1
|
Cao Y, Ning B, Tian Y, Lan T, Chu Y, Ren F, Wang Y, Meng Q, Li J, Jia B, Chang Z. CREPT Disarms the Inhibitory Activity of HDAC1 on Oncogene Expression to Promote Tumorigenesis. Cancers (Basel) 2022; 14:cancers14194797. [PMID: 36230720 PMCID: PMC9562184 DOI: 10.3390/cancers14194797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary It has been proposed that highly expressed HDAC1 (histone deacetylases 1) removes the acetyl group from the histones at the promoter regions of tumor suppressor genes to block their expression in tumors. We here revealed the underlying mechanism that HDAC1 differentially regulates the expression of oncogenes and tumor suppressors. In detail, we found that HDAC1 is unable to occupy the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with an oncoprotein, CREPT (cell cycle-related and expression-elevated protein in tumor). Abstract Histone deacetylases 1 (HDAC1), an enzyme that functions to remove acetyl molecules from ε-NH3 groups of lysine in histones, eliminates the histone acetylation at the promoter regions of tumor suppressor genes to block their expression during tumorigenesis. However, it remains unclear why HDAC1 fails to impair oncogene expression. Here we report that HDAC1 is unable to occupy at the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with CREPT (cell cycle-related and expression-elevated protein in tumor, also named RPRD1B), an oncoprotein highly expressed in tumors. We observed that CREPT competed with HDAC1 for binding to oncogene (such as CCND1, CLDN1, VEGFA, PPARD and BMP4) promoters but not the tumor suppressor gene (such as p21 and p27) promoters by a chromatin immunoprecipitation (ChIP) qPCR experiment. Using immunoprecipitation experiments, we deciphered that CREPT specifically occupied at the oncogene promoter via TCF4, a transcription factor activated by Wnt signaling. In addition, we performed a real-time quantitative PCR (qRT-PCR) analysis on cells that stably over-expressed CREPT and/or HDAC1, and we propose that HDAC1 inhibits CREPT to activate oncogene expression under Wnt signaling activation. Our findings revealed that HDAC1 functions differentially on tumor suppressors and oncogenes due to its interaction with the oncoprotein CREPT.
Collapse
Affiliation(s)
- Yajun Cao
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Bobin Ning
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
| | - Ye Tian
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Tingwei Lan
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yunxiang Chu
- Department of Gastroenterology, Emergency General Hospital, Beijing 100028, China
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qingyu Meng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
| | - Jun Li
- Qingda Cell Biotech Inc., Beijing 100084, China
| | - Baoqing Jia
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
- Correspondence: (B.J.); (Z.C.); Tel.: +86-(10)-62773624 (B.J.); +86-(10)-62785076 (Z.C.)
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
- Correspondence: (B.J.); (Z.C.); Tel.: +86-(10)-62773624 (B.J.); +86-(10)-62785076 (Z.C.)
| |
Collapse
|
2
|
Venkat Ramani MK, Yang W, Irani S, Zhang Y. Simplicity is the Ultimate Sophistication-Crosstalk of Post-translational Modifications on the RNA Polymerase II. J Mol Biol 2021; 433:166912. [PMID: 33676925 PMCID: PMC8184622 DOI: 10.1016/j.jmb.2021.166912] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
The highly conserved C-terminal domain (CTD) of the largest subunit of RNA polymerase II comprises a consensus heptad (Y1S2P3T4S5P6S7) repeated multiple times. Despite the simplicity of its sequence, the essential CTD domain orchestrates eukaryotic transcription and co-transcriptional processes, including transcription initiation, elongation, and termination, and mRNA processing. These distinct facets of the transcription cycle rely on specific post-translational modifications (PTM) of the CTD, in which five out of the seven residues in the heptad repeat are subject to phosphorylation. A hypothesis termed the "CTD code" has been proposed in which these PTMs and their combinations generate a sophisticated landscape for spatiotemporal recruitment of transcription regulators to Pol II. In this review, we summarize the recent experimental evidence understanding the biological role of the CTD, implicating a context-dependent theme that significantly enhances the ability of accurate transcription by RNA polymerase II. Furthermore, feedback communication between the CTD and histone modifications coordinates chromatin states with RNA polymerase II-mediated transcription, ensuring the effective and accurate conversion of information into cellular responses.
