1
|
Venhorst J, Hanemaaijer R, Dulos R, Caspers MPM, Toet K, Attema J, de Ruiter C, Kalkman G, Rouhani Rankouhi T, de Jong JCBC, Verschuren L. Integrating text mining with network models for successful target identification: in vitro validation in MASH-induced liver fibrosis. Front Pharmacol 2024; 15:1442752. [PMID: 39399467 PMCID: PMC11466758 DOI: 10.3389/fphar.2024.1442752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/28/2024] [Indexed: 10/15/2024] Open
Abstract
An in silico target discovery pipeline was developed by including a directional and weighted molecular disease network for metabolic dysfunction-associated steatohepatitis (MASH)-induced liver fibrosis. This approach integrates text mining, network biology, and artificial intelligence/machine learning with clinical transcriptome data for optimal translational power. At the mechanistic level, the critical components influencing disease progression were identified from the disease network using in silico knockouts. The top-ranked genes were then subjected to a target efficacy analysis, following which the top-5 candidate targets were validated in vitro. Three targets, including EP300, were confirmed for their roles in liver fibrosis. EP300 gene-silencing was found to significantly reduce collagen by 37%; compound intervention studies performed in human primary hepatic stellate cells and the hepatic stellate cell line LX-2 showed significant inhibition of collagen to the extent of 81% compared to the TGFβ-stimulated control (1 μM inobrodib in LX-2 cells). The validated in silico pipeline presents a unique approach for the identification of human-disease-mechanism-relevant drug targets. The directionality of the network ensures adherence to physiologically relevant signaling cascades, while the inclusion of clinical data boosts its translational power and ensures identification of the most relevant disease pathways. In silico knockouts thus provide crucial molecular insights for successful target identification.
Collapse
Affiliation(s)
- Jennifer Venhorst
- Biomedical and Digital Health, The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, Netherlands
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Remon Dulos
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Martien P. M. Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Karin Toet
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Joline Attema
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Christa de Ruiter
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Gino Kalkman
- Biomedical and Digital Health, The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, Netherlands
| | - Tanja Rouhani Rankouhi
- Biomedical and Digital Health, The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, Netherlands
| | - Jelle C. B. C. de Jong
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
2
|
Liu X, Wang J, Li F, Timchenko N, Tsai RYL. Transcriptional control of a stem cell factor nucleostemin in liver regeneration and aging. PLoS One 2024; 19:e0310219. [PMID: 39259742 PMCID: PMC11389944 DOI: 10.1371/journal.pone.0310219] [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: 05/23/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPβ. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Institute of Biosciences and Technology, Texas A&M Health, Houston, TX, United States of America
- Department of Translational Medical Sciences, Texas A&M University School of Medicine, Bryan, TX, United States of America
| | - Junying Wang
- Institute of Biosciences and Technology, Texas A&M Health, Houston, TX, United States of America
| | - Fang Li
- Institute of Biosciences and Technology, Texas A&M Health, Houston, TX, United States of America
| | - Nikolai Timchenko
- Department of Surgery, Cincinnati Children Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States of America
| | - Robert Y L Tsai
- Institute of Biosciences and Technology, Texas A&M Health, Houston, TX, United States of America
- Department of Translational Medical Sciences, Texas A&M University School of Medicine, Bryan, TX, United States of America
| |
Collapse
|
3
|
Zhu YN, Pan F, Gan XW, Liu Y, Wang WS, Sun K. The Role of DNMT1 and C/EBPα in the Regulation of CYP11A1 Expression During Syncytialization of Human Placental Trophoblasts. Endocrinology 2023; 165:bqad195. [PMID: 38146648 DOI: 10.1210/endocr/bqad195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
Progesterone synthesized in the placenta is essential for pregnancy maintenance. CYP11A1 is a key enzyme in progesterone synthesis, and its expression increases greatly during trophoblast syncytialization. However, the underlying mechanism remains elusive. Here, we demonstrated that passive demethylation of CYP11A1 promoter accounted for the upregulation of CYP11A1 expression during syncytialization with the participation of the transcription factor C/EBPα. We found that the methylation rate of a CpG locus in the CYP11A1 promoter was significantly reduced along with decreased DNA methyltransferase 1 (DNMT1) expression and its enrichment at the CYP11A1 promoter during syncytialization. DNMT1 overexpression not only increased the methylation of this CpG locus in the CYP11A1 promoter, but also decreased CYP11A1 expression and progesterone production. In silico analysis disclosed multiple C/EBPα binding sites in both CYP11A1 and DNMT1 promoters. C/EBPα expression and its enrichments at both the DNMT1 and CYP11A1 promoters were significantly increased during syncytialization. Knocking-down C/EBPα expression increased DNMT1 while it decreased CYP11A1 expression during syncytialization. Conclusively, C/EBPα plays a dual role in the regulation of CYP11A1 during syncytialization. C/EBPα not only drives CYP11A1 expression directly, but also indirectly through downregulation of DNMT1, which leads to decreased methylation in the CpG locus of the CYP11A1 promoter, resulting in increased progesterone production during syncytialization.
