51
|
A negative reciprocal regulatory axis between cyclin D1 and HNF4α modulates cell cycle progression and metabolism in the liver. Proc Natl Acad Sci U S A 2020; 117:17177-17186. [PMID: 32631996 DOI: 10.1073/pnas.2002898117] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocyte nuclear factor 4α (HNF4α) is a master regulator of liver function and a tumor suppressor in hepatocellular carcinoma (HCC). In this study, we explore the reciprocal negative regulation of HNF4α and cyclin D1, a key cell cycle protein in the liver. Transcriptomic analysis of cultured hepatocyte and HCC cells found that cyclin D1 knockdown induced the expression of a large network of HNF4α-regulated genes. Chromatin immunoprecipitation-sequencing (ChIP-seq) demonstrated that cyclin D1 inhibits the binding of HNF4α to thousands of targets in the liver, thereby diminishing the expression of associated genes that regulate diverse metabolic activities. Conversely, acute HNF4α deletion in the liver induces cyclin D1 and hepatocyte cell cycle progression; concurrent cyclin D1 ablation blocked this proliferation, suggesting that HNF4α maintains proliferative quiescence in the liver, at least, in part, via repression of cyclin D1. Acute cyclin D1 deletion in the regenerating liver markedly inhibited hepatocyte proliferation after partial hepatectomy, confirming its pivotal role in cell cycle progression in this in vivo model, and enhanced the expression of HNF4α target proteins. Hepatocyte cyclin D1 gene ablation caused markedly increased postprandial liver glycogen levels (in a HNF4α-dependent fashion), indicating that the cyclin D1-HNF4α axis regulates glucose metabolism in response to feeding. In AML12 hepatocytes, cyclin D1 depletion led to increased glucose uptake, which was negated if HNF4α was depleted simultaneously, and markedly elevated glycogen synthesis. To summarize, mutual repression by cyclin D1 and HNF4α coordinately controls the cell cycle machinery and metabolism in the liver.
Collapse
|
52
|
Zhang X, Xu Y, Bai Q, Li X, Han J, Hou Y, Ji Y, Zhang Z. Inhibition of LXR signaling by SULT2B1b promotes liver regeneration after partial hepatectomy in mouse models of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2020; 319:G87-G96. [PMID: 32475129 DOI: 10.1152/ajpgi.00380.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydroxysteroid sulfotransferase 2B1b (SULT2B1b) plays a critical role in hepatic energy homeostasis. Liver X receptors (LXRs) are implicated in multiple physiological functions, including the inhibition of hepatocyte proliferation and regulation of fatty acid and cholesterol metabolism. We have previously reported that SULT2B1b promotes hepatocyte proliferation by inactivating LXR signaling in vivo and in vitro, leading to our hypothesis that SULT2B1b promotes fatty liver regeneration. In the present study, female C57BL/6 and S129 mice were fed a high-fat diet for 8 wk to establish a nonalcoholic fatty liver disease (NAFLD) mouse model. 70% partial hepatectomy (PH) was performed to induce liver regeneration. Our experiments revealed that the SULT2B1b overexpression significantly promotes the regeneration of hepatocytes in NAFLD C57BL/6 mice after PH, increasing liver regrowth by 11% within 1 day, and then by 21%, 33%, and 24% by 2, 3, and 5 days post-PH, respectively. Compared with the wild-type NAFLD S129 mice, SULT2B1 deletion NAFLD S129 mice presented reduced hepatocyte regeneration at postoperative day 2, as verified by decreased liver regrowth (37.4% vs. 46.1%, P < 0.05) and the results of immunohistochemical staining, quantitative real-time polymerase chain reaction, and Western blot analysis. Moreover, LXRα signaling and SULT2B1b expression are highly correlated in the regeneration of NAFLD mouse liver; SULT2B1b overexpression suppresses LXRα signaling, while the LXRα-signaling agonist T0901317 blocks SULT2B1b-induced hepatocyte regeneration in NAFLD mouse liver. Thus, the upregulation of SULT2B1b may promote hepatocyte regeneration via the suppression of LXRα activation in NAFLD mice, providing a potential strategy for improving hepatic-steatosis-related liver regeneration disorders.NEW & NOTEWORTHY This study demonstrates for the first time that hydroxysteroid sulfotransferase 2B1b (SULT2B1b) overexpression promotes the regeneration of fatty liver after partial hepatectomy in mice with nonalcoholic fatty liver disease, while reducing triglyceride accumulation in the regenerative fatty liver. Liver X receptor signaling may be crucial in the SULT2B1b-mediated regeneration of fatty liver. Thus, SULT2B1b may be a potential target for treating hepatic steatosis-related liver regeneration disorders.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yuyin Xu
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Han
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
53
|
Early Transcriptional Changes within Liver, Adrenal Gland, and Lymphoid Tissues Significantly Contribute to Ebola Virus Pathogenesis in Cynomolgus Macaques. J Virol 2020; 94:JVI.00250-20. [PMID: 32213610 DOI: 10.1128/jvi.00250-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/05/2023] Open
Abstract
Ebola virus (EBOV) continues to pose a significant threat to human health, as evidenced by the 2013-2016 epidemic in West Africa and the ongoing outbreak in the Democratic Republic of the Congo. EBOV causes hemorrhagic fever, organ damage, and shock culminating in death, with case fatality rates as high as 90%. This high lethality combined with the paucity of licensed medical countermeasures makes EBOV a critical human pathogen. Although EBOV infection results in significant damage to the liver and the adrenal glands, little is known about the molecular signatures of injury in these organs. Moreover, while changes in peripheral blood cells are becoming increasingly understood, the host responses within organs and lymphoid tissues remain poorly characterized. To address this knowledge gap, we tracked longitudinal transcriptional changes in tissues collected from EBOV-Makona-infected cynomolgus macaques. Following infection, both liver and adrenal glands exhibited significant and early downregulation of genes involved in metabolism, coagulation, hormone synthesis, and angiogenesis; upregulated genes were associated with inflammation. Analysis of lymphoid tissues showed early upregulation of genes that play a role in innate immunity and inflammation and downregulation of genes associated with cell cycle and adaptive immunity. Moreover, transient activation of innate immune responses and downregulation of humoral immune responses in lymphoid tissues were confirmed with flow cytometry. Together, these data suggest that the liver, adrenal gland, and lymphatic organs are important sites of EBOV infection and that dysregulating the function of these vital organs contributes to the development of Ebola virus disease.IMPORTANCE Ebola virus (EBOV) remains a high-priority pathogen since it continues to cause outbreaks with high case fatality rates. Although it is well established that EBOV results in severe organ damage, our understanding of tissue injury in the liver, adrenal glands, and lymphoid tissues remains limited. We begin to address this knowledge gap by conducting longitudinal gene expression studies in these tissues, which were collected from EBOV-infected cynomolgus macaques. We report robust and early gene expression changes within these tissues, indicating they are primary sites of EBOV infection. Furthermore, genes involved in metabolism, coagulation, and adaptive immunity were downregulated, while inflammation-related genes were upregulated. These results indicate significant tissue damage consistent with the development of hemorrhagic fever and lymphopenia. Our study provides novel insight into EBOV-host interactions and elucidates how host responses within the liver, adrenal glands, and lymphoid tissues contribute to EBOV pathogenesis.
Collapse
|
54
|
Yuan X, Lu H, Zhao A, Ding Y, Min Q, Wang R. Transcriptional regulation of CYP3A4 by nuclear receptors in human hepatocytes under hypoxia. Drug Metab Rev 2020; 52:225-234. [PMID: 32270716 DOI: 10.1080/03602532.2020.1733004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/17/2020] [Indexed: 02/08/2023]
Abstract
The human hepatic cytochrome P-450 3A4 (CYP3A4), recognized as a multifunctional enzyme, has a wide range of substrates including commonly used drugs. Previous investigations demonstrated that the expression of CYP3A4 in human hepatocytes could be regulated by some nuclear receptors (NRs) at transcriptional level under diverse situations. The significance of oxygen on CYP3A4-mediated metabolism seems notable while the regulatory mode of CYP3A4 in the particular case still remains elusive. Recently, striking evidence has emerged that both CYP3A4 and its regulator NR could be inhibited by exposure to hypoxia. Therefore, it is of great importance to elucidate whether and how these NRs act in the transcriptional regulation of CYP3A4 in human hepatocytes under hypoxic conditions. In this review, we mainly summarized transcriptional regulation of the pivotal enzyme CYP3A4 by NRs and explored the possible regulatory pathways of CYP3A4 via these major NRs under hypoxia, expecting to provide favorable evidence for further clinical guidance under such pathological situations.
Collapse
Affiliation(s)
- Xuechun Yuan
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
- College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hui Lu
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Anpeng Zhao
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Yidan Ding
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
- College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qiong Min
- Pharmacy department, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Rong Wang
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
- College of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
55
|
Bolleyn J, Rombaut M, Nair N, Branson S, Heymans A, Chuah M, VandenDriessche T, Rogiers V, De Kock J, Vanhaecke T. Genetic and Epigenetic Modification of Rat Liver Progenitor Cells via HNF4α Transduction and 5' Azacytidine Treatment: An Integrated miRNA and mRNA Expression Profile Analysis. Genes (Basel) 2020; 11:E486. [PMID: 32365562 PMCID: PMC7291069 DOI: 10.3390/genes11050486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Neonatal liver-derived rat epithelial cells (rLEC) from biliary origin are liver progenitor cells that acquire a hepatocyte-like phenotype upon sequential exposure to hepatogenic growth factors and cytokines. Undifferentiated rLEC express several liver-enriched transcription factors, including the hepatocyte nuclear factors (HNF) 3β and HNF6, but not the hepatic master regulator HNF4α. In this study, we first investigated the impact of the ectopic expression of HNF4α in rLEC on both mRNA and microRNA (miR) level by means of microarray technology. We found that HNF4α transduction did not induce major changes to the rLEC phenotype. However, we next investigated the influence of DNA methyl transferase (DNMT) inhibition on the phenotype of undifferentiated naïve rLEC by exposure to 5' azacytidine (AZA), which was found to have a significant impact on rLEC gene expression. The transduction of HNF4α or AZA treatment resulted both in significantly downregulated C/EBPα expression levels, while the exposure of the cells to AZA had a significant effect on the expression of HNF3β. Computationally, dysregulated miRNAs were linked to target mRNAs using the microRNA Target Filter function of Ingenuity Pathway Analysis. We found that differentially regulated miRNA-mRNA target associations predict ectopic HNF4α expression in naïve rLEC to interfere with cell viability and cellular maturation (miR-19b-3p/NR4A2, miR30C-5p/P4HA2, miR328-3p/CD44) while it predicts AZA exposure to modulate epithelial/hepatic cell proliferation, apoptosis, cell cycle progression and the differentiation of stem cells (miR-18a-5p/ESR1, miR-503-5p/CCND1). Finally, our computational analysis predicts that the combination of HNF4α transduction with subsequent AZA treatment might cause changes in hepatic cell proliferation and maturation (miR-18a-5p/ESR1, miR-503-5p/CCND1, miR-328-3p/CD44) as well as the apoptosis (miR-16-5p/BCL2, miR-17-5p/BCL2, miR-34a-5p/BCL2 and miR-494-3p/HMOX1) of naïve rLEC.
Collapse
Affiliation(s)
- Jennifer Bolleyn
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Matthias Rombaut
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Nisha Nair
- Department of Gene Therapy and Regenerative Medicine (GTRM), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (N.N.); (M.C.); (T.V.)
| | - Steven Branson
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Anja Heymans
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Marinee Chuah
- Department of Gene Therapy and Regenerative Medicine (GTRM), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (N.N.); (M.C.); (T.V.)