Collapse
Affiliation(s)
| | - Wanjie Yang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, United States
| | - Seema Irani
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, United States
| | - Yan Zhang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, United States; The Institute for Cellular and Molecular Biology. University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
3
|
Yang G, Wang Y, Xiao J, Zhao F, Qiu J, Liu Y, Chen G, Cao Z, You L, Zheng L, Zhang T, Zhao Y. CREPT serves as a biomarker of poor survival in pancreatic ductal adenocarcinoma. Cell Oncol (Dordr) 2021; 44:345-355. [PMID: 33125631 DOI: 10.1007/s13402-020-00569-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies. Cell-cycle-related and expression-elevated protein in tumor (CREPT) plays an important role in the phosphorylation of RNA Pol II, and has been implicated in the development of several types of cancer. As yet, however, there have been no reports on its role in PDAC. Here, we aimed to explore the value of CREPT as a prognostic biomarker in PDAC. METHODS CREPT expression was assessed by immunohistochemistry (IHC) on a tissue microarray containing samples from 375 PDAC patients. Kaplan-Meier and Cox regression analyses were performed to explore the independent prognostic value of CREPT expression for the disease-free survival (DFS) and overall survival (OS) of PDAC patients. A Cell Counting Kit-8 (CCK8) assay was used to determine the growth rates and gemcitabine sensitivities of PDAC cells, while a Transwell assay was used to determine the migration and invasion abilities of PDAC cells. Subcutaneous xenografts were used to explore the effect of CREPT expression on tumor growth in vivo. RESULTS We found that CREPT is highly expressed in tumor tissues and may serve as an independent prognostic biomarker for DFS and OS of PDAC patients. In vitro assays revealed that CREPT expression promotes the proliferation, migration, invasion and gemcitabine resistance of PDAC cells, and in vivo assays showed that CREPT expression knockdown led to inhibition of PDAC tumor growth. CONCLUSIONS We conclude that high CREPT expression enhances the proliferation, migration, invasion and gemcitabine resistance of PDAC cells. In addition, we conclude that CREPT may serve as an independent prognostic biomarker and therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yicheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Jianchun Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Guangyu Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
4
|
Zhai W, Ye X, Wang Y, Feng Y, Wang Y, Lin Y, Ding L, Yang L, Wang X, Kuang Y, Fu X, Eugene Chin Y, Jia B, Zhu B, Ren F, Chang Z. CREPT/RPRD1B promotes tumorigenesis through STAT3-driven gene transcription in a p300-dependent manner. Br J Cancer 2021; 124:1437-1448. [PMID: 33531691 PMCID: PMC8039031 DOI: 10.1038/s41416-021-01269-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 11/14/2020] [Accepted: 01/05/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Signal transducer and activator of transcription 3 (STAT3) has been shown to upregulate gene transcription during tumorigenesis. However, how STAT3 initiates transcription remains to be exploited. This study is to reveal the role of CREPT (cell cycle-related and elevated-expression protein in tumours, or RPRD1B) in promoting STAT3 transcriptional activity. METHODS BALB/c nude mice, CREPT overexpression or deletion cells were employed for the assay of tumour formation, chromatin immunoprecipitation, assay for transposase-accessible chromatin using sequencing. RESULTS We demonstrate that CREPT, a recently identified oncoprotein, enhances STAT3 transcriptional activity to promote tumorigenesis. CREPT expression is positively correlated with activation of STAT3 signalling in tumours. Deletion of CREPT led to a decrease, but overexpression of CREPT resulted in an increase, in STAT3-initiated tumour cell proliferation, colony formation and tumour growth. Mechanistically, CREPT interacts with phosphorylated STAT3 (p-STAT3) and facilitates p-STAT3 to recruit p300 to occupy at the promoters of STAT3-targeted genes. Therefore, CREPT and STAT3 coordinately facilitate p300-mediated acetylation of histone 3 (H3K18ac and H3K27ac), further augmenting RNA polymerase II recruitment. Accordingly, depletion of p300 abolished CREPT-enhanced STAT3 transcriptional activity. CONCLUSIONS We propose that CREPT is a co-activator of STAT3 for recruiting p300. Our study provides an alternative strategy for the therapy of cancers related to STAT3.
Collapse
Affiliation(s)
- Wanli Zhai
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Joint Center for Life Sciences, School of Life Science, Tsinghua University, Beijing, China
| | - Xiongjun Ye
- Urology and Lithotripsy Center, Peking University People's Hospital, Beijing, China
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Yarui Feng
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Ying Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Yuting Lin
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Joint Center for Life Sciences, School of Life Science, Tsinghua University, Beijing, China
| | - Lidan Ding
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Liu Yang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Xuning Wang
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Yanshen Kuang
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xinyuan Fu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Beijing, China
| | - Y Eugene Chin
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Baoqing Jia
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Bingtao Zhu
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China.
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China.
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Current understanding of CREPT and p15RS, carboxy-terminal domain (CTD)-interacting proteins, in human cancers. Oncogene 2020; 40:705-716. [PMID: 33239754 DOI: 10.1038/s41388-020-01544-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 12/27/2022]
Abstract
CREPT and p15RS, also named RPRD1B and RPRD1A, are RPRD (regulation of nuclear pre-mRNA-domain-containing) proteins containing C-terminal domain (CTD)-interacting domain (CID), which mediates the binding to the CTD of Rpb1, the largest subunit of RNA polymerase II (RNAPII). CREPT and p15RS are highly conserved, with a common yeast orthologue Rtt103. Intriguingly, human CREPT and p15RS possess opposite functions in the regulation of cell proliferation and tumorigenesis. While p15RS inhibits cell proliferation, CREPT promotes cell cycle and tumor growth. Aberrant expression of both CREPT and p15RS was found in numerous types of cancers. At the molecular level, both CREPT and p15RS were reported to regulate gene transcription by interacting with RNAPII. However, CREPT also exerts a key function in the processes linked to DNA damage repairs. In this review, we summarized the recent studies regarding the biological roles of CREPT and p15RS, as well as the molecular mechanisms underlying their activities. Fully revealing the mechanisms of CREPT and p15RS functions will not only provide new insights into understanding gene transcription and maintenance of DNA stability in tumors, but also promote new approach development for tumor diagnosis and therapy.
Collapse
|
6
|
Ma D, Zou Y, Chu Y, Liu Z, Liu G, Chu J, Li M, Wang J, Sun SY, Chang Z. A cell-permeable peptide-based PROTAC against the oncoprotein CREPT proficiently inhibits pancreatic cancer. Am J Cancer Res 2020; 10:3708-3721. [PMID: 32206117 PMCID: PMC7069095 DOI: 10.7150/thno.41677] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
Cancers remain a threat to human health due to the lack of effective therapeutic strategies. Great effort has been devoted to the discovery of drug targets to treat cancers, but novel oncoproteins still need to be unveiled for efficient therapy. Methods: We show that CREPT is highly expressed in pancreatic cancer and is associated with poor disease-free survival. CREPT overexpression promotes but CREPT deletion blocks colony formation and proliferation of pancreatic cancer cells. To provide a proof of concept for CREPT as a new target for the inhibition of pancreatic cancer, we designed a cell-permeable peptide-based proteolysis targeting chimera (PROTAC), named PRTC, based on the homodimerized leucine-zipper-like motif in the C-terminus domain of CREPT to induce its degradation in vivo. Results: PRTC has high affinity for CREPT, with Kd = 0.34 +/- 0.11 μM and is able to permeate into cells because of the attached membrane-transportable peptide RRRRK. PRTC effectively induces CREPT degradation in a proteasome-dependent manner. Intriguingly, PRTC inhibits colony formation, cell proliferation, and motility in pancreatic cancer cells and ultimately impairs xenograft tumor growth, comparable to the effect of CREPT deletion. Conclusions: PRTC-induced degradation of CREPT leads to inhibition of tumor growth, which is promising for the development of new drugs against pancreatic cancer. In addition, using an interacting motif based on the dimerized structure of proteins may be a new way to design a PROTAC aiming at degrading any protein without known interacting small molecules or peptides.
