1
|
Zhang Y, Jiang X, Wu D, Huang H, Jia G, Zhao G. Sema4D deficiency enhances glucose tolerance through GLUT2 retention in hepatocytes. J Transl Med 2024; 22:864. [PMID: 39334386 PMCID: PMC11429007 DOI: 10.1186/s12967-024-05694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The glucose transporter 2 (GLUT2) is constitutively expressed in pancreatic beta cells and hepatocytes of mice. It is the most important receptor in glucose-stimulated insulin release and hepatic glucose transport. The Sema4D is a signalin receptor on cell membranes. The correlation between Sema4D and GLUT2 has not been reported previously. We investigated whether knockdown of Sema4D could exert a hypoglycemic effect based on the increased GLUT2 expression in Sema4D -/- mice hepatocytes. METHODS The glucose tolerance test and insulin tolerance test in sema4D -/- and sema4D +/+ mice were compared before and after streptozotocin (STZ) injection; the expression of GLUT2 content on the membrane surface of both groups was verified by Western blot. Then, the levels of insulin and C-peptide in the serum of the two groups of mice after STZ injection were measured by ELISA; the differentially expressed mRNAs in the liver of the two groups of mice were analyzed by transcriptomic analysis; then the differences in the expression of GLUT2, glycogen, insulin and glucagon in the two groups of mice were compared by tissue section staining. Finally, metabolomics analysis was performed to analyze the metabolites differentially expressed in the two groups of mice. KEY FINDINGS First, Sema4D -/- male mice exhibited significantly greater glucose tolerance than wild-type mice in a hyperglycemic environment. Secondly, Sema4D -/- mice had more retained GLUT2 in liver membranes after STZ injection according to an immunofluorescence assay. After STZ injection, Sema4D -/- male mice did not exhibit fasting hyperinsulinemia like wild-type mice. Finally, analysis of metabolomic and immunohistochemical data also revealed that Sema4D -/- mice produce hypoglycemic effects by enhancing the pentose phosphate pathway, but not glycogen synthesis. CONCLUSIONS Thus, Sema4D may play an important role in the regulation of glucose homeostasis by affecting GLUT2 synthesis.
Collapse
Affiliation(s)
- Yanling Zhang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Xiaomei Jiang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Dongsong Wu
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Hao Huang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West Second Section, First Ring Road, Chengdu, 610072, China.
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
2
|
Zhang J, Huang Y, Li H, Xu P, Liu Q, Sun Y, Zhang Z, Wu T, Tang Q, Jia Q, Xia Y, Xu Y, Jing X, Li J, Mo L, Xie W, Qu A, He J, Li Y. B3galt5 functions as a PXR target gene and regulates obesity and insulin resistance by maintaining intestinal integrity. Nat Commun 2024; 15:5919. [PMID: 39004626 PMCID: PMC11247088 DOI: 10.1038/s41467-024-50198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Pregnane X receptor (PXR) has been reported to regulate glycolipid metabolism. The dysfunction of intestinal barrier contributes to metabolic disorders. However, the role of intestinal PXR in metabolic diseases remains largely unknown. Here, we show that activation of PXR by tributyl citrate (TBC), an intestinal-selective PXR agonist, improves high fat diet (HFD)-induced obesity. The metabolic benefit of intestinal PXR activation is associated with upregulation of β-1,3 galactosyltransferase 5 (B3galt5). Our results reveal that B3galt5 mainly expresses in the intestine and is a direct PXR transcriptional target. B3galt5 knockout exacerbates HFD-induced obesity, insulin resistance and inflammation. Mechanistically, B3galt5 is essential to maintain the integrity of intestinal mucus barrier. B3galt5 ablation impairs the O-glycosylation of mucin2, destabilizes the mucus layer, and increases intestinal permeability. Furthermore, B3galt5 deficiency abolishes the beneficial effect of intestinal PXR activation on metabolic disorders. Our results suggest the intestinal-selective PXR activation regulates B3galt5 expression and maintains metabolic homeostasis, making it a potential therapeutic strategy in obesity.
Collapse
Affiliation(s)
- Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ya Huang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Pharmacy, GuiQian International General Hospital, Guiyang, China
| | - Hong Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qinhui Liu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Disease, Kunming, Yunnan Province, China
| | - Zijing Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tong Wu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qin Tang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan Xia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiandan Jing
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiahui Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
Yan L, Yan Y, Yang K, Chang Q, Zhang L. Metabolomics reveals dysregulated all-trans retinoic acid and polyunsaturated fatty acid metabolism contribute to PXR-induced hepatic steatosis in mice. Toxicol Lett 2024; 398:150-160. [PMID: 38971454 DOI: 10.1016/j.toxlet.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China.
| | - Yachun Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China
| | - Kun Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Qi Chang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
4
|
Peng R, Song C, Gou S, Liu H, Kang H, Dong Y, Xu Y, Hu P, Cai K, Feng Q, Guan H, Li F. Gut Clostridium sporogenes-derived indole propionic acid suppresses osteoclast formation by activating pregnane X receptor. Pharmacol Res 2024; 202:107121. [PMID: 38431091 DOI: 10.1016/j.phrs.2024.107121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Bone homeostasis is maintained by osteoclast-mediated bone resorption and osteoblast-mediated bone formation. A dramatic decrease in estrogen levels in postmenopausal women leads to osteoclast overactivation, impaired bone homeostasis, and subsequent bone loss. Changes in the gut microbiome affect bone mineral density. However, the role of the gut microbiome in estrogen deficiency-induced bone loss and its underlying mechanism remain unknown. In this study, we found that the abundance of Clostridium sporogenes (C. spor.) and its derived metabolite, indole propionic acid (IPA), were decreased in ovariectomized (OVX) mice. In vitro assays suggested that IPA suppressed osteoclast differentiation and function. At the molecular level, IPA suