1
|
Ofosu-Boateng M, Shaik F, Choi S, Ekuban FA, Gebreyesus LH, Twum E, Nnamani D, Yeyeodu ST, Yadak N, Collier DM, Gyamfi MA. High-fat diet induced obesity promotes inflammation, oxidative stress, and hepatotoxicity in female FVB/N mice. Biofactors 2024; 50:572-591. [PMID: 38183321 PMCID: PMC11178471 DOI: 10.1002/biof.2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/04/2023] [Indexed: 01/08/2024]
Abstract
Although obesity and subsequent liver injury are increasingly prevalent in women, female mouse models have generally shown resistance to high-fat diet (HFD)-induced obesity. We evaluated control and HFD-fed male and female FVB/N mice, a strain well-suited to transgenic analyses, for phenotypic, histological, and molecular markers related to control of glucose, lipids, and inflammation in serum, liver, and perigonadal white adipose tissues. Unlike many mouse models, HFD-fed FVB/N females gained more perigonadal and mesenteric fat mass and overall body weight than their male counterparts, with increased hepatic expression of lipogenic PPARγ target genes (Cd36, Fsp27, and Fsp27β), oxidative stress genes and protein (Nqo1 and CYP2E1), inflammatory gene (Mip-2), and the pro-fibrotic gene Pai-1, along with increases in malondialdehyde and serum ALT levels. Further, inherent to females (independently of HFD), hepatic antioxidant heme oxygenase-1 (HMOX1, HO-1) protein levels were reduced compared to their male counterparts. In contrast, males may have been relatively protected from HFD-induced oxidative stress and liver injury by elevated mRNA and protein levels of hepatic antioxidants BHMT and Gpx2, increased fatty acid oxidation genes in liver and adipocytes (Pparδ), despite disorganized and inflamed adipocytes. Thus, female FVB/N mice offer a valuable preclinical, genetically malleable model that recapitulates many of the features of diet-induced obesity and liver damage observed in human females.
Collapse
Affiliation(s)
- Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Fathima Shaik
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
| | - Frederick A. Ekuban
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Lidya H. Gebreyesus
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Elizabeth Twum
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Daniel Nnamani
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Susan T. Yeyeodu
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
- Charles River Discovery Services, Durham, NC, 27709
| | - Nour Yadak
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Daniel M. Collier
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Maxwell A. Gyamfi
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
| |
Collapse
|
2
|
Wang F, Liu J, Hernandez R, Park SH, Lai YJ, Wang S, Blumberg B, Zhou C. Adipocyte-Derived PXR Signaling Is Dispensable for Diet-Induced Obesity and Metabolic Disorders in Mice. Drug Metab Dispos 2023; 51:1207-1215. [PMID: 37230767 PMCID: PMC10449100 DOI: 10.1124/dmd.123.001311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/21/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Pregnane X receptor (PXR) is a xenobiotic receptor that can be activated by numerous chemicals including endogenous hormones, dietary steroids, pharmaceutical agents, and environmental chemicals. PXR has been established to function as a xenobiotic sensor to coordinately regulate xenobiotic metabolism by regulating the expression of many enzymes and transporters required for xenobiotic metabolism. Recent studies have implicated a potentially important role for PXR in obesity and metabolic disease beyond xenobiotic metabolism, but how PXR action in different tissues or cell types contributes to obesity and metabolic disorders remains elusive. To investigate the role of adipocyte PXR in obesity, we generated a novel adipocyte-specific PXR deficient mouse model (PXRΔAd). Notably, we found that loss of adipocyte PXR did not affect food intake, energy expenditure, and obesity in high-fat diet-fed male mice. PXRΔAd mice also had similar obesity-associated metabolic disorders including insulin resistance and hepatic steatosis as control littermates. PXR deficiency in adipocytes did not affect expression of key adipose genes in PXRΔAd mice. Our findings suggest that adipocyte PXR signaling may be dispensable in diet-induced obesity and metabolic disorders in mice. Further studies are needed to understand the role of PXR signaling in obesity and metabolic disorders in the future. SIGNIFICANCE STATEMENT: The authors demonstrate that deficiency of adipocyte pregnane X receptor (PXR) does not affect diet-induced obesity or metabolic disorders in mice and infers that adipocyte PXR signaling may not play a key role in diet-induced obesity. More studies are needed to understand the tissue-specific role of PXR in obesity.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Jingwei Liu
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Rebecca Hernandez
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Ying-Jing Lai
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Bruce Blumberg
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky (F.W., S.-H.P., S.W.); Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California (J.L., R.H., Y.-J.L., C.Z.); and Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California (B.B.)
| |
Collapse
|
3
|
Krøyer Rasmussen M, Thøgersen R, Horsbøl Lindholm P, Bertram HC, Pilegaard H. Hepatic PGC-1α has minor regulatory effect on the transcriptome and metabolome during high fat high fructose diet and exercise. Gene 2022; 851:147039. [DOI: 10.1016/j.gene.2022.147039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
|
4
|
Dutta M, Lim JJ, Cui JY. Pregnane X Receptor and the Gut-Liver Axis: A Recent Update. Drug Metab Dispos 2022; 50:478-491. [PMID: 34862253 PMCID: PMC11022899 DOI: 10.1124/dmd.121.000415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023] Open
Abstract
It is well-known that the pregnane X receptor (PXR)/Nr1i2 is a critical xenobiotic-sensing nuclear receptor enriched in liver and intestine and is responsible for drug-drug interactions, due to its versatile ligand binding domain (LBD) and target genes involved in xenobiotic biotransformation. PXR can be modulated by various xenobiotics including pharmaceuticals, nutraceuticals, dietary factors, and environmental chemicals. Microbial metabolites such as certain secondary bile acids (BAs) and the tryptophan metabolite indole-3-propionic acid (IPA) are endogenous PXR activators. Gut microbiome is increasingly recognized as an important regulator for host xenobiotic biotransformation and intermediary metabolism. PXR regulates and is regulated by the gut-liver axis. This review summarizes recent research advancements leveraging pharmaco- and toxico-metagenomic approaches that have redefined the previous understanding of PXR. Key topics covered in this review include: (1) genome-wide investigations on novel PXR-target genes, novel PXR-DNA interaction patterns, and novel PXR-targeted intestinal bacteria; (2) key PXR-modulating activators and suppressors of exogenous and endogenous sources; (3) novel bidirectional interactions between PXR and gut microbiome under physiologic, pathophysiological, pharmacological, and toxicological conditions; and (4) modifying factors of PXR-signaling including species and sex differences and time (age, critical windows of exposure, and circadian rhythm). The review also discusses critical knowledge gaps and important future research topics centering around PXR. SIGNIFICANCE STATEMENT: This review summarizes recent research advancements leveraging O'mics approaches that have redefined the previous understanding of the xenobiotic-sensing nuclear receptor pregnane X receptor (PXR). Key topics include: (1) genome-wide investigations on novel PXR-targeted host genes and intestinal bacteria as well as novel PXR-DNA interaction patterns; (2) key PXR modulators including microbial metabolites under physiological, pathophysiological, pharmacological, and toxicological conditions; and (3) modifying factors including species, sex, and time.