Collapse
|
7
|
Yin H, Cao Q, Zhao H, Wang S, Chen W, Zhang X, Chang Z, Xu T, Ye X. Expression of CREPT is associated with poor prognosis of patients with renal cell carcinoma. Oncol Lett 2019; 18:4789-4797. [PMID: 31611989 PMCID: PMC6781659 DOI: 10.3892/ol.2019.10831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
Cell-cycle-associated and expression-elevated protein in tumor (CREPT) functions as a cell cycle modulator that enhances the transcription of cyclin D1 by interacting with RNA polymerase II. CREPT has been identified to be overexpressed in various human cancer types; however, the expression and significance of CREPT in renal cell carcinoma (RCC) has remained largely elusive. In the present study, increased expression of CREPT was identified in 46.7% RCC tissues compared with adjacent normal tissue (31.1%; P=0.032) using immunohistochemistry. Furthermore, overexpression of CREPT was significantly associated with the Tumor-Node-Metastasis stage (χ2=11.967, P=0.001) and Fuhrman grade (χ2=15.453, P<0.001). In addition, increased expression of CREPT was associated with poor overall survival (P=0.021) and disease-free survival (P=0.015) of patients according to Kaplan-Meier analysis. Cellular function assays demonstrated that knockdown of CREPT in the 786-O and 769P RCC cell lines suppressed their proliferative, colony formation, migratory and invasive capacity and led to cell cycle arrest in the G1 phase. In addition, the western blotting analysis demonstrated that CREPT may control the cell cycle through downregulation of cyclin D1 and c-myc. Collectively, the overexpression of CREPT was indicated to be a negative prognostic factor for RCC, and CREPT may serve as a novel therapeutic target for the treatment of RCC.
Collapse
Affiliation(s)
- Huaqi Yin
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Qingfei Cao
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Haiyue Zhao
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Shenheng Wang
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Weinan Chen
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Xiaowei Zhang
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, P.R. China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| | - Xiongjun Ye
- Department of Urology, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China.,Urology and Lithotripsy Center, Peking University People's Hospital, The Second Clinical Medical College of Peking University, Beijing 100044, P.R. China
| |
Collapse
|
8
|
Ali I, Ruiz DG, Ni Z, Johnson JR, Zhang H, Li PC, Khalid MM, Conrad RJ, Guo X, Min J, Greenblatt J, Jacobson M, Krogan NJ, Ott M. Crosstalk between RNA Pol II C-Terminal Domain Acetylation and Phosphorylation via RPRD Proteins. Mol Cell 2019; 74:1164-1174.e4. [PMID: 31054975 DOI: 10.1016/j.molcel.2019.04.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/26/2019] [Accepted: 03/30/2019] [Indexed: 01/01/2023]
Abstract
Post-translational modifications of the RNA polymerase II C-terminal domain (CTD) coordinate the transcription cycle. Crosstalk between different modifications is poorly understood. Here, we show how acetylation of lysine residues at position 7 of characteristic heptad repeats (K7ac)-only found in higher eukaryotes-regulates phosphorylation of serines at position 5 (S5p), a conserved mark of polymerases initiating transcription. We identified the regulator of pre-mRNA-domain-containing (RPRD) proteins as reader proteins of K7ac. K7ac enhanced CTD peptide binding to the CTD-interacting domain (CID) of RPRD1A and RPRD1B proteins in isothermal calorimetry and molecular modeling experiments. Deacetylase inhibitors increased K7ac- and decreased S5-phosphorylated polymerases, consistent with acetylation-dependent S5 dephosphorylation by an RPRD-associated S5 phosphatase. Consistent with this model, RPRD1B knockdown increased S5p but enhanced K7ac, indicating that RPRD proteins recruit K7 deacetylases, including HDAC1. We also report autoregulatory crosstalk between K7ac and S5p via RPRD proteins and their interactions with acetyl- and phospho-eraser proteins.