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine (GTRM), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (N.N.); (M.C.); (T.V.)
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-cosmetology (IVTD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.B.); (M.R.); (S.B.); (A.H.); (V.R.); (T.V.)
| |
Collapse
|
56
|
Li ZH, Xie ZY, Ouyang XX, Huang KZ, Yu XP, Zhao YL, Zhang YH, Zhu DH, Yu J, Li LJ. Assessment of biological functions for C3A cells interacting with adverse environments of liver failure plasma. Hepatobiliary Pancreat Dis Int 2020; 19:129-137. [PMID: 31704080 DOI: 10.1016/j.hbpd.2019.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND For its better differentiated hepatocyte phenotype, C3A cell line has been utilized in bioartificial liver system. However, up to now, there are only a few of studies working at the metabolic alternations of C3A cells under the culture conditions with liver failure plasma, which mainly focus on carbohydrate metabolism, total protein synthesis and ureagenesis. In this study, we investigated the effects of acute liver failure plasma on the growth and biological functions of C3A cells, especially on CYP450 enzymes. METHODS C3A cells were treated with fresh DMEM medium containing 10% FBS, fresh DMEM medium containing 10% normal plasma and acute liver failure plasma, respectively. After incubation, the C3A cells were assessed for cell viabilities, lactate dehydrogenase leakage, gene transcription, protein levels, albumin secretion, ammonia metabolism and CYP450 enzyme activities. RESULTS Cell viabilities decreased 15%, and lactate dehydrogenase leakage had 1.3-fold elevation in acute liver failure plasma group. Gene transcription exhibited up-regulation, down-regulation or stability for different hepatic genes. In contrast, protein expression levels for several CYP450 enzymes kept constant, while the CYP450 enzyme activities decreased or remained stable. Albumin secretion reduced about 48%, and ammonia accumulation increased approximately 41%. CONCLUSIONS C3A cells cultured with acute liver failure plasma showed mild inhibition of cell viabilities, reduction of albumin secretion, and increase of ammonia accumulation. Furthermore, CYP450 enzymes demonstrated various alterations on gene transcription, protein expression and enzyme activities.
Collapse
Affiliation(s)
- Zu-Hong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhong-Yang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiao-Xi Ouyang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kai-Zhou Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiao-Peng Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ya-Lei Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yan-Hong Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Dan-Hua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lan-Juan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
57
|
Hao J, Qi T, Zhu X, Chen J. Comparative Proteomic Analyses of the Liver in D-Galactosamine-Sensitized Mice Treated with Different Toll-Like Receptor Agonists. Proteomics 2020; 20:e1900393. [PMID: 32131144 DOI: 10.1002/pmic.201900393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/13/2020] [Indexed: 01/03/2023]
Abstract
Acute liver failure (ALF) is a severe consequence of abrupt hepatocyte injury and has lethal outcomes. Three toll-like receptor agonists, including polyinosinic-polycytidylic acid (poly(I:C)), lipopolysaccharide (LPS), and cytosine-phosphate-guanine (CpG) DNA, cause acute and severe hepatitis, respectively, in D-galactosamine (D-GalN)-sensitized mice. However, the molecular differences among three ALF models (LPS/D-GalN, poly(I:C)/D-GalN, and CpG DNA/D-GalN), are unclear. Here, tandem mass tag based quantitative proteomic analyses of three ALF mouse models are performed. 52 common differentially expressed proteins (DEPs) are identified, in three ALF groups, compared to the control. Gene ontology analyses show that among the common DEPs, ten proteins are involved in immune system process, and 39 proteins in metabolic process. Among 80,195, and 23 specifically-expressed proteins in poly(I:C)/D-GalN, LPS/D-GalN, and CpG DNA/D-GalN groups, LPS/D-GalN-specific proteins are mostly distributed in the endoplasmic reticulum and more enriched in metabolic pathways, whereas poly (I:C)/D-GalN-specific proteins are mainly in the membrane and CpG DNA/D-GalN-specific proteins are related to the ribosome structural composition. In conclusion, the common and specific DEPs in three ALF mouse models at molecular level are identified; and determined a close-to-complete reference map of mouse liver proteins which will be useful for clinical diagnosis and treatment of liver failure in humans.
Collapse
Affiliation(s)
- Jun Hao
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Tingting Qi
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xiaoying Zhu
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jinjun Chen
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
58
|
Thang SK, Chen PY, Gao WY, Wu MJ, Pan MH, Yen JH. Xanthohumol Suppresses NPC1L1 Gene Expression through Downregulation of HNF-4α and Inhibits Cholesterol Uptake in Caco-2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11119-11128. [PMID: 31525874 DOI: 10.1021/acs.jafc.9b05221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Xanthohumol (Xan) is a prenylated chalcone mainly found in hops; it has been demonstrated to function against hypercholesterolemia, hyperlipidemia, and atherosclerosis. In this study, we focused on the hypocholesterolemic effect of Xan on cholesterol uptake and the underlying molecular mechanisms of Xan in human intestinal Caco-2 cells. The microarray data showed that Niemann-Pick C1-like 1 (NPC1L1), an essential transporter for dietary cholesterol absorption, was significantly downregulated in Xan-treated Caco-2 cells. We demonstrated that Xan (10 and 20 μM) suppressed the mRNA and protein expression of NPC1L1 by 0.65 ± 0.12-fold and 0.54 ± 0.15-fold and 0.72 ± 0.04-fold and 0.44 ± 0.12-fold, respectively, compared to that of the vehicle-treated Caco-2 cells. Moreover, Xan (10 and 20 μM) significantly inhibited cholesterol uptake by approximately 12 and 32% in Caco-2 cells. NPC1L1 promoter activity was significantly suppressed by Xan, and a DNA element within the NPC1L1 promoter involved in Xan-mediated NPC1L1 reduction located between the -120 and -20 positions was identified. Moreover, Xan markedly decreased the mRNA and protein levels of hepatocyte nuclear factor 4α (HNF-4α), a critical activator of NPC1L1 transcription, and subsequently attenuated HNF-4α/NPC1L1 promoter complex formation, resulting in the suppression of NPC1L1 gene expression. Finally, we demonstrated that Xan markedly abolished lovastatin-induced NPC1L1 overexpression in Caco-2 cells. These findings reveal that Xan suppresses NPC1L1 expression via downregulation of HNF-4α and exerts inhibitory effects on cholesterol uptake in the intestinal Caco-2 cells. Our findings suggest Xan could serve as a potential cholesterol-lowering agent and supplement for statin therapy.
Collapse
Affiliation(s)
- Sang Kim Thang
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
| | - Pei-Yi Chen
- Center of Medical Genetics , Hualien Tzu Chi Hospital, Buddhist Tzu Chi Foundation , Hualien 970 , Taiwan
| | - Wan-Yun Gao
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| | - Ming-Jiuan Wu
- Department of Biotechnology , Chia-Nan University of Pharmacy and Science , Tainan 717 , Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology , National Taiwan University , Taipei 10617 , Taiwan
| | - Jui-Hung Yen
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| |
Collapse
|
59
|
Rajan M, Anderson CP, Rindler PM, Romney SJ, Ferreira dos Santos MC, Gertz J, Leibold EA. NHR-14 loss of function couples intestinal iron uptake with innate immunity in C. elegans through PQM-1 signaling. eLife 2019; 8:e44674. [PMID: 31532389 PMCID: PMC6777940 DOI: 10.7554/elife.44674] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023] Open
Abstract
Iron is essential for survival of most organisms. All organisms have thus developed mechanisms to sense, acquire and sequester iron. In C. elegans, iron uptake and sequestration are regulated by HIF-1. We previously showed that hif-1 mutants are developmentally delayed when grown under iron limitation. Here we identify nhr-14, encoding a nuclear receptor, in a screen conducted for mutations that rescue the developmental delay of hif-1 mutants under iron limitation. nhr-14 loss upregulates the intestinal metal transporter SMF-3 to increase iron uptake in hif-1 mutants. nhr-14 mutants display increased expression of innate immune genes and DAF-16/FoxO-Class II genes, and enhanced resistance to Pseudomonas aeruginosa. These responses are dependent on the transcription factor PQM-1, which localizes to intestinal cell nuclei in nhr-14 mutants. Our data reveal how C. elegans utilizes nuclear receptors to regulate innate immunity and iron availability, and show iron sequestration as a component of the innate immune response.
Collapse
Affiliation(s)
- Malini Rajan
- Department of Medicine, Division of HematologyUniversity of UtahSalt Lake CityUnited States
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
| | - Cole P Anderson
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
- Department of Oncological SciencesUniversity of UtahSalt Lake CityUnited States
| | - Paul M Rindler
- Department of Medicine, Division of HematologyUniversity of UtahSalt Lake CityUnited States
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
| | - Steven Joshua Romney
- Department of Medicine, Division of HematologyUniversity of UtahSalt Lake CityUnited States
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
| | - Maria C Ferreira dos Santos
- Department of Medicine, Division of HematologyUniversity of UtahSalt Lake CityUnited States
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
| | - Jason Gertz
- Department of Oncological SciencesUniversity of UtahSalt Lake CityUnited States
- Huntsman Cancer InstituteUniversity of UtahSalt Lake CityUnited States
| | - Elizabeth A Leibold
- Department of Medicine, Division of HematologyUniversity of UtahSalt Lake CityUnited States
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUnited States
- Department of Oncological SciencesUniversity of UtahSalt Lake CityUnited States
| |
Collapse
|
60
|
Huck I, Gunewardena S, Espanol-Suner R, Willenbring H, Apte U. Hepatocyte Nuclear Factor 4 Alpha Activation Is Essential for Termination of Liver Regeneration in Mice. Hepatology 2019; 70:666-681. [PMID: 30520062 PMCID: PMC6551324 DOI: 10.1002/hep.30405] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022]
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) is critical for hepatic differentiation. Recent studies have highlighted its role in inhibition of hepatocyte proliferation and tumor suppression. However, the role of HNF4α in liver regeneration (LR) is not known. We hypothesized that hepatocytes modulate HNF4α activity when navigating between differentiated and proliferative states during LR. Western blotting analysis revealed a rapid decline in nuclear and cytoplasmic HNF4α protein levels, accompanied with decreased target gene expression, within 1 hour after two-thirds partial hepatectomy (post-PH) in C57BL/6J mice. HNF4α protein expression did not recover to pre-PH levels until day 3. Hepatocyte-specific deletion of HNF4α (HNF4α-KO [knockout]) in mice resulted in 100% mortality post-PH, despite increased proliferative marker expression throughout regeneration. Sustained loss of HNF4α target gene expression throughout regeneration indicated that HNF4α-KO mice were unable to compensate for loss of HNF4α transcriptional activity. Deletion of HNF4α resulted in sustained proliferation accompanied by c-Myc and cyclin D1 overexpression and a complete deficiency of hepatocyte function after PH. Interestingly, overexpression of degradation-resistant HNF4α in hepatocytes delayed, but did not prevent, initiation of regeneration after PH. Finally, adeno-associated virus serotype 8 (AAV8)-mediated reexpression of HNF4α in hepatocytes of HNF4α-KO mice post-PH restored HNF4α protein levels, induced target gene expression, and improved survival of HNF4α-KO mice post-PH. Conclusion: In conclusion, these data indicate that HNF4α reexpression following initial decrease is critical for hepatocytes to exit from cell cycle and resume function during the termination phase of LR. These results indicate the role of HNF4α in LR and have implications for therapy of liver failure.
