1
|
Kashyap J, Chhabra A, Kumari N, Tyagi RK. Nuclear localization signal in nuclear receptor VDR facilitates the mitotic genome bookmarking by involving distinct amino acid residues. Mol Cell Endocrinol 2024; 589:112233. [PMID: 38616036 DOI: 10.1016/j.mce.2024.112233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/22/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Mitotic genome-bookmarking preserves epigenetic information, re-establishing progenitor's gene expression profile through transcription factors, chromatin remodelers, and histone modifiers, thereby regulating cell fate and lineage commitment post-mitotically in progeny cells. Our recent study revealed that the constitutive association of VDR with mitotic chromatin involves its DNA-binding domain. However, amino acid residues in this domain, crucial for genome bookmarking, remain elusive. This study demonstrates that nuclear localization signal (NLS) residues between 49 and 55 amino acids in VDR are essential for receptor-chromatin interaction during mitosis. Furthermore, it is revealed that both bipartite nature of VDR-NLS region and N-terminally located positively charged arginine residues are critical for its 'genome-bookmarking' property. Since mitotic chromatin association of heterodimeric partner RXR depends on VDR-chromatin association, interventions in VDR binding also abort RXR-chromatin interaction. Overall, this study documents the mechanistic details underlying VDR-chromatin interactions in genome-bookmarking behavior, potentially aiding in comprehending VDR-mediated diseases attributed to certain SNPs.
Collapse
Affiliation(s)
- Jyoti Kashyap
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ayushi Chhabra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Neha Kumari
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India; Special Centre for Systems Medicine (Concurrent Faculty), Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
2
|
Rakateli L, Huchzermeier R, van der Vorst EPC. AhR, PXR and CAR: From Xenobiotic Receptors to Metabolic Sensors. Cells 2023; 12:2752. [PMID: 38067179 PMCID: PMC10705969 DOI: 10.3390/cells12232752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Traditionally, xenobiotic receptors are known for their role in chemical sensing and detoxification, as receptor activation regulates the expression of various key enzymes and receptors. However, recent studies have highlighted that xenobiotic receptors also play a key role in the regulation of lipid metabolism and therefore function also as metabolic sensors. Since dyslipidemia is a major risk factor for various cardiometabolic diseases, like atherosclerosis and non-alcoholic fatty liver disease, it is of major importance to understand the molecular mechanisms that are regulated by xenobiotic receptors. In this review, three major xenobiotic receptors will be discussed, being the aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Specifically, this review will focus on recent insights into the metabolic functions of these receptors, especially in the field of lipid metabolism and the associated dyslipidemia.
Collapse
Affiliation(s)
- Leonida Rakateli
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Rosanna Huchzermeier
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (L.R.); (R.H.)
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
3
|
Lan H, Zhang Y, Fan M, Wu B, Wang C. Pregnane X receptor as a therapeutic target for cholestatic liver injury. Drug Metab Rev 2023; 55:371-387. [PMID: 37593784 DOI: 10.1080/03602532.2023.2248680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
Cholestatic liver injury (CLI) is caused by toxic bile acids (BAs) accumulation in the liver and can lead to inflammation and liver fibrosis. The mechanisms underlying CLI development remain unclear, and this disease has no effective cure. However, regulating BA synthesis and homeostasis represents a promising therapeutic strategy for CLI treatment. Pregnane X receptor (PXR) plays an essential role in the metabolism of endobiotics and xenobiotics via the transcription of metabolic enzymes and transporters, which can ultimately modulate BA homeostasis and exert anticholestatic effects. Furthermore, recent studies have demonstrated that PXR exhibits antifibrotic and anti-inflammatory properties, providing novel insights into treating CLI. Meanwhile, several drugs have been identified as PXR agonists that improve CLI. Nevertheless, the precise role of PXR in CLI still needs to be fully understood. This review summarizes how PXR improves CLI by ameliorating cholestasis, inhibiting inflammation, and reducing fibrosis and discusses the progress of promising PXR agonists for treating CLI.
Collapse
Affiliation(s)
- Huan Lan
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Ying Zhang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Minqi Fan
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Bingxin Wu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Caiyan Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
4
|
Niu X, Wu T, Yin Q, Gu X, Li G, Zhou C, Ma M, Su L, Tang S, Tian Y, Yang M, Cui H. Combination of Paclitaxel and PXR Antagonist SPA70 Reverses Paclitaxel-Resistant Non-Small Cell Lung Cancer. Cells 2022; 11:3094. [PMID: 36231056 PMCID: PMC9563422 DOI: 10.3390/cells11193094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
Abstract
Paclitaxel (PTX) is one of the most efficient drugs for late-stage non-small cell lung cancer (NSCLC) patients. However, most patients gradually develop resistance to PTX with long-term treatments. The identification of new strategies to reverse PTX resistance in NSCLC is crucially important for the treatment. PTX is an agonist for the pregnane X receptor (PXR) which regulates PTX metabolism. Antagonizing PXR, therefore, may render the NSCLC more sensitive to the PTX treatment. In this study, we investigated the PXR antagonist SPA70 and its role in PTX treatment of NSCLC. In vitro, SPA70 and PTX synergistically inhibited cell growth, migration and invasion in both paclitaxel-sensitive and paclitaxel-resistant A549 and H460 lung cancer cells. Mechanistically, we found PTX and SPA70 cotreatment disassociated PXR from ABCB1 (MDR1, P-gp) promoter, thus inhibiting P-gp expression. Furthermore, the combination regimen synergistically enhanced the interaction between PXR and Tip60, which abrogated Tip60-mediated α-tubulin acetylation, leading to mitosis defect, S-phase arrest and necroptosis/apoptosis. Combination of PXT and SPA70 dramatically inhibited tumor growth in a paclitaxel-resistant A549/TR xenograft tumor model. Taken together, we showed that SPA70 reduced the paclitaxel resistance of NSCLC. The combination regimen of PTX and SPA70 could be potential novel candidates for the treatment of taxane-resistant lung cancer.
Collapse
Affiliation(s)
- Xiaxia Niu
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qishuang Yin
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xinsheng Gu
- College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China
| | - Gege Li
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Changlong Zhou
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Mei Ma
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Li Su
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shu Tang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Ming Yang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
5
|
Abstract
Mifepristone (Mif), an effective synthetic steroidal antiprogesterone drug, is widely used for medical abortion and pregnancy prevention. Due to its anti-glucocorticoid effect, high-dose Mif is also used to treat Cushing's syndrome. Mif was reported to active pregnane X receptor (PXR) in vitro and PXR can induce hepatomegaly via activation and interaction with yes-associated protein (YAP) pathway. High-dose Mif was reported to induce hepatomegaly in rats and mice, but the underlying mechanism remains unclear. Here, the role of PXR was studied in Mif-induced hepatomegaly in C57BL/6 mice and Pxr-knockout mice. The results demonstrated that high-dose Mif (100 mg · kg-1 · d-1, i.p.) treatment for 5 days significantly induced hepatomegaly with enlarged hepatocytes and promoted proliferation, but low dose of Mif (5 mg · kg-1 · d-1, i.p.) cannot induce hepatomegaly. The dual-luciferase reporter gene assays showed that Mif can activate human PXR in a concentration-dependent manner. In addition, Mif could promote nuclear translocation of PXR and YAP, and significantly induced the expression of PXR, YAP, and their target proteins such as CYP3A11, CYP2B10, UGT1A1, ANKRD, and CTGF. However, Mif (100 mg · kg-1 · d-1, i.p.) failed to induce hepatomegaly in Pxr-knockout mice, as well as hepatocyte enlargement and proliferation, further indicating that Mif-induced hepatomegaly is PXR-dependent. In summary, this study demonstrated that PXR-mediated Mif-induced hepatomegaly in mice probably via activation of YAP pathway. This study provides new insights in Mif-induced hepatomegaly, and provides novel evidence on the crucial function of PXR in liver enlargement and regeneration.
Collapse
|
6
|
Rogers RS, Parker A, Vainer PD, Elliott E, Sudbeck D, Parimi K, Peddada VP, Howe PG, D’Ambrosio N, Ruddy G, Stackable K, Carney M, Martin L, Osterholt T, Staudinger JL. The Interface between Cell Signaling Pathways and Pregnane X Receptor. Cells 2021; 10:cells10113262. [PMID: 34831484 PMCID: PMC8617909 DOI: 10.3390/cells10113262] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Highly expressed in the enterohepatic system, pregnane X receptor (PXR, NR1I2) is a well-characterized nuclear receptor (NR) that regulates the expression of genes in the liver and intestines that encode key drug metabolizing enzymes and drug transporter proteins in mammals. The net effect of PXR activation is to increase metabolism and clear drugs and xenobiotics from the body, producing a protective effect and mediating clinically significant drug interaction in patients on combination therapy. The complete understanding of PXR biology is thus important for the development of safe and effective therapeutic strategies. Furthermore, PXR activation is now known to specifically transrepress the inflammatory- and nutrient-signaling pathways of gene expression, thereby providing a mechanism for linking these signaling pathways together with enzymatic drug biotransformation pathways in the liver and intestines. Recent research efforts highlight numerous post-translational modifications (PTMs) which significantly influence the biological function of PXR. However, this thrust of research is still in its infancy. In the context of gene-environment interactions, we present a review of the recent literature that implicates PXR PTMs in regulating its clinically relevant biology. We also provide a discussion of how these PTMs likely interface with each other to respond to extracellular cues to appropriately modify PXR activity.
