1
|
Dvořák Z, Li H, Mani S. Microbial Metabolites as Ligands to Xenobiotic Receptors: Chemical Mimicry as Potential Drugs of the Future. Drug Metab Dispos 2023; 51:219-227. [PMID: 36184080 PMCID: PMC9900867 DOI: 10.1124/dmd.122.000860] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/28/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023] Open
Abstract
Xenobiotic receptors, such as the pregnane X receptor, regulate multiple host physiologic pathways including xenobiotic metabolism, certain aspects of cellular metabolism, and innate immunity. These ligand-dependent nuclear factors regulate gene expression via genomic recognition of specific promoters and transcriptional activation of the gene. Natural or endogenous ligands are not commonly associated with this class of receptors; however, since these receptors are expressed in a cell-type specific manner in the liver and intestines, there has been significant recent effort to characterize microbially derived metabolites as ligands for these receptors. In general, these metabolites are thought to be weak micromolar affinity ligands. This journal anniversary minireview focuses on recent efforts to derive potentially nontoxic microbial metabolite chemical mimics that could one day be developed as drugs combating xenobiotic receptor-modifying pathophysiology. The review will include our perspective on the field and recommend certain directions for future research. SIGNIFICANCE STATEMENT: Xenobiotic receptors (XRs) regulate host drug metabolism, cellular metabolism, and immunity. Their presence in host intestines allows them to function not only as xenosensors but also as a response to the complex metabolic environment present in the intestines. Specifically, this review focuses on describing microbial metabolite-XR interactions and the translation of these findings toward discovery of novel chemical mimics as potential drugs of the future for diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Zdeněk Dvořák
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Hao Li
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
2
|
Dai Y, Luo J, Xiang E, Guo Q, He Z, Gong Z, Sun X, Kou H, Xu K, Fan C, Liu J, Qiu S, Wang Y, Wang H, Guo Y. Prenatal Exposure to Retrorsine Induces Developmental Toxicity and Hepatotoxicity of Fetal Rats in a Sex-Dependent Manner: The Role of Pregnane X Receptor Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3219-3231. [PMID: 33685126 DOI: 10.1021/acs.jafc.0c06748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are a type of natural phytotoxin that contaminate food and feed and become an environmental health risk to humans and livestock. PAs exert toxicity that requires metabolic activation by cytochrome P450 (CYP) 3A, and case reports showed that fetuses are quite susceptible to PAs toxicity. The aim of this study was to explore the characteristics of developmental toxicity and fetal hepatotoxicity induced by retrorsine (RTS, a typcial toxic PA) and the underlying mechanism. Pregnant Wistar rats were intragastrically administered with 20 mg/(kg·day) RTS from gestation day (GD) 9 to 20. Results showed that prenatal RTS exposure lowered fetal bodyweights, reduced hepatocyte numbers, and potentiated hepatic apoptosis in fetuses, particularly females. Simutaneously, RTS increased CYP3A expression and pregnane X receptor (PXR) activation in female fetal liver. We further confirmed that RTS was a PXR agonist in LO2 and HepG2 cell lines. Furthermore, agonism or antagonism of androgen receptor (AR) either induced or blocked RTS-mediated PXR activation, respectively. As a PXR agonist, RTS toxicity was exacerbated in female fetus due to the increased CYP3A induction and self-metabolism, while the inhibitory effect of AR on PXR activation reduced the susceptibility of male fetus to RTS. Our findings indicated that PXR may be a potential therapeutic target for PA toxicity.