Collapse
Affiliation(s)
- Ya-Nan Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
- Center for Reproductive Medicine, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, Hubei 441021, P.R. China
| | - Fan Pan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
| | - Xiao-Wen Gan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
| | - Yun Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P.R. China
| |
Collapse
|
4
|
Yang L, Wang X, Yu XX, Yang L, Zhou BC, Yang J, Xu CR. The default and directed pathways of hepatoblast differentiation involve distinct epigenomic mechanisms. Dev Cell 2023; 58:1688-1700.e6. [PMID: 37490911 DOI: 10.1016/j.devcel.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/01/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
The effectiveness of multiomics analyses in defining cell differentiation pathways during development is ambiguous. During liver development, hepatoblasts follow a default or directed pathway to differentiate into hepatocytes or cholangiocytes, respectively, and this provides a practical model to address this issue. Our study discovered that promoter-associated histone modifications and chromatin accessibility dynamics, rather than enhancer-associated histone modifications, effectively delineated the "default vs. directed" process of hepatoblast differentiation. Histone H3K27me3 on bivalent promoters is associated with this asymmetric differentiation strategy in mice and humans. We demonstrated that Ezh2 and Jmjd3 exert opposing regulatory roles in hepatoblast-cholangiocyte differentiation. Additionally, active enhancers, regulated by P300, correlate with the development of both hepatocytes and cholangiocytes. This research proposes a model highlighting the division of labor between promoters and enhancers, with promoter-associated chromatin modifications governing the "default vs. directed" differentiation mode of hepatoblasts, whereas enhancer-associated modifications primarily dictate the progressive development processes of hepatobiliary lineages.
Collapse
Affiliation(s)
- Li Yang
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xin Wang
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Xin-Xin Yu
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Lu Yang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; School of Life Sciences, Peking University, Beijing 100871, China; State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bi-Chen Zhou
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jing Yang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; School of Life Sciences, Peking University, Beijing 100871, China; State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Cheng-Ran Xu
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China.
| |
Collapse
|
5
|
Xiong YJ, Zhu Y, Liu YL, Zhao YF, Shen X, Zuo WQ, Lin F, Liang ZQ. P300 Participates in Ionizing Radiation-Mediated Activation of Cathepsin L by Mutant p53. J Pharmacol Exp Ther 2021; 378:276-286. [PMID: 34253647 DOI: 10.1124/jpet.121.000639] [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: 04/01/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Our previous studies have shown that cathepsin L (CTSL) is involved in the ability of tumors to resist ionizing radiation (IR), but the specific mechanisms responsible for this remain unknown. We report here that mutant p53 (mut-p53) is involved in IR-induced transcription of CTSL. We found that irradiation caused activation of CTSL in mut-p53 cell lines, whereas there was almost no activation in p53 wild-type cell lines. Additionally, luciferase reporter gene assay results demonstrated that IR induced the p53 binding region on the CTSL promoter. We further demonstrated that the expression of p300 and early growth response factor-1 (Egr-1) was upregulated in mut-p53 cell lines after IR treatment. Accordingly, the expression of Ac-H3, Ac-H4, AcH3K9 was upregulated after IR treatment in mut-p53 cell lines, whereas histone deacetylase (HDAC) 4 and HDAC6 were reciprocally decreased. Moreover, knockdown of either Egr-1 or p300 abolished the binding of mut-p53 to the promoter of CTSL. Chromatin immunoprecipitation assay results showed that the IR-activated transcription of CTSL was dependent on p300. To further delineate the clinical relevance of interactions between Egr-1/p300, mut-p53, and CTSL, we accessed primary tumor samples to evaluate the relationships between mut-p53, CTSL, and Egr-1/p300 ex vivo. The results support the notion that mut-p53 is correlated with CTSL transcription involving the Egr-1/p300 pathway. Taken together, the results of our study revealed that p300 is an important target in the process of IR-induced transcription of CTSL, which confirms that CTSL participates in mut-p53 gain-of-function. SIGNIFICANCE STATEMENT: Transcriptional activation of cathepsin L by ionizing radiation required the involvement of mutated p53 and Egr-1/p300. Interference with Egr-1 or p300 could inhibit the expression of cathepsin L induced by ionizing radiation. The transcriptional activation of cathepsin L by p300 may be mediated by p53 binding sites on the cathepsin L promoter.