Collapse
Affiliation(s)
- Robert S. Rogers
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Annemarie Parker
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Phill D. Vainer
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Elijah Elliott
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Dakota Sudbeck
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Kaushal Parimi
- Thomas Jefferson Independent Day School, Joplin, MO 64801, USA;
| | - Venkata P. Peddada
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Parker G. Howe
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Nick D’Ambrosio
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Gregory Ruddy
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Kaitlin Stackable
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Megan Carney
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Lauren Martin
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Thomas Osterholt
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
| | - Jeff L. Staudinger
- Division of Basic Sciences, Farber-McIntire Campus, College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, USA; (R.S.R.); (A.P.); (P.D.V.); (E.E.); (D.S.); (V.P.P.); (P.G.H.); (G.R.); (K.S.); (M.C.); (L.M.); (T.O.)
- Correspondence:
| |
Collapse
|
7
|
El-Ghiaty MA, El-Kadi AO. Arsenic: Various species with different effects on cytochrome P450 regulation in humans. EXCLI JOURNAL 2021; 20:1184-1242. [PMID: 34512225 PMCID: PMC8419240 DOI: 10.17179/excli2021-3890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022]
Abstract
Arsenic is well-recognized as one of the most hazardous elements which is characterized by its omnipresence throughout the environment in various chemical forms. From the simple inorganic arsenite (iAsIII) and arsenate (iAsV) molecules, a multitude of more complex organic species are biologically produced through a process of metabolic transformation with biomethylation being the core of this process. Because of their differential toxicity, speciation of arsenic-based compounds is necessary for assessing health risks posed by exposure to individual species or co-exposure to several species. In this regard, exposure assessment is another pivotal factor that includes identification of the potential sources as well as routes of exposure. Identification of arsenic impact on different physiological organ systems, through understanding its behavior in the human body that leads to homeostatic derangements, is the key for developing strategies to mitigate its toxicity. Metabolic machinery is one of the sophisticated body systems targeted by arsenic. The prominent role of cytochrome P450 enzymes (CYPs) in the metabolism of both endobiotics and xenobiotics necessitates paying a great deal of attention to the possible effects of arsenic compounds on this superfamily of enzymes. Here we highlight the toxicologically relevant arsenic species with a detailed description of the different environmental sources as well as the possible routes of human exposure to these species. We also summarize the reported findings of experimental investigations evaluating the influence of various arsenicals on different members of CYP superfamily using human-based models.
Collapse
Affiliation(s)
- Mahmoud A. El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O.S. El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Zhao YY, Yao XP, Jiao TY, Tian JN, Gao Y, Fan SC, Chen PP, Jiang YM, Zhou YY, Chen YX, Yang X, Huang M, Bi HC. Schisandrol B promotes liver enlargement via activation of PXR and YAP pathways in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153520. [PMID: 33662920 DOI: 10.1016/j.phymed.2021.153520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/28/2021] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Schisandrol B (SolB) is one of the bioactive components from a traditional Chinese medicine Schisandra chinensis or Schisandra sphenanthera. It has been demonstrated that SolB exerts hepatoprotective effects against drug-induced liver injury and promotes liver regeneration. It was found that SolB can induce hepatomegaly but the involved mechanisms remain unknown. PURPOSE This study aimed to explore the mechanisms involved in SolB-induced hepatomegaly. METHODS Male C57BL/6 mice were injected intraperitoneally with SolB (100 mg/kg) for 5 days. Serum and liver samples were collected for biochemical and histological analyses. The mechanisms of SolB were investigated by qRT-PCR and western blot analyses, luciferase reporter gene assays and immunofluorescence. RESULTS SolB significantly increased hepatocyte size and proliferation, and then promoted liver enlargement without liver injury and inflammation. SolB transactivated human PXR, activated PXR in mice and upregulated hepatic expression of its downstream proteins, such as CYP3A11, CYP2B10 and UGT1A1. SolB also significantly enhanced nuclear translocation of PXR and YAP in human cell lines. YAP signal pathway was activated by SolB in mice. CONCLUSION These findings demonstrated that SolB can significantly induce liver enlargement, which is associated with the activation of PXR and YAP pathways.
Collapse
Affiliation(s)
- Ying-Yuan Zhao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xin-Peng Yao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ting-Ying Jiao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jia-Ning Tian
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yue Gao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shi-Cheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Pan-Pan Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yi-Ming Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yan-Ying Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yi-Xin Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiao Yang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hui-Chang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
9
|
Wang F, Wang H, Wu Y, Wang L, Zhang L, Ye X, Peng D, Chen W. Activation of Pregnane X Receptor-Cytochrome P450s Axis: A Possible Reason for the Enhanced Accelerated Blood Clearance Phenomenon of PEGylated Liposomes In Vivo. Drug Metab Dispos 2019; 47:785-793. [PMID: 31118196 DOI: 10.1124/dmd.119.086769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/16/2019] [Indexed: 12/30/2022] Open
Abstract
Recently, we reported that repeated injection of PEGylated liposomes (PEG-L) at certain intervals to the same rat lead to the disappearance of their long-circulation properties, referred to as the "accelerated blood clearance (ABC) phenomenon". Evidence from our recent studies suggested that cytochrome P450s (P450s) contribute to induction of the ABC phenomenon, a possibility that had been previously ignored. However, few details are known about the mechanism for induction of P450s. The present study was undertaken to investigate the roles in the ABC phenomenon of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), the major upstream transcriptional regulators of the P450 genes, including CYP3A1, CYP2C6, and CYP1A2. The results demonstrated that expression of rat PXR and CAR was significantly increased in the ABC phenomenon and was accompanied by elevated CYP3A1, CYP2C6, and CYP1A2 levels. Further findings revealed that PXR but not CAR protein was substantially upregulated in the hepatocyte nucleus, together with marked nuclear colocalization of the PXR-retinoid X receptor alpha (RXRα) transcriptionally active heterodimer, indicating that nuclear translocation of PXR was induced in the ABC phenomenon, whereas nuclear translocation of CAR was not observed. Notably, pretreatment with the specific PXR inducer dexamethasone significantly induced accelerated systemic clearance of the subsequent injection of PEG-L, associating with increased nuclear colocalization of PXR-RXRα These results revealed that the induction of P450s in the ABC phenomenon may be attributable largely to the activation of PXR induced by sequential injections of PEG-L, thus confirming the crucial involvement of the PXR-P450s axis in promoting the ABC phenomenon. SIGNIFICANCE STATEMENT: The results of this study revealed that the induction of P450s in the ABC phenomenon may be largely attributable to the activation of PXR induced by sequential injections of PEG-L, thus confirming the crucial involvement of the PXR-P450s axis in promoting the ABC phenomenon. The data may help to extend our insights into 1) the role of P450s, which are regulated by the liver-enriched nuclear receptor PXR, in the ABC phenomenon, and 2) the therapeutic potential of targeting the PXR-P450 axis for reducing the magnitude of the ABC phenomenon in clinical practice.
Collapse
Affiliation(s)
- Fengling Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Huihui Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Yifan Wu
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Lei Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Ling Zhang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Xi Ye
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Daiyin Peng
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Weidong Chen
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| |
Collapse
|
10
|
Chen K, Zhong J, Hu L, Li R, Du Q, Cai J, Li Y, Gao Y, Cui X, Yang X, Wu X, Yao L, Dai J, Wang Y, Jin H. The Role of Xenobiotic Receptors on Hepatic Glycolipid Metabolism. Curr Drug Metab 2019; 20:29-35. [PMID: 30227815 DOI: 10.2174/1389200219666180918152241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/13/2018] [Accepted: 08/20/2018] [Indexed: 01/14/2023]
Abstract
Background:
PXR (Pregnane X Receptor) and CAR (Constitutive Androstane Receptor) are termed as
xenobiotic receptors, which are known as core factors in regulation of the transcription of metabolic enzymes and
drug transporters. However, accumulating evidence has shown that PXR and CAR exert their effects on energy metabolism
through the regulation of gluconeogenesis, lipogenesis and β-oxidation. Therefore, in this review, we are
trying to summary recent advances to show how xenobiotic receptors regulate energy metabolism.
Methods:
A structured search of databases has been performed by using focused review topics. According to conceptual
framework, the main idea of research literature was summarized and presented.
Results:
For introduction of each receptor, the general introduction and the critical functions in hepatic glucose and
lipid metabolism have been included. Recent important studies have shown that CAR acts as a negative regulator of
lipogenesis, gluconeogenesis and β -oxidation. PXR activation induces lipogenesis, inhibits gluconeogenesis and
inhabits β-oxidation.
Conclusion:
In this review, the importance of xenobiotic receptors in hepatic glucose and lipid metabolism has been
confirmed. Therefore, PXR and CAR may become new therapeutic targets for metabolic syndrome, including obesity
and diabetes. However, further research is required to promote the clinical application of this new energy metabolism
function of xenobiotic receptors.
Collapse
Affiliation(s)
- Ke Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinwei Zhong
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin Hu
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruliu Li
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qun Du
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiazhong Cai
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanwu Li
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Gao
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaona Cui
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoying Yang
- Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaojie Wu
- Department of Immunology, Binzhou Medical University, Yantai, Shangdong, China
| | - Lu Yao
- Jilin Medical University, Jilin, China
| | - Juji Dai
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Wang
- Department of Otolaryngology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiyong Jin
- Department of Otolaryngology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Jiang Y, Feng D, Ma X, Fan S, Gao Y, Fu K, Wang Y, Sun J, Yao X, Liu C, Zhang H, Xu L, Liu A, Gonzalez FJ, Yang Y, Gao B, Huang M, Bi H. Pregnane X Receptor Regulates Liver Size and Liver Cell Fate by Yes-Associated Protein Activation in Mice. Hepatology 2019; 69:343-358. [PMID: 30048004 PMCID: PMC6324985 DOI: 10.1002/hep.30131] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 05/30/2018] [Indexed: 12/13/2022]
Abstract
Activation of pregnane X receptor (PXR), a nuclear receptor that controls xenobiotic and endobiotic metabolism, is known to induce liver enlargement, but the molecular signals and cell types responding to PXR-induced hepatomegaly remain unknown. In this study, the effect of PXR activation on liver enlargement and cell change was evaluated in several strains of genetically modified mice and animal models. Lineage labeling using AAV-Tbg-Cre-treated Rosa26EYFP mice or Sox9-CreERT , Rosa26EYFP mice was performed and Pxr-null mice or AAV Yap short hairpin RNA (shRNA)-treated mice were used to confirm the role of PXR or yes-associated protein (YAP). Treatment with selective PXR activators induced liver enlargement and accelerated regeneration in wild-type (WT) and PXR-humanized mice, but not in Pxr-null mice, by increase of cell size, induction of a regenerative hybrid hepatocyte (HybHP) reprogramming, and promotion of hepatocyte and HybHP proliferation. Mechanistically, PXR interacted with YAP and PXR activation induced nuclear translocation of YAP. Blockade of YAP abolished PXR-induced liver enlargement in mice. Conclusion: These findings revealed a function of PXR in enlarging liver size and changing liver cell fate by activation of the YAP signaling pathway. These results have implications for understanding the physiological functions of PXR and suggest the potential for manipulation of liver size and liver cell fate.