Collapse
Affiliation(s)
- Ian Huck
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Sumedha Gunewardena
- Department of Biostatistics University of Kansas Medical Center, Kansas City, KS
| | | | - Holger Willenbring
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research,Liver Center, Division of Transplantation, University of California San Francisco, San Francisco, CA,Department of Surgery, Division of Transplantation, University of California San Francisco, San Francisco, CA
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
61
|
Xie P, Hu X, Li D, Xie S, Zhou Z, Meng X, Shan H. Bioluminescence Imaging of Transplanted Mesenchymal Stem Cells by Overexpression of Hepatocyte Nuclear Factor4α: Tracking Biodistribution and Survival. Mol Imaging Biol 2019; 21:44-53. [PMID: 29761416 DOI: 10.1007/s11307-018-1204-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE The purposes of this study were to construct immortalized human bone marrow mesenchymal stem cells (UE7T-13) with overexpression of the hepatocyte nuclear factor4α (hHNF4α) and luciferase2-mKate2 dual-fusion reporter gene, further investigate their impact on treating acute liver injury (ALI) in rats, and track their biodistribution and survival by bioluminescence imaging (BLI). PROCEDURES The hHNF4α and luciferase2-mKate2 genes were transduced by a lentiviral vector into UE7T-13 cells (named E7-hHNF4α-R cells), and expression was verified by immunofluorescence, RT-PCR, and flow cytometry. E7-hGFP-R cells expressing the luciferase2-mKate2/hGFP gene served as a negative group. A correlation between the bioluminescence signal and cell number was detected by BLI. The ALI rats were established and divided into three groups: PBS, E7-hGFP-R, and E7-hHNF4α-R. After transplantation of 2.0 × 106 cells, BLI was used to dynamically track their biodistribution and survival. The restoration of biological functions was assessed by serum biochemical and histological analyses. RESULTS Stable high-level expression of hHNF4α and mKate2 protein was established in the E7-hHNF4α-R cells in vitro. The E7-hHNF4α-R cells strongly expressed hGFP, hHNF4α, and mKate2 proteins, and the hHNF4α gene. hGFP-mKate2 dual-positive cell expression reached approximately 93 %. BLI verified that a linear relationship existed between the cell number and bioluminescence signal (R2 = 0.9991). The cells improved liver function in vivo after transplantation into the ALI rat liver, as evidenced by the fact that AST and ALT temporarily returned to normal levels in the recipient ALI rats. The presence of the transplanted E7-hGFP-R and E7-hHNF4α-R cells in recipient rat livers was confirmed by BLI and immunohistochemistry. However, the cells were cleared by the immune system a short time after transplantation into ALI rats with a normal immune system. CONCLUSION Our data revealed that the E7-hHNF4α-R cells can transiently improve damaged liver function and were rapidly cleared by the immune system. In addition, BLI is a useful tool to track transplanted cell biodistribution and survival.
Collapse
Affiliation(s)
- Peiyi Xie
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojun Hu
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China
| | - Dan Li
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China.,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China
| | - Sidong Xie
- The Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyang Zhou
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Hong Shan
- Guang Dong Provincial Engineering Research Center of Molecular Imaging, Zhuhai, China. .,Interventional Medicine Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China. .,Interventional Radiology Institute, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
62
|
Goud ESK, Pandey M, Singh C, Vedamurthy GV, Singh D, Onteru SK. Effect of Dioxins in Milk on the 3D Cultured Primary Buffalo Hepatocyte Model System. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8007-8019. [PMID: 31268702 DOI: 10.1021/acs.jafc.9b03384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cow and human milk have been reported to contain dioxins ranging from 0.023 to 26.46 and 0.88 to 19 pg/g of fat, respectively. However, the toxic effects of the dioxins in the milk in this range of concentrations were not explored. Therefore, considering the outbred livestock tissues as better models than inbred laboratory animals, the present study targeted to study the effect of dioxins present in the milk on three-dimensionally (3D) cultured buffalo primary hepatocyte spheroids. The spheroids were treated with a model dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), directly and also through milk fat at different concentrations (i.e, 0.02-20 pg/mL) for 24 h. Among the liver-cell-specific (ALB, HNF4α, and AFP) genes, a similar ALB and upregulated HNF4α expression at all treatments indicated the functional and transcriptionally active hepatocyte spheroids. Supportingly, no significant difference in the antiapoptotic gene expression between the treatments of milk fat and milk fat containing dioxins indicated the survivability of the spheroids during dioxin treatments. Among the selected TCDD responsive (CYP1A1, CYP1A2, AHR, CYP1B1, and TIPARP) genes, a nonsignificant increasing trend of the CYP1A1 expression was observed from 0.2 to 10 pg/mL of TCDD concentration through milk fat. This pattern was similar to the reported insensitive response of human primary hepatocytes toward dioxins than that of rat primary hepatocytes. This may indicate that the buffalo hepatocyte spheroids could be better models than rats for TCDD hepatotoxic studies. Further, TCDD in the milk in the range of 0.02-20 pg/mL concentration may not be very hepatotoxic.
Collapse
Affiliation(s)
- Emmagouni Sharath Kumar Goud
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Mamta Pandey
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Chhama Singh
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Gowdar Veerappa Vedamurthy
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics and Systems Biology Lab, Animal Biochemistry Division , ICAR-National Dairy Research Institute , Karnal 132001 , India
| |
Collapse
|
63
|
Sun Q, Xu W, Ji S, Qin Y, Liu W, Hu Q, Zhang Z, Liu M, Yu X, Xu X. Role of hepatocyte nuclear factor 4 alpha in cell proliferation and gemcitabine resistance in pancreatic adenocarcinoma. Cancer Cell Int 2019; 19:49. [PMID: 30867652 PMCID: PMC6398265 DOI: 10.1186/s12935-019-0767-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 02/28/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Hepatocyte nuclear factor 4α (HNF4α) is a tissue-specific transcription factor that regulates the expression of numerous genes in hepatocytes and pancreatic β cells. HNF4α has been reported to affect cell proliferation and chemoresistance in several cancers. However, the role of HNF4α in pancreatic adenocarcinoma (PDAC) has not been studied extensively and remains unclear. METHODS By utilizing immunohistochemical (IHC) staining, we measured the expression of HNF4α in PDAC tissues. By silencing HNF4α in PDAC cell lines, we assessed the impact of HNF4α on pancreatic cancer cell proliferation and gemcitabine sensitivity. We used CCK8 and colony formation assays to examine the effect of HNF4α on cell proliferation. A flow cytometry assay was used to assess cell apoptosis. The expression of gemcitabine-related genes was detected by quantitative real‑time PCR (qRT-PCR) and Western blotting. IHC was utilized to assess the correlation between HNF4α and human equilibrative nucleoside transporter 1 (hENT1) expression in PDAC patients. Chromatin immunoprecipitation (ChIP) and dual‑luciferase reporter assays were used to confirm that hENT1 is a target gene of HNF4α. RESULTS Increased HNF4α expression was detected in PDAC tissues; patients with higher HNF4α expression displayed worse prognosis. To elucidate the function of HNF4α, we examined its role in pancreatic cancer cell proliferation, apoptosis and gemcitabine resistance. In HNF4α-silenced Capan-1 and MiaPaCa-2 cells, we observed decreased cell proliferation and increased sensitivity to gemcitabine compared to those of controls. The mechanism of HNF4α in gemcitabine-related chemosensitivity was then explored. In response to HNF4α silencing, the expression levels of gemcitabine-related proteins, hENT1 and deoxycytidine kinase (dCK) were significantly increased. Additionally, hENT1 was negatively correlated with HNF4α in PDAC tissue samples. Moreover, we identified hENT1 as a downstream target of HNF4α. CONCLUSION HNF4α is a prognostic marker for overall survival, is required for pancreatic cancer cell proliferation and promotes resistance to gemcitabine by downregulating hENT1. Therefore, targeting HNF4α might reverse gemcitabine resistance and provide novel treatment strategies for PDAC.
Collapse
Affiliation(s)
- Qiqing Sun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| |
Collapse
|
64
|
Singh P, Tung SP, Han EH, Lee IK, Chi YI. Dimerization defective MODY mutations of hepatocyte nuclear factor 4α. Mutat Res 2019; 814:1-6. [PMID: 30648609 DOI: 10.1016/j.mrfmmm.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/13/2018] [Accepted: 01/08/2019] [Indexed: 10/27/2022]
Abstract
HNF4α is a culprit gene product for a monogenic and dominantly-inherited form of diabetes, referred to as MODY1 (Maturity Onset Diabetes of the Young type 1). Reduced HNF4α activities have been linked to impaired insulin secretion and β-cell function. Numerous mutations have been identified from the patients and they have been instructive as to the individual residue's role in protein structure-function and dysfunction. As a member of the nuclear receptor (NR) superfamily, HNF4α is made of characteristic modular domains and it functions exclusively as a homodimer despite its sequence homology to RXR, a common heterodimer partner of non-steroidal NRs. Transcription factors commonly dimerize to enhance their molecular functions mainly by facilitating the recognition of double helix target DNAs that display an intrinsic pseudo-2-fold symmetry and the recruitment of the remainder of the main transcriptional machinery. HNF4α is no exception and its dimerization is maintained by the ligand binding domain (LBD) mainly through the leucine-zipper-like interactions at the stalk of two interacting helices. Although many MODY1 mutations have been previously characterized, including DNA binding disruptors, ligand binding disruptors, coactivator binding disruptors, and protein stability disruptors, protein dimerization disruptors have not been formally reported. In this report, we present a set of data for the two MODY1 mutations found right at the dimerization interface (L332 P and L328del mutations) which clearly exhibit the disruptive effects of directly affecting dimerization, protein stability, and transcriptional activities. These data reinforced the fact that MODY mutations are loss-of-function mutations and HNF4α dimerization is essential for its optimal function and normal physiology.
Collapse
Affiliation(s)
- Puja Singh
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Shu-Ping Tung
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Eun Hee Han
- Drug & Disease Target Group, Division of Life Science, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Young-In Chi
- Section of Structural Biology, Hormel Institute, University of Minnesota, Austin, MN, United States.
| |
Collapse
|
65
|
Guo S, Lu H. Novel mechanisms of regulation of the expression and transcriptional activity of hepatocyte nuclear factor 4α. J Cell Biochem 2019; 120:519-532. [PMID: 30191603 PMCID: PMC7745837 DOI: 10.1002/jcb.27407] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Hepatocyte nuclear factor 4α (HNF4α) is a master regulator of development and function of digestive tissues. The HNF4A gene uses two separate promoters P1 and P2, with P1 products predominant in adult liver, whereas P2 products prevalent in fetal liver, pancreas, and liver/colon cancer. To date, the mechanisms for the regulation of HNF4A and the dynamic switch of P1-HNF4α and P2-HNF4α during ontogenesis and carcinogenesis are still obscure. Our study validated the previously reported self-stimulation of P1-HNF4α but invalidated the reported synergism between HNF4α and HNF1α. HNF4A-AS1, a long noncoding RNA, is localized between the P2 and P1 promoters of HNF4A. We identified critical roles of P1-HNF4α in regulating the expression of HNF4A-AS1 and its mouse ortholog Hnf4a-os. Paired box 6 (PAX6), a master regulator of pancreas development overexpressed in colon cancer, cooperated with HNF1α to induce P2-HNF4α but antagonized HNF4α in HNF4A-AS1 expression. Thus, PAX6 may be important in determining ontogenic and carcinogenic changes of P2-HNF4α and HNF4A-AS1 in the pancreas and intestine. We also interrogated transactivation activities on multiple gene targets by multiple known and novel HNF4α mutants identified in patients with maturity onset diabetes of the young 1 (MODY1) and liver cancer. Particularly, HNF4α-D78A and HNF4α-G79S, two mutants found in liver cancer with mutations in DNA-binding domain, displayed highly gene-specific transactivation activities. Interestingly, HNF4α-Q277X, a MODY1 truncation mutant, antagonized the transactivation activities of HNF1α and farnesoid X receptor, key regulators of insulin secretion. Taken together, our study provides novel mechanistic insights regarding the transcriptional regulation and transactivation activity of HNF4α in digestive tissues.