Collapse
Affiliation(s)
- Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Jinyuan Luo
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province, China
| | - E Xiang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Qi Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Zheng He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zheng Gong
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Xiaoxiang Sun
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Hao Kou
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei Province, China
| | - Kequan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Chengpeng Fan
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
| | - Jie Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Shuaikai Qiu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Yanqing Wang
- Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei Province, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071 Hubei Province, China
| |
Collapse
|
3
|
Female-specific activation of pregnane X receptor mediates sex difference in fetal hepatotoxicity by prenatal monocrotaline exposure. Toxicol Appl Pharmacol 2020; 406:115137. [PMID: 32682830 DOI: 10.1016/j.taap.2020.115137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 01/06/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are a group of hepatic toxicant widely present in plants. Cytochrome P450 (CYP) 3A plays a key role in metabolic activation of PAs to generate electrophilic metabolites, which is the main cause of hepatotoxicity. We have previously demonstrated the sex difference in developmental toxicity and hepatotoxicity in fetal rats exposed to monocrotaline (MCT), a representative toxic PA. The aim of this study was to explore the underlying mechanism. 20 mg·kg-1·d-1 MCT was intragastrically given to pregnant Wistar rats from gestation day 9 to 20. CYP3As expression and pregnane X receptor (PXR) activation were specifically enhanced in female fetal liver. After MCT treatment, we also observed a significant increase of CYP3As expression in LO2 cells (high PXR level) or hPXR-transfected HepG2 cells (low PXR level). Employing hPXR and CYP3A4 dual-luciferase reporter gene assay, we confirmed the agonism effect of MCT on PXR-dependent transcriptional activity of CYP3A4. Agonism and antagonism of the androgen receptor (AR) either induced or blocked MCT-induced PXR activation, respectively. This study was the first report identifying that MCT served as PXR agonist to induce CYP3A expression. CYP3A induction may increase self-metabolic activation of MCT and subsequently lead to more severe hepatotoxicity in female fetus. While in male, during the intrauterine period, activated AR by testosterone secretion from developing testes represses MCT-induced PXR activation and CYP3A induction, which may partially protect male fetus from MCT-induced hepatotoxicity.
Collapse
|
4
|
Xing Y, Yan J, Niu Y. PXR: a center of transcriptional regulation in cancer. Acta Pharm Sin B 2020; 10:197-206. [PMID: 32082968 PMCID: PMC7016272 DOI: 10.1016/j.apsb.2019.06.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/10/2019] [Accepted: 06/20/2019] [Indexed: 02/05/2023] Open
Abstract
Pregnane X receptor (PXR, NR1I2) is a prototypical member of the nuclear receptor superfamily. PXR can be activated by both endobiotics and xenobiotics. As a key xenobiotic receptor, the cellular function of PXR is mostly exerted by its binding to the regulatory gene sequences in a ligand-dependent manner. Classical downstream target genes of PXR participate in xenobiotic responses, such as detoxification, metabolism and inflammation. Emerging evidence also implicates PXR signaling in the processes of apoptosis, cell cycle arrest, proliferation, angiogenesis and oxidative stress, which are closely related to cancer. Here, we discussed, in addition to the characterization of PXR per se, the biological function and regulatory mechanism of PXR signaling in cancer, and its potential for the targeted prevention and therapeutics.
Collapse
Affiliation(s)
- Yaqi Xing
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Jiong Yan
- Center for Pharmacogenetics, University of Pittsburgh, PA 15261, USA
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
- Corresponding author.
| |
Collapse
|
5
|
Luo J, Yang X, Qiu S, Li X, Xiang E, Fang Y, Wang Y, Zhang L, Wang H, Zheng J, Guo Y. Sex difference in monocrotaline-induced developmental toxicity and fetal hepatotoxicity in rats. Toxicology 2019; 418:32-40. [PMID: 30825512 DOI: 10.1016/j.tox.2019.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022]
Abstract
Pyrrolizidine alkaloids (PAs) are a class of hepatic toxins widely existing in plants. Cytochromes P450 (CYP) mediates PA bioactivation and toxicities in mammals. It has been reported that PAs can induce developmental toxicity, but systematic research is lacking. In this study, we investigated developmental toxicity of monocrotaline (MCT) in rats. Pregnant rats were administered with MCT (20 mg/kg) intragastrically from gestation day 9 to 20, followed by determination of changes in fetal growth, hepatic morphology, serum biochemical indices, and indicators of hepatocytes apoptosis. MCT was found to induce developmental toxicity and fetal hepatotoxicity, particularly in female fetuses. Metabolic activation was also studied by examination of bioactivation efficiency of MCT in fetal liver microsomes, serum MCT, pyrrole-protein adduction derived from MCT, and hepatic CYP3 A expression of fetuses in vivo. Male fetuses showed greater basal MCT bioactivation than that of female fetuses, but continuous exposure to MCT caused a selective CYP3 A induction in female fetuses, which may contribute to the sex difference in MCT-induced developmental toxicity.