Collapse
Affiliation(s)
- Yiming Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shicheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Yue Gao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Kaili Fu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Ying Wang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahong Sun
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Xinpeng Yao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Conghui Liu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Huizhen Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Leqian Xu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo, China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yingzi Yang
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China.,Correspondence to: Hui-chang Bi, Ph.D., School of Pharmaceutical Sciences, Sun Yat-sen University, 132# Waihuandong Road, Guangzhou University City, Guangzhou 510006, P. R. China, Phone: +86-20-39943470, Fax: +86-20-39943000,
| |
Collapse
|
12
|
Yamasaki Y, Kobayashi K, Inaba A, Uehara D, Tojima H, Kakizaki S, Chiba K. Indirect activation of pregnane X receptor in the induction of hepatic CYP3A11 by high-dose rifampicin in mice. Xenobiotica 2018; 48:1098-1105. [PMID: 29095659 DOI: 10.1080/00498254.2017.1400128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/29/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Although it is thought that RIF is not a ligand of rodent PXR, treatment with high-dose RIF (e.g. more than 20 mg/kg) increases the expression of CYP3A in the mouse liver. In this study, we investigated whether the induction of CYP3A by high-dose RIF in the mouse liver is mediated via indirect activation of mouse PXR (mPXR). The results showed that high-dose RIF increased the expression of CYP3A11 and other PXR-target genes in the liver of wild-type mice but not PXR-knockout mice. However, the results of reporter gene and ligand-dependent assembly assays showed that RIF does not activate mPXR in a ligand-dependent manner. In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. In conclusion, the present study suggests that high-dose RIF stimulates nuclear translocation of mPXR in the liver of mice by indirect activation, resulting in the transactivation of Cyp3a11 and other PXR-target genes.
Collapse
Affiliation(s)
- Yuki Yamasaki
- a Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba , Japan and
| | - Kaoru Kobayashi
- a Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba , Japan and
| | - Asumi Inaba
- a Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba , Japan and
| | - Daisuke Uehara
- b Division of Gastroenterology and Hepatology, Department of Internal Medicine, Graduate School of Medicine, Gunma University , Maebashi , Japan
| | - Hiroki Tojima
- b Division of Gastroenterology and Hepatology, Department of Internal Medicine, Graduate School of Medicine, Gunma University , Maebashi , Japan
| | - Satoru Kakizaki
- b Division of Gastroenterology and Hepatology, Department of Internal Medicine, Graduate School of Medicine, Gunma University , Maebashi , Japan
| | - Kan Chiba
- a Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University , Chiba , Japan and
| |
Collapse
|
13
|
Mackowiak B, Hodge J, Stern S, Wang H. The Roles of Xenobiotic Receptors: Beyond Chemical Disposition. Drug Metab Dispos 2018; 46:1361-1371. [PMID: 29759961 PMCID: PMC6124513 DOI: 10.1124/dmd.118.081042] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/07/2018] [Indexed: 02/06/2023] Open
Abstract
Over the past 20 years, the ability of the xenobiotic receptors to coordinate an array of drug-metabolizing enzymes and transporters in response to endogenous and exogenous stimuli has been extensively characterized and well documented. The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are the xenobiotic receptors that have received the most attention since they regulate the expression of numerous proteins important to drug metabolism and clearance and formulate a central defensive mechanism to protect the body against xenobiotic challenges. However, accumulating evidence has shown that these xenobiotic sensors also control many cellular processes outside of their traditional realms of xenobiotic metabolism and disposition, including physiologic and/or pathophysiologic responses in energy homeostasis, cell proliferation, inflammation, tissue injury and repair, immune response, and cancer development. This review will highlight recent advances in studying the noncanonical functions of xenobiotic receptors with a particular focus placed on the roles of CAR and PXR in energy homeostasis and cancer development.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Jessica Hodge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| |
Collapse
|
14
|
Rana M, Dash AK, Ponnusamy K, Tyagi RK. Nuclear localization signal region in nuclear receptor PXR governs the receptor association with mitotic chromatin. Chromosome Res 2018; 26:255-276. [DOI: 10.1007/s10577-018-9583-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022]
|
15
|
Tebbens JD, Azar M, Friedmann E, Lanzendörfer M, Pávek P. Mathematical Models in the Description of Pregnane X Receptor (PXR)-Regulated Cytochrome P450 Enzyme Induction. Int J Mol Sci 2018; 19:ijms19061785. [PMID: 29914136 PMCID: PMC6032247 DOI: 10.3390/ijms19061785] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
The pregnane X receptor (PXR) is a drug/xenobiotic-activated transcription factor of crucial importance for major cytochrome P450 xenobiotic-metabolizing enzymes (CYP) expression and regulation in the liver and the intestine. One of the major target genes regulated by PXR is the cytochrome P450 enzyme (CYP3A4), which is the most important human drug-metabolizing enzyme. In addition, PXR is supposed to be involved both in basal and/or inducible expression of many other CYPs, such as CYP2B6, CYP2C8, 2C9 and 2C19, CYP3A5, CYP3A7, and CYP2A6. Interestingly, the dynamics of PXR-mediated target genes regulation has not been systematically studied and we have only a few mechanistic mathematical and biologically based models describing gene expression dynamics after PXR activation in cellular models. Furthermore, few indirect mathematical PKPD models for prediction of CYP3A metabolic activity in vivo have been built based on compartmental models with respect to drug–drug interactions or hormonal crosstalk. Importantly, several negative feedback loops have been described in PXR regulation. Although current mathematical models propose these adaptive mechanisms, a comprehensive mathematical model based on sufficient experimental data is still missing. In the current review, we summarize and compare these models and address some issues that should be considered for the improvement of PXR-mediated gene regulation modelling as well as for our better understanding of the quantitative and spatial dynamics of CYPs expression.
Collapse
Affiliation(s)
- Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Malek Azar
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| | - Elfriede Friedmann
- Department of Applied Mathematics, Faculty of Mathematics and Computer Sciences, Mathematikon, University Heidelberg, Im Neuenheimer Feld 205, D-69120 Heidelberg, Germany.
| | - Martin Lanzendörfer
- Institute of Hydrogeology, Engineering Geology and Applied Geophysics, Faculty of Science, Charles University, Albertov 6, 128 43 Praha 2, Czech Republic.
| | - Petr Pávek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic.
| |
Collapse
|
16
|
Oladimeji PO, Chen T. PXR: More Than Just a Master Xenobiotic Receptor. Mol Pharmacol 2018; 93:119-127. [PMID: 29113993 PMCID: PMC5767680 DOI: 10.1124/mol.117.110155] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022] Open
Abstract
Pregnane X receptor (PXR) is a nuclear receptor considered to be a master xenobiotic receptor that coordinately regulates the expression of genes encoding drug-metabolizing enzymes and drug transporters to essentially detoxify and eliminate xenobiotics and endotoxins from the body. In the past several years, the function of PXR in the regulation of xenobiotic metabolism has been extensively studied, and the role of PXR as a xenobiotic sensor has been well established. It is now clear, however, that PXR plays many other roles in addition to its xenobiotic-sensing function. For instance, recent studies have discovered previously unidentified roles of PXR in inflammatory response, cell proliferation, and cell migration. PXR also contributes to the dysregulation of these processes in diseases states. These recent discoveries of the role of PXR in the physiologic and pathophysiologic conditions of other cellular processes provides the possibility of novel targets for drug discovery. This review highlights areas of PXR regulation that require further clarification and summarizes the recent progress in our understanding of the nonxenobiotic functions of PXR that can be explored for relevant therapeutic applications.
Collapse
Affiliation(s)
- Peter O Oladimeji
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
17
|
Dash AK, Yende AS, Jaiswal B, Tyagi RK. Heterodimerization of Retinoid X Receptor with Xenobiotic Receptor partners occurs in the cytoplasmic compartment: Mechanistic insights of events in living cells. Exp Cell Res 2017; 360:337-346. [DOI: 10.1016/j.yexcr.2017.09.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 01/30/2023]
|
18
|
Yokobori K, Kobayashi K, Azuma I, Akita H, Chiba K. Intracellular localization of pregnane X receptor in HepG2 cells cultured by the hanging drop method. Drug Metab Pharmacokinet 2017; 32:265-272. [DOI: 10.1016/j.dmpk.2017.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 08/05/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022]
|
19
|
Pavek P. Pregnane X Receptor (PXR)-Mediated Gene Repression and Cross-Talk of PXR with Other Nuclear Receptors via Coactivator Interactions. Front Pharmacol 2016; 7:456. [PMID: 27932985 PMCID: PMC5122737 DOI: 10.3389/fphar.2016.00456] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
Pregnane X receptor is a ligand-activated nuclear receptor (NR) that mainly controls inducible expression of xenobiotics handling genes including biotransformation enzymes and drug transporters. Nowadays it is clear that PXR is also involved in regulation of intermediate metabolism through trans-activation and trans-repression of genes controlling glucose, lipid, cholesterol, bile acid, and bilirubin homeostasis. In these processes PXR cross-talks with other NRs. Accumulating evidence suggests that the cross-talk is often mediated by competing for common coactivators or by disruption of coactivation and activity of other transcription factors by the ligand-activated PXR. In this respect mainly PXR-CAR and PXR-HNF4α interference have been reported and several cytochrome P450 enzymes (such as CYP7A1 and CYP8B1), phase II enzymes (SULT1E1, Gsta2, Ugt1a1), drug and endobiotic transporters (OCT1, Mrp2, Mrp3, Oatp1a, and Oatp4) as well as intermediate metabolism enzymes (PEPCK1 and G6Pase) have been shown as down-regulated genes after PXR activation. In this review, I summarize our current knowledge of PXR-mediated repression and coactivation interference in PXR-controlled gene expression regulation.
Collapse
Affiliation(s)
- Petr Pavek
- Department of Pharmacology and Toxicology and Centre for Drug Development, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague Hradec Kralove, Czechia
| |
Collapse
|
20
|
A SUMO-acetyl switch in PXR biology. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1170-1182. [PMID: 26883953 DOI: 10.1016/j.bbagrm.2016.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/22/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
Abstract
Post-translational modification (PTM) of nuclear receptor superfamily members regulates various aspects of their biology to include sub-cellular localization, the repertoire of protein-binding partners, as well as their stability and mode of degradation. The nuclear receptor pregnane X receptor (PXR, NR1I2) is a master-regulator of the drug-inducible gene expression in liver and intestine. The PXR-mediated gene activation program is primarily recognized to increase drug metabolism, drug transport, and drug efflux pathways in these tissues. The activation of PXR also has important implications in significant human diseases including inflammatory bowel disease and cancer. Our recent investigations reveal that PXR is modified by multiple PTMs to include phosphorylation, SUMOylation, and ubiquitination. Using both primary cultures of hepatocytes and cell-based assays, we show here that PXR is modified through acetylation on lysine residues. Further, we show that increased acetylation of PXR stimulates its increased SUMO-modification to support active transcriptional suppression. Pharmacologic inhibition of lysine de-acetylation using trichostatin A (TSA) alters the sub-cellular localization of PXR in cultured hepatocytes, and also has a profound impact upon PXR transactivation capacity. Both the acetylation and SUMOylation status of the PXR protein is affected by its ability to associate with the lysine de-acetylating enzyme histone de-acetylase (HDAC)3 in a complex with silencing mediator of retinoic acid and thyroid hormone receptor (SMRT). Taken together, our data support a model in which a SUMO-acetyl 'switch' occurs such that acetylation of PXR likely stimulates SUMO-modification of PXR to promote the active repression of PXR-target gene expression. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
21
|
Mackowiak B, Wang H. Mechanisms of xenobiotic receptor activation: Direct vs. indirect. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1130-1140. [PMID: 26877237 DOI: 10.1016/j.bbagrm.2016.02.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/05/2016] [Accepted: 02/06/2016] [Indexed: 12/31/2022]
Abstract
The so-called xenobiotic receptors (XRs) have functionally evolved into cellular sensors for both endogenous and exogenous stimuli by regulating the transcription of genes encoding drug-metabolizing enzymes and transporters, as well as those involving energy homeostasis, cell proliferation, and/or immune responses. Unlike prototypical steroid hormone receptors, XRs are activated through both direct ligand-binding and ligand-independent (indirect) mechanisms by a plethora of structurally unrelated chemicals. This review covers research literature that discusses direct vs. indirect activation of XRs. A particular focus is centered on the signaling control of the constitutive androstane receptor (CAR), the pregnane X receptor (PXR), and the aryl hydrocarbon receptor (AhR). We expect that this review will shed light on both the common and distinct mechanisms associated with activation of these three XRs. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, United States.
| |
Collapse
|
22
|
Ginkgolide B protects human umbilical vein endothelial cells against xenobiotic injuries via PXR activation. Acta Pharmacol Sin 2016; 37:177-86. [PMID: 26775663 DOI: 10.1038/aps.2015.124] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
AIM Pregnane X receptor (PXR) is a nuclear receptor that regulates a number of genes encoding drug metabolism enzymes and transporters and plays a key role in xeno- and endobiotic detoxification. Ginkgolide B has shown to increase the activity of PXR. Here we examined whether ginkgolide B activated PXR and attenuated xenobiotic-induced injuries in endothelial cells. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with ginkgolide B. The expression of PXR, CYP3A4, MDR1, VCAM-1, E-selectin and caspase-3 were quantified with qRT-PCR and Western blot analysis. Cell apoptosis was analyzed with flow cytometry. Fluorescently labeled human acute monocytic leukemia cells (THP-1 cells) were used to examine cell adhesion. RESULTS Ginkgolide B (30-300 μmol/L) did not change the mRNA and protein levels of PXR in the cells, but dose-dependently increased nuclear translocation of PXR protein. Ginkgolide B increased the expression of CYP3A4 and MDR1 in the cells, which was partially reversed by pretreatment with the selective PXR signaling antagonist sulforaphane, or transfection with PXR siRNA. Functionally, ginkgolide B dose-dependently attenuated doxorubicin- or staurosporine-induced apoptosis, which was reversed by transfection with PXR siRNA. Moreover, ginkgolide B suppressed TNF-α-induced THP-1 cell adhesion and TNF-α-induced expression of vascular adhesion molecule 1 (VCAM-1) and E-selectin in the cells, which was also reversed by transfection with PXR siRNA. CONCLUSION Ginkgolide B exerts anti-apoptotic and anti-inflammatory effects on endothelial cells via PXR activation, suggesting that a PXR-mediated endothelial detoxification program may be important for protecting endothelial cells from xeno- and endobiotic-induced injuries.
Collapse
|
23
|
Litwa E, Rzemieniec J, Wnuk A, Lason W, Krzeptowski W, Kajta M. RXRα, PXR and CAR xenobiotic receptors mediate the apoptotic and neurotoxic actions of nonylphenol in mouse hippocampal cells. J Steroid Biochem Mol Biol 2016; 156:43-52. [PMID: 26643981 DOI: 10.1016/j.jsbmb.2015.11.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/21/2015] [Accepted: 11/26/2015] [Indexed: 12/26/2022]
Abstract
In the present study, we investigated the role of the retinoid X receptor (RXR), the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR), in the apoptotic and toxic effects of nonylphenol in mouse primary neuronal cell cultures. Our study demonstrated that nonylphenol activated caspase-3 and induced lactate dehydrogenase (LDH) release in hippocampal cells, which was accompanied by an increase in the mRNA expression and protein levels of RXRα, PXR and CAR. Nonylphenol stimulated Rxra, Pxr, and Car mRNA expression. These effects were followed by increase in the protein levels of particular receptors. Immunofluorescence labeling revealed the cellular distribution of RXRα, PXR and CAR in hippocampal neurons in response to nonylphenol, shortening of neurites and cytoplasmic shrinking, as indicated by MAP2 staining. It also showed NP-induced translocation of receptor-specific immunofluorescence from cytoplasm to the nucleus. The use of specific siRNAs demonstrated that Rxra-, Pxr-, and Car-siRNA-transfected cells were less vulnerable to nonylphenol-induced activation of caspase-3 and LDH, thus confirming the key involvement of RXRα/PXR/CAR signaling pathways in the apoptotic and neurotoxic actions of nonylphenol. These new data give prospects for the targeting xenobiotic nuclear receptors to protect the developing nervous system against endocrine disrupting chemicals.
Collapse
Affiliation(s)
- E Litwa
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - J Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - A Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - W Lason
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - W Krzeptowski
- Department of Cell Biology and Imaging, Confocal Microscopy Laboratory, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland
| | - M Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland.
| |
Collapse
|
24
|
Sugatani J, Noguchi Y, Hattori Y, Yamaguchi M, Yamazaki Y, Ikari A. Threonine-408 Regulates the Stability of Human Pregnane X Receptor through Its Phosphorylation and the CHIP/Chaperone-Autophagy Pathway. Drug Metab Dispos 2016; 44:137-50. [PMID: 26534988 DOI: 10.1124/dmd.115.066308] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/02/2015] [Indexed: 11/22/2022] Open
Abstract
The human pregnane X receptor (hPXR) is a xenobiotic-sensing nuclear receptor that transcriptionally regulates drug metabolism-related genes. The aim of the present study was to elucidate the mechanism by which hPXR is regulated through threonine-408. A phosphomimetic mutation at threonine-408 (T408D) reduced the transcriptional activity of hPXR and its protein stability in HepG2 and SW480 cells in vitro and mouse livers in vivo. Proteasome inhibitors (calpain inhibitor I and MG132) and Hsp90 inhibitor geldanamycin, but not Hsp70 inhibitor pifithrin-μ, increased wild-type (WT) hPXR in the nucleus. The translocation of the T408D mutant to the nucleus was significantly reduced even in the presence of proteasome inhibitors, whereas the complex of yellow fluorescent protein (YFP)-hPXR T408D mutant with heat shock cognate protein 70/heat shock protein 70 and carboxy terminus Hsp70-interacting protein (CHIP; E3 ligase) was similar to that of the WT in the cytoplasm. Treatment with pifithrin-μ and transfection with anti-CHIP small-interfering RNA reduced the levels of CYP3A4 mRNA induced by rifampicin, suggesting the contribution of Hsp70 and CHIP to the transactivation of hPXR. Autophagy inhibitor 3-methyladenine accumulated YFP-hPXR T408D mutant more efficiently than the WT in the presence of proteasome inhibitor lactacystin, and the T408D mutant colocalized with the autophagy markers, microtubule-associated protein 1 light chain 3 and p62, which were contained in the autophagic cargo. Lysosomal inhibitor chloroquine caused the marked accumulation of the T408D mutant in the cytoplasm. Protein kinase C (PKC) directly phosphorylated the threonine-408 of hPXR. These results suggest that hPXR is regulated through its phosphorylation at threonine-408 by PKC, CHIP/chaperone-dependent stability check, and autophagic degradation pathway.