Collapse
Affiliation(s)
- Shangdong Guo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S
| |
Collapse
|
66
|
Vilas-Boas V, Cooreman A, Gijbels E, Van Campenhout R, Gustafson E, Ballet S, Annaert P, Cogliati B, Vinken M. Primary hepatocytes and their cultures for the testing of drug-induced liver injury. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 85:1-30. [PMID: 31307583 DOI: 10.1016/bs.apha.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury is a major reason for discontinuation of drug development and withdrawal of drugs from the market. Intensive efforts in the last decades have focused on the establishment and finetuning of liver-based in vitro models for reliable prediction of hepatotoxicity triggered by drug candidates. Of those, primary hepatocytes and their cultures still are considered the gold standard, as they provide an acceptable reflection of the hepatic in vivo situation. Nevertheless, these in vitro systems cope with gradual deterioration of the differentiated morphological and functional phenotype. The present paper gives an overview of traditional and more recently introduced strategies to counteract this dedifferentiation process in an attempt to set up culture models that can be used for long-term testing purposes. The relevance and applicability of such optimized cultures of primary hepatocytes for the testing of drug-induced cholestatic liver injury is demonstrated.
Collapse
Affiliation(s)
- Vânia Vilas-Boas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Axelle Cooreman
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Gijbels
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emma Gustafson
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
67
|
Khojasteh SC, Miller GP, Mitra K, Rietjens IMCM. Biotransformation and bioactivation reactions - 2017 literature highlights *. Drug Metab Rev 2018; 50:221-255. [PMID: 29954222 DOI: 10.1080/03602532.2018.1473875] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
This annual review is the third one to highlight recent advances in the study and assessment of biotransformations and bioactivations ( Table 1 ). We followed the same format as the previous years with selection and authoring each section (see Baillie et al. 2016 ; Khojasteh et al. 2017 ). We acknowledge that many universities no longer train students in mechanistic biotransformation studies reflecting a decline in the investment for those efforts by public funded granting institutions. We hope this work serves as a resource to appreciate the knowledge gained each year to understand and hopefully anticipate toxicological outcomes dependent on biotransformations and bioactivations. This effort itself also continues to evolve. I am pleased that Drs. Rietjens and Miller have again contributed to this annual review. We would like to welcome Kaushik Mitra as an author for this year's issue, and we thank Deepak Dalvie for his contributions to last year's edition. We have intentionally maintained a balance of authors such that two come from an academic setting and two come from industry. As always, please drop us a note if you find this review helpful. We would be pleased to hear your opinions of our commentary, and we extend an invitation to anyone who would like to contribute to a future edition of this review.
Collapse
Affiliation(s)
- S Cyrus Khojasteh
- a Department of Drug Metabolism and Pharmacokinetics , Genentech, Inc , South San Francisco , CA , USA
| | - Grover P Miller
- b Department of Biochemistry and Molecular Biology , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Kaushik Mitra
- c Department of Safety Assessment and Laboratory Animal Resources , Merck Research Laboratories (MRL), Merck & Co., Inc , West Point , PA , USA
| | | |
Collapse
|
68
|
Ten Years' Experience with the CYP2D6 Activity Score: A Perspective on Future Investigations to Improve Clinical Predictions for Precision Therapeutics. J Pers Med 2018; 8:jpm8020015. [PMID: 29673183 PMCID: PMC6023391 DOI: 10.3390/jpm8020015] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/06/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
The seminal paper on the CYP2D6 Activity Score (AS) was first published ten years ago and, since its introduction in 2008, it has been widely accepted in the field of pharmacogenetics. This scoring system facilitates the translation of highly complex CYP2D6 diplotype data into a patient’s phenotype to guide drug therapy and is at the core of all CYP2D6 gene/drug pair guidelines issued by the Clinical Pharmacogenetics Implementation Consortium (CPIC). The AS, however, only explains a portion of the variability observed among individuals and ethnicities. In this review, we provide an overview of sources in addition to CYP2D6 genotype that contribute to the variability in CYP2D6-mediated drug metabolism and discuss other factors, genetic and non-genetic, that likely contribute to the observed variability in CYP2D6 enzymatic activity.
Collapse
|
69
|
Bi Y, Shi X, Zhu J, Guan X, Garbacz WG, Huang Y, Gao L, Yan J, Xu M, Ren S, Ren S, Liu Y, Ma X, Li S, Xie W. Regulation of Cholesterol Sulfotransferase SULT2B1b by Hepatocyte Nuclear Factor 4α Constitutes a Negative Feedback Control of Hepatic Gluconeogenesis. Mol Cell Biol 2018; 38:e00654-17. [PMID: 29378829 PMCID: PMC5854833 DOI: 10.1128/mcb.00654-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/04/2018] [Accepted: 01/18/2018] [Indexed: 01/10/2023] Open
Abstract
The cholesterol sulfotransferase SULT2B1b converts cholesterol to cholesterol sulfate (CS). We previously reported that SULT2B1b inhibits hepatic gluconeogenesis by antagonizing the gluconeogenic activity of hepatocyte nuclear factor 4α (HNF4α). In this study, we showed that the SULT2B1b gene is a transcriptional target of HNF4α, which led to our hypothesis that the induction of SULT2B1b by HNF4α represents a negative feedback to limit the gluconeogenic activity of HNF4α. Indeed, downregulation of Sult2B1b enhanced the gluconeogenic activity of HNF4α, which may have been accounted for by the increased acetylation of HNF4α as a result of decreased expression of the HNF4α deacetylase sirtuin 1 (Sirt1). The expression of Sult2B1b was also induced by HNF4α upon fasting, and the Sult2B1b null (Sult2B1b-/-) mice showed increased gluconeogenic gene expression and an elevated fasting glucose level, suggesting that SULT2B1b also plays a restrictive role in HNF4α-mediated fasting-responsive gluconeogenesis. We also developed thiocholesterol, a hydrolysis-resistant derivative of CS, which showed superior activity to that of the native CS in inhibiting gluconeogenesis and improving insulin sensitivity in high-fat-diet-induced diabetic mice. We conclude that the HNF4α-SULT2B1b-CS axis represents a key endogenous mechanism to prevent uncontrolled gluconeogenesis. Thiocholesterol may be used as a therapeutic agent to manage hyperglycemia.
Collapse
Affiliation(s)
- Yuhan Bi
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiongjie Shi
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Junjie Zhu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiudong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wojciech G Garbacz
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yixian Huang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Li Gao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Jiong Yan
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shunlin Ren
- Department of Medicine, Virginia Commonwealth University, Veterans Affairs McGuire Medical Center, Richmond, Virginia, USA
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Xiaochao Ma
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Song Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
70
|
de Boussac H, Gondeau C, Briolotti P, Duret C, Treindl F, Römer M, Fabre JM, Herrero A, Ramos J, Maurel P, Templin M, Gerbal-Chaloin S, Daujat-Chavanieu M. Epidermal Growth Factor Represses Constitutive Androstane Receptor Expression in Primary Human Hepatocytes and Favors Regulation by Pregnane X Receptor. Drug Metab Dispos 2018; 46:223-236. [PMID: 29269410 DOI: 10.1124/dmd.117.078683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
Growth factors have key roles in liver physiology and pathology, particularly by promoting cell proliferation and growth. Recently, it has been shown that in mouse hepatocytes, epidermal growth factor receptor (EGFR) plays a crucial role in the activation of the xenosensor constitutive androstane receptor (CAR) by the antiepileptic drug phenobarbital. Due to the species selectivity of CAR signaling, here we investigated epidermal growth factor (EGF) role in CAR signaling in primary human hepatocytes. Primary human hepatocytes were incubated with CITCO, a human CAR agonist, or with phenobarbital, an indirect CAR activator, in the presence or absence of EGF. CAR-dependent gene expression modulation and PXR involvement in these responses were assessed upon siRNA-based silencing of the genes that encode CAR and PXR. EGF significantly reduced CAR expression and prevented gene induction by CITCO and, to a lower extent, by phenobarbital. In the absence of EGF, phenobarbital and CITCO modulated the expression of 144 and 111 genes, respectively, in primary human hepatocytes. Among these genes, only 15 were regulated by CITCO and one by phenobarbital in a CAR-dependent manner. Conversely, in the presence of EGF, CITCO and phenobarbital modulated gene expression only in a CAR-independent and PXR-dependent manner. Overall, our findings suggest that in primary human hepatocytes, EGF suppresses specifically CAR signaling mainly through transcriptional regulation and drives the xenobiotic response toward a pregnane X receptor (PXR)-mediated mechanism.
Collapse
Affiliation(s)
- Hugues de Boussac
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Claire Gondeau
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Philippe Briolotti
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Cédric Duret
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Fridolin Treindl
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Michael Römer
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Jean-Michel Fabre
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Astrid Herrero
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Jeanne Ramos
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Patrick Maurel
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Markus Templin
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Sabine Gerbal-Chaloin
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Martine Daujat-Chavanieu
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| |
Collapse
|
71
|
Conjunction of G-quadruplex and stem-loop in the 5' untranslated region of mouse hepatocyte nuclear factor 4-alpha1 mediates strong inhibition of protein expression. Mol Cell Biochem 2018; 446:73-81. [PMID: 29332143 DOI: 10.1007/s11010-018-3274-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
Hepatocyte nuclear factor 4-alpha (HNF4α) is a well-established master regulator of liver development and function. Restoration of HNF4α can treat multiple liver disorders and liver cancers. To date, HNF4α is still "undruggable" due to lack of known activating ligands. Thus, understanding the regulatory mechanism of HNF4α expression may help develop an alternative approach to modulate HNF4α protein levels. G-quadruplexes (G4) are non-canonical stable secondary structures discovered mostly in the promoters of oncogenes. Recent genome-wide studies demonstrate the enrichment of G4s in the 5' untranslated region (UTR). By protoporphyrin IX-binding assay and circular dichroism spectrum, we validated the presence of a chemically highly stable 4-ring G4 within the 5' UTR of mouse Hnf4a1. Our real-time PCR and Western blot data showed that the Hnf4a1 5' UTR caused a remarkable translational suppression regardless of a moderate effect on Hnf4a1 mRNA levels. The subsequent deletion/mutation analysis of Hnf4a1 5' UTR using dual-luciferase reporter assays further demonstrated that although the disruption of the chemically highly stable 4-ring G4 resulted in a marked attenuation of inhibition, the G4 alone only weakly inhibited translation. Likewise, disruption of a long stem-loop adjacent to the 4-ring G4 markedly attenuated translational inhibition, although the stem-loop alone only exerted a weak inhibitory effect. Thus, the tight conjunction of G4s and an adjacent stem-loop within the Hnf4a1 5' UTR was both necessary and sufficient to mediate the very strong translational repression. Our results establish a novel working model that a chemically stable G4 may require co-factors to be bio-stable for exerting biological functions.