Collapse
Affiliation(s)
- Ya-Jie Xiong
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Ying Zhu
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Ya-Li Liu
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Yi-Fan Zhao
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Xiao Shen
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Wen-Qing Zuo
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Fang Lin
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Zhong-Qin Liang
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| |
Collapse
|
6
|
Gao J, Wei B, Liu M, Hirsova P, Sehrawat TS, Cao S, Hu X, Xue F, Yaqoob U, Kang N, Cui H, Pomerantz WCK, Kostallari E, Shah VH. Endothelial p300 Promotes Portal Hypertension and Hepatic Fibrosis Through C-C Motif Chemokine Ligand 2-Mediated Angiocrine Signaling. Hepatology 2021; 73:2468-2483. [PMID: 33159815 PMCID: PMC8102654 DOI: 10.1002/hep.31617] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS During liver fibrosis, liver sinusoidal endothelial cells (LSECs) release angiocrine signals to recruit inflammatory cells into the liver. p300, a master regulator of gene transcription, is associated with pathological inflammatory response. Therefore, we examined how endothelial p300 regulates angiocrine signaling and inflammation related to portal hypertension and fibrogenesis. APPROACH AND RESULTS CCl4 or partial inferior vena cava ligation (pIVCL) was used to induce liver injury. Mice with LSEC-specific p300 deletion (p300LSECΔ/Δ ) or C-C motif chemokine ligand 2 (Ccl2) deficiency, nuclear factor kappa B (NFκB)-p50 knockout mice, and bromodomain containing 4 (BRD4) inhibitors in wild-type mice were used to investigate mechanisms of inflammation regulation. Leukocytes were analyzed by mass cytometry by time-of-flight. Epigenetic histone marks were modified by CRISPR endonuclease-deficient CRISPR-associated 9-fused with the Krüppel associated box domain (CRISPR-dCas9-KRAB)-mediated epigenome editing. Portal pressure and liver fibrosis were reduced in p300LSECΔ/Δ mice compared to p300fl/fl mice following liver injury. Accumulation of macrophages was also reduced in p300LSECΔ/Δ mouse livers. Ccl2 was the most up-regulated chemokine in injured LSECs, but its increase was abrogated in p300LSECΔ/Δ mice. While the macrophage accumulation was increased in NFκB-p50 knockout mice with enhanced NFκB activity, it was reduced in mice with LSEC-specific Ccl2 deficiency and mice treated with specific BRD4 inhibitors. In vitro, epigenome editing of CCL2 enhancer and promoter regions by CRISPR-dCas9-KRAB technology repressed TNFα-induced CCL2 transcription through H3K9 trimethylation. In contrast, TNFα activated CCL2 transcription by promoting p300 interaction with NFκB and BRD4, leading to histone H3 lysine 27 acetylation at CCL2 enhancer and promoter regions. CONCLUSIONS In summary, endothelial p300 interaction with NFκB and BRD4 increases CCL2 expression, leading to macrophage accumulation, portal hypertension, and liver fibrosis. Inhibition of p300 and its binding partners might serve as therapy in the treatment of liver diseases.