Collapse
Affiliation(s)
- Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
5
|
Kim S, Choi S, Dutta M, Asubonteng JO, Polunas M, Goedken M, Gonzalez FJ, Cui JY, Gyamfi MA. Pregnane X receptor exacerbates nonalcoholic fatty liver disease accompanied by obesity- and inflammation-prone gut microbiome signature. Biochem Pharmacol 2021; 193:114698. [PMID: 34303710 PMCID: PMC9135326 DOI: 10.1016/j.bcp.2021.114698] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease due to the current epidemics of obesity and diabetes. The pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor known for trans-activating liver genes involved in drug metabolism and transport, and more recently implicated in energy metabolism. The gut microbiota can modulate the host xenobiotic biotransformation and contribute to the development of obesity. While the male sex confers a higher risk for NAFLD than women before menopause, the mechanism remains unknown. We hypothesized that the presence of PXR promotes obesity by modifying the gut-liver axis in a sex-specific manner. Male and female C57BL/6 (wild-type/WT) and PXR-knockout (PXR-KO) mice were fed control or high-fat diet (HFD) for 16-weeks. Serum parameters, liver histopathology, transcriptomic profiling, 16S-rDNA sequencing, and bile acid (BA) metabolomics were performed. PXR enhanced HFD-induced weight gain, hepatic steatosis and inflammation especially in males, accompanied by PXR-dependent up-regulation in hepatic genes involved in microbial response, inflammation, oxidative stress, and cancer; PXR-dependent increase in intestinal Firmicutes/Bacteroides ratio (hallmark of obesity) and the pro-inflammatory Lactobacillus, as well as a decrease in the anti-obese Allobaculum and the anti-inflammatory Bifidobacterum, with a PXR-dependent reduction of beneficial BAs in liver. The resistance to NAFLD in females may be explained by PXR-dependent decrease in pro-inflammatory bacteria (Ruminococcus gnavus and Peptococcaceae). In conclusion, PXR exacerbates hepatic steatosis and inflammation accompanied by obesity- and inflammation-prone gut microbiome signature, suggesting that gut microbiome may contribute to PXR-mediated exacerbation of NAFLD.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jeffrey O Asubonteng
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Marianne Polunas
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Michael Goedken
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
6
|
Hukkanen J, Hakkola J. PXR and 4β-Hydroxycholesterol Axis and the Components of Metabolic Syndrome. Cells 2020; 9:cells9112445. [PMID: 33182477 PMCID: PMC7696146 DOI: 10.3390/cells9112445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 01/10/2023] Open
Abstract
Pregnane X receptor (PXR) activation has been found to regulate glucose and lipid metabolism and affect obesity in response to high-fat diets. PXR also modulates vascular tone. In fact, PXR appears to regulate multiple components of metabolic syndrome. In most cases, the effect of PXR action is harmful to metabolic health, and PXR can be hypothesized to play an important role in metabolic disruption elicited by exposure to endocrine-disrupting chemicals. The majority of the data on the effects of PXR activation on metabolic health come from animal and cell culture experiments. However, randomized, placebo-controlled, human trials indicate that the treatment with PXR ligands impairs glucose tolerance and increases 24-h blood pressure and heart rate. In addition, plasma 4β-hydroxycholesterol (4βHC), formed under the control of PXR in the liver, is associated with lower blood pressure in healthy volunteers. Furthermore, 4βHC regulates cholesterol transporters in peripheral tissues and may activate the beneficial reverse HDL cholesterol transport. In this review, we discuss the current knowledge on the role of PXR and the PXR–4βHC axis in the regulation of components of metabolic syndrome.
Collapse
Affiliation(s)
- Janne Hukkanen
- Research Unit of Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, POB 5000, FI-90014 Oulu, Finland
- Correspondence: (J.H.); (J.H.); Tel.: +358-8-3156212 (J.H.); +358-294-485235 (J.H.)
| | - Jukka Hakkola
- Research Unit of Biomedicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, POB 5000, FI-90014 Oulu, Finland
- Correspondence: (J.H.); (J.H.); Tel.: +358-8-3156212 (J.H.); +358-294-485235 (J.H.)
| |
Collapse
|
7
|
Sfera A, Osorio C, Diaz EL, Maguire G, Cummings M. The Other Obesity Epidemic-Of Drugs and Bugs. Front Endocrinol (Lausanne) 2020; 11:488. [PMID: 32849279 PMCID: PMC7411001 DOI: 10.3389/fendo.2020.00488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic psychiatric patients with schizophrenia and related disorders are frequently treatment-resistant and may require higher doses of psychotropic drugs to remain stable. Prolonged exposure to these agents increases the risk of weight gain and cardiometabolic disorders, leading to poorer outcomes and higher medical cost. It is well-established that obesity has reached epidemic proportions throughout the world, however it is less known that its rates are two to three times higher in mentally ill patients compared to the general population. Psychotropic drugs have emerged as a major cause of weight gain, pointing to an urgent need for novel interventions to attenuate this unintended consequence. Recently, the gut microbial community has been linked to psychotropic drugs-induced obesity as these agents were found to possess antimicrobial properties and trigger intestinal dysbiosis, depleting Bacteroidetes phylum. Since germ-free animals exposed to psychotropics have not demonstrated weight gain, altered commensal flora composition is believed to be necessary and sufficient to induce dysmetabolism. Conversely, not only do psychotropics disrupt the composition of gut microbiota but the later alter the metabolism of the former. Here we review the role of gut bacterial community in psychotropic drugs metabolism and dysbiosis. We discuss potential biomarkers reflecting the status of Bacteroidetes phylum and take a closer look at nutritional interventions, fecal microbiota transplantation, and transcranial magnetic stimulation, strategies that may lower obesity rates in chronic psychiatric patients.
Collapse
Affiliation(s)
- Adonis Sfera
- Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
- *Correspondence: Adonis Sfera
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Eddie Lee Diaz
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Gerald Maguire
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | - Michael Cummings
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| |
Collapse
|
8
|
Hepatic PGC-1α is not essential for fasting-induced cytochrome p450 regulation in mouse liver. Biochem Pharmacol 2019; 172:113736. [PMID: 31786263 DOI: 10.1016/j.bcp.2019.113736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022]
Abstract
Fasting has been shown to regulate the expression of the cytochrome p450 (CYP) enzyme system in the liver. However, the exact mechanism behind the fasting-induced regulation of the CYP's remains unknown. In the present study we tested the hypothesis that the peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), which is a key-regulator of energy metabolism, is responsible for the fasting-induced regulation of the CYP's. Lox/lox and liver specific PGC-1α (LKO) mice of both sexes, fasted for 18 h and the content of the CYP's as well as the hepatic metabolome was assessed. Fasting increased the mRNA content of Cyp2a4, Cyp2e1, Cyp3a11 and Cyp4a10. The fasting-induced response in Cyp4a10 mRNA content was different between lox/lox and LKO mice, while the absence of PGC-1α had no effect on the fasting-induced response for the other Cyp's. Moreover, the fasting-induced response in mRNA content of Sirtinus 1 and Perilipin 2 was different between lox/lox and LKO mice. Only the CYP1A isoform showed a fasting-induced response at the protein level. Absence of hepatic PGC-1α had no effect on the apparent metabolome, where fasting vs fed was the only discriminate in the following multivariate analysis. In conclusion, hepatic PGC-1α is not essential for the fasting-induced regulation of hepatic CYP's.