Collapse
Affiliation(s)
- Junko Sugatani
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Yuji Noguchi
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Yoshiki Hattori
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Masahiko Yamaguchi
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Yasuhiro Yamazaki
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| | - Akira Ikari
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan
| |
Collapse
|
25
|
Wang YM, Chai SC, Lin W, Chai X, Elias A, Wu J, Ong SS, Pondugula SR, Beard JA, Schuetz EG, Zeng S, Xie W, Chen T. Serine 350 of human pregnane X receptor is crucial for its heterodimerization with retinoid X receptor alpha and transactivation of target genes in vitro and in vivo. Biochem Pharmacol 2015; 96:357-68. [PMID: 26119819 DOI: 10.1016/j.bcp.2015.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
The human pregnane X receptor (hPXR), a member of the nuclear receptor superfamily, senses xenobiotics and controls the transcription of genes encoding drug-metabolizing enzymes and transporters. The regulation of hPXR's transcriptional activation of its target genes is important for xenobiotic detoxification and endobiotic metabolism, and hPXR dysregulation can cause various adverse drug effects. Studies have implicated the putative phosphorylation site serine 350 (Ser(350)) in regulating hPXR transcriptional activity, but the mechanism of regulation remains elusive. Here we investigated the transactivation of hPXR target genes in vitro and in vivo by hPXR with a phosphomimetic mutation at Ser(350) (hPXR(S350D)). The S350D phosphomimetic mutation reduced the endogenous expression of cytochrome P450 3A4 (an hPXR target gene) in HepG2 and LS180 cells. Biochemical assays and structural modeling revealed that Ser(350) of hPXR is crucial for formation of the hPXR-retinoid X receptor alpha (RXRα) heterodimer. The S350D mutation abrogated heterodimerization in a ligand-independent manner, impairing hPXR-mediated transactivation. Further, in a novel humanized transgenic mouse model expressing the hPXR(S350D) transgene, we demonstrated that the S350D mutation alone is sufficient to impair hPXR transcriptional activity in mouse liver. This transgenic mouse model provides a unique tool to investigate the regulation and function of hPXR, including its non-genomic function, in vivo. Our finding that phosphorylation regulates hPXR activity has implications for development of novel hPXR antagonists and for safety evaluation during drug development.
Collapse
Affiliation(s)
- Yue-Ming Wang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiaojuan Chai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ayesha Elias
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Su Sien Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Satyanarayana R Pondugula
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jordan A Beard
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Erin G Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Su Zeng
- Department of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
26
|
Hiura Y, Satsu H, Hamada M, Shimizu M. Analysis of flavonoids regulating the expression of UGT1A1 via xenobiotic receptors in intestinal epithelial cells. Biofactors 2014; 40:336-45. [PMID: 24375494 DOI: 10.1002/biof.1153] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/11/2013] [Indexed: 12/23/2022]
Abstract
UDP-glucuronosyltransferase (UGT) 1A1 is one of the major metabolic enzymes for the detoxification of harmful xenobiotics in intestines, and its expression is regulated by transcription factors like the aryl hydrocarbon receptor (AhR) and the pregnane X receptor (PXR). A screening assay using real-time PCR showed that baicalein and 3-hydroxyflavone induced human UGT1A1 mRNA expression in LS180 cells. Experimental results confirmed that these flavonoids increased UGT1A protein expression as well as its enzymatic activity. The results indicated that baicalein and 3-hydroxyflavone increased the transcriptional activity of UGT1A1 via AhR and PXR, respectively. Observation via immunofluorescence microscopy suggested that baicalein and 3-hydroxyflavone further induced nuclear translocation of AhR and PXR, respectively. In addition, direct interaction between baicalein and AhR or 3-hydroxyflavone and PXR were observed using the quartz crystal microbalance method. These results elucidate the molecular mechanism of flavonoid-induced UGT1A1 gene expression via xenobiotic receptors in the intestines.
Collapse
Affiliation(s)
- Yuto Hiura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | | | | | | |
Collapse
|
27
|
Šíma M, Netíková I, Slanař O. Pregnane xenobiotic receptors and their effect on drug elimination from the organism. Prague Med Rep 2014; 114:205-13. [PMID: 24485337 DOI: 10.14712/23362936.2014.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Nuclear receptors are intracellular proteins which, having been activated by their more or less specific ligands, regulate (usually increase) the transcription of target genes. They thus participate in a regulation of a number of physiologic functions. Some of them - especially pregnane xenobiotic receptors - serve primarily as protection of the organism from the xenobiotic intoxication. This is because many xenobiotics activate their function which consists in increasing the gene expression of enzymes involved in the metabolism of xenobiotics and detoxication drug transporters. Clarification of these mechanisms enabled the understanding of the substance of many drug-drug interactions observed in the clinical practice. Polymorphism of the nuclear receptors appears to be one of the causes of the interindividual variability in response to drug administration.
Collapse
Affiliation(s)
- M Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - I Netíková
- Institute of Pharmacology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - O Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech RepublicInstitute of Pharmacology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
28
|
Shear stress activation of nuclear receptor PXR in endothelial detoxification. Proc Natl Acad Sci U S A 2013; 110:13174-9. [PMID: 23878263 DOI: 10.1073/pnas.1312065110] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endothelial cells (ECs) are constantly exposed to xenobiotics and endobiotics or their metabolites, which perturb EC function, as well as to shear stress, which plays a crucial role in vascular homeostasis. Pregnane X receptor (PXR) is a nuclear receptor and a key regulator of the detoxification of xeno- and endobiotics. Here we show that laminar shear stress (LSS), the atheroprotective flow, activates PXR in ECs, whereas oscillatory shear stress, the atheroprone flow, suppresses PXR. LSS activation of PXR in cultured ECs led to the increased expression of a PXR target gene, multidrug resistance 1 (MDR1). An in vivo study using rats showed that the expression of MDR1 was significantly higher in the endothelium from the descending thoracic aorta, where flow is mostly laminar, than from the inner curvature of aortic arch, where flow is disturbed. Functionally, LSS-activated PXR protects ECs from apoptosis triggered by doxorubicin via the induction of MDR1 and other detoxification genes. PXR also suppressed the expression of proinflammatory adhesion molecules and monocyte adhesion in response to TNF-α and lipopolysaccharide. Overexpression of a constitutively active PXR in rat carotid arteries potently attenuated proinflammatory responses. In addition, cDNA microarray revealed a large number of the PXR-activated endothelial genes whose products are responsible for major steps of detoxification, including phase I and II metabolizing enzymes and transporters. These detoxification genes in ECs are induced by LSS in ECs in a PXR-dependent manner. In conclusion, our results indicate that PXR represents a flow-activated detoxification system to protect ECs against damage by xeno- and endobiotics.
Collapse
|
29
|
Sharma D, Lau AJ, Sherman MA, Chang TK. Agonism of human pregnane X receptor by rilpivirine and etravirine: Comparison with first generation non-nucleoside reverse transcriptase inhibitors. Biochem Pharmacol 2013; 85:1700-11. [DOI: 10.1016/j.bcp.2013.04.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/02/2013] [Accepted: 04/02/2013] [Indexed: 11/24/2022]
|
30
|
Qiao E, Ji M, Wu J, Ma R, Zhang X, He Y, Zha Q, Song X, Zhu LW, Tang J. Expression of the PXR gene in various types of cancer and drug resistance. Oncol Lett 2013; 5:1093-1100. [PMID: 23599746 PMCID: PMC3628904 DOI: 10.3892/ol.2013.1149] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/02/2013] [Indexed: 01/13/2023] Open
Abstract
Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily of ligand-regulated transcription factors. PXR is a key xenobiotic receptor that regulates the expression of genes implicated in drug metabolism, detoxification and clearance, including drug metabolizing enzymes and transporters, suggesting that it is significant in the drug resistance of cancer cells. PXR is expressed in a wide range of tissues in the human body. Studies have demonstrated that PXR is expressed in a variety of tumor types, correlating not only with drug resistance but also with the cell proliferation, apoptosis and prognosis of cancer. The purpose of the present review is to provide a comprehensive review of PXR and its potential roles in multidrug resistance and the biological characteristics of PXR-positive tumors.
Collapse
Affiliation(s)
- Enqi Qiao
- Department of General Surgery, Jiangsu Cancer Hospital, Affiliated to Nanjing Medical University, Nanjing 210009
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lemmen J, Tozakidis IE, Galla HJ. Pregnane X receptor upregulates ABC-transporter Abcg2 and Abcb1 at the blood-brain barrier. Brain Res 2013; 1491:1-13. [DOI: 10.1016/j.brainres.2012.10.060] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/17/2012] [Accepted: 10/30/2012] [Indexed: 10/27/2022]
|
32
|
Honma M, Kozawa M, Suzuki H. Methods for the quantitative evaluation and prediction of CYP enzyme induction using human in vitro systems. Expert Opin Drug Discov 2012; 5:491-511. [PMID: 22823132 DOI: 10.1517/17460441003762717] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD For successful drug development, it is important to investigate the potency of candidate drugs causing drug-drug interactions (DDI) during the early stages of development. The most common mechanisms of DDIs are the inhibition and induction of CYP enzymes. Therefore, it is important to develop co.mpounds with lower potencies for CYP enzyme induction. AREAS COVERED IN THIS REVIEW The aim of the present paper is to present an overview of the current knowledge of CYP induction mechanisms, particularly focusing on the transcriptional gene activation mediated by pregnane X receptor, aryl hydrocarbon receptor and constitutive androstane receptor. The adoptable options of in vitro assay methods for evaluating CYP induction are also summarized. Finally, we introduce a method for the quantitative prediction of CYP3A4 induction considering the turnover of CYP3A4 mRNA and protein in hepatocytes based on the data obtained from a reporter gene assay. WHAT THE READER WILL GAIN In order to predict in vivo CYP enzyme induction quantitatively based on in vitro information, an understanding of the physiological induction mechanisms and the features of each in vitro assay system is essential. We also present the estimation method of in vivo CYP induction potency of each compound based on the in vitro data which are routinely obtained but not necessarily utilized maximally in pharmaceutical companies. TAKE HOME MESSAGE It is desirable to select compounds with lower potencies for the inductive effect. For this purpose, an accurate prioritization procedure to evaluate the induction potency of each compound in a quantitative manner considering the pharmacologically effective concentration of each compound is necessary.