Collapse
|
72
|
Guo S, Lu H. Conjunction of potential G-quadruplex and adjacent cis-elements in the 5' UTR of hepatocyte nuclear factor 4-alpha strongly inhibit protein expression. Sci Rep 2017; 7:17444. [PMID: 29234104 PMCID: PMC5727235 DOI: 10.1038/s41598-017-17629-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/28/2017] [Indexed: 01/11/2023] Open
Abstract
Hepatocyte nuclear factor 4-alpha (HNF4α) is a well established master regulator of liver development and function. We identified the in vitro presence of a stable secondary structure, G-quadruplex (G4) in the 5' UTR of P1-HNF4A, the predominant HNF4α isoform(s) in adult liver. Our data suggest that the cooperation of G4 and the adjacent putative protein-binding sites within the 5' UTR was necessary and sufficient to mediate a strong translational repression. This was supported by analysis of deleted/mutated 5'UTRs and two native regulatory single-nucleotide polymorphisms in the 5'UTR. Additional results indicated that G4 motifs in the 5' UTRs of other liver-enriched transcription factors also inhibited protein expression. Moreover, pyridostatin, a G4 ligand, specifically potentiated the translational suppressing effect of P1-HNF4A-5' UTR. In summary, the present study provides the first evidence of the presence of G4 in human P1-HNF4A-5' UTR in vitro, and establishes a novel working model of strong inhibition of protein translation via interactions of G4 with potential RNA-binding proteins (RBPs). The protein expression of the tumor suppressor HNF4α may be inhibited by interactions of RBPs with the G4 motif in the 5' UTR to promote cell proliferation during liver development and carcinogenesis.
Collapse
Affiliation(s)
- Shangdong Guo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, United States
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, United States.
| |
Collapse
|
73
|
Ranjan A, Ansari SA. Therapeutic potential of Mediator complex subunits in metabolic diseases. Biochimie 2017; 144:41-49. [PMID: 29061530 DOI: 10.1016/j.biochi.2017.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/16/2017] [Indexed: 01/16/2023]
Abstract
The multisubunit Mediator is an evolutionary conserved transcriptional coregulatory complex in eukaryotes. It is needed for the transcriptional regulation of gene expression in general as well as in a gene specific manner. Mediator complex subunits interact with different transcription factors as well as components of RNA Pol II transcription initiation complex and in doing so act as a bridge between gene specific transcription factors and general Pol II transcription machinery. Specific interaction of various Mediator subunits with nuclear receptors (NRs) and other transcription factors involved in metabolism has been reported in different studies. Evidences indicate that ligand-activated NRs recruit Mediator complex for RNA Pol II-dependent gene transcription. These NRs have been explored as therapeutic targets in different metabolic diseases; however, they show side-effects as targets due to their overlapping involvement in different signaling pathways. Here we discuss the interaction of various Mediator subunits with transcription factors involved in metabolism and whether specific interaction of these transcription factors with Mediator subunits could be potentially utilized as therapeutic strategy in a variety of metabolic diseases.
Collapse
Affiliation(s)
- Amol Ranjan
- Stowers Institute for Medical Research, 1000 E, 50th Street, Kansas City, MO, 64110, USA
| | - Suraiya A Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, UAE University, AlAin, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
74
|
Inoue J, Ikeda S, Kanayama T, Sato R. The flavonoid derivative 4′-nitro-6-hydroxyflavone suppresses the activity of HNF4α and stimulates the degradation of HNF4α protein through the activation of AMPK. Biosci Biotechnol Biochem 2017; 81:1548-1552. [DOI: 10.1080/09168451.2017.1325316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Hepatocyte nuclear factor 4α (HNF4α) is a nuclear receptor that regulates the expression of genes involved in a number of critical metabolic pathways. The modulation of HNF4α activity is thought to be a promising drug target pathway for hyperlipidemia. To identify compounds that reduce the activity of HNF4α, we conducted luciferase reporter assays using the promoter region of microsomal triglyceride transfer protein (MTP) gene, which contains an HNF4α-responsive element. Using this system, we show here that the flavonoid derivative 4′-nitro-6-hydroxyflavone (NOHF) suppresses MTP promoter activity. Treatment with NOHF caused a decrease in the expression of the HNF4α target gene. We also identified that NOHF triggers the AMP-activated protein kinase (AMPK) and accelerates the degradation of HNF4α protein. Knock-down of AMPK diminishes the effect of NOHF. These results indicate that NOHF is an AMPK activator and attenuates the transcriptional activity of HNF4α, at least in part, by accelerating HNF4α protein degradation.
Collapse
Affiliation(s)
- Jun Inoue
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shiori Ikeda
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohiko Kanayama
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
75
|
Shen C, Zhang B, Liu Z, Tang Y, Zhang Y, Wang S, Guo Y, Ding Y, Wang S, Ding M. Effects of ABCB1, ABCC2, UGT2B7 and HNF4α genetic polymorphisms on oxcarbazepine concentrations and therapeutic efficacy in patients with epilepsy. Seizure 2017; 51:102-106. [PMID: 28837897 DOI: 10.1016/j.seizure.2017.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The aim of the study is to investigate the effects of ABCB1, ABCC2, UGT2B7 and HNF4α genetic polymorphisms on plasma oxcarbazepine (OXC) concentrations and therapeutic efficacy in Han Chinese patients with epilepsy. METHODS We recruited 116 Han Chinese patients with epilepsy who were receiving OXC monotherapy. Blood samples were taken and OXC levels were measured. The polymorphisms of ABCB1 rs1045642, ABCC2 rs2273697, UGT2B7 rs7439366, and HNF4α rs2071197 were determined. The therapeutic efficacy of OXC at the 1-year time-point was assessed. Data analysis was performed using IBM SPSS Statistics 22.0. RESULTS The genetic polymorphism of ABCB1 rs1045642 was found to be associated with normalized OXC concentration and therapeutic efficacy in patients with epilepsy (P<0.05). As for UGT2B7 rs7439366, the allele polymorphism exhibited a correlation with treatment outcome, but not OXC concentration. The polymorphisms of ABCC2 rs2273697 and HNF4α rs2071197 was not associated with OXC concentrations and therapeutic efficacy. CONCLUSION These results suggested that ABCB1 rs1045642 and UGT2B7 rs7439366 may affect OXC pharmacokinetics and therapeutic efficacy in Han Chinese patients with epilepsy. However, further studies in larger populations and other ethnic groups are required.
Collapse
Affiliation(s)
- Chunhong Shen
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Bijun Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhirong Liu
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yelei Tang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yinxi Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Shan Wang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yi Guo
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yao Ding
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Shuang Wang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Meiping Ding
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
76
|
Hanging Drop, A Best Three-Dimensional (3D) Culture Method for Primary Buffalo and Sheep Hepatocytes. Sci Rep 2017; 7:1203. [PMID: 28446763 PMCID: PMC5430879 DOI: 10.1038/s41598-017-01355-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/27/2017] [Indexed: 02/07/2023] Open
Abstract
Livestock, having close resemblance to humans, could be a better source of primary hepatocytes than rodents. Herein, we successfully developed three-dimensional (3D) culturing system for primary sheep and buffalo hepatocytes. The 3D-structures of sheep hepatocytes were formed on the fifth-day and maintained until the tenth-day on polyHEMA-coated plates and in hanging drops with William’s E media (HDW). Between the cultured and fresh cells, we observed a similar expression of GAPDH, HNF4α, ALB, CYP1A1, CK8 and CK18. Interestingly, a statistically significant increase was noted in the TAT, CPS, AFP, AAT, GSP and PCNA expression. In buffalo hepatocytes culture, 3D-like structures were formed on the third-day and maintained until the sixth-day on polyHEMA and HDW. The expression of HNF4α, GSP, CPS, AFP, AAT, PCNA and CK18 was similar between cultured and fresh cells. Further, a statistically significant increase in the TAT and CK8 expression, and a decrease in the GAPDH, CYP1A1 and ALB expression were noted. Among the culture systems, HDW maintained the liver transcript markers more or less similar to the fresh hepatocytes of the sheep and buffalo for ten and six days, respectively. Taken together, hanging drop is an efficient method for 3D culturing of primary sheep and buffalo hepatocytes.
Collapse
|
77
|
Chen R, Feng Y, Wu J, Song Y, Li H, Shen Q, Li D, Zhang J, Lu Z, Xiao H, Zhang Y. Metformin attenuates angiotensin II-induced TGFβ1 expression by targeting hepatocyte nuclear factor-4-α. Br J Pharmacol 2017; 175:1217-1229. [PMID: 28230250 DOI: 10.1111/bph.13753] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/26/2017] [Accepted: 02/13/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Metformin, a small molecule, antihyperglycaemic agent, is a well-known activator of AMP-activated protein kinase (AMPK) and protects against cardiac fibrosis. However, the underlying mechanisms remain elusive. TGFβ1 is a key cytokine mediating cardiac fibrosis. Here, we investigated the effects of metformin on TGFβ1 production induced by angiotensin II (AngII) and the underlying mechanisms. EXPERIMENTAL APPROACH Wild-type and AMPKα2-/- C57BL/6 mice were injected s.c. with metformin or saline and infused with AngII (3 mg·kg-1 ·day-1 ) for 7 days. Adult mouse cardiac fibroblasts (CFs) were isolated for in vitro experiments. KEY RESULTS In CFs, metformin inhibited AngII-induced TGFβ1 expression via AMPK activation. Analysis using bioinformatics predicted a potential hepatocyte nuclear factor 4α (HNF4α)-binding site in the promoter region of the Tgfb1 gene. Overexpressing HNF4α increased TGFβ1 expression in CFs. HNF4α siRNA attenuated AngII-induced TGFβ1 production and cardiac fibrosis in vitro and in vivo. Metformin inhibited the AngII-induced increases in HNF4α protein expression and binding to the Tgfb1 promoter in CFs. In vivo, metformin blocked the AngII-induced increase in cardiac HNF4α protein levels in wild-type mice but not in AMPKα2-/- mice. Consequently, metformin inhibited AngII-induced TGFβ1 production and cardiac fibrosis in wild-type mice but not in AMPKα2-/- mice. CONCLUSIONS AND IMPLICATIONS HNF4α mediates AngII-induced TGFβ1 transcription and cardiac fibrosis. Metformin inhibits AngII-induced HNF4α expression via AMPK activation, thus decreasing TGFβ1 transcription and cardiac fibrosis. These findings reveal a novel antifibrotic mechanism of action of metformin and identify HNF4α as a new potential therapeutic target for cardiac fibrosis. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Ruifei Chen
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yenan Feng
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Jimin Wu
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yao Song
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Hao Li
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Qiang Shen
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Dan Li
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Jianshu Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Zhizhen Lu
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Han Xiao
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| |
Collapse
|
78
|
The transcriptional activity of hepatocyte nuclear factor 4 alpha is inhibited via phosphorylation by ERK1/2. PLoS One 2017; 12:e0172020. [PMID: 28196117 PMCID: PMC5308853 DOI: 10.1371/journal.pone.0172020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/30/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) nuclear receptor is a master regulator of hepatocyte development, nutrient transport and metabolism. HNF4α is regulated both at the transcriptional and post-transcriptional levels by different mechanisms. Several kinases (PKA, PKC, AMPK) were shown to phosphorylate and decrease the activity of HNF4α. Activation of the ERK1/2 signalling pathway, inducing proliferation and survival, inhibits the expression of HNF4α. However, based on our previous results we hypothesized that HNF4α is also regulated at the post-transcriptional level by ERK1/2. Here we show that ERK1/2 is capable of directly phosphorylating HNF4α in vitro at several phosphorylation sites including residues previously shown to be targeted by other kinases, as well. Furthermore, we also demonstrate that phosphorylation of HNF4α leads to a reduced trans-activational capacity of the nuclear receptor in luciferase reporter gene assay. We confirm the functional relevance of these findings by demonstrating with ChIP-qPCR experiments that 30-minute activation of ERK1/2 leads to reduced chromatin binding of HNF4α. Accordingly, we have observed decreasing but not disappearing binding of HNF4α to the target genes. In addition, 24-hour activation of the pathway further decreased HNF4α chromatin binding to specific loci in ChIP-qPCR experiments, which confirms the previous reports on the decreased expression of the HNF4a gene due to ERK1/2 activation. Our data suggest that the ERK1/2 pathway plays an important role in the regulation of HNF4α-dependent hepatic gene expression.