Collapse
Affiliation(s)
- Jinyuan Luo
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - Xiaojing Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, People's Republic of China.
| | - Shuaikai Qiu
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - Xia Li
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - E Xiang
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - Yan Fang
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - Yanqing Wang
- Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China.
| | - Li Zhang
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China.
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, People's Republic of China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China; Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China.
| | - Yu Guo
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, People's Republic of China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, People's Republic of China.
| |
Collapse
|
6
|
Leach DA, Powell SM, Bevan CL. WOMEN IN CANCER THEMATIC REVIEW: New roles for nuclear receptors in prostate cancer. Endocr Relat Cancer 2016; 23:T85-T108. [PMID: 27645052 DOI: 10.1530/erc-16-0319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 12/20/2022]
Abstract
Prostate cancer has, for decades, been treated by inhibiting androgen signalling. This is effective in the majority of patients, but inevitably resistance develops and patients progress to life-threatening metastatic disease - hence the quest for new effective therapies for 'castrate-resistant' prostate cancer (CRPC). Studies into what pathways can drive tumour recurrence under these conditions has identified several other nuclear receptor signalling pathways as potential drivers or modulators of CRPC.The nuclear receptors constitute a large (48 members) superfamily of transcription factors sharing a common modular functional structure. Many of them are activated by the binding of small lipophilic molecules, making them potentially druggable. Even those for which no ligand exists or has yet been identified may be tractable to activity modulation by small molecules. Moreover, genomic studies have shown that in models of CRPC, other nuclear receptors can potentially drive similar transcriptional responses to the androgen receptor, while analysis of expression and sequencing databases shows disproportionately high mutation and copy number variation rates among the superfamily. Hence, the nuclear receptor superfamily is of intense interest in the drive to understand how prostate cancer recurs and how we may best treat such recurrent disease. This review aims to provide a snapshot of the current knowledge of the roles of different nuclear receptors in prostate cancer - a rapidly evolving field of research.
Collapse
Affiliation(s)
- Damien A Leach
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| | - Sue M Powell
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of CancerImperial Centre for Translational & Experimental Medicine, Imperial, College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
7
|
De Mattia E, Cecchin E, Roncato R, Toffoli G. Pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors as emerging players in cancer precision medicine. Pharmacogenomics 2016; 17:1547-71. [DOI: 10.2217/pgs-2016-0095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Great research effort has been focused on elucidating the contribution of host genetic variability on pharmacological outcomes in cancer. Nuclear receptors have emerged as mediators between environmental stimuli and drug pharmacokinetics and pharmacodynamics. The pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors have been reported to regulate transcription of genes that encode drug metabolizing enzymes and transporters. Altered nuclear receptor expression has been shown to affect the metabolism and pharmacological profile of traditional chemotherapeutics and targeted agents. Accordingly, polymorphic variants in these genes have been studied as pharmacogenetic markers of outcome variability. This review summarizes the state of knowledge about the roles played by pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factor expression and genetics as predictive markers of anticancer drug toxicity and efficacy, which can improve cancer precision medicine.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Rossana Roncato
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| |
Collapse
|
8
|
Pondugula SR, Pavek P, Mani S. Pregnane X Receptor and Cancer: Context-Specificity is Key. NUCLEAR RECEPTOR RESEARCH 2016; 3. [PMID: 27617265 DOI: 10.11131/2016/101198] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pregnane X receptor (PXR) is an adopted orphan nuclear receptor that is activated by a wide-range of endobiotics and xenobiotics, including chemotherapy drugs. PXR plays a major role in the metabolism and clearance of xenobiotics and endobiotics in liver and intestine via induction of drug-metabolizing enzymes and drug-transporting proteins. However, PXR is expressed in several cancer tissues and the accumulating evidence strongly points to the differential role of PXR in cancer growth and progression as well as in chemotherapy outcome. In cancer cells, besides regulating the gene expression of enzymes and proteins involved in drug metabolism and transport, PXR also regulates other genes involved in proliferation, metastasis, apoptosis, anti-apoptosis, inflammation, and oxidative stress. In this review, we focus on the differential role of PXR in a variety of cancers, including prostate, breast, ovarian, endometrial, and colon. We also discuss the future directions to further understand the differential role of PXR in cancer, and conclude with the need to identify novel selective PXR modulators to target PXR in PXR-expressing cancers.
