukuoka 812-8582, Japan
| | - Anthony N Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| |
Collapse
|
50
|
Liu Z, Merkurjev D, Yang F, Li W, Oh S, Friedman MJ, Song X, Zhang F, Ma Q, Ohgi KA, Krones A, Rosenfeld MG. Enhancer activation requires trans-recruitment of a mega transcription factor complex. Cell 2015; 159:358-73. [PMID: 25303530 DOI: 10.1016/j.cell.2014.08.027] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 12/31/2022]
Abstract
Enhancers provide critical information directing cell-type-specific transcriptional programs, regulated by binding of signal-dependent transcription factors and their associated cofactors. Here, we report that the most strongly activated estrogen (E2)-responsive enhancers are characterized by trans-recruitment and in situ assembly of a large 1-2 MDa complex of diverse DNA-binding transcription factors by ERα at ERE-containing enhancers. We refer to enhancers recruiting these factors as mega transcription factor-bound in trans (MegaTrans) enhancers. The MegaTrans complex is a signature of the most potent functional enhancers and is required for activation of enhancer RNA transcription and recruitment of coactivators, including p300 and Med1. The MegaTrans complex functions, in part, by recruiting specific enzymatic machinery, exemplified by DNA-dependent protein kinase. Thus, MegaTrans-containing enhancers represent a cohort of functional enhancers that mediate a broad and important transcriptional program and provide a molecular explanation for transcription factor clustering and hotspots noted in the genome.
Collapse
Affiliation(s)
- Zhijie Liu
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Daria Merkurjev
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Graduate Program in Bioinformatics and System Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Feng Yang
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenbo Li
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Soohwan Oh
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Graduate Program in Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Meyer J Friedman
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiaoyuan Song
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; CAS Key Laboratory of Brain Function and Disease and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Feng Zhang
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qi Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Graduate Program in Bioinformatics and System Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kenneth A Ohgi
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna Krones
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|