Collapse
Affiliation(s)
- Masashi Honma
- The University of Tokyo Hospital, Faculty of Medicine, Department of Pharmacy, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan +81 3 3815 5411 ; +81 3 3816 6159 ;
| | | | | |
Collapse
|
33
|
Sesamin: A Naturally Occurring Lignan Inhibits CYP3A4 by Antagonizing the Pregnane X Receptor Activation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:242810. [PMID: 22645625 PMCID: PMC3356939 DOI: 10.1155/2012/242810] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/30/2012] [Accepted: 02/06/2012] [Indexed: 12/02/2022]
Abstract
Inconsistent expression and regulation of drug-metabolizing enzymes (DMEs) are common causes of adverse drug effects in some drugs with a narrow therapeutic index (TI). An important cytochrome, cytochrome P450 3A4 (CYP3A4), is predominantly regulated by a nuclear receptor, pregnane X receptor (PXR). Sesamin, a major lignan constituent in sesame seeds and oil, exhibits a variety of biological functions; however, the effect of sesamin on the modulation of CYP3A4 is not well understood. In this study, the effects of sesamin on the PXR-CYP3A4 pathway were characterized, as well as the underlying mechanisms of those effects. Sesamin potently attenuated CYP3A4 induction in a dose-dependent manner by blocking the activation of PXR. The PXR inducer-mediated inhibition of CYP3A4 was further evidenced by the ability of sesamin to attenuate the effects of several PXR ligands in the CYP3A4 reporter assay. Further mechanistic studies showed that sesamin inhibited PXR by interrupting the interacting with coregulators. These results may lead to the development of new therapeutic and dietary approaches to reduce the frequency of inducer-drug interaction. Sesamin was established as a novel inhibitor of PXR and may be useful for modulating DMEs expression and drug efficacies. Modification of CYP3A4 expression and activity by consumption of sesamin may have important implications for drug safety.
Collapse
|
34
|
Wang YM, Ong SS, Chai SC, Chen T. Role of CAR and PXR in xenobiotic sensing and metabolism. Expert Opin Drug Metab Toxicol 2012; 8:803-17. [PMID: 22554043 DOI: 10.1517/17425255.2012.685237] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The xenobiotic detoxification system, which protects the human body from external chemicals, comprises drug-metabolizing enzymes and transporters whose expressions are regulated by pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). The progress made in a large number of recent studies calls for a timely review to summarize and highlight these key discoveries. AREAS COVERED This review summarizes recent advances in elucidating the roles of PXR and CAR in the xenobiotic detoxification system. It also highlights the progress in understanding the regulation of PXR and CAR activity at the post-translational levels, as well as the structural basis for the regulation of these two xenobiotic sensors. EXPERT OPINION Future efforts are needed to discover novel agonists and antagonists with species and isoform selectivity, to systematically understand the regulation of PXR and CAR at multiple levels (transcriptional, post-transcriptional and post-translational levels) in response to xenobiotics exposure, and to solve the structures of the full-length receptors, which will be enabled by improved protein expression and purification techniques and approaches. In addition, more efforts will be needed to validate PXR and CAR as disease-related therapeutic targets and thus expand their roles as master xenobiotic sensors.
Collapse
Affiliation(s)
- Yue-Ming Wang
- St. Jude Children's Research Hospital, Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
35
|
Chen Y, Tang Y, Guo C, Wang J, Boral D, Nie D. Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters. Biochem Pharmacol 2012; 83:1112-26. [PMID: 22326308 PMCID: PMC3339266 DOI: 10.1016/j.bcp.2012.01.030] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 01/23/2012] [Accepted: 01/25/2012] [Indexed: 01/18/2023]
Abstract
Chemotherapy is one of the three most common treatment modalities for cancer. However, its efficacy is limited by multidrug resistant cancer cells. Drug metabolizing enzymes (DMEs) and efflux transporters promote the metabolism, elimination, and detoxification of chemotherapeutic agents. Consequently, elevated levels of DMEs and efflux transporters reduce the therapeutic effectiveness of chemotherapeutics and, often, lead to treatment failure. Nuclear receptors, especially pregnane X receptor (PXR, NR1I2) and constitutive androstane activated receptor (CAR, NR1I3), are increasingly recognized for their role in xenobiotic metabolism and clearance as well as their role in the development of multidrug resistance (MDR) during chemotherapy. Promiscuous xenobiotic receptors, including PXR and CAR, govern the inducible expressions of a broad spectrum of target genes that encode phase I DMEs, phase II DMEs, and efflux transporters. Recent studies conducted by a number of groups, including ours, have revealed that PXR and CAR play pivotal roles in the development of MDR in various human carcinomas, including prostate, colon, ovarian, and esophageal squamous cell carcinomas. Accordingly, PXR/CAR expression levels and/or activation statuses may predict prognosis and identify the risk of drug resistance in patients subjected to chemotherapy. Further, PXR/CAR antagonists, when used in combination with existing chemotherapeutics that activate PXR/CAR, are feasible and promising options that could be utilized to overcome or, at least, attenuate MDR in cancer cells.
Collapse
Affiliation(s)
- Yakun Chen
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794, United States
| | | | | | | | | | | |
Collapse
|
36
|
Chan GNY, Hoque MT, Cummins CL, Bendayan R. Regulation of P-glycoprotein by orphan nuclear receptors in human brain microvessel endothelial cells. J Neurochem 2011; 118:163-75. [DOI: 10.1111/j.1471-4159.2011.07288.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Tamási V, Monostory K, Prough RA, Falus A. Role of xenobiotic metabolism in cancer: involvement of transcriptional and miRNA regulation of P450s. Cell Mol Life Sci 2011; 68:1131-46. [PMID: 21184128 PMCID: PMC11115005 DOI: 10.1007/s00018-010-0600-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 11/04/2010] [Accepted: 11/18/2010] [Indexed: 02/06/2023]
Abstract
Cytochrome P450 enzymes (P450s) are important targets in cancer, due to their role in xenobiotic metabolism. Since P450s are the "bridges" between the environment and our body, their function can be linked in many ways to carcinogenesis: they activate dietary and environmental components to ultimate carcinogens (i), the cancer tissue maintains its drug resistance with altered expression of P450s (ii), P450s metabolize (sometimes activate) drugs used for cancer treatment (iii) and they are potential targets for anticancer therapy (iiii). These highly polymorphic enzymes are regulated at multiple molecular levels. Regulation is as important as genetic difference in the existing individual variability in P450 activity. In this review, examples of the transcriptional (DNA methylation, histone modification, modulation by xenosensors) and post-transcriptional (miRNA) regulation will be presented and thereby introduce potential molecular targets at which the metabolism of anticancer drugs, the elimination of cancerogenes or the progress of carcinogenesis could be affected.
Collapse
Affiliation(s)
- Viola Tamási
- Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, PO Box 370, Budapest, 1445, Hungary.
| | | | | | | |
Collapse
|
38
|
Bonamassa B, Liu D. Nonviral gene transfer as a tool for studying transcription regulation of xenobiotic metabolizing enzymes. Adv Drug Deliv Rev 2010; 62:1250-6. [PMID: 20713102 PMCID: PMC2991602 DOI: 10.1016/j.addr.2010.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/05/2010] [Accepted: 08/10/2010] [Indexed: 12/19/2022]
Abstract
Numerous xenobiotic metabolizing enzymes are regulated by nuclear receptors at transcriptional level. The challenge we currently face is to understand how a given nuclear receptor interacts with its xenobiotics, migrates into nucleus, binds to the xenobiotic response element of a target gene, and regulates transcription. Toward this end, new methods have been developed to introduce the nuclear receptor gene into appropriate cells and study its activity in activating reporter gene expression under the control of a promoter containing xenobiotic response elements. The goal of this review is to critically examine the gene transfer methods currently available. We concentrate on the gene transfer mechanism, advantages and limitations of each method when employed for nuclear receptor-mediated gene regulation studies. It is our hope that the information provided highlights the importance of gene transfer in studying the mechanisms by which our body eliminates the potentially harmful substances and maintains the homeostasis.
Collapse
Affiliation(s)
- Barbara Bonamassa
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, 527 Salk Hall, 3501 Terrace Street, Pittsburgh, PA15261, United States
| | | |
Collapse
|
39
|
Li H, Wang H. Activation of xenobiotic receptors: driving into the nucleus. Expert Opin Drug Metab Toxicol 2010; 6:409-26. [PMID: 20113149 DOI: 10.1517/17425251003598886] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE OF THE FIELD Xenobiotic receptors (XRs) play pivotal roles in regulating the expression of genes that determine the clearance and detoxification of xenobiotics, such as drugs and environmental chemicals. Recently, it has become increasingly evident that most XRs shuttle between the cytoplasm and nucleus, and activation of such receptors is directly associated with xenobiotic-induced nuclear import. AREAS COVERED IN THIS REVIEW The scope of this review covers research literature that discusses nuclear translocation and activation of XRs, as well as unpublished data generated from this laboratory. Specific emphasis is given to the constitutive androstane receptor (CAR), the pregnane X receptor and the aryl hydrocarbon receptor. WHAT THE READERS WILL GAIN A number of molecular chaperons presumably associated with cellular localization of XRs have been identified. Primary hepatocyte cultures have been established as a unique model retaining inactive CAR in the cytoplasm. Moreover, several splicing variants of human CAR exhibit altered cellular localization and chemical activation. TAKE HOME MESSAGE Nuclear accumulation is an essential step in the activation of XRs. Although great strides have been made, much remains to be understood concerning the mechanisms underlying intracellular localization and trafficking of XRs, which involve both direct ligand-binding and indirect pathways.