Collapse
|
9
|
Barretto SA, Lasserre F, Fougerat A, Smith L, Fougeray T, Lukowicz C, Polizzi A, Smati S, Régnier M, Naylies C, Bétoulières C, Lippi Y, Guillou H, Loiseau N, Gamet-Payrastre L, Mselli-Lakhal L, Ellero-Simatos S. Gene Expression Profiling Reveals that PXR Activation Inhibits Hepatic PPARα Activity and Decreases FGF21 Secretion in Male C57Bl6/J Mice. Int J Mol Sci 2019; 20:ijms20153767. [PMID: 31374856 PMCID: PMC6696478 DOI: 10.3390/ijms20153767] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 01/25/2023] Open
Abstract
The pregnane X receptor (PXR) is the main nuclear receptor regulating the expression of xenobiotic-metabolizing enzymes and is highly expressed in the liver and intestine. Recent studies have highlighted its additional role in lipid homeostasis, however, the mechanisms of these regulations are not fully elucidated. We investigated the transcriptomic signature of PXR activation in the liver of adult wild-type vs. Pxr-/- C57Bl6/J male mice treated with the rodent specific ligand pregnenolone 16α-carbonitrile (PCN). PXR activation increased liver triglyceride accumulation and significantly regulated the expression of 1215 genes, mostly xenobiotic-metabolizing enzymes. Among the down-regulated genes, we identified a strong peroxisome proliferator-activated receptor α (PPARα) signature. Comparison of this signature with a list of fasting-induced PPARα target genes confirmed that PXR activation decreased the expression of more than 25 PPARα target genes, among which was the hepatokine fibroblast growth factor 21 (Fgf21). PXR activation abolished plasmatic levels of FGF21. We provide a comprehensive signature of PXR activation in the liver and identify new PXR target genes that might be involved in the steatogenic effect of PXR. Moreover, we show that PXR activation down-regulates hepatic PPARα activity and FGF21 circulation, which could participate in the pleiotropic role of PXR in energy homeostasis.
Collapse
Affiliation(s)
- Sharon Ann Barretto
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Frédéric Lasserre
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Anne Fougerat
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Lorraine Smith
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Tiffany Fougeray
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Céline Lukowicz
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Arnaud Polizzi
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Sarra Smati
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Marion Régnier
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Claire Naylies
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Colette Bétoulières
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Yannick Lippi
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Laurence Gamet-Payrastre
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Laila Mselli-Lakhal
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France
| | - Sandrine Ellero-Simatos
- Institut National de la Recherche Agronomique (INRA), UMR1331 Toxalim, F31-027 Toulouse CEDEX 3, France.
| |
Collapse
|
10
|
The Sex-Gender Effects in the Road to Tailored Botanicals. Nutrients 2019; 11:nu11071637. [PMID: 31319627 PMCID: PMC6682902 DOI: 10.3390/nu11071637] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022] Open
Abstract
Phenols are a wide family of phytochemicals that are characterized by large chemical diversity and are considered to bioactive molecules of foods, beverages, and botanicals. Although they have a multitude of biological actions, their beneficial effects are rarely evidenced in clinical research with high scientific rigor. This may occur due to the presence of numerous confounders, such as the modulation of phenol bioavailability, which can be regulated by microbiota, age, sex-gender. Sex-gender is an important determinant of health and well-being, and has an impact on environmental and occupational risks, access to health care, disease prevalence, and treatment outcomes. In addition, xenobiotic responses may be strongly influenced by sex-gender. This review describes how sex–gender differentially influences the activities of phenols also in some critical periods of women life such as pregnancy and lactation, considering also the sex of fetuses and infants. Thus, sex–gender is a variable that must be carefully considered and should be used to propose directions for future research on the road to tailored medicine and nutrition.
Collapse
|
11
|
Zeng H, Lin Y, Gong J, Lin S, Gao J, Li C, Feng Z, Zhang H, Zhang J, Li Y, Yu C. CYP3A suppression during diet-induced nonalcoholic fatty liver disease is independent of PXR regulation. Chem Biol Interact 2019; 308:185-193. [PMID: 31132328 DOI: 10.1016/j.cbi.2019.05.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/19/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Cytochrome P450 3A (CYP3A) activity is inhibited, and its expression is suppressed during many diseases, including nonalcoholic fatty liver disease (NAFLD). However, the mechanism is controversial. Here, we report that PXR may not take part in the downregulation of CYP3A during NAFLD. Hepatic CYP3A11 (major subtype of mouse CYP3A) mRNA and protein expression was significantly decreased in both mice fed a high-fat diet (HFD) for 8 weeks and palmitate (PA)-treated mouse primary hepatocytes. Similarly, in HepG2 cells, PA treatment significantly suppressed the CYP3A4 (major subtype of human CYP3A) mRNA level and promoter transcription activity. However, Western blotting analysis found an induction of PXR nuclear translocation during NAFLD in both in vivo and in vitro models. Moreover, immunofluorescence determination also found nuclear translocation effect of PXR by PA stimulation in HepG2 cells. In addition, the siRNA knockdown of PXR did not affect the suppressive effects of PA on the CYP3A4 promoter transcription activity and mRNA levels in HepG2 cells. Similarly, PXR knockdown also did not affect the suppressive effects of PA on CYP3A11 mRNA and protein expression levels in mouse primary hepatoctyes. Taken together, the results showed that the suppressive effect of CYP3A transcription was independent of PXR regulation.