Collapse
Affiliation(s)
- Jinhang Gao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.,Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Wei
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.,Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengfei Liu
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Sheng Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Xiao Hu
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Fei Xue
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Usman Yaqoob
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Ningling Kang
- Tumor Microenvironment and Metastasis Section, The Hormel Institute, University of Minnesota, Austin, MN
| | - Huarui Cui
- Department of Chemistry, University of Minnesota, Minneapolis, MN
| | | | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
7
|
E3 ubiquitin ligase UBR5 promotes pancreatic cancer growth and aerobic glycolysis by downregulating FBP1 via destabilization of C/EBPα. Oncogene 2020; 40:262-276. [PMID: 33122826 DOI: 10.1038/s41388-020-01527-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 11/08/2022]
Abstract
Pancreatic cancer is one of the most fatal cancers in humans. While it thrives in a state of malnutrition, the mechanism by which pancreatic cancer cells adapt to metabolic stress through metabolic reprogramming remains unclear. Here, we showed that UBR5, an E3 ubiquitin ligase, was significantly upregulated in pancreatic cancer patient samples compared to the levels in adjacent normal tissues. Levels of UBR5 were closely related to a malignant phenotype and shorter survival among pancreatic cancer patients. Multivariate analyses also revealed that UBR5 overexpression was an independent predictor of poor outcomes among patients with pancreatic cancer. Functional assays revealed that UBR5 contributes to the growth of pancreatic cancer cells by inducing aerobic glycolysis. Furthermore, we demonstrated that UBR5 knockdown increased levels of fructose-1,6-bisphosphatase (FBP1), an important negative regulator in the process of aerobic glycolysis in many cancers. We found a significant negative correlation between levels of UBR5 and FBP1, further demonstrating that UBR5-induced aerobic glycolysis is dependent on FBP1 in pancreatic cancer cells. Mechanistically, UBR5 regulates FBP1 expression by modulating C/EBPα, directly binding to C/EBPα, and promoting its ubiquitination and degradation. Together, these results identify a mechanism used by pancreatic cancer cells to survive the nutrient-poor tumour microenvironment and also provide insight regarding the role of UBR5 in pancreatic cancer cell adaptation to metabolic stresses.
Collapse
|
8
|
Zhang R, Li J, Cui X. Tissue distribution, excretion, and metabolism of 2,6-di-tert-butyl-hydroxytoluene in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 739:139862. [PMID: 32544679 DOI: 10.1016/j.scitotenv.2020.139862] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 05/14/2023]
Abstract
As one typical synthetic phenolic antioxidant, 2,6-di-tert-butyl-hydroxytoluene (BHT) has been widely adopted in food and other human products, and considered as an emerging contaminant due to its toxic effects. Understanding bioaccumulation and metabolism of BHT is crucial to evaluate its environmental fate and toxicity. In this study, the tissue distribution, excretion, and metabolism of BHT in mice were investigated. It was shown that BHT was prone to be accumulated in metabolism-related organs (i.e., liver and kidney) with AUC0-120 h (area under the concentration-time curve from 0 to 120 h) values of 206 h·μg/g in liver and 162 h·μg/g in kidney. For metabolites, 2,6-di-tert-butyl-4-hydroxy-4-methyl-2,5-cyclohexadione (BHT-quinol) was preferentially accumulated in liver, while 3,5-di-tert-butyl-4-hydroxybenzoic acid (BHT-COOH) was the major metabolite in excreta. The major excretion of BHT and metabolites was through feces with a value of 25.1 ± 0.16% of the initial dose compared with urine of 1.27 ± 0.05%. The possible metabolic pathways of BHT were elucidated as the oxidation of the para-methyl, tert-butyl groups, and aromatic ring based on the known and identified unknown metabolites by HPLC-Q-TOF-MS/MS. The preferred accumulation of BHT and metabolites in liver implies their potential hepatotoxicity. Results here also suggested that considering the distribution and excretion of metabolites can better assess BHT's fate and risk in mammals.
Collapse
Affiliation(s)
- Ruirui Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Juying Li
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xinyi Cui
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
9
|
Macchi F, Sadler KC. Unraveling the Epigenetic Basis of Liver Development, Regeneration and Disease. Trends Genet 2020; 36:587-597. [PMID: 32487496 DOI: 10.1016/j.tig.2020.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022]
Abstract
A wealth of studies over several decades has revealed an epigenetic prepattern that determines the competence of cellular differentiation in the developing liver. More recently, studies focused on the impact of epigenetic factors during liver regeneration suggest that an epigenetic code in the quiescent liver may establish its regenerative potential. We review work on the pioneer factors and other chromatin remodelers that impact the gene expression patterns instructing hepatocyte and biliary cell specification and differentiation, along with the requirement of epigenetic regulatory factors for hepatic outgrowth. We then explore recent studies involving the role of epigenetic regulators, Arid1a and Uhrf1, in efficient activation of proregenerative genes during liver regeneration, thus highlighting the epigenetic mechanisms of liver disease and tumor development.