Collapse
Affiliation(s)
- Haishan Li
- University of Maryland School of Pharmacy, Department of Pharmaceutical Sciences, 20 Penn Street, Baltimore, MD 21201, USA
| | | |
Collapse
|
40
|
Chen Y, Tang Y, Robbins GT, Nie D. Camptothecin attenuates cytochrome P450 3A4 induction by blocking the activation of human pregnane X receptor. J Pharmacol Exp Ther 2010; 334:999-1008. [PMID: 20504912 PMCID: PMC2939670 DOI: 10.1124/jpet.110.168294] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 05/25/2010] [Indexed: 01/25/2023] Open
Abstract
Differential regulation of drug-metabolizing enzymes (DMEs) is a common cause of adverse drug effects in cancer therapy. Due to the extremely important role of cytochrome P450 3A4 (CYP3A4) in drug metabolism and the dominant regulation of human pregnane X receptor (hPXR) on CYP3A4, finding inhibitors for hPXR could provide a unique tool to control drug efficacies in cancer therapy. Camptothecin (CPT) was demonstrated as a novel and potent inhibitor (IC(50) = 0.58 microM) of an hPXR-mediated transcriptional regulation on CYP3A4 in this study. In contrast, one of its analogs, irinotecan (CPT-11), was found to be an hPXR agonist in the same tests. CPT disrupted the interaction of hPXR with steroid receptor coactivator-1 but had effects on neither the competition of ligand binding nor the formation of the hPXR and retinoid X receptor alpha heterodimer, nor the interaction between the regulatory complex and DNA-responsive elements. CPT treatment resulted in delayed metabolism of nifedipine in human hepatocytes treated with rifampicin, suggesting a potential prevention of drug-drug interactions between CYP3A4 inducers and CYP3A4-metabolized drugs. Because CPT is the leading compound of topoisomerase I inhibitors, which comprise a quickly developing class of anticancer agents, the findings indicate the potential of a new class of compounds to modify hPXR activity as agonists/inhibitors and are important in the development of CPT analogs.
Collapse
Affiliation(s)
- Yakun Chen
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine and SimmonsCooper Cancer Institute, Springfield, IL 62794-9626, USA
| | | | | | | |
Collapse
|
41
|
Interplay between cholesterol and drug metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2010; 1814:146-60. [PMID: 20570756 DOI: 10.1016/j.bbapap.2010.05.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/14/2022]
Abstract
Cholesterol biosynthetic and metabolic pathways contain several branching points towards physiologically active molecules, such as coenzyme Q, vitamin D, glucocorticoid and steroid hormones, oxysterols, or bile acids. Sophisticated regulatory mechanisms are involved in maintenance of the homeostasis of not only cholesterol but also other cholesterogenic molecules. In addition to endogenous cues, cholesterol homeostasis needs to accommodate also to exogenous cues that are imported into the body, such as chemicals and medications. Steroid and nuclear receptors together with sterol regulatory element-binding protein (SREBP) mediate the fine tuning of biosynthetic and metabolic routes as well as transports of cholesterol and its derivatives. Similarly, drug/xenobiotic metabolism is the subject to the feedback regulation of cytochrome P450 enzymes and transporters. The regulatory mechanisms that maintain the homeostasis of cholesterogenic molecules and are involved in drug metabolism share similarities. Cholesterol and cholesterogenic compounds (bile acids, glucocorticoids, vitamin D, etc.) regulate the xenosensor signaling in drug-mediated induction of the major drug-metabolizing cytochrome P450 enzymes. The key cellular receptors, pregnane X receptor (PXR), constitutive androstane receptor (CAR), vitamin D receptor (VDR), and glucocorticoid receptor (GR) provide a functional cross-talk between the pathways maintaining cholesterol homeostasis and controlling the expression of drug-metabolizing enzymes. These receptors serve as metabolic sensors, resulting in a coordinate regulation of cholesterogenic compounds metabolism and of the defense against xenobiotic and endobiotic toxicity. Herein we present a comprehensive review of functional interactions between cholesterol homeostasis and drug metabolism involving the main nuclear and steroid receptors.
Collapse
|
42
|
Takeyama D, Miki Y, Fujishima F, Suzuki T, Akahira JI, Hata S, Miyata G, Satomi S, Sasano H. Steroid and xenobiotic receptor in human esophageal squamous cell carcinoma: a potent prognostic factor. Cancer Sci 2010; 101:543-9. [PMID: 19860844 PMCID: PMC11158420 DOI: 10.1111/j.1349-7006.2009.01380.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Steroid and xenobiotic receptor (SXR) is a nuclear receptor activated by diverse exogenous and endogenous compounds and has been demonstrated to play a pivotal role in detoxification through its regulation of various metabolizing enzymes and transporters. Recent studies also demonstrated the potential roles of SXR in the regulation of apoptosis and inflammation in various carcinoma cells, but the status of SXR in human esophageal squamous cell carcinoma (ESCC) has not been examined. Therefore, in this study, we performed immunohistochemical and quantitative RT-PCR evaluations in human ESCC in order to clarify its biological and clinical significance. We first immunolocalized SXR in 73 human ESCC cases. SXR immunoreactivity was detected in the nuclei, or in both nuclei and cytoplasm of carcinoma cells (98%, 20% of cases, respectively). The status of nuclear SXR immunoreactivity was inversely correlated with histological grade, lymph node status, ki67/MIB1 labeling index, and positively correlated with retinoid X receptor alpha status. In addition, high nuclear SXR expression was significantly correlated with favorable clinical outcome of the patients. Multivariate analysis further demonstrated SXR status in carcinoma cells as an independent favorable prognostic factor of the patients. Results of quantitative RT-PCR study demonstrated that SXR mRNA expression was detected in three of five cases, and was marked higher in the cancerous tissue than non-neoplastic tissue of these patients. This is the first study to demonstrate the status of SXR in human ESCC and the results suggest that SXR is a potent favorable prognostic factor of human ESCC.
Collapse
Affiliation(s)
- Daisuke Takeyama
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Boobis A, Watelet JB, Whomsley R, Benedetti MS, Demoly P, Tipton K. Drug interactions. Drug Metab Rev 2009; 41:486-527. [PMID: 19601724 DOI: 10.1080/10837450902891550] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Drugs for allergy are often taken in combination with other drugs, either to treat allergy or other conditions. In common with many pharmaceuticals, most such drugs are subject to metabolism by P450 enzymes and to transmembrane transport. This gives rise to considerable potential for drug-drug interactions, to which must be added consideration of drug-diet interactions. The potential for metabolism-based drug interactions is increasingly being taken into account during drug development, using a variety of in silico and in vitro approaches. Prediction of transporter-based interactions is not as advanced. The clinical importance of a drug interaction will depend upon a number of factors, and it is important to address concerns quantitatively, taking into account the therapeutic index of the compound.
Collapse
Affiliation(s)
- Alan Boobis
- Department of Experimental Medicine and Toxicology, Division of Medicine, Imperial College London, Hammersmith Campus, London.
| | | | | | | | | | | |
Collapse
|
44
|
Plant N, Aouabdi S. Nuclear receptors: the controlling force in drug metabolism of the liver? Xenobiotica 2009; 39:597-605. [PMID: 19622002 DOI: 10.1080/00498250903098218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The body is in a constant battle to achieve homeostasis; indeed, the robustness with which it can respond to moves away from homeostasis is a vital part in the survival of the organism as a whole. There thus exists a need for a network of sensors that are able to capture, interpret, and respond to alterations in chemical levels that move the body away from homeostasis and this applies to both endogenous and exogenous chemicals. With respect to external chemicals (xenobiotics), this xenosensing is often carried out through specific interactions with cellular receptors. The phenomenon of 'xenosensing' has attracted much interest of late, whereby xenobiotics interact with receptors resulting in the activation of a battery of genes mediating oxidative drug metabolism, conjugation, and transport, thereby enhancing the elimination of the xenobiotic by the organism. However, this beneficial response is counterbalanced by the increasingly recognized role of nuclear receptors in mediating drug-drug interactions via enzyme induction or the production of toxicity through interaction with endogenous pathways. This review will focus on the role of nuclear receptors in mediating these effects, and how such knowledge will contribute to a mechanism-based risk assessment for xenobiotics.
Collapse
Affiliation(s)
- N Plant
- Centre for Toxicology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU27XH, UK.
| | | |
Collapse
|
45
|
Pondugula SR, Dong H, Chen T. Phosphorylation and protein-protein interactions in PXR-mediated CYP3A repression. Expert Opin Drug Metab Toxicol 2009; 5:861-73. [PMID: 19505191 PMCID: PMC2719259 DOI: 10.1517/17425250903012360] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The expression of drug-metabolizing enzymes CYPs is controlled by pregnane X receptor (PXR), and, therefore, understanding how PXR modulates CYP expression is important to minimize adverse drug interactions, one type of preventable adverse drug reaction. OBJECTIVE We review the mechanisms of PXR-mediated repression of CYP expression. METHODS We discuss the clinical implications of CYP repression and the role of signal cross-talks, including protein-protein interactions and phosphorylation of PXR and coregulators, in inhibiting PXR and repressing CYP expression. RESULTS/CONCLUSION Kinases such as cyclin-dependent kinase 2, protein kinase A, PKC and 70 kDa form of ribosomal protein S6 kinase repress CYP expression by phosphorylating and inhibiting PXR. Growth factor signaling represses CYP expression by phosphorylating and inhibiting forkhead in rhabdomyosarcoma, a co-activator of PXR. During inflammation, NF-kappaB represses both PXR and CYP expression through protein-protein interactions with the PXR pathway.