Collapse
Affiliation(s)
- Hang Zeng
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yiming Lin
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jiande Gong
- Department of Gastroenterology, Yinzhou People's Hospital, Ningbo, 315040, China
| | - Sisi Lin
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Jianguo Gao
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Chunxiao Li
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zemin Feng
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hong Zhang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jie Zhang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Youming Li
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
12
|
Heintz MM, Kumar R, Rutledge MM, Baldwin WS. Cyp2b-null male mice are susceptible to diet-induced obesity and perturbations in lipid homeostasis. J Nutr Biochem 2019; 70:125-137. [PMID: 31202118 DOI: 10.1016/j.jnutbio.2019.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/26/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Obesity is an endemic problem in the United States and elsewhere, and data indicate that in addition to overconsumption, exposure to specific chemicals enhances obesity. CYP2B metabolizes multiple endo- and xenobiotics, and recent data suggests that repression of Cyp2b activity increases dyslipidemia and age-onset obesity, especially in males. To investigate the role played by Cyp2b in lipid homeostasis and obesity, we treated wildtype and Cyp2b-null mice with a normal (ND) or 60% high-fat diet (HFD) for 10 weeks and determined metabolic and molecular changes. Male HFD-fed Cyp2b-null mice weigh 15% more than HFD-fed wildtype mice, primarily due to an increase in white adipose tissue (WAT); however, Cyp2b-null female mice did not demonstrate greater body mass or WAT. Serum parameters indicate increased ketosis, leptin and cholesterol in HFD-fed Cyp2b-null male mice compared to HFD-fed wildtype mice. Liver triglycerides and liver:serum triglyceride ratios were higher than their similarly treated wildtype counterparts in Cyp2b-null male mice, indicating a role for Cyp2b in fatty acid metabolism regardless of diet. Furthermore, RNAseq demonstrates that hepatic gene expression in ND-fed Cyp2b-null male mice is similar to HFD-fed WT male mice, suggestive of fatty liver disease progression and a role for Cyp2b in lipid homeostasis. Females did not show as demonstrative changes in liver health, and significantly fewer changes in gene expression, as well as gene expression associated with liver disease. Overall our data indicates that the repression or inhibition of CYP2B may exacerbate metabolic disorders and cause obesity by perturbing fatty acid metabolism, especially in males.
Collapse
Affiliation(s)
- Melissa M Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634
| | - Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC 29634
| | | | - William S Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634; Biological Sciences, Clemson University, Clemson, SC 29634.
| |
Collapse
|
13
|
Knebel C, Buhrke T, Süssmuth R, Lampen A, Marx-Stoelting P, Braeuning A. Pregnane X receptor mediates steatotic effects of propiconazole and tebuconazole in human liver cell lines. Arch Toxicol 2019; 93:1311-1322. [PMID: 30989312 DOI: 10.1007/s00204-019-02445-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022]
Abstract
Triazoles are commonly used fungicides which show liver toxicity in rodent studies. While hepatocellular hypertrophy is the most prominent finding, some triazoles have also been reported to cause hepatocellular steatosis. The aim of our study was to elucidate molecular mechanisms of triazole-mediated steatosis. Therefore, we used the two triazoles propiconazole (Pi) and tebuconazole (Te) as test compounds in in vitro assays using the human hepatocarcinoma cell lines HepG2 and HepaRG. Triglyceride accumulation was measured using the Adipored assay and by a gas-chromatographic method. Reporter gene analyses were used to assess the ability of Pi and Te to activate nuclear receptors, which are described as the molecular initiators in the adverse outcome pathway (AOP) for liver steatosis. The expression of steatosis-associated genes was investigated by RT-PCR. Mechanistic analyses of triazole-mediated steatosis were performed using HepaRG subclones that are deficient in different nuclear receptors. Pi and Te both interacted with the constitutive androstane receptor (CAR), the peroxisome proliferator-activated receptor alpha (PPARα), and the pregnane X receptor (PXR). Both compounds induced expression of steatosis-related genes and cellular triglyceride accumulation. The knockout of PXR in HepaRG cells, but not the CAR knockout, abolished triazole-induced triglyceride accumulation, thus underlining the crucial role of PXR in hepatic steatosis resulting from exposure to these fungicides. In conclusion, our findings provide new insight into the molecular mechanisms of steatosis induction by triazole fungicides and identify PXR as a critical mediator of this process.
Collapse
Affiliation(s)
- Constanze Knebel
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Thorsten Buhrke
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Roderich Süssmuth
- Institute of Chemistry, Technical University Berlin, Straße des 17.Juni 124, 10623, Berlin, Germany
| | - Alfonso Lampen
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|
14
|
Knudsen JG, Bertholdt L, Gudiksen A, Gerbal-Chaloin S, Rasmussen MK. Skeletal Muscle Interleukin-6 Regulates Hepatic Cytochrome P450 Expression: Effects of 16-Week High-Fat Diet and Exercise. Toxicol Sci 2019; 162:309-317. [PMID: 29177473 DOI: 10.1093/toxsci/kfx258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
High-fat diet (HFD) induces several changes to the pathways regulating energy homeostasis and changes the expression of the hepatic cytochrome p450 (Cyp) enzyme-system. Despite these pervious findings, it is still unclear how the effects of HFD and especially HFD in combination with treadmill running affect hepatic Cyp expression. In this study, we investigated the mRNA and protein expression of selected Cyp's in mice subjected to 16 weeks of HFD and treadmill running. To understand the regulatory mechanisms behind the exercise-induced reversion of the HFD-induced changes in Cyp expression, we used a model in which the exercise-induced myokine and known regulator of hepatic Cyp's, interleukin-6 (IL-6), were knocked out specifically in skeletal muscle. We found that HFD increased the mRNA expression of Cyp1a1 and Cyp4a10, and decreased the expression of Cyp2a4, Cyp2b10, Cyp2e1, and Cyp3a11. HFD in combination with treadmill running reversed the HFD increase in Cyp4a10 mRNA expression. In addition, we observed increased Cyp1a and Cyp3a protein expression as an effect of exercise, whereas Cyp2b expression was lowered as an effect of HFD. IL-6 effected the response in Cyp3a11 and Cyp1a expression. We observed no changes in the content of the aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, or peroxisome proliferation activator receptor alpha. In conclusion, we show that both HFD and exercise in HFD-fed animals can regulate hepatic Cyp expression and that changes in Cyp3a in response to HFD and exercise are dependent on skeletal muscular IL-6.
Collapse
Affiliation(s)
- Jakob G Knudsen
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Lærke Bertholdt
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Anders Gudiksen
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | | | | |
Collapse
|
15
|
Tran M, Liu Y, Huang W, Wang L. Nuclear receptors and liver disease: Summary of the 2017 basic research symposium. Hepatol Commun 2018; 2:765-777. [PMID: 30129636 PMCID: PMC6049066 DOI: 10.1002/hep4.1203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/03/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
The nuclear receptor superfamily contains important transcriptional regulators that play pleiotropic roles in cell differentiation, development, proliferation, and metabolic processes to govern liver physiology and pathology. Many nuclear receptors are ligand-activated transcription factors that regulate the expression of their target genes by modulating transcriptional activities and epigenetic changes. Additionally, the protein complex associated with nuclear receptors consists of a multitude of coregulators, corepressors, and noncoding RNAs. Therefore, acquiring new information on nuclear receptors may provide invaluable insight into novel therapies and shed light on new interventions to reduce the burden and incidence of liver diseases. (Hepatology Communications 2018;2:765-777).