Collapse
Affiliation(s)
- Filippo Macchi
- Program in Biology, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
10
|
Chromatin dynamics during liver regeneration. Semin Cell Dev Biol 2020; 97:38-46. [DOI: 10.1016/j.semcdb.2019.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/12/2019] [Accepted: 03/28/2019] [Indexed: 12/15/2022]
|
11
|
Li X, Zhang Q, Zhang M, Luo Y, Fu Y. Downregulation of nuclear ING3 expression and translocalization to cytoplasm promotes tumorigenesis and progression in head and neck squamous cell carcinoma (HNSCC). Histol Histopathol 2019; 35:681-690. [PMID: 31886514 DOI: 10.14670/hh-18-197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
ING3 (inhibitor of growth gene 3) is a member of the ING gene family, and is considered as a candidate tumor suppressor gene. In order to explore the roles of ING3 in tumorigenesis and cancer progression of head and neck squamous cell carcinoma (HNSCC), ING3 expression was assessed in 173 cases of HNSCC by immunohistochemistry. The expression of ING3 was also compared to clinicopathological variables, and the expression of several tumorigenic markers. Nuclear expression of ING3 in HNSCC was significantly lower than that in dysplasia and normal epithelium, and was negatively correlated with a poor-differentiated status, T staging and TNM staging. In contrast, cytoplasmic expression of ING3 was significantly increased in HNSCC, and was statistically associated with lymph node metastasis and 14-3-3η expression. In addition, nuclear expression of ING3 was positively correlated with the expression of p300, p21 and acetylated p53. In conclusion, decreases in nuclear ING3 may play important roles in tumorigenesis, progression and tumor differentiation in HNSCC. Increases in cytoplasmic ING3 may be due to 14-3-3η binding and may also be involved in malignant progression. Nuclear ING3 may modulate the transactivation of target genes, promoting apoptosis through interactions with p300 and p21. Moreover, ING3 may interact with p300 to upregulate the level of acetylation of p53, and promote p53-mediated cell cycle arrest, senescence and/or apoptosis. Therefore, ING3 may be a potential tumor suppressor and a possible therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Xiaohan Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qun Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yusong Luo
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yaping Fu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Protective Effects of Oridonin on Acute Liver Injury via Impeding Posttranslational Modifications of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4) in the Toll-Like Receptor 4 (TLR4) Signaling Pathway. Mediators Inflamm 2019; 2019:7634761. [PMID: 31611735 PMCID: PMC6757283 DOI: 10.1155/2019/7634761] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 08/09/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Recent researches have demonstrated that inflammation-related diseases are effectively regulated by posttranslational modifications (PTMs) including phosphorylation and acetylation. Our previous study found a new acetyltransferase inhibitor, oridonin, which had a protective effect on acute liver injury (ALI). In the present study, we further investigated its protective mechanism against D-galactosamine (D-Gal) combined with lipopolysaccharide- (LPS-) induced ALI in mice. Methods Intraperitoneal injections of LPS (40 μg/mouse)/D-Gal (5 mg/mouse) were given to the mice, and the experimental group was pretreated with intraperitoneal injection of oridonin (0.2 mg/mouse). To elucidate the protective mechanism of oridonin, we collected liver specimens and used RNA-sequencing (RNA-Seq) analysis. We focused on the genes that were upregulated by LPS/D-Gal and downregulated after pretreatment with oridonin. The downregulated genes examined by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were further verified by real-time polymerase chain reaction (PCR) and western blot. Results GO analysis showed that genes that were downregulated after pretreatment with oridonin were extremely concentrated in immune response, chemotaxis, and inflammatory response. Real-time PCR confirmed that the expression of these genes was upregulated by LPS/D-Gal induction and reduced after treatment with oridonin, which was consistent with RNA-Seq results. KEGG pathway analysis showed a significantly enriched downregulated gene that was present in the Toll-like receptor (TLR) 4 signaling cascade. Our results manifested that phosphorylation levels of upstream signaling molecules in the TLR4 signaling cascade, including extracellular signal-regulated kinase (ERK), P38, and IκB, were significantly inhibited by oridonin. Furthermore, LPS/D-Gal stimulation triggered posttranslational modifications of related gene loci in the TLR4 signaling pathway, including phosphorylation of IL-1 receptor-associated kinase 4 (IRAK4 T345/S346) and acetylation of IRAK4 (K34). However, after treatment with oridonin, the modification pattern of IRAK4 expression stimulated by LPS/D-Gal was suggestively attenuated. Conclusion Our study revealed that the protective effects of oridonin on LPS/D-Gal-induced ALI mediated by inhibition of the PTMs of IRAK4, including phosphorylation of T345/S346 and acetylation of K34.