Collapse
Affiliation(s)
- Satyanarayana R Pondugula
- St. Jude Children's Research Hospital, Department of Chemical Biology and Therapeutics, 262 Danny Thomas Place, Mail Stop 1000, Memphis, TN 38105, USA
| | | | | |
Collapse
|
46
|
Chen Y, Goldstein JA. The transcriptional regulation of the human CYP2C genes. Curr Drug Metab 2009; 10:567-78. [PMID: 19702536 PMCID: PMC2808111 DOI: 10.2174/138920009789375397] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 07/14/2009] [Indexed: 01/09/2023]
Abstract
In humans, four members of the CYP2C subfamily (CYP2C8, CYP2C9, CYP2C18, and CYP2C19) metabolize more than 20% of all therapeutic drugs as well as a number of endogenous compounds. The CYP2C enzymes are found predominantly in the liver, where they comprise approximately 20% of the total cytochrome P450. A variety of xenobiotics such as phenobarbital, rifampicin, and hyperforin have been shown to induce the transcriptional expression of CYP2C genes in primary human hepatocytes and to increase the metabolism of CYP2C substrates in vivo in man. This induction can result in drug-drug interactions, drug tolerance, and therapeutic failure. Several drug-activated nuclear receptors including CAR, PXR, VDR, and GR recognize drug responsive elements within the 5' flanking promoter region of CYP2C genes to mediate the transcriptional upregulation of these genes in response to xenobiotics and steroids. Other nuclear receptors and transcriptional factors including HNF4alpha, HNF3gamma, C/EBPalpha and more recently RORs, have been reported to regulate the constitutive expression of CYP2C genes in liver. The maximum transcriptional induction of CYP2C genes appears to be achieved through a coordinative cross-talk between drug responsive nuclear receptors, hepatic factors, and coactivators. The transcriptional regulatory mechanisms of the expression of CYP2C genes in extrahepatic tissues has received less study, but these may be altered by perturbations from pathological conditions such as ischemia as well as some of the receptors mentioned above.
Collapse
Affiliation(s)
- Yuping Chen
- Laboratory of Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | - Joyce A. Goldstein
- Laboratory of Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| |
Collapse
|
47
|
Matic M, Nakhel S, Lehnert AM, Polly P, Clarke SJ, Robertson GR. A novel approach to investigate the subcellular distribution of nuclear receptors in vivo. NUCLEAR RECEPTOR SIGNALING 2009; 7:e004. [PMID: 19471583 PMCID: PMC2686083 DOI: 10.1621/nrs.07004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 04/03/2009] [Indexed: 01/06/2023]
Abstract
Subcellular compartmentalisation and the intracellular movement of nuclear receptors are major regulatory steps in executing their transcriptional function. Though significant progress has been made in understanding these regulatory processes in cultured mammalian cells, such results have rarely been confirmed within cells of a living mammal. This article describes a simple, time-efficient approach to study the nuclear versus cytoplasmic accumulation of nuclear receptors and the regions of nuclear receptor proteins that govern subcellular trafficking within hepatocytes of live mice. Pregnane X receptor, a xenobiotic-activated member of the nuclear receptor family, was used to exemplify the approach. Using dual-labeled wild-type and mutant PXR expression constructs, we outline their in vivo delivery, simultaneous cellular expression, visualization and categorical classification within hepatocytes of live mice. Using this approach, we identified three mutants that had an altered subcellular distribution in the presence and absence of a PXR ligand. This novel in vivo method complements the current cell culture-based experimental systems in protein subcellular localisation studies.
Collapse
Affiliation(s)
- Marko Matic
- Cancer Pharmacology Unit, ANZAC Research Institute, Concord RG Hospital, Australia
| | | | | | | | | | | |
Collapse
|
48
|
di Masi A, De Marinis E, Ascenzi P, Marino M. Nuclear receptors CAR and PXR: Molecular, functional, and biomedical aspects. Mol Aspects Med 2009; 30:297-343. [PMID: 19427329 DOI: 10.1016/j.mam.2009.04.002] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 04/28/2009] [Indexed: 12/31/2022]
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors sharing a common evolutionary history and having similar sequence features at the protein level. Selective ligand(s) for some NRs is not known, therefore these NRs have been named "orphan receptors". Whenever ligands have been recognized for any of the orphan receptor, it has been categorized and grouped as "adopted" orphan receptor. This group includes the constitutive androstane receptor (CAR) and the pregnane X receptor (PXR). They function as sensors of toxic byproducts derived from endogenous metabolites and of exogenous chemicals, in order to enhance their elimination. This unique function of CAR and PXR sets them apart from the steroid hormone receptors. The broad response profile has established that CAR and PXR are xenobiotic sensors that coordinately regulate xenobiotic clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. In the past few years, research has revealed new and mostly unsuspected roles for CAR and PXR in modulating hormone, lipid, and energy homeostasis as well as cancer and liver steatosis. The purpose of this review is to highlight the structural and molecular bases of CAR and PXR impact on human health, providing information on mechanisms through which diet, chemical exposure, and environment ultimately impact health and disease.
Collapse
Affiliation(s)
- Alessandra di Masi
- Department of Biology, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | | | | | | |
Collapse
|
49
|
Ott M, Fricker G, Bauer B. Pregnane X receptor (PXR) regulates P-glycoprotein at the blood-brain barrier: functional similarities between pig and human PXR. J Pharmacol Exp Ther 2009; 329:141-9. [PMID: 19147857 DOI: 10.1124/jpet.108.149690] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pharmacotherapy of central nervous system (CNS) disorders is impaired by the drug efflux transporter, P-glycoprotein, which limits drug penetration across the blood-brain barrier into the CNS. One strategy to increase brain drug levels is to modulate P-glycoprotein regulation. This approach requires understanding of the mechanisms that control transporter expression and function. One mechanism through which P-glycoprotein is regulated is the nuclear receptor, pregnane X receptor (PXR). Xenobiotics including drugs activate PXR and induce P-glycoprotein, which potentially affects pharmacokinetics/pharmacodynamics of coadministered drugs. Because rodent models are not suitable to predict xenobiotic interactions with human PXR, in a porcine model, we studied functional similarities between pig and human PXR. We used brain capillary endothelial cells from pig to study the effect of PXR activation on P-glycoprotein. To activate PXR, we used the PXR ligands, rifampicin, hyperforin, and pregnenolone-16alpha-carbonitrile (PCN), and measured abcb1 mRNA with quantitative polymerase chain reaction, P-glycoprotein expression with Western blotting, and P-glycoprotein transport activity with a calcein assay. We provide first proof of principle that the human PXR ligands, rifampicin and hyperforin, but not the rodent PXR ligand, PCN, activate pig PXR at the blood-brain barrier and induce mRNA, protein expression, and transport activity of P-glycoprotein. Our data indicate functional similarities between human and pig PXR that suggest the pig model could be useful for predicting xenobiotic-PXR interactions in humans. Because PXR is crucial in controlling drug efflux transporters, our findings will contribute to a better understanding of the regulation of blood-brain barrier function, which could potentially have important clinical implications for the treatment of CNS disorders.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology
- Animals
- Blood-Brain Barrier/physiology
- Blotting, Western
- Bridged Bicyclo Compounds/pharmacology
- Cell Separation
- Cells, Cultured
- Endothelial Cells/drug effects
- Fluoresceins/metabolism
- Humans
- Immunohistochemistry
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phloroglucinol/analogs & derivatives
- Phloroglucinol/pharmacology
- Pregnane X Receptor
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Steroid/drug effects
- Receptors, Steroid/genetics
- Receptors, Steroid/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Rifampin/pharmacology
- Swine
- Terpenes/pharmacology
- Up-Regulation/drug effects
- Xenobiotics/pharmacology
Collapse
Affiliation(s)
- Melanie Ott
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | | | | |
Collapse
|
50
|
Zhou C, Verma S, Blumberg B. The steroid and xenobiotic receptor (SXR), beyond xenobiotic metabolism. NUCLEAR RECEPTOR SIGNALING 2009; 7:e001. [PMID: 19240808 PMCID: PMC2646121 DOI: 10.1621/nrs.07001] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 12/12/2008] [Indexed: 12/31/2022]
Abstract
The steroid and xenobiotic receptor (SXR) (also known as pregnane X receptor or PXR) is a nuclear hormone receptor activated by a diverse array of endogenous hormones, dietary steroids, pharmaceutical agents, and xenobiotic compounds. SXR has an enlarged, flexible, hydrophobic ligand binding domain (LBD) which is remarkably divergent across mammalian species and SXR exhibits considerable differences in its pharmacology among mammals. The broad response profile of SXR has led to the development of "the steroid and xenobiotic sensor hypothesis". SXR has been established as a xenobiotic sensor that coordinately regulates xenobiotic clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. In the past few years, research has revealed new and mostly unsuspected roles for SXR in modulating inflammation, bone homeostasis, vitamin D metabolism, lipid homeostasis, energy homeostasis and cancer. The identification of SXR as a xenobiotic sensor has provided an important tool for studying new mechanisms through which diet, chemical exposure, and environment ultimately impact health and disease. The discovery and pharmacological development of new PXR modulators might represent an interesting and innovative therapeutic approach to combat various diseases.
Collapse
Affiliation(s)
- Changcheng Zhou
- Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University, New York, New York, USA.
| | | | | |
Collapse
|