Collapse
Affiliation(s)
- Melanie Tran
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Yanjun Liu
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Li Wang
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT.,Veterans Affairs Connecticut Healthcare System West Haven CT.,Department of Internal Medicine, Section of Digestive Diseases Yale University New Haven CT
| |
Collapse
|
16
|
Kumar R, Litoff EJ, Boswell WT, Baldwin WS. High fat diet induced obesity is mitigated in Cyp3a-null female mice. Chem Biol Interact 2018; 289:129-140. [PMID: 29738703 PMCID: PMC6717702 DOI: 10.1016/j.cbi.2018.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/16/2018] [Accepted: 05/01/2018] [Indexed: 12/30/2022]
Abstract
Recent studies indicate a role for the constitutive androstane receptor (CAR), pregnane X-receptor (PXR), and hepatic xenobiotic detoxifying CYPs in fatty liver disease or obesity. Therefore, we examined whether Cyp3a-null mice show increased obesity and fatty liver disease following 8-weeks of exposure to a 60% high-fat diet (HFD). Surprisingly, HFD-fed Cyp3a-null females fed a HFD gained 50% less weight than wild-type (WT; B6) females fed a HFD. In contrast, Cyp3a-null males gained more weight than WT males, primarily during the first few weeks of HFD-treatment. Cyp3a-null females also recovered faster than WT females from a glucose tolerance test; males showed no difference in glucose tolerance between the groups. Serum concentrations of the anti-obesity hormone, adiponectin are 60% higher and β-hydroxybutyrate levels are nearly 50% lower in Cyp3a-null females than WT females, in agreement with reduced weight gain, faster glucose response, and reduced ketogenesis. In contrast, Cyp3a-null males have higher liver triglyceride concentrations and lipidomic analysis indicates an increase in phosphatidylinositol, phosphatidylserine and sphingomyelin. None of these changes were observed in females. Last, Pxr, Cyp2b, and IL-6 expression increased in Cyp3a-null females following HFD-treatment. Cyp2b and Fatp1 increased, while Pxr, Cpt1a, Srebp1 and Fasn decreased in Cyp3a-null males following a HFD, indicating compensatory biochemical responses in male (and to a lesser extent) female mice fed a HFD. In conclusion, lack of Cyp3a has a positive effect on acclimation to a HFD in females as it improves weight gain, glucose response and ketosis.
Collapse
Affiliation(s)
- Ramiya Kumar
- Biological Sciences, Clemson University, Clemson, SC 29634, United States
| | - Elizabeth J Litoff
- Biological Sciences, Clemson University, Clemson, SC 29634, United States
| | - W Tyler Boswell
- Biological Sciences, Clemson University, Clemson, SC 29634, United States
| | - William S Baldwin
- Biological Sciences, Clemson University, Clemson, SC 29634, United States; Environmental Toxicology Program, Clemson University, Clemson, SC 29634, United States.
| |
Collapse
|
17
|
Hakkola J, Bernasconi C, Coecke S, Richert L, Andersson TB, Pelkonen O. Cytochrome P450 Induction and Xeno-Sensing Receptors Pregnane X Receptor, Constitutive Androstane Receptor, Aryl Hydrocarbon Receptor and Peroxisome Proliferator-Activated Receptor α at the Crossroads of Toxicokinetics and Toxicodynamics. Basic Clin Pharmacol Toxicol 2018. [DOI: 10.1111/bcpt.13004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology; Faculty of Medicine; University of Oulu; Oulu Finland
- Medical Research Center Oulu; University of Oulu; Oulu Finland
| | | | - Sandra Coecke
- European Commission Joint Research Centre; EURL ECVAM; Ispra Italy
| | | | - Tommy B. Andersson
- Drug Metabolism and Pharmacokinetics; Cardiovascular and Metabolic Diseases; IMED Biotech Unit; AstraZeneca; Gothenburg Sweden
- Department of Physiology and Pharmacology; Section of Pharmacogenetics; Karolinska Institutet; Stockholm Sweden
| | - Olavi Pelkonen
- Research Unit of Biomedicine, Pharmacology and Toxicology; Faculty of Medicine; University of Oulu; Oulu Finland
- Medical Research Center Oulu; University of Oulu; Oulu Finland
| |
Collapse
|
18
|
Choi S, Gyamfi AA, Neequaye P, Addo S, Gonzalez FJ, Gyamfi MA. Role of the pregnane X receptor in binge ethanol-induced steatosis and hepatotoxicity. J Pharmacol Exp Ther 2018; 365:165-178. [PMID: 29431616 DOI: 10.1124/jpet.117.244665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/27/2018] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Abstract
The pregnane X receptor (PXR, NR1I2) is a xenobiotic-sensing nuclear receptor that defends against toxic agents. We have shown that PXR promotes chronic ethanol (EtOH)-induced steatosis. Therefore, we examined the role of PXR in binge EtOH-induced hepatotoxicity. Male wild type (WT) and Pxr-null mice were orally administered three binge doses of EtOH (4.5 g/kg, every 12 hours) and euthanized four hours after the final dose. Pxr-null mice displayed higher basal mRNA levels of hepatic lipogenic transcription factor sterol regulatory element binding protein 1c (Srebp-1c) and its target stearoyl-CoA desaturase 1 (Scd1) and the lipid peroxide detoxifying aldo-keto reductase 1b7 (Akr1b7) and higher protein levels of EtOH-metabolizing alcohol dehydrogenase 1 (ADH1). In both genotypes, binge EtOH-induced triglyceride accumulation was associated with inhibition of fatty acid β-oxidation and upregulation of Srebp-1c- regulated lipogenic genes and hepatic CYP2E1 protein. Unexpectedly, gene expression of Cyp2b10, a constitutive androstane receptor target gene, implicated in EtOH hepatotoxicity, was PXR-dependent upregulated by binge EtOH. Also, PXR-dependent was the binge EtOH-induced inhibition of hepatic Akr1b8 mRNA, and protein levels of aldehyde dehydrogenase (ALDH) 1A1 and anti-apoptotic Bcl-2, but increased pro-apoptotic Bax protein expression, leading to increases in residual EtOH concentration and the cellular oxidative stress marker, malondialdehyde. In contrast, Pxr-null mice displayed increased Akr1b7 gene and ADH1 protein expression and hypertriglyceridemia following binge EtOH exposure. Taken together, this study demonstrates that PXR ablation prevents EtOH induced upregulation of Cyp2b10 and that PXR potentiates binge EtOH-induced oxidative stress and inhibition of EtOH catabolism, but protects against alcoholic hyperlipidemia.