Collapse
Affiliation(s)
- Satyanarayana R Pondugula
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL 36849, USA; Auburn University Research Initiative in Cancer, Auburn University, Auburn, AL 36849, USA
| | - Petr Pavek
- Faculty of Pharmacy in Hradec Králové, Charles University in Prague, Heyrovského 1203, Hradec Králové 500 05, Czech Republic, European Union
| | - Sridhar Mani
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
9
|
Cecchin E, De Mattia E, Toffoli G. Nuclear receptors and drug metabolism for the personalization of cancer therapy. Expert Opin Drug Metab Toxicol 2016; 12:291-306. [DOI: 10.1517/17425255.2016.1141196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| |
Collapse
|
10
|
Kotiya D, Rana M, Subbarao N, Puri N, Tyagi RK. Transcription regulation of nuclear receptor PXR: Role of SUMO-1 modification and NDSM in receptor function. Mol Cell Endocrinol 2016; 420:194-207. [PMID: 26549688 DOI: 10.1016/j.mce.2015.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 11/01/2015] [Accepted: 11/01/2015] [Indexed: 01/13/2023]
Abstract
Pregnane & Xenobiotic Receptor (PXR) is one of the 48 members of the nuclear receptor superfamily of ligand-modulated transcription factors. PXR plays an important role in metabolism and elimination of diverse noxious endobiotics and xenobiotics. Like in case of some nuclear receptors its function may also be differentially altered, positively or negatively, by various post-translational modifications. In this context, regulation of PXR function by SUMOylation is the subject of present investigation. Here, we report that human PXR is modified by SUMO-1 resulting in its enhanced transcriptional activity. RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. In addition, mammalian two-hybrid assay exhibited enhanced interaction between PXR and co-activator SRC-1. EMSA results revealed that SUMOylation has no influence on the DNA binding ability of PXR. In silico analysis suggested that PXR protein contains four putative SUMOylation sites, centered at K108, K129, K160 and K170. In addition to this, we identified the presence of NDSM (Negative charge amino acid Dependent SUMOylation Motif) in PXR. Substitution of all its four putative lysine residues along with NDSM abolished the effect of SUMO-1-mediated transactivation function of PXR. Furthermore, we show that interaction between PXR and E2-conjugation enzyme UBCh9, an important step for implementation of SUMOylation event, was reduced in case of NDSM mutant PXRD115A. Overall, our results suggest that SUMOylation at specific sites on PXR protein are involved in enhancement of transcription function of this receptor.
Collapse
Affiliation(s)
- Deepak Kotiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Manjul Rana
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - N Subbarao
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
11
|
The tumor suppressor TERE1 (UBIAD1) prenyltransferase regulates the elevated cholesterol phenotype in castration resistant prostate cancer by controlling a program of ligand dependent SXR target genes. Oncotarget 2014; 4:1075-92. [PMID: 23919967 PMCID: PMC3759667 DOI: 10.18632/oncotarget.1103] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Castrate-Resistant Prostate Cancer (CRPC) is characterized by persistent androgen receptor-driven tumor growth in the apparent absence of systemic androgens. Current evidence suggests that CRPC cells can produce their own androgens from endogenous sterol precursors that act in an intracrine manner to stimulate tumor growth. The mechanisms by which CRPC cells become steroidogenic during tumor progression are not well defined. Herein we describe a novel link between the elevated cholesterol phenotype of CRPC and the TERE1 tumor suppressor protein, a prenyltransferase