Collapse
Affiliation(s)
- Ibraheem Ali
- J. David Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Diego Garrido Ruiz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zuyao Ni
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | | | - Heng Zhang
- Structural Genomics Consortium, University of Toronto, ON, Canada
| | - Pao-Chen Li
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mir M Khalid
- J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ryan J Conrad
- J. David Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Xinghua Guo
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Jinrong Min
- Structural Genomics Consortium, University of Toronto, ON, Canada
| | | | - Matthew Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA; California Institute for Quantitative Biosciences (QBC), University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nevan J Krogan
- J. David Gladstone Institutes, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences (QBC), University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melanie Ott
- J. David Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
9
|
Fan X, Zhao J, Ren F, Wang Y, Feng Y, Ding L, Zhao L, Shang Y, Li J, Ni J, Jia B, Liu Y, Chang Z. Dimerization of p15RS mediated by a leucine zipper-like motif is critical for its inhibitory role on Wnt signaling. J Biol Chem 2018; 293:7618-7628. [PMID: 29618509 DOI: 10.1074/jbc.ra118.001969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/27/2018] [Indexed: 01/31/2023] Open
Abstract
We previously demonstrated that p15RS, a newly discovered tumor suppressor, inhibits Wnt/β-catenin signaling by interrupting the formation of β-catenin·TCF4 complex. However, it remains unclear how p15RS helps exert such an inhibitory effect on Wnt signaling based on its molecular structure. In this study, we reported that dimerization of p15RS is required for its inhibition on the transcription regulation of Wnt-targeted genes. We found that p15RS forms a dimer through a highly conserved leucine zipper-like motif in the coiled-coil terminus domain. In particular, residues Leu-248 and Leu-255 were identified as being responsible for p15RS dimerization, as mutation of these two leucines into prolines disrupted the homodimer formation of p15RS and weakened its suppression of Wnt signaling. Functional studies further confirmed that mutations of p15RS at these residues results in diminishment of its inhibition on cell proliferation and tumor formation. We therefore concluded that dimerization of p15RS governed by the leucine zipper-like motif is critical for its inhibition of Wnt/β-catenin signaling and tumorigenesis.
Collapse
Affiliation(s)
- Xuanzi Fan
- From the State Key Laboratory of Membrane Biology, School of Medicine and.,the School of Life Sciences, Tsinghua University, Beijing 100084
| | - Juan Zhao
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Fangli Ren
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Yinyin Wang
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Yarui Feng
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Lidan Ding
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Linpeng Zhao
- the Department of Cell Biology, College of Life Sciences, Key Laboratory for Cell Proliferation and Regulation Biology of Ministry of Education, Beijing Normal University, Beijing 100875
| | - Yu Shang
- the Department of Cell Biology, College of Life Sciences, Key Laboratory for Cell Proliferation and Regulation Biology of Ministry of Education, Beijing Normal University, Beijing 100875
| | - Jun Li
- the Institute of Immunology, PLA, The Third Military Medical University, Chongqing 400038, and
| | - Jianquan Ni
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Baoqing Jia
- the Department of General Surgery/Pathology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yule Liu
- the School of Life Sciences, Tsinghua University, Beijing 100084
| | - Zhijie Chang
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| |
Collapse
|
10
|
Inhibition of CREPT restrains gastric cancer growth by regulation of cycle arrest, migration and apoptosis via ROS-regulated p53 pathway. Biochem Biophys Res Commun 2018; 496:1183-1190. [PMID: 29402413 DOI: 10.1016/j.bbrc.2018.01.167] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 01/27/2018] [Indexed: 01/07/2023]
Abstract
CREPT (cell-cycle related and expression-elevated protein in tumor) was reported to be associated with growth of several human cancers; however, its clinical significance and regulatory mechanism still remain unclear in human gastric cancer. In the present study, we found CREPT was significantly increased in gastric cancer tissues compared to the matched adjacent normal tissues. CREPT silence inhibited the proliferation of gastric cancer cells through inducing G0/G1 phase cell cycle arrest, which was linked to the reduction of Cyclin D1 and Cyclin D-dependent kinase 4 (CDK4), and the elevation of p53 and p21. In addition, CREPT knockdown (KD) decreased migration of gastric cancer cells through up-regulating E-cadherin and down-regulating vimentin, N-cadherin and matrix metalloproteinase 1 (MMP-1) expressions. Further, CREPT KD induced apoptosis in gastric cancer cells, as evidenced by the increase of cleaved Caspase-3 and poly (ADP-ribose) polymerase (PARP). Intriguingly, suppressing p53 expressions significantly abolished CREPT silence-induced apoptosis, and reduction of cell viability. Moreover, CREPT KD caused reactive oxygen species (ROS) generation using discounted cash flow (DCF) analysis, which was reversed by ROS scavenger, N-acetyl-l-cysteine (NAC), pretreatment. Of note, NAC pretreatment abrogated apoptotic cell death in CREPT KD gastric cancer cells. In vivo, suppressing CREPT reduced the gastric tumor growth in gastric cancer xenograft models. Altogether, our results provided a novel insight into CREPT in regulating gastric cancer progression through apoptosis regulated by ROS/p53 pathways.