Collapse
|
19
|
Hassani-Nezhad-Gashti F, Rysä J, Kummu O, Näpänkangas J, Buler M, Karpale M, Hukkanen J, Hakkola J. Activation of nuclear receptor PXR impairs glucose tolerance and dysregulates GLUT2 expression and subcellular localization in liver. Biochem Pharmacol 2018; 148:253-264. [PMID: 29309761 DOI: 10.1016/j.bcp.2018.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022]
Abstract
Pregnane X receptor (PXR) is a nuclear receptor that senses chemical environment and is activated by numerous clinically used drugs and environmental contaminants. Previous studies have indicated that several drugs known to activate PXR appear to induce glucose intolerance. We now aimed to reveal the role of PXR in drug-induced glucose intolerance and characterize the mechanisms involved. We used PXR knockout mice model to investigate the significance of this nuclear receptor in the regulation of glucose tolerance. PXR ligand pregnenolone-16ɑ-carbonitrile (PCN) impaired glucose tolerance in the wildtype mice but not in the PXR knockout mice. Furthermore, DNA microarray and bioinformatics analysis of differentially expressed genes and glucose metabolism relevant pathways in PCN treated primary hepatocytes indicated that PXR regulates genes involved in glucose uptake. PCN decreased the expression of glucose transporter 2 (GLUT2) in mouse liver and in the wildtype mouse hepatocytes but not in the PXR knockout cells. Data mining of published chromatin immunoprecipitation-sequencing results indicate that Glut2 gene is a direct PXR target. Furthermore, PCN induced internalization of GLUT2 protein from the plasma membrane to the cytosol in the liver in vivo and repressed glucose uptake in the primary hepatocytes. Our results indicate that the activation of PXR impairs glucose tolerance and thus PXR represents a novel diabetogenic pathway. PXR activation dysregulates GLUT2 function by two different mechanisms. These findings may partly explain the diabetogenic effects of medications and environmental contaminants.
Collapse
Affiliation(s)
- Fatemeh Hassani-Nezhad-Gashti
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Outi Kummu
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Juha Näpänkangas
- Department of Pathology, Cancer Research and Translational Medicine Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Marcin Buler
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Mikko Karpale
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Janne Hukkanen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Internal Medicine, Research Unit of Internal Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| |
Collapse
|
20
|
Choi S, Neequaye P, French SW, Gonzalez FJ, Gyamfi MA. Pregnane X receptor promotes ethanol-induced hepatosteatosis in mice. J Biol Chem 2017; 293:1-17. [PMID: 29123032 DOI: 10.1074/jbc.m117.815217] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
The pregnane X receptor (PXR, NR1I2) is a xenobiotic-sensing nuclear receptor that modulates the metabolic response to drugs and toxic agents. Both PXR activation and deficiency promote hepatic triglyceride accumulation, a hallmark feature of alcoholic liver disease. However, the molecular mechanism of PXR-mediated activation of ethanol (EtOH)-induced steatosis is unclear. Here, using male wildtype (WT) and Pxr-null mice, we examined PXR-mediated regulation of chronic EtOH-induced hepatic lipid accumulation and hepatotoxicity. EtOH ingestion for 8 weeks significantly (1.8-fold) up-regulated Pxr mRNA levels in WT mice. The EtOH exposure also increased mRNAs encoding hepatic constitutive androstane receptor (3-fold) and its target, Cyp2b10 (220-fold), in a PXR-dependent manner. Furthermore, WT mice had higher serum EtOH levels and developed hepatic steatosis characterized by micro- and macrovesicular lipid accumulation. Consistent with the development of steatosis, lipogenic gene induction was significantly increased in WT mice, including sterol regulatory element-binding protein 1c target gene fatty-acid synthase (3.0-fold), early growth response-1 (3.2-fold), and TNFα (3.0-fold), whereas the expression of peroxisome proliferator-activated receptor α target genes was suppressed. Of note, PXR deficiency suppressed these changes and steatosis. Protein levels, but not mRNAs levels, of EtOH-metabolizing enzymes, including alcohol dehydrogenase 1, aldehyde dehydrogenase 1A1, and catalase, as well as the microsomal triglyceride transfer protein, involved in regulating lipid output were higher in Pxr-null than in WT mice. These findings establish that PXR signaling contributes to ALD development and suggest that PXR antagonists may provide a new approach for ALD therapy.
Collapse
Affiliation(s)
- Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Prince Neequaye
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Samuel W French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California 90509
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707.
| |
Collapse
|
21
|
Oladimeji PO, Chen T. PXR: More Than Just a Master Xenobiotic Receptor. Mol Pharmacol 2017; 93:119-127. [PMID: 29113993 DOI: 10.1124/mol.117.110155] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [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
|
22
|
Tanaka N, Aoyama T, Kimura S, Gonzalez FJ. Targeting nuclear receptors for the treatment of fatty liver disease. Pharmacol Ther 2017; 179:142-157. [PMID: 28546081 DOI: 10.1016/j.pharmthera.2017.05.011] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ligand-activated nuclear receptors, including peroxisome proliferator-activated receptor alpha (PPARα), pregnane X receptor, and constitutive androstane receptor, were first identified as key regulators of the responses against chemical toxicants. However, numerous studies using mouse disease models and human samples have revealed critical roles for these receptors and others, such as PPARβ/δ, PPARγ, farnesoid X receptor (FXR), and liver X receptor (LXR), in maintaining nutrient/energy homeostasis in part through modulation of the gut-liver-adipose axis. Recently, disorders associated with disrupted nutrient/energy homeostasis, e.g., obesity, metabolic syndrome, and non-alcoholic fatty liver disease (NAFLD), are increasing worldwide. Notably, in NAFLD, a progressive subtype exists, designated as non-alcoholic steatohepatitis (NASH) that is characterized by typical histological features resembling alcoholic steatohepatitis (ASH), and NASH/ASH are recognized as major causes of hepatitis virus-unrelated liver cirrhosis and hepatocellular carcinoma. Since hepatic steatosis is basically caused by an imbalance between fat/energy influx and utilization, abnormal signaling of these nuclear receptors contribute to the pathogenesis of fatty liver disease. Standard therapeutic interventions have not been fully established for fatty liver disease, but some new agents that activate or inhibit nuclear receptor signaling have shown promise as possible therapeutic targets. In this review, we summarize recent findings on the roles of nuclear receptors in fatty liver disease and discuss future perspectives to develop promising pharmacological strategies targeting nuclear receptors for NAFLD/NASH.
Collapse
Affiliation(s)
- Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Nagano, Japan.