that synthesizes vitamin K-2, which is a potent endogenous ligand for the SXR nuclear hormone receptor. We show that 50% of primary and metastatic prostate cancer specimens exhibit a loss of TERE1 expression and we establish a correlation between TERE1 expression and cholesterol in the LnCaP-C81 steroidogenic cell model of the CRPC. LnCaP-C81 cells also lack TERE1 protein, and show elevated cholesterol synthetic rates, higher steady state levels of cholesterol, and increased expression of enzymes in the de novo cholesterol biosynthetic pathways than the non-steroidogenic prostate cancer cells. C81 cells also show decreased expression of the SXR nuclear hormone receptor and a panel of directly regulated SXR target genes that govern cholesterol efflux and steroid catabolism. Thus, a combination of increased synthesis, along with decreased efflux and catabolism likely underlies the CRPC phenotype: SXR might coordinately regulate this phenotype. Moreover, TERE1 controls synthesis of vitamin K-2, which is a potent endogenous ligand for SXR activation, strongly suggesting a link between TERE1 levels, K-2 synthesis and SXR target gene regulation. We demonstrate that following ectopic TERE1 expression or induction of endogenous TERE1, the elevated cholesterol levels in C81 cells are reduced. Moreover, reconstitution of TERE1 expression in C81 cells reactivates SXR and switches on a suite of SXR target genes that coordinately promote both cholesterol efflux and androgen catabolism. Thus, loss of TERE1 during tumor progression reduces K-2 levels resulting in reduced transcription of SXR target genes. We propose that TERE1 controls the CPRC phenotype by regulating the endogenous levels of Vitamin K-2 and hence the transcriptional control of a suite of steroidogenic genes via the SXR receptor. These data implicate the TERE1 protein as a previously unrecognized link affecting cholesterol and androgen accumulation that could govern acquisition of the CRPC phenotype.
Collapse
|
12
|
Poutiainen PK, Huhtala T, Jääskeläinen T, Petsalo A, Küblbeck J, Kaikkonen S, Palvimo JJ, Raunio H, Närvänen A, Peräkylä M, Juvonen RO, Honkakoski P, Laatikainen R, Pulkkinen JT. Preclinical pharmacology of FL442, a novel nonsteroidal androgen receptor modulator. Mol Cell Endocrinol 2014; 387:8-18. [PMID: 24565895 DOI: 10.1016/j.mce.2014.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/14/2014] [Accepted: 02/15/2014] [Indexed: 11/29/2022]
Abstract
The preclinical profiles of two most potent compounds of our recently published cycloalkane[d]isoxazole pharmacophore-based androgen receptor (AR) modulators, FL442 (4-(3a,4,5,6,7,7a-hexahydro-benzo[d]isoxazol-3-yl)-2-(trifluoromethyl)benzonitrile) and its nitro analog FL425 (3-(4-nitro-3-(trifluoromethyl)phenyl)-3a,4,5,6,7,7a-hexahydrobenzo[d]isoxazole), were explored to evaluate their druggability for the treatment of AR dependent prostate cancer. The studies revealed that both compounds are selective to AR over other closely related steroid hormone receptors and that FL442 exhibits equal inhibition efficiency towards the androgen-responsive LNCaP prostate cancer cell line as the most widely used antiandrogen bicalutamide and the more recently discovered enzalutamide. Notably, FL442 maintains antiandrogenic activity with enzalutamide-activated AR mutant F876L. In contrast to bicalutamide, FL442 does not stimulate the VCaP prostate cancer cells which express elevated levels of the AR. Distribution analyses showed that [(14)CN]FL442 accumulates strongly in the mouse prostate. In spite of its low plasma concentration obtained by intraperitoneal administration, FL442 significantly inhibited LNCaP xenograft tumor growth. These findings provide a preclinical proof for FL442 as a promising AR targeted candidate for a further optimization.