Collapse
|
11
|
Abstract
RNA polymerase II contains a long C-terminal domain (CTD) that regulates interactions at the site of transcription. The CTD architecture remains poorly understood due to its low sequence complexity, dynamic phosphorylation patterns, and structural variability. We used integrative structural biology to visualize the architecture of the CTD in complex with Rtt103, a 3'-end RNA-processing and transcription termination factor. Rtt103 forms homodimers via its long coiled-coil domain and associates densely on the repetitive sequence of the phosphorylated CTD via its N-terminal CTD-interacting domain. The CTD-Rtt103 association opens the compact random coil structure of the CTD, leading to a beads-on-a-string topology in which the long rod-shaped Rtt103 dimers define the topological and mobility restraints of the entire assembly. These findings underpin the importance of the structural plasticity of the CTD, which is templated by a particular set of CTD-binding proteins.
Collapse
|
12
|
Liang Z, Feng Q, Xu L, Li S, Zhou L. CREPT regulated by miR-138 promotes breast cancer progression. Biochem Biophys Res Commun 2017; 493:263-269. [PMID: 28893536 DOI: 10.1016/j.bbrc.2017.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 01/21/2023]
Abstract
CREPT (also known as RPRD1B) function as an oncogene and is highly expressed in several kinds of cancers. However, the distribution and clinical significance of CREPT in breast cancer (BC) still not clarified. In this study, we found that the CREPT expression is greatly upregulated in BC tissues and cell lines. Moreover, the CREPT expression was significantly associated with tumor differentiation and metastasis. Next, the functional assay of CREPT showed that CREPT could promote BC proliferation and invasion both in vitro and in vivo. Dual-luciferase reporter assay indicated that miR-138 regulated the expression of CREPT by binding to its 3'-UTR. miR-138 is downregulated and inversely correlated with CREPT expression in BCs. Overexpression of miR-138 suppressed tumor growth and invasion, these effects could be reversed by re-expressing CREPT. Mechanistically, CREPT regulated β-catenin/TCF4/cyclin D1 pathway in BC. In conclusion, the data suggested that miR-138/CREPT involved BC progression, providing potential therapeutic targets for BC.
Collapse
Affiliation(s)
- Zhi Liang
- Department of General Surgery, Yantaishan Hospital, Yantai City, Shandong Province, 264000, PR China.
| | - Qi Feng
- The 21 Ward of General Surgery, Daqing Oil Field General Hospital, Daqing, Heilongjiang, 163000, China.
| | - Licheng Xu
- Department of General Surgery, Yantaishan Hospital, Yantai City, Shandong Province, 264000, PR China.
| | - Shuyan Li
- Department of General Surgery, Yantaishan Hospital, Yantai City, Shandong Province, 264000, PR China.
| | - Lei Zhou
- Department of General Surgery, People's Hospital of Anqiu City, Anqiu City, Shandong Province, 262100, PR China.