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Nagano, Japan
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
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: 46] [Impact Index Per Article: 5.1] [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
|
24
|
RNA-Seq reveals common and unique PXR- and CAR-target gene signatures in the mouse liver transcriptome. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1198-1217. [PMID: 27113289 DOI: 10.1016/j.bbagrm.2016.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022]
Abstract
The pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known xenobiotic-sensing nuclear receptors with overlapping functions. However, there lacks a quantitative characterization to distinguish between the PXR and CAR target genes and signaling pathways in the liver. The present study performed a transcriptomic comparison of the PXR- and CAR-targets using RNA-Seq in livers of adult wild-type mice that were treated with the prototypical PXR ligand PCN (200mg/kg, i.p. once daily for 4days in corn oil) or the prototypical CAR ligand TCPOBOP (3mg/kg, i.p., once daily for 4days in corn oil). At the given doses, TCPOBOP differentially regulated many more genes (2125) than PCN (212), and 147 of the same genes were differentially regulated by both chemicals. As expected, the top pathways differentially regulated by both PCN and TCPOBOP were involved in xenobiotic metabolism, and they also up-regulated genes involved in retinoid metabolism, but down-regulated genes involved in inflammation and iron homeostasis. Regarding unique pathways, PXR activation appeared to overlap with the aryl hydrocarbon receptor signaling, whereas CAR activation appeared to overlap with the farnesoid X receptor signaling, acute-phase response, and mitochondrial dysfunction. The mRNAs of differentially regulated drug-processing genes (DPGs) partitioned into three patterns, namely TCPOBOP-induced, PCN-induced, as well as TCPOBOP-suppressed gene clusters. The cumulative mRNAs of the differentially regulated DPGs, phase-I and -II enzymes, as well as efflux transporters were all up-regulated by both PCN and TCPOBOPOP, whereas the cumulative mRNAs of the uptake transporters were down-regulated only by TCPOBOP. The absolute mRNA abundance in control and receptor-activated conditions was examined in each DPG category to predict the contribution of specific DPG genes in the PXR/CAR-mediated pharmacokinetic responses. The preferable differential regulation by TCPOBOP in the entire hepatic transcriptome correlated with a marked change in the expression of many DNA and histone epigenetic modifiers. In conclusion, the present study has revealed known and novel, as well as common and unique targets of PXR and CAR in mouse liver following pharmacological activation using their prototypical ligands. Results from this study will further support the role of these receptors in regulating the homeostasis of xenobiotic and intermediary metabolism in the liver, and aid in distinguishing between PXR and CAR signaling at various physiological and pathophysiological conditions. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
25
|
Hakkola J, Rysä J, Hukkanen J. Regulation of hepatic energy metabolism by the nuclear receptor PXR. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1072-1082. [PMID: 27041449 DOI: 10.1016/j.bbagrm.2016.03.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/30/2022]
Abstract
The pregnane X receptor (PXR) is a nuclear receptor that is traditionally thought to be specialized for sensing xenobiotic exposure. In concurrence with this feature PXR was originally identified to regulate drug-metabolizing enzymes and transporters. During the last ten years it has become clear that PXR harbors broader functions. Evidence obtained both in experimental animals and humans indicate that ligand-activated PXR regulates hepatic glucose and lipid metabolism and affects whole body metabolic homeostasis. Currently, the consequences of PXR activation on overall metabolic health are not yet fully understood and varying results on the effect of PXR activation or knockout on metabolic disorders and weight gain have been published in mouse models. Rifampicin and St. John's wort, the prototypical human PXR agonists, impair glucose tolerance in healthy volunteers. Chronic exposure to PXR agonists could potentially represent a risk factor for diabetes and metabolic syndrome. 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)
- Jukka Hakkola
- Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Janne Hukkanen
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Research Unit of Internal Medicine, University of Oulu, Oulu, Finland; Department of Internal Medicine, Oulu University Hospital, Oulu, Finland; Biocenter Oulu, Oulu, Finland
| |
Collapse
|
26
|
Xie YQ, Zhang JZ, Zhang H, Peng L, Zhou S, Li LZ, Fang BJ, Lyu CZ. Pregnane receptor gene polymorphisms, pathogenic bacteria distribution and drug sensitivity, and TCM syndrome differentiation in patients with cholelithiasis. ASIAN PAC J TROP MED 2016; 9:307-312. [DOI: 10.1016/j.apjtm.2016.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/20/2016] [Accepted: 03/01/2016] [Indexed: 10/22/2022] Open
|
27
|
Mo L, Shen J, Liu Q, Zhang Y, Kuang J, Pu S, Cheng S, Zou M, Jiang W, Jiang C, Qu A, He J. Irisin Is Regulated by CAR in Liver and Is a Mediator of Hepatic Glucose and Lipid Metabolism. Mol Endocrinol 2016; 30:533-42. [PMID: 27007446 DOI: 10.1210/me.2015-1292] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Irisin, a hormone proteolytically processed from fibronectin type III domain-containing protein 5 (FNDC5), has been reported to induce the browning of sc adipocytes by increasing the level of uncoupling protein 1. In this study, we showed that activation of the nuclear receptor constitutive androstane receptor induced FNDC5 mRNA expression in the liver and increased the circulating level of irisin in mice. FNDC5/irisin is a direct transcriptional target of constitutive androstane receptor. Hepatic-released irisin functioned as a paracrine/autocrine factor that inhibited lipogenesis and gluconeogenesis via the Adenosine 5'-monophosphate (AMP)-activated protein kinase pathway. Adenovirus-overexpressed irisin improved hepatic steatosis and insulin resistance in genetic-induced obese mice. Irisin transgenic mice were also protected against high-fat diet-induced obesity and insulin resistance. In conclusion, our results reveal a novel pathway in regulating FNDC5/irisin expression and identify a physiological role for this hepatic hormone in glucose and lipid homeostasis.
Collapse
Affiliation(s)
- Li Mo
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Jing Shen
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Qinhui Liu
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Yuwei Zhang
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Jiangying Kuang
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Shiyun Pu
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Shihai Cheng
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Min Zou
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Wei Jiang
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Changtao Jiang
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Aijuan Qu
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| | - Jinhan He
- Center of Gerontology and Geriatrics (L.M.), Department of Pharmacy (J.S., J.K., S.P., S.C., M.Z., J.H.), Laboratory of Clinical Pharmacy and Adverse Drug Reaction (Q.L., J.H.), Division of Endocrinology and Metabolism (Y.Z.), Molecular Medicine Research Center (W.J.), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041 Sichuan, China; Department of Physiology and Pathophysiology (C.J.), School of Basic Medical Sciences, Peking University, Beijing 100871; and Department of Physiology and Pathophysiology (A.Q.), School of Basic Medical Sciences, Capital Medical University, Beijing 100069
| |
Collapse
|
28
|
Cave MC, Clair HB, Hardesty JE, Falkner KC, Feng W, Clark BJ, Sidey J, Shi H, Aqel BA, McClain CJ, Prough RA. Nuclear receptors and nonalcoholic fatty liver disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1083-1099. [PMID: 26962021 DOI: 10.1016/j.bbagrm.2016.03.002] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Nuclear receptors are transcription factors which sense changing environmental or hormonal signals and effect transcriptional changes to regulate core life functions including growth, development, and reproduction. To support this function, following ligand-activation by xenobiotics, members of subfamily 1 nuclear receptors (NR1s) may heterodimerize with the retinoid X receptor (RXR) to regulate transcription of genes involved in energy and xenobiotic metabolism and inflammation. Several of these receptors including the peroxisome proliferator-activated receptors (PPARs), the pregnane and xenobiotic receptor (PXR), the constitutive androstane receptor (CAR), the liver X receptor (LXR) and the farnesoid X receptor (FXR) are key regulators of the gut:liver:adipose axis and serve to coordinate metabolic responses across organ systems between the fed and fasting states. Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease and may progress to cirrhosis and even hepatocellular carcinoma. NAFLD is associated with inappropriate nuclear receptor function and perturbations along the gut:liver:adipose axis including obesity, increased intestinal permeability with systemic inflammation, abnormal hepatic lipid metabolism, and insulin resistance. Environmental chemicals may compound the problem by directly interacting with nuclear receptors leading to metabolic confusion and the inability to differentiate fed from fasting conditions. This review focuses on the impact of nuclear receptors in the pathogenesis and treatment of NAFLD. Clinical trials including PIVENS and FLINT demonstrate that nuclear receptor targeted therapies may lead to the paradoxical dissociation of steatosis, inflammation, fibrosis, insulin resistance, dyslipidemia and obesity. Novel strategies currently under development (including tissue-specific ligands and dual receptor agonists) may be required to separate the beneficial effects of nuclear receptor activation from unwanted metabolic side effects. The impact of nuclear receptor crosstalk in NAFLD is likely to be profound, but requires further elucidation. 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)
- Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, KY 40202, USA.