Collapse
Affiliation(s)
- Pekka K Poutiainen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Tuulia Huhtala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Tiina Jääskeläinen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Institute of Dentistry, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Aleksanteri Petsalo
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, P.O. Box 5000, FI-90014, Finland
| | - Jenni Küblbeck
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Sanna Kaikkonen
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Hannu Raunio
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ale Närvänen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Mikael Peräkylä
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Risto O Juvonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Reino Laatikainen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Juha T Pulkkinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
13
|
De Mattia E, Dreussi E, Cecchin E, Toffoli G. Pharmacogenetics of the nuclear hormone receptors: the missing link between environment and drug effects? Pharmacogenomics 2013; 14:2035-54. [DOI: 10.2217/pgs.13.214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In the last decade, genetic variations in ABC/SLC transporters and phase I/II enzymes have raised pharmacogenetic markers as being predictive to the attention of researchers in the field of personalized medicine in oncology. However, it is becoming evident that the sequence variations in these genes cannot address by themselves the sharp interindividual variability in drug effects. Recently, nuclear receptors (NRs), including pregnane X receptor, constitutive androstane receptor, retinoid X receptor, farnesoid X receptor, liver X receptor, vitamin D receptor, peroxisome proliferator-activated receptors and HNF4A, have demonstrated key roles in regulating transporter and metabolic gene expression in response to xeno/endobiotics, as well as antineoplastic drugs. These findings attracted interest to the genetics of the NRs for their possible role in influencing the metabolism and pharmacological profiles of chemotherapeutics. In this review, we aim to summarize the most recent findings in the innovative field of NR pharmacogenetics and findings in how they could integrate with more traditional markers in order to improve drug treatment personalization.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Eva Dreussi
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico–National Cancer Institute, Via Franco Gallini, 2, 33081, Aviano, Italy
| |
Collapse
|
14
|
Pregnane xenobiotic receptor in cancer pathogenesis and therapeutic response. Cancer Lett 2012; 328:1-9. [PMID: 22939994 DOI: 10.1016/j.canlet.2012.08.030] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/20/2012] [Accepted: 08/22/2012] [Indexed: 01/24/2023]
Abstract
Pregnane xenobiotic receptor (PXR) is an orphan nuclear receptor that regulates the metabolism of endobiotics and xenobiotics. PXR is promiscuous and unique in that it is activated by a diverse group of xenochemicals, including therapeutic anticancer drugs and naturally-occurring endocrine disruptors. PXR has been predominantly studied to understand its regulatory role in xenobiotic clearance in liver and intestine via induction of drug metabolizing enzymes and drug transporters. PXR, however, is widely expressed and has functional implications in other normal and malignant tissues, including breast, prostate, ovary, endometrium and bone. The differential expression of PXR and its target genes in cancer tissues has been suggested to determine the prognosis of chemotherapeutic outcome. In addition, the emerging evidence points to the implications of PXR in regulating apoptotic and antiapoptotic as well as growth factor signaling that promote tumor proliferation and metastasis. In this review, we highlight the recent progress made in understanding the role of PXR in cancer, discuss the future directions to further understand the mechanistic role of PXR in cancer, and conclude with the need to identify novel selective PXR modulators.
Collapse
|
15
|
Chaturvedi NK, Kumar S, Negi S, Tyagi RK. Endocrine disruptors provoke differential modulatory responses on androgen receptor and pregnane and xenobiotic receptor: potential implications in metabolic disorders. Mol Cell Biochem 2010; 345:291-308. [PMID: 20830510 DOI: 10.1007/s11010-010-0583-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 08/28/2010] [Indexed: 12/21/2022]
Abstract
A systematic comparison of the impact of some potential endocrine disruptors (EDs) on modulation of androgen receptor (AR) and pregnane and xenobiotic receptor (PXR) function was conducted in a multi-step analysis. Promoter-reporter-based transcription assays were performed in conjunction with receptor dynamic studies in living cells that helped implicating the suspected EDs for their deleterious effects. We demonstrate that most of the selected EDs not only inhibit AR transcriptional activity, but also alter its subcellular dynamics. Conversely, some of these anti-androgenic compounds were potent activator of xeno-sensing nuclear receptor, PXR. Interestingly, agonist-activated AR that associates with the mitotic chromatin fails to achieve this association when bound to anti-androgenic EDs. Conclusively, most EDs (except BCH) behaved like pure antagonist for AR while as agonist for PXR. Subsequent experiments with DDT treatment in mice model indicated that in testis AR and its regulated genes PEM and ODC levels are down-regulated, whereas in liver of same mice PEM is up-regulated while AR and ODC remain unchanged. On the contrary, PXR and its regulated genes CYP3A11 and MDR1 levels in mice liver were up-regulated while in testis PXR remained unchanged, CYP3A11 up-regulated and MDR1 were down-regulated. Based on a novel "Biopit" concept it is speculated that long-term exposure to endocrine disrupting chemicals may influence the epigenetic profile of target cells via transcription factors thereby making them vulnerable to onset of chemically induced endocrine-related malignancies or metabolic disorders.
Collapse
|