| |
Collapse
|
13
|
Liu C, Zhang Y, Li J, Wang Y, Ren F, Zhou Y, Wu Y, Feng Y, Zhou Y, Su F, Jia B, Wang D, Chang Z. p15RS/RPRD1A (p15INK4b-related sequence/regulation of nuclear pre-mRNA domain-containing protein 1A) interacts with HDAC2 in inhibition of the Wnt/β-catenin signaling pathway. J Biol Chem 2015; 290:9701-13. [PMID: 25697359 PMCID: PMC4392270 DOI: 10.1074/jbc.m114.620872] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Indexed: 02/05/2023] Open
Abstract
We previously reported that p15RS (p15INK4b-related sequence), a regulation of nuclear pre-mRNA domain containing protein, inhibited Wnt signaling by interrupting the formation of the β-catenin·TCF4 complex. However, how p15RS functions as an intrinsic repressor to repress transcription remains unclear. In this study, we show that p15RS, through a specific interaction with HDAC2 (histone deacetylase 2), a deacetylase that regulates gene transcription, maintains histone H3 in a deacetylated state in the promoter region of Wnt-targeted genes where β-catenin·TCF4 is bound. We observed that histone deacetylase inhibitors impair the ability of p15RS in inhibiting Wnt/β-catenin signaling. Depletion of HDAC2 markedly disabled p15RS inhibition of Wnt/β-catenin-mediated transcription. Interestingly, overexpression of p15RS decreases the level of acetylated histone H3 in the c-MYC promoter. Finally, we demonstrate that p15RS significantly enhances the association of HDAC2 and TCF4 and enhances the occupancy of HDAC2 to DNA, resulting in the deacetylation of histone H3 and the failure of β-catenin interaction. We propose that p15RS acts as an intrinsic transcriptional repressor for Wnt/β-catenin-mediated gene transcription at least partially through recruiting HDAC2 to occupy the promoter and maintaining deacetylated histone H3.
Collapse
Affiliation(s)
- Chunxiao Liu
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Yanquan Zhang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Jun Li
- the Department of Immunology, Third Military Medical School, Chongqing 610041, China
| | - Yinyin Wang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Fangli Ren
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Yifan Zhou
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China
| | - Yinyuan Wu
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Yarui Feng
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Yu Zhou
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China
| | - Fuqin Su
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China
| | - Baoqing Jia
- the Departments of Surgical Oncology and Pathology, Chinese PLA General Hospital, Beijing 100853, China, and
| | - Dong Wang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China, the Bioinformatics Division and Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| | - Zhijie Chang
- From the State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, China, the State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China,
| |
Collapse
|
14
|
Zhang Y, Liu C, Duan X, Ren F, Li S, Jin Z, Wang Y, Feng Y, Liu Z, Chang Z. CREPT/RPRD1B, a recently identified novel protein highly expressed in tumors, enhances the β-catenin·TCF4 transcriptional activity in response to Wnt signaling. J Biol Chem 2014; 289:22589-22599. [PMID: 24982424 DOI: 10.1074/jbc.m114.560979] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
CREPT (cell cycle-related and expression elevated protein in tumor)/RPRD1B (regulation of nuclear pre-mRNA domain-containing protein 1B), highly expressed during tumorigenesis, was shown to enhance transcription of CCND1 and to promote cell proliferation by interacting with RNA polymerase II. However, which signaling pathway is involved in CREPT-mediated activation of gene transcription remains unclear. In this study, we reveal that CREPT participates in transcription of the Wnt/β-catenin signaling activated genes through the β-catenin and the TCF4 complex. Our results demonstrate that CREPT interacts with both β-catenin and TCF4, and enhances the association of β-catenin with TCF4, in response to Wnt stimulation. Furthermore, CREPT was shown to occupy at TCF4 binding sites (TBS) of the promoters of Wnt-targeted genes under Wnt stimulation. Interestingly, depletion of CREPT resulted in decreased occupancy of β-catenin on TBS, and over-expression of CREPT enhances the activity of the β-catenin·TCF4 complex to initiate transcription of Wnt target genes, which results in up-regulated cell proliferation and invasion. Our study suggests that CREPT acts as an activator to promote transcriptional activity of the β-catenin·TCF4 complex in response to Wnt signaling.
Collapse
Affiliation(s)
- Yanquan Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Chunxiao Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Xiaolin Duan
- The Second People's Hospital of Zhuhai, Guangdong 519000, China
| | - Fangli Ren
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Shan Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084
| | - Zhe Jin
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Yinyin Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Yarui Feng
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| | - Zewen Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084
| | - Zhijie Chang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084,; State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, and
| |
Collapse
|
15
|
Shi Y. A glimpse of life science research at Tsinghua. SCIENCE CHINA-LIFE SCIENCES 2013; 57:1-3. [PMID: 24369355 DOI: 10.1007/s11427-013-4594-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Yigong Shi
- School of Life Sciences, Tsinghua University, Beijing, 100084, China,
| |
Collapse
|