Collapse
|
79
|
Lu H, Lei X, Liu J, Klaassen C. Regulation of hepatic microRNA expression by hepatocyte nuclear factor 4 alpha. World J Hepatol 2017; 9:191-208. [PMID: 28217257 PMCID: PMC5295159 DOI: 10.4254/wjh.v9.i4.191] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/02/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To uncover the role of hepatocyte nuclear factor 4 alpha (HNF4α) in regulating hepatic expression of microRNAs.
METHODS Microarray and real-time PCR were used to determine hepatic expression of microRNAs in young-adult mice lacking Hnf4α expression in liver (Hnf4α-LivKO). Integrative genomics viewer software was used to analyze the public chromatin immunoprecipitation-sequencing datasets for DNA-binding of HNF4α, RNA polymerase-II, and histone modifications to loci of microRNAs in mouse liver and human hepatoma cells. Dual-luciferase reporter assay was conducted to determine effects of HNF4α on the promoters of mouse and human microRNAs as well as effects of microRNAs on the untranslated regions (3’UTR) of two genes in human hepatoma cells.
RESULTS Microarray data indicated that most microRNAs remained unaltered by Hnf4α deficiency in Hnf4α-LivKO mice. However, certain liver-predominant microRNAs were down-regulated similarly in young-adult male and female Hnf4α-LivKO mice. The down-regulation of miR-101, miR-192, miR-193a, miR-194, miR-215, miR-802, and miR-122 as well as induction of miR-34 and miR-29 in male Hnf4α-LivKO mice were confirmed by real-time PCR. Analysis of public chromatin immunoprecipitation-sequencing data indicates that HNF4α directly binds to the promoters of miR-101, miR-122, miR-194-2/miR-192 and miR-193, which is associated with histone marks of active transcription. Luciferase reporter assay showed that HNF4α markedly activated the promoters of mouse and human miR-101b/miR-101-2 and the miR-194/miR-192 cluster. Additionally, miR-192 and miR-194 significantly decreased activities of luciferase reporters for the 3’UTR of histone H3F3 and chromodomain helicase DNA binding protein 1 (CHD1), respectively, suggesting that miR-192 and miR-194 might be important in chromosome remodeling through directly targeting H3F3 and CHD1.
CONCLUSION HNF4α is essential for hepatic basal expression of a group of liver-enriched microRNAs, including miR-101, miR-192, miR-193a, miR-194 and miR-802, through which HNF4α may play a major role in the post-transcriptional regulation of gene expression and maintenance of the epigenome in liver.
Collapse
|
80
|
Pan X, Ning M, Jeong H. Transcriptional Regulation of CYP2D6 Expression. Drug Metab Dispos 2017; 45:42-48. [PMID: 27698228 PMCID: PMC5193069 DOI: 10.1124/dmd.116.072249] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/29/2016] [Indexed: 01/04/2023] Open
Abstract
CYP2D6-mediated drug metabolism exhibits large interindividual variability. Although genetic variations in the CYP2D6 gene are well known contributors to the variability, the sources of CYP2D6 variability in individuals of the same genotype remain unexplained. Accumulating data indicate that transcriptional regulation of CYP2D6 may account for part of CYP2D6 variability. Yet, our understanding of factors governing transcriptional regulation of CYP2D6 is limited. Recently, mechanistic studies of increased CYP2D6-mediated drug metabolism in pregnancy revealed two transcription factors, small heterodimer partner (SHP) and Krüppel-like factor 9, as a transcriptional repressor and an activator, respectively, of CYP2D6. Chemicals that increase SHP expression (e.g., retinoids and activators of farnesoid X receptor) were shown to downregulate CYP2D6 expression in the humanized mice as well as in human hepatocytes. This review summarizes the series of studies on the transcriptional regulation of CYP2D6 expression, potentially providing a basis to better understand the large interindividual variability in CYP2D6-mediated drug metabolism.
Collapse
Affiliation(s)
- Xian Pan
- Department of Biopharmaceutical Sciences (X.P., M.N., H.J.), and Department of Pharmacy Practice (H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Miaoran Ning
- Department of Biopharmaceutical Sciences (X.P., M.N., H.J.), and Department of Pharmacy Practice (H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Hyunyoung Jeong
- Department of Biopharmaceutical Sciences (X.P., M.N., H.J.), and Department of Pharmacy Practice (H.J.), College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
81
|
Yang L, Li P, Yang W, Ruan X, Kiesewetter K, Zhu J, Cao H. Integrative Transcriptome Analyses of Metabolic Responses in Mice Define Pivotal LncRNA Metabolic Regulators. Cell Metab 2016; 24:627-639. [PMID: 27667668 PMCID: PMC5181118 DOI: 10.1016/j.cmet.2016.08.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 06/23/2016] [Accepted: 08/25/2016] [Indexed: 12/22/2022]
Abstract
To systemically identify long noncoding RNAs (lncRNAs) regulating energy metabolism, we performed transcriptome analyses to simultaneously profile mRNAs and lncRNAs in key metabolic organs in mice under pathophysiologically representative metabolic conditions. Of 4,759 regulated lncRNAs, function-oriented filters yield 359 tissue-specifically regulated and metabolically sensitive lncRNAs that are predicted by lncRNA-mRNA correlation analyses to function in diverse aspects of energy metabolism. Specific regulations of liver metabolically sensitive lncRNAs (lncLMS) by nutrients, metabolic hormones, and key transcription factors were further defined in primary hepatocytes. Combining genome-wide screens, bioinformatics function predictions, and cell-based analyses, we developed an integrative roadmap to identify lncRNA metabolic regulators. An lncLMS was experimentally confirmed in mice to suppress lipogenesis by forming a negative feedback loop in the SREBP1c pathway. Taken together, this study supports that a class of lncRNAs function as important metabolic regulators and establishes a framework for systemically investigating the role of lncRNAs in physiological homeostasis.
Collapse
Affiliation(s)
- Ling Yang
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ping Li
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenjing Yang
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiangbo Ruan
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kurtis Kiesewetter
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Zhu
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haiming Cao
- Center for Molecular Medicine, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
82
|
Crosstalk of HNF4 α with extracellular and intracellular signaling pathways in the regulation of hepatic metabolism of drugs and lipids. Acta Pharm Sin B 2016; 6:393-408. [PMID: 27709008 PMCID: PMC5045537 DOI: 10.1016/j.apsb.2016.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
The liver is essential for survival due to its critical role in the regulation of metabolic homeostasis. Metabolism of xenobiotics, such as environmental chemicals and drugs by the liver protects us from toxic effects of these xenobiotics, whereas metabolism of cholesterol, bile acids (BAs), lipids, and glucose provide key building blocks and nutrients to promote the growth or maintain the survival of the organism. As a well-established master regulator of liver development and function, hepatocyte nuclear factor 4 alpha (HNF4α) plays a critical role in regulating a large number of key genes essential for the metabolism of xenobiotics, metabolic wastes, and nutrients. The expression and activity of HNF4α is regulated by diverse hormonal and signaling pathways such as growth hormone, glucocorticoids, thyroid hormone, insulin, transforming growth factor-β, estrogen, and cytokines. HNF4α appears to play a central role in orchestrating the transduction of extracellular hormonal signaling and intracellular stress/nutritional signaling onto transcriptional changes in the liver. There have been a few reviews on the regulation of drug metabolism, lipid metabolism, cell proliferation, and inflammation by HNF4α. However, the knowledge on how the expression and transcriptional activity of HNF4α is modulated remains scattered. Herein I provide comprehensive review on the regulation of expression and transcriptional activity of HNF4α, and how HNF4α crosstalks with diverse extracellular and intracellular signaling pathways to regulate genes essential in liver pathophysiology.
Collapse
|
83
|
De Mattia E, Cecchin E, Roncato R, Toffoli G. Pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors as emerging players in cancer precision medicine. Pharmacogenomics 2016; 17:1547-71. [DOI: 10.2217/pgs-2016-0095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Great research effort has been focused on elucidating the contribution of host genetic variability on pharmacological outcomes in cancer. Nuclear receptors have emerged as mediators between environmental stimuli and drug pharmacokinetics and pharmacodynamics. The pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors have been reported to regulate transcription of genes that encode drug metabolizing enzymes and transporters. Altered nuclear receptor expression has been shown to affect the metabolism and pharmacological profile of traditional chemotherapeutics and targeted agents. Accordingly, polymorphic variants in these genes have been studied as pharmacogenetic markers of outcome variability. This review summarizes the state of knowledge about the roles played by pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factor expression and genetics as predictive markers of anticancer drug toxicity and efficacy, which can improve cancer precision medicine.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Rossana Roncato
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| |
Collapse
|
84
|
Zhang Q, Koser SL, Donkin SS. Propionate induces the bovine cytosolic phosphoenolpyruvate carboxykinase promoter activity. J Dairy Sci 2016; 99:6654-6664. [DOI: 10.3168/jds.2016-11103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/30/2016] [Indexed: 01/26/2023]
|
85
|
Chaudhari P, Tian L, Deshmukh A, Jang YY. Expression kinetics of hepatic progenitor markers in cellular models of human liver development recapitulating hepatocyte and biliary cell fate commitment. Exp Biol Med (Maywood) 2016; 241:1653-62. [PMID: 27390263 DOI: 10.1177/1535370216657901] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Due to the limitations of research using human embryos and the lack of a biological model of human liver development, the roles of the various markers associated with liver stem or progenitor cell potential in humans are largely speculative, and based on studies utilizing animal models and certain patient tissues. Human pluripotent stem cell-based in vitro multistage hepatic differentiation systems may serve as good surrogate models for mimicking normal human liver development, pathogenesis and injury/regeneration studies. Here, we describe the implications of various liver stem or progenitor cell markers and their bipotency (i.e. hepatocytic- and biliary-epithelial cell differentiation), based on the pluripotent stem cell-derived model of human liver development. Future studies using the human cellular model(s) of liver and biliary development will provide more human relevant biological and/or pathological roles of distinct markers expressed in heterogeneous liver stem/progenitor cell populations.
Collapse
Affiliation(s)
- Pooja Chaudhari
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore 21205, USA Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore 21205, USA Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore 21205, USA
| | - Lipeng Tian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore 21205, USA
| | - Abhijeet Deshmukh
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore 21205, USA
| | - Yoon-Young Jang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore 21205, USA Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore 21205, USA Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore 21205, USA
| |
Collapse
|
86
|
Rasmussen MK, Theil PK, Oksbjerg N. Constitutive expression of cytochrome P450 in foetal and adult porcine livers-Effects of body weight. Toxicol Lett 2016; 258:87-92. [PMID: 27320961 DOI: 10.1016/j.toxlet.2016.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 01/08/2023]
Abstract
The liver hosts a great number of enzymatically driven processes, including detoxification. The super-family of enzymes named cytochrome P450 (CYP) is the major participant in that process. The expression of CYPs is affected by several factors including life-stage (foetal vs. adult). In the present study we investigated the impact of birth-weight (high or low birth weight) and life-stage on constitutive expression of porcine hepatic CYP1A1, CYP1A2, CYP2A19, CYP2B22, CYP2C33, CYP2D25, CYP2E1 and CYP3A29, as well as the transcription factors controlling their expression; aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, C/EBP and hepatocyte nuclear factors 1 and 4. Both RT-PCR and western blotting showed a marked increase in the expression of the adult pigs compared with prenatal pigs. Moreover, CYP2E1 mRNA expression was 7.5 fold higher in foetuses with low birth weight compared with foetuses with high birth weight. Gender did not affect the mRNA expression within the different life-stages. These results indicate a similarity to what is observed in humans and porcine foetuses may therefore be a model for humans when studying expression of CYPs.