Collapse
|
13
|
Histone deacetylase inhibitors promote ATP2A3 gene expression in hepatocellular carcinoma cells: p300 as a transcriptional regulator. Int J Biochem Cell Biol 2019; 113:8-16. [DOI: 10.1016/j.biocel.2019.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 11/24/2022]
|
14
|
Yan JK, Zhang T, Dai LN, Gu BL, Zhu J, Yan WH, Cai W, Wang Y. CELF1/p53 axis: a sustained antiproliferative signal leading to villus atrophy under total parenteral nutrition. FASEB J 2018; 33:3378-3391. [PMID: 30514107 DOI: 10.1096/fj.201801695r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intestinal villus atrophy is a major complication of total parenteral nutrition (TPN). Our previous study revealed that TPN-induced villus atrophy is accompanied by elevated expression of CUGBP, Elav-like family member 1 (CELF1); however, its mechanism of action has not been fully understood. Herein, we report a pivotal role of CELF1/p53 axis, which induces a sustained antiproliferative signal, leading to suppressed proliferation of intestinal epithelial cells (IECs). By using a rat model of TPN, we found synchronous upregulation of CELF1 and p53 in jejunum mucosa, accompanied by a 51% decrease in crypt cell proliferation rate. By using HCT-116 cells as an IEC model in vitro, we found that the expression of CELF1 altered dynamically in parallel to proliferation rate, suggesting a self-adaptive expression pattern in IECs in vitro. Furthermore, ectopic overexpression of CELF1 elicited a significant antiproliferative effect in HCT-116, Caco-2, and IEC-6 cells, whereas knockdown of CELF1 elicited a significant proproliferative effect. Moreover, cell-cycle assay revealed that ectopic overexpression of CELF1 induced sustained G2 arrest and G1 arrest in HCT-116 and IEC-6 cells, respectively, which could be abolished by p53 silencing. Mechanistically, polysomal profiling and nascent protein analysis revealed that regulation of p53 by CELF1 was mediated through accelerating its protein translation in polysomes. Taken together, our findings revealed a sustained suppression of IEC proliferation evoked by CELF1/p53 axis, which may be a potential therapeutic target for the treatment of TPN-induced villus atrophy.-Yan, J.-K., Zhang, T., Dai, L.-N., Gu, B.-L., Zhu, J., Yan, W.-H., Cai, W., Wang, Y. CELF1/p53 axis: a sustained antiproliferative signal leading to villus atrophy under total parenteral nutrition.
Collapse
Affiliation(s)
- Jun-Kai Yan
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Tian Zhang
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and
| | - Li-Na Dai
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and
| | - Bei-Lin Gu
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Jie Zhu
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and
| | - Wei-Hui Yan
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and
| | - Wei Cai
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Ying Wang
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; and.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| |
Collapse
|
15
|
D’Souza AM, Jiang Y, Cast A, Valanejad L, Wright M, Lewis K, Kumbaji M, Shah S, Smithrud D, Karns R, Shin S, Timchenko N. Gankyrin Promotes Tumor-Suppressor Protein Degradation to Drive Hepatocyte Proliferation. Cell Mol Gastroenterol Hepatol 2018; 6:239-255. [PMID: 30109252 PMCID: PMC6083020 DOI: 10.1016/j.jcmgh.2018.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/18/2018] [Indexed: 12/19/2022]
Abstract
Background & Aims Uncontrolled liver proliferation is a key characteristic of liver cancer; however, the mechanisms by which this occurs are not well understood. Elucidation of these mechanisms is necessary for the development of better therapy. The oncogene Gankyrin (Gank) is overexpressed in both hepatocellular carcinoma and hepatoblastoma. The aim of this work was to determine the role of Gank in liver proliferation and elucidate the mechanism by which Gank promotes liver proliferation. Methods We generated Gank liver-specific knock-out (GLKO) mice and examined liver biology and proliferation after surgical resection and liver injury. Results Global profiling of gene expression in GLKO mice showed significant changes in pathways involved in liver cancer and proliferation. Investigations of liver proliferation after partial hepatectomy and CCl4 treatment showed that GLKO mice have dramatically inhibited proliferation of hepatocytes at early stages after surgery and injury. In control LoxP mice, liver proliferation was characterized by Gank-mediated reduction of tumor-suppressor proteins (TSPs). The failure of GLKO hepatocytes to proliferate is associated with a lack of down-regulation of these proteins. Surprisingly, we found that hepatic progenitor cells of GLKO mice start proliferation at later stages and restore the original size of the liver at 14 days after partial hepatectomy. To examine the proliferative activities of Gank in cancer cells, we used a small molecule, cjoc42, to inhibit interactions of Gank with the 26S proteasome. These studies showed that Gank triggers degradation of TSPs and that cjoc42-mediated inhibition of Gank increases levels of TSPs and inhibits proliferation of cancer cells. Conclusions These studies show that Gank promotes hepatocyte proliferation by elimination of TSPs. This work provides background for the development of Gank-mediated therapy for the treatment of liver cancer. RNA sequencing data can be accessed in the NCBI Gene Expression Omnibus: GSE104395.