| | - Heather B Clair
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Josiah E Hardesty
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - K Cameron Falkner
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Barbara J Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jennifer Sidey
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bashar A Aqel
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Scottsdale, AZ 85054, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA; The KentuckyOne Health Jewish Hospital Liver Transplant Program, Louisville, KY 40202, USA
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
29
|
Zhou C. Novel functions of PXR in cardiometabolic disease. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1112-1120. [PMID: 26924429 DOI: 10.1016/j.bbagrm.2016.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/17/2022]
Abstract
Cardiometabolic disease emerges as a worldwide epidemic and there is urgent need to understand the molecular mechanisms underlying this chronic disease. The chemical environment to which we are exposed has significantly changed in the past few decades and recent research has implicated its contribution to the development of many chronic human diseases. However, the mechanisms of how exposure to chemicals contributes to the development of cardiometabolic disease are poorly understood. Numerous chemicals have been identified as ligands for the pregnane X receptor (PXR), a nuclear receptor functioning as a xenobiotic sensor to coordinately regulate xenobiotic metabolism via transcriptional regulation of xenobiotic-detoxifying enzymes and transporters. In the past decade, the function of PXR in the regulation of xenobiotic metabolism has been extensively studied by many laboratories and the role of PXR as a xenobiotic sensor has been well-established. The identification of PXR as a xenobiotic sensor has provided an important tool for the study of new mechanisms through which xenobiotic exposure impacts human chronic diseases. Recent studies have revealed novel and unexpected roles of PXR in modulating obesity, insulin sensitivity, lipid homeostasis, atherogenesis, and vascular functions. These studies suggest that PXR signaling may contribute significantly to the pathophysiological effects of many known xenobiotics on cardiometabolic disease in humans. The discovery of novel functions of PXR in cardiometabolic disease not only contributes to our understanding of "gene-environment interactions" in predisposing individuals to chronic diseases but also provides strong evidence to inform future risk assessment for relevant chemicals. 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)
- Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
30
|
Acetylation of lysine 109 modulates pregnane X receptor DNA binding and transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1155-1169. [PMID: 26855179 DOI: 10.1016/j.bbagrm.2016.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/19/2016] [Accepted: 01/26/2016] [Indexed: 12/31/2022]
Abstract
Pregnane X receptor (PXR) is a major transcriptional regulator of xenobiotic metabolism and transport pathways in the liver and intestines, which are critical for protecting organisms against potentially harmful xenobiotic and endobiotic compounds. Inadvertent activation of drug metabolism pathways through PXR is known to contribute to drug resistance, adverse drug-drug interactions, and drug toxicity in humans. In both humans and rodents, PXR has been implicated in non-alcoholic fatty liver disease, diabetes, obesity, inflammatory bowel disease, and cancer. Because of PXR's important functions, it has been a therapeutic target of interest for a long time. More recent mechanistic studies have shown that PXR is modulated by multiple PTMs. Herein we provide the first investigation of the role of acetylation in modulating PXR activity. Through LC-MS/MS analysis, we identified lysine 109 (K109) in the hinge as PXR's major acetylation site. Using various biochemical and cell-based assays, we show that PXR's acetylation status and transcriptional activity are modulated by E1A binding protein (p300) and sirtuin 1 (SIRT1). Based on analysis of acetylation site mutants, we found that acetylation at K109 represses PXR transcriptional activity. The mechanism involves loss of RXRα dimerization and reduced binding to cognate DNA response elements. This mechanism may represent a promising therapeutic target using modulators of PXR acetylation levels. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
31
|
Woolsey SJ, Mansell SE, Kim RB, Tirona RG, Beaton MD. CYP3A Activity and Expression in Nonalcoholic Fatty Liver Disease. Drug Metab Dispos 2015; 43:1484-90. [DOI: 10.1124/dmd.115.065979] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/29/2015] [Indexed: 12/16/2022] Open
|
32
|
Spruiell K, Gyamfi AA, Yeyeodu ST, Richardson RM, Gonzalez FJ, Gyamfi MA. Pregnane X Receptor-Humanized Mice Recapitulate Gender Differences in Ethanol Metabolism but Not Hepatotoxicity. J Pharmacol Exp Ther 2015; 354:459-70. [PMID: 26159875 DOI: 10.1124/jpet.115.224295] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 07/08/2015] [Indexed: 12/20/2022] Open
Abstract
Both human and rodent females are more susceptible to developing alcoholic liver disease following chronic ethanol (EtOH) ingestion. However, little is known about the relative effects of acute EtOH exposure on hepatotoxicity in female versus male mice. The nuclear receptor pregnane X receptor (PXR; NR1I2) is a broad-specificity sensor with species-specific responses to toxic agents. To examine the effects of the human PXR on acute EtOH toxicity, the responses of male and female PXR-humanized (hPXR) transgenic mice administered oral binge EtOH (4.5 g/kg) were analyzed. Basal differences were observed between hPXR males and females in which females expressed higher levels of two principal enzymes responsible for EtOH metabolism, alcohol dehydrogenase 1 and aldehyde dehydrogenase 2, and two key mediators of hepatocyte replication and repair, cyclin D1 and proliferating cell nuclear antigen. EtOH ingestion upregulated hepatic estrogen receptor α, cyclin D1, and CYP2E1 in both genders, but differentially altered lipid and EtOH metabolism. Consistent with higher basal levels of EtOH-metabolizing enzymes, blood EtOH was more rapidly cleared in hPXR females. These factors combined to provide greater protection against EtOH-induced liver injury in female hPXR mice, as revealed by markers for liver damage, lipid peroxidation, and endoplasmic reticulum stress. These results indicate that female hPXR mice are less susceptible to acute binge EtOH-induced hepatotoxicity than their male counterparts, due at least in part to the relative suppression of cellular stress and enhanced expression of enzymes involved in both EtOH metabolism and hepatocyte proliferation and repair in hPXR females.
Collapse
Affiliation(s)
- Krisstonia Spruiell
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| | - Afua A Gyamfi
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| | - Susan T Yeyeodu
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| | - Ricardo M Richardson
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| | - Frank J Gonzalez
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina (K.S., A.A.G., S.T.Y., R.M.R., M.A.G.); and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (F.J.G.)
| |
Collapse
|