Collapse
|
87
|
Fu ZD, Selwyn FP, Cui JY, Klaassen CD. RNA Sequencing Quantification of Xenobiotic-Processing Genes in Various Sections of the Intestine in Comparison to the Liver of Male Mice. Drug Metab Dispos 2016; 44:842-56. [PMID: 27048750 PMCID: PMC4885488 DOI: 10.1124/dmd.115.068270] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/04/2016] [Indexed: 12/31/2022] Open
Abstract
Previous reports on tissue distribution of xenobiotic-processing genes (XPGs) have limitations, because many non-cytochrome P450 phase I enzymes have not been investigated, and one cannot compare the real mRNA abundance of multiple XPGs using conventional quantification methods. Therefore, this study aimed to quantify and compare the mRNA abundance of all major XPGs in the liver and intestine using RNA sequencing. The mRNA profiles of 304 XPGs, including phase I, phase II enzymes, phase II cosubstrate synthetic enzymes, xenobiotic transporters, as well as xenobiotic-related transcription factors, were systematically examined in the liver and various sections of the intestine in adult male C57BL/6J mice. By two-way hierarchical clustering, over 80% of the XPGs had tissue-divergent expression, which partitioned into liver-predominant, small intestine-predominant, and large intestine-predominant patterns. Among the genes, 54% were expressed highest in the liver, 21% in the duodenum, 4% in the jejunum, 6% in the ileum, and 15% in the large intestine. The highest-expressed XPG in the liver was Mgst1; in the duodenum, Cyp3a11; in the jejunum and ileum, Ces2e; and in the large intestine, Cyp2c55. Interestingly, XPGs in the same family usually exhibited highly different tissue distribution patterns, and many XPGs were almost exclusively expressed in one tissue and minimally expressed in others. In conclusion, the present study is among the first and the most comprehensive investigations of the real mRNA abundance and tissue-divergent expression of all major XPGs in mouse liver and intestine, which aids in understanding the tissue-specific biotransformation and toxicity of drugs and other xenobiotics.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Felcy Pavithra Selwyn
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Curtis D Klaassen
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
88
|
Ebert B, Kisiela M, Maser E. Transcriptional regulation of human and murine short-chain dehydrogenase/reductases (SDRs) – an in silico approach. Drug Metab Rev 2016; 48:183-217. [DOI: 10.3109/03602532.2016.1167902] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Bettina Ebert
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Michael Kisiela
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Edmund Maser
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
89
|
Tracy TS, Chaudhry AS, Prasad B, Thummel KE, Schuetz EG, Zhong XB, Tien YC, Jeong H, Pan X, Shireman LM, Tay-Sontheimer J, Lin YS. Interindividual Variability in Cytochrome P450-Mediated Drug Metabolism. Drug Metab Dispos 2016; 44:343-51. [PMID: 26681736 PMCID: PMC4767386 DOI: 10.1124/dmd.115.067900] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/16/2015] [Indexed: 12/24/2022] Open
Abstract
The cytochrome P450 (P450) enzymes are the predominant enzyme system involved in human drug metabolism. Alterations in the expression and/or activity of these enzymes result in changes in pharmacokinetics (and consequently the pharmacodynamics) of drugs that are metabolized by this set of enzymes. Apart from changes in activity as a result of drug-drug interactions (by P450 induction or inhibition), the P450 enzymes can exhibit substantial interindividual variation in basal expression and/or activity, leading to differences in the rates of drug elimination and response. This interindividual variation can result from a myriad of factors, including genetic variation in the promoter or coding regions, variation in transcriptional regulators, alterations in microRNA that affect P450 expression, and ontogenic changes due to exposure to xenobiotics during the developmental and early postnatal periods. Other than administering a probe drug or cocktail of drugs to obtain the phenotype or conducting a genetic analysis to determine genotype, methods to determine interindividual variation are limited. Phenotyping via a probe drug requires exposure to a xenobiotic, and genotyping is not always well correlated with phenotype, making both methodologies less than ideal. This article describes recent work evaluating the effect of some of these factors on interindividual variation in human P450-mediated metabolism and the potential utility of endogenous probe compounds to assess rates of drug metabolism among individuals.
Collapse
Affiliation(s)
- Timothy S Tracy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Amarjit S Chaudhry
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Kenneth E Thummel
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Erin G Schuetz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Yun-Chen Tien
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Hyunyoung Jeong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Xian Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Laura M Shireman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Jessica Tay-Sontheimer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| | - Yvonne S Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky (T.S.T.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (B.P., K.E.T., L.M.S., J.T.-S., Y.S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z., Y.-C.T); and Departments of Pharmacy Practice and Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois (H.J., X.P.)
| |
Collapse
|
90
|
Shah RR, Gaedigk A, LLerena A, Eichelbaum M, Stingl J, Smith RL. CYP450 genotype and pharmacogenetic association studies: a critical appraisal. Pharmacogenomics 2016; 17:259-75. [DOI: 10.2217/pgs.15.172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite strong pharmacological support, association studies using genotype-predicted phenotype as a variable have yielded conflicting or inconclusive evidence to promote personalized pharmacotherapy. Unless the patient is a genotypic poor metabolizer, imputation of patient's metabolic capacity (or metabolic phenotype), a major factor in drug exposure-related clinical response, is a complex and highly challenging task because of limited number of alleles interrogated, population-specific differences in allele frequencies, allele-specific substrate-selectivity and importantly, phenoconversion mediated by co-medications and inflammatory co-morbidities that modulate the functional activity of drug metabolizing enzymes. Furthermore, metabolic phenotype and clinical outcomes are not binary functions; there is large intragenotypic and intraindividual variability. Therefore, the ability of association studies to identify relationships between genotype and clinical outcomes can be greatly enhanced by determining phenotype measures of study participants and/or by therapeutic drug monitoring to correlate drug concentrations with genotype and actual metabolic phenotype. To facilitate improved analysis and reporting of association studies, we propose acronyms with the prefixes ‘g’ (genotype-predicted phenotype) and ‘m’ (measured metabolic phenotype) to better describe this important variable of the study subjects. Inclusion of actually measured metabolic phenotype, and when appropriate therapeutic drug monitoring, promises to reveal relationships that may not be detected by using genotype alone as the variable.
Collapse
Affiliation(s)
| | - Andrea Gaedigk
- Clinical Pharmacology, Toxicology &, Therapeutic Innovation, Children's Mercy-Kansas City, 2401 Gillham Rd, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri-Kansas City, MO, USA
| | - Adrián LLerena
- CICAB Clinical Research Centre, Extremadura University Hospital & Medical School, Badajoz, Spain
| | - Michel Eichelbaum
- Dr. Margarete Fischer-Bosch – Institut für Klinische Pharmakologie, 70376 Stuttgart Auerbachstr., 112 Germany
| | - Julia Stingl
- Centre for Translational Medicine, University of Bonn Medical School, Bonn, Germany
| | - Robert L Smith
- Department of Surgery & Cancer, Faculty of Medicine, Imperial College, South Kensington Campus, London, UK
| |
Collapse
|
91
|
Chikada H, Ito K, Yanagida A, Nakauchi H, Kamiya A. The basic helix-loop-helix transcription factor, Mist1, induces maturation of mouse fetal hepatoblasts. Sci Rep 2015; 5:14989. [PMID: 26456005 PMCID: PMC4601036 DOI: 10.1038/srep14989] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022] Open
Abstract
Hepatic stem/progenitor cells, hepatoblasts, have a high proliferative ability and can differentiate into mature hepatocytes and cholangiocytes. Therefore, these cells are considered to be useful for regenerative medicine and drug screening for liver diseases. However, it is problem that in vitro maturation of hepatoblasts is insufficient in the present culture system. In this study, a novel regulator to induce hepatic differentiation was identified and the molecular function of this factor was examined in embryonic day 13 hepatoblast culture with maturation factor, oncostatin M and extracellular matrices. Overexpression of the basic helix-loop-helix type transcription factor, Mist1, induced expression of mature hepatocytic markers such as carbamoyl-phosphate synthetase1 and several cytochrome P450 (CYP) genes in this culture system. In contrast, Mist1 suppressed expression of cholangiocytic markers such as Sox9, Sox17, Ck19, and Grhl2. CYP3A metabolic activity was significantly induced by Mist1 in this hepatoblast culture. In addition, Mist1 induced liver-enriched transcription factors, CCAAT/enhancer-binding protein α and Hepatocyte nuclear factor 1α, which are known to be involved in liver functions. These results suggest that Mist1 partially induces mature hepatocytic expression and function accompanied by the down-regulation of cholangiocytic markers.
Collapse
Affiliation(s)
- Hiromi Chikada
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Keiichi Ito
- Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-4 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-4 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-4 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, California 94305-5461, USA
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
92
|
Li J, Inoue J, Choi JM, Nakamura S, Yan Z, Fushinobu S, Kamada H, Kato H, Hashidume T, Shimizu M, Sato R. Identification of the Flavonoid Luteolin as a Repressor of the Transcription Factor Hepatocyte Nuclear Factor 4α. J Biol Chem 2015; 290:24021-35. [PMID: 26272613 DOI: 10.1074/jbc.m115.645200] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 01/14/2023] Open
Abstract
Hepatocyte nuclear factor 4α (HNF4α) is a nuclear receptor that regulates the expression of genes involved in the secretion of apolipoprotein B (apoB)-containing lipoproteins and in glucose metabolism. In the present study, we identified a naturally occurring flavonoid, luteolin, as a repressor of HNF4α by screening for effectors of the human microsomal triglyceride transfer protein (MTP) promoter. Luciferase reporter gene assays revealed that the activity of the MTP gene promoter was suppressed by luteolin and that the mutation of HNF4α-binding element abolished luteolin responsiveness. Luteolin treatment caused a significant decrease in the mRNA levels of HNF4α target genes in HepG2 cells and inhibited apoB-containing lipoprotein secretion in HepG2 and differentiated Caco2 cells. The interaction between luteolin and HNF4α was demonstrated using absorption spectrum analysis and luteolin-immobilized beads. Luteolin did not affect the DNA binding of HNF4α to the promoter region of its target genes but suppressed the acetylation level of histone H3 in the promoter region of certain HNF4α target genes. Short term treatment of mice with luteolin significantly suppressed the expression of HNF4α target genes in the liver. In addition, long term treatment of mice with luteolin significantly suppressed their diet-induced obesity and improved their serum glucose and lipid parameters. Importantly, long term luteolin treatment lowered serum VLDL and LDL cholesterol and serum apoB protein levels, which was not accompanied by fat accumulation in the liver. These results suggest that the flavonoid luteolin ameliorates an atherogenic lipid profile in vivo that is likely to be mediated through the inactivation of HNF4α.