Collapse
Key Words
- 2D, 2-dimensional
- BrdU, bromodeoxyuridine
- C/EBP, CCAAT/enhancer binding protein
- CUGBP1, CUG triplet repeat binding protein 1
- Cancer
- Co-IP, co-immunoprecipitation
- DEN, diethylnitrosamine
- FXR, farnesoid X receptor
- GLKO, Gankyrin liver-specific knock-out
- Gank, Gankyrin
- HCC, hepatocellular carcinoma
- HNF4α, hepatocyte nuclear factor 4α
- LKO, liver-specific knock-out
- Liver
- Opn, osteopontin
- PCNA, proliferating cell nuclear antigen
- PH, partial hepatectomy
- Progenitor Cells
- Proliferation
- RT-PCR, reverse-transcriptase polymerase chain reaction
- Rb, retinoblastoma
- TSP, tumor-suppressor protein
- Tumor-Suppressor Proteins
- UPS, ubiquitin proteasome system
- WT, wild-type
- cDNA, complementary DNA
- mRNA, messenger RNA
Collapse
Affiliation(s)
- Amber M. D’Souza
- Department of Oncology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Yanjun Jiang
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas
| | - Ashley Cast
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Leila Valanejad
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Mary Wright
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Kyle Lewis
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Meenasri Kumbaji
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Sheeniza Shah
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio
| | - David Smithrud
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio
| | - Rebekah Karns
- Department of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Soona Shin
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Nikolai Timchenko
- Department of Surgery, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
16
|
Hwang JT, Choi HK, Kim SH, Chung S, Hur HJ, Park JH, Chung MY. Hypolipidemic Activity of Quercus acutissima Fruit Ethanol Extract is Mediated by Inhibition of Acetylation. J Med Food 2017; 20:542-549. [PMID: 28581876 DOI: 10.1089/jmf.2016.3912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The acetylation of histone and nonhistone proteins is associated with adipogenesis. The objective of the present study was to investigate whether an ethanol extract of Quercus acutissima fruit (QF) exhibits antiobesity effects through inhibition of acetylation in 3T3-L1 preadipocytes and high fat diet (HFD)-fed obese mice. We observed that QF acts as a histone acetyltransferase (HAT) inhibitor and that QF (400 μg/mL) markedly inhibits the activity of p300 and CREB-binding protein. QF (200 μg/mL) significantly attenuated lipid accumulation without apparent toxicity, which is likely attributable to a decrease in the expressions of lipogenic proteins, including fatty acid synthase, peroxisome proliferator-activated receptor gamma, sterol regulatory element-binding protein 1, and CCAAT-enhancer-binding proteins alpha that were otherwise increased by MDI (a hormonal cocktail containing methyl isobutylmethylxanthine, dexamethasone, and insulin). MDI increased the acetylation of total lysine residues in whole 3T3-L1 cell lysate, an effect that was reversed by QF treatment (200 μg/mL). To further confirm the antiobesity activity of QF, mice were fed with HFD supplemented with QF at 50 and 200 mg/kg body weight. Mice fed with HFD exhibited increased masses of body, liver, and retroperitoneal fat, an effect that was suppressed in the presence of QF supplementation. QF-mediated decreases in body weight were attributable to a decrease in the average size of lipid droplets, as well as lipid accumulation in retroperitoneal fat and the liver, respectively. QF-mediated reductions in the size of the lipid droplets in the retroperitoneal fat tissue were likely associated with decreased expression of DGAT2. Taken together, our observations suggest that QF acts as an HAT inhibitor and attenuates adipogenesis in 3T3-L1 preadipocytes, resulting in the mitigation of HFD-induced obesity.