Collapse
Affiliation(s)
- Juan Li
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan
| | - Jun Inoue
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan,
| | - Jung-Min Choi
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan
| | - Shugo Nakamura
- the Department of Biotechnology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Zhen Yan
- the Department of Biotechnology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Shinya Fushinobu
- the Department of Biotechnology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Haruhiko Kamada
- the Laboratory of Biopharmaceutical Research, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Hisanori Kato
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan, the Corporate Sponsored Research Program "Food for Life," Organization for Interdisciplinary Research Projects, University of Tokyo, Tokyo, 113-8657, Japan, and
| | - Tsutomu Hashidume
- the Institute of Gerontology, University of Tokyo, Tokyo 113-8656, Japan
| | - Makoto Shimizu
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan
| | - Ryuichiro Sato
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 1-1-1 Yayoi, 113-8657, Japan,
| |
Collapse
|
93
|
Muñoz A, Villagrán M, Guzmán P, Solíz C, Gatica M, Aylwin C, Sweet K, Maldonado M, Escobar E, Reyes AM, Toledo JR, Sánchez O, Oñate SA, Carlos Vera J, Rivas CI. Cis-regulatory elements involved in species-specific transcriptional regulation of the SVCT1 gene in rat and human hepatoma cells. Free Radic Biol Med 2015; 85:183-96. [PMID: 25933589 DOI: 10.1016/j.freeradbiomed.2015.04.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/09/2015] [Accepted: 04/20/2015] [Indexed: 01/06/2023]
Abstract
Ascorbic acid is transported into cells by the sodium-coupled vitamin C transporters (SVCTs). Recently, we obtained evidence of differential regulation of SVCT expression in response to acute oxidative stress in cells from species that differ in their capacity to synthesize vitamin C, with a marked decrease in SVCT1 mRNA and protein levels in rat hepatoma cells that was not observed in human hepatoma cells. To better understand the regulatory aspects involved, we performed a structural and functional analysis of the proximal promoter of the SVCT1 rat gene. We cloned a 1476-bp segment containing the proximal promoter of the rat SVCT1 gene and generated deletion-derived truncated promoters of decreasing sizes and mutant promoters by modification of consensus binding sites for transcription factors by site-directed mutagenesis. We next analyzed their capacity to direct the transcription of a reporter gene after transfection into rat H4IIE and human HepG2 hepatoma cells, in experiments involving the coexpression of transcription factors whose consensus binding sequences are present in the SVCT1 promoter. This analysis revealed the presence of two critical cis-regulatory elements of the transcriptional activity of the rat SVCT1 gene promoter, sites containing consensus sequences for the binding of the transcription factors Bach1 and HNF4 that are not present in equivalent locations in the human SVCT1 gene promoter. Moreover, a consensus site for HNF1 that is crucial for the regulation of the human SVCT1 promoter is present in the SVCT1 rat promoter but has no effect on its transcriptional activity. These findings imply that regulation of vitamin C metabolism in the rat, a species with the capacity to synthesize large amounts of ascorbic acid, may differ from that of humans, a species that must obtain ascorbic acid from the diet through a transport mechanism that depends on proper SVCT1 expression.
Collapse
Affiliation(s)
- Alejandra Muñoz
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Marcelo Villagrán
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Paula Guzmán
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Carlos Solíz
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Marcell Gatica
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Carlos Aylwin
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Karen Sweet
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Mafalda Maldonado
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Elizabeth Escobar
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Alejandro M Reyes
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja, Valdivia, Chile
| | - Jorge R Toledo
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Oliberto Sánchez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Sergio A Oñate
- Departamento de Especialidades Médicas, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile
| | - Juan Carlos Vera
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile.
| | - Coralia I Rivas
- Departamento de Fisiopatología, Facultad de Medicina, Universidad de Concepción, Concepción, Bio-Bio 4070386, Chile.
| |
Collapse
|
94
|
Zheng H, Yang S, Yang Y, Yuan SX, Wu FQ, Wang LL, Yan HL, Sun SH, Zhou WP. Epigenetically silenced long noncoding-SRHC promotes proliferation of hepatocellular carcinoma. J Cancer Res Clin Oncol 2015; 141:1195-203. [PMID: 25512078 DOI: 10.1007/s00432-014-1871-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/05/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE To investigate the expression of SRHC and the role of SRHC in the pathogenesis of hepatocellular carcinoma (HCC). METHODS We analyzed HCC samples and matched non-tumor liver tissues (controls) collected from 81 patients who underwent hepatectomy in Shanghai, China. The expression levels of SRHC were determined by quantitative reverse-transcription polymerase chain reaction. Statistical analyses were used to associate the levels of SRHC with tumor features and patient outcomes. RESULTS We found that a lower SRHC expression level was significantly more frequent in tissues with a high serum a-fetoprotein level (positive, >20 µg/L, P = 0.004) and a low degree of differentiated tumors (poorly differentiated, P = 0.017). Furthermore, we found that the promoter region of SRHC contains a CpG-rich island and that SRHC is down-regulated in tumors by DNA methylation. CONCLUSION Here, we identified a new long noncoding RNA designated as SRHC that is capable of inhibiting cancer proliferation and is down-regulated in tumors at least partly by DNA methylation.
Collapse
Affiliation(s)
- Hao Zheng
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
|
96
|
Lin X, Jacobi S, Odle J. Transplacental induction of fatty acid oxidation in term fetal pigs by the peroxisome proliferator-activated receptor alpha agonist clofibrate. J Anim Sci Biotechnol 2015; 6:11. [PMID: 25883783 PMCID: PMC4399351 DOI: 10.1186/s40104-015-0010-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/10/2015] [Indexed: 12/23/2022] Open
Abstract
Background To induce peroxisomal proliferator-activated receptor α (PPARα) expression and increase milk fat utilization in pigs at birth, the effect of maternal feeding of the PPARα agonist, clofibrate (2-(4-chlorophenoxy)-2-methyl-propanoic acid, ethyl ester), on fatty acid oxidation was examined at full-term delivery (0 h) and 24 h after delivery in this study. Each group of pigs (n = 10) was delivered from pregnant sows fed a commercial diet with or without 0.8% clofibrate for the last 7 d of gestation. Blood samples were collected from the utero-ovarian artery of the sows and the umbilical cords of the pigs as they were removed from the sows by C-section on day 113 of gestation. Results HPLC analysis identified that clofibric acid was present in the plasma of the clofibrate-fed sow (~4.2 μg/mL) and its offspring (~1.5 μg/mL). Furthermore, the maternal-fed clofibrate had no impact on the liver weight of the pigs at 0 h and 24 h, but hepatic fatty acid oxidation examined in fresh homogenates showed that clofibrate increased (P < 0.01) 14C-accumulation in CO2 and acid soluble products 2.9-fold from [1-14C]-oleic acid and 1.6-fold from [1-14C]-lignoceric acid respectively. Correspondingly, clofibrate increased fetal hepatic carnitine palmitoyltransferase (CPT) and acyl-CoA oxidase (ACO) activities by 36% and 42% over controls (P < 0.036). The mRNA abundance of CPT I was 20-fold higher in pigs exposed to clofibrate (P < 0.0001) but no differences were detected for ACO and PPARα mRNA between the two groups. Conclusion These data demonstrate that dietary clofibrate is absorbed by the sow, crosses the placental membrane, and enters fetal circulation to induce hepatic fatty acid oxidation by increasing the CPT and ACO activities of the newborn.
Collapse
Affiliation(s)
- Xi Lin
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Box 7621, Raleigh, NC 27695 USA
| | - Sheila Jacobi
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Box 7621, Raleigh, NC 27695 USA
| | - Jack Odle
- Laboratory of Developmental Nutrition, Department of Animal Sciences, North Carolina State University, Box 7621, Raleigh, NC 27695 USA
| |
Collapse
|
97
|
Nishikawa T, Bell A, Brooks JM, Setoyama K, Melis M, Han B, Fukumitsu K, Handa K, Tian J, Kaestner KH, Vodovotz Y, Locker J, Soto-Gutierrez A, Fox IJ. Resetting the transcription factor network reverses terminal chronic hepatic failure. J Clin Invest 2015; 125:1533-44. [PMID: 25774505 DOI: 10.1172/jci73137] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/22/2015] [Indexed: 12/16/2022] Open
Abstract
The cause of organ failure is enigmatic for many degenerative diseases, including end-stage liver disease. Here, using a CCl4-induced rat model of irreversible and fatal hepatic failure, which also exhibits terminal changes in the extracellular matrix, we demonstrated that chronic injury stably reprograms the critical balance of transcription factors and that diseased and dedifferentiated cells can be returned to normal function by re-expression of critical transcription factors, a process similar to the type of reprogramming that induces somatic cells to become pluripotent or to change their cell lineage. Forced re-expression of the transcription factor HNF4α induced expression of the other hepatocyte-expressed transcription factors; restored functionality in terminally diseased hepatocytes isolated from CCl4-treated rats; and rapidly reversed fatal liver failure in CCl4-treated animals by restoring diseased hepatocytes rather than replacing them with new hepatocytes or stem cells. Together, the results of our study indicate that disruption of the transcription factor network and cellular dedifferentiation likely mediate terminal liver failure and suggest reinstatement of this network has therapeutic potential for correcting organ failure without cell replacement.
Collapse
|
98
|
Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet 2015; 30:30-51. [DOI: 10.1016/j.dmpk.2014.12.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/24/2023]
|
99
|
Probing the effect of MODY mutations near the co-activator-binding pocket of HNF4α. Biosci Rep 2015; 31:411-9. [PMID: 21323639 DOI: 10.1042/bsr20110013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
HNF4α (hepatocyte nuclear factor 4α) is a culprit gene product for a monogenic and dominantly inherited form of diabetes, referred to as MODY (maturity onset diabetes of the young). As a member of the NR (nuclear receptor) superfamily, HNF4α recruits transcriptional co-activators such as SRC-1α (steroid receptor co-activator-1α) and PGC-1α (peroxisome-proliferator-activated receptor γ co-activator-1α) through the LXXLL-binding motifs for its transactivation, and our recent crystal structures of the complex provided the molecular details and the mechanistic insights into these co-activator recruitments. Several mutations have been identified from the MODY patients and, among these, point mutations can be very instructive site-specific measures of protein function and structure. Thus, in the present study, we probed the functional effects of the two MODY point mutations (D206Y and M364R) found directly near the LXXLL motif-binding site by conducting a series of experiments on their structural integrity and specific functional roles such as overall transcription, ligand selectivity, target gene recognition and co-activator recruitment. While the D206Y mutation has a subtle effect, the M364R mutation significantly impaired the overall transactivation by HNF4α. These functional disruptions are mainly due to their reduced ability to recruit co-activators and lowered protein stability (only with M364R mutation), while their DNA-binding activities and ligand selectivities are preserved. These results confirmed our structural predictions and proved that MODY mutations are loss-of-function mutations leading to impaired β-cell function. These findings should help target selective residues for correcting mutational defects or modulating the overall activity of HNF4α as a means of therapeutic intervention.
Collapse
|
100
|
Gouliarmou V, Pelkonen O, Coecke S. Differentiation-Promoting Medium Additives for Hepatocyte Cultivation and Cryopreservation. Methods Mol Biol 2015; 1250:143-159. [PMID: 26272140 DOI: 10.1007/978-1-4939-2074-7_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Isolated primary hepatocytes are considered as the reference system for in vitro hepatic methods. Following the isolation of primary hepatocytes from liver tissue, an unfavorable process named dedifferentiation is initiated leading to the attenuation of the hepatocellular phenotype both at the morphological and functional level. Freshly isolated hepatocytes can be used immediately or can be cryopreserved for future purposes. Currently, a number of antidedifferentiation strategies exist to extend the life span of isolated hepatocytes. The addition of differentiation-promoting compounds to the hepatocyte culture medium is the oldest and simplest antidedifferentiation approach applied. In the present chapter, the most commonly used medium additives for cultivation and cryopreservation of primary hepatocytes are reviewed.
Collapse
Affiliation(s)
- Varvara Gouliarmou
- EURL ECVAM, Systems Toxicology Unit, Institute for Health and Consumer Protection, European Commission, Joint Research Center, Via Fermi 2749, Ispra, 21027, Italy
| | | | | |
Collapse
|