Collapse
Affiliation(s)
- Jin-Taek Hwang
- 1 Korea Food Research Institute , Seongnam, Korea.,2 Department of Food Biotechnology, Korea University of Science & Technology , Daejeon, Korea
| | | | - Sung Hee Kim
- 1 Korea Food Research Institute , Seongnam, Korea
| | | | | | - Jae Ho Park
- 1 Korea Food Research Institute , Seongnam, Korea.,2 Department of Food Biotechnology, Korea University of Science & Technology , Daejeon, Korea
| | - Min-Yu Chung
- 1 Korea Food Research Institute , Seongnam, Korea
| |
Collapse
|
17
|
CUG-binding protein 1 regulates HSC activation and liver fibrogenesis. Nat Commun 2016; 7:13498. [PMID: 27853137 PMCID: PMC5118555 DOI: 10.1038/ncomms13498] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 10/07/2016] [Indexed: 12/17/2022] Open
Abstract
Excessive activation of hepatic stellate cells (HSCs) is a key step in liver fibrogenesis. Here we report that CUG-binding protein 1 (CUGBP1) expression is elevated in HSCs and positively correlates with liver fibrosis severity in human liver biopsies. Transforming growth factor-beta (TGF-β) selectively increases CUGBP1 expression in cultured HSCs in a p38 mitogen-activated protein kinase (MAPK)-dependent manner. Knockdown of CUGBP1 inhibits alpha smooth muscle actin (α-SMA) expression and promotes interferon gamma (IFN-γ) production in HSCs in vitro. We further show that CUGBP1 specifically binds to the 3′ untranslated region (UTR) of human IFN-γ mRNA and promotes its decay. In mice, knockdown of CUGBP1 alleviates, whereas its overexpression exacerbates, bile duct ligation (BDL)-induced hepatic fibrosis. Therefore, CUGBP1-mediated IFN-γ mRNA decay is a key event for profibrotic TGF-β-dependent activation of HSCs, and inhibiting CUGBP1 to promote IFN-γ signalling in activated HSCs could be a novel strategy to treat liver fibrosis. Activation of hepatic stellate cells is a critical event in the development of fibrosis, which is driven by TGF-beta and inhibited by IFN-gamma. Here Wu et al. show that the RNA binding protein CUGBP1 is increased by TGF-beta signalling and promotes IFN-gamma mRNA degradation.
Collapse
|
18
|
Wang C, Yang S, Huang J, Chen S, Li Y, Li Q. Activation of corticotropin releasing factor receptors up regulates collagen production by hepatic stellate cells via promoting p300 expression. Biol Chem 2016; 397:437-44. [PMID: 26756093 DOI: 10.1515/hsz-2015-0233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022]
Abstract
Liver fibrosis is characterized with the over expression and excessive accumulation of extracellular matrix proteins, including collagens. The causative factors in the over production of collagens are not fully understood. This study aims to test a hypothesis that activation of corticotropin releasing factor receptors up regulates the expression of collagen in hepatic stellate cells. In this study, human hepatic stellate cell line, LX-2 cells were cultured. Expression of collagens by LX-2 cells was assessed by real time RT-PCR, Western blotting. The results showed that, upon exposure to urocortin in the culture, LX-2 cells (a human hepatic stellate cell line) increased the expression of collagen IV (Col4) markedly. The exposure to urocortin also enhanced the levels of pTip60, H3K9, RNA polymerase II and forkhead box protein 3 at the collagen promoter locus as well as increase in the expression of Col4 mRNA and protein in the cells. Blocking p300 efficiently suppressed the urocortin-induced Col4 expression in LX-2 cells and unveiled an apoptosis-inducing effect of urocortin. In conclusion, activation of CRF receptors is capable of enforcing the production of Col4 by LX-2 cells via up regulating the p300 pathway, which may contribute to the development of liver fibrosis.
Collapse
|
19
|
Li GB, Huang LY, Li H, Ji S, Li LL, Yang SY. Identification of new p300 histone acetyltransferase inhibitors from natural products by a customized virtual screening method. RSC Adv 2016. [DOI: 10.1039/c6ra11240d] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The natural compounds NP-2, NP-3, NP-9, and NP-15 were found to be potent p300 HAT inhibitors by a customized structure-based virtual screening method.
Collapse
Affiliation(s)
- Guo-Bo Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- China
| | - Lu-Yi Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- China
| | - Hui Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- China
| | - Sen Ji
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- China
| | - Lin-Li Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu
- China
| | - Sheng-Yong Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy
- West China Hospital
- West China Medical School
- Sichuan University
- China
| |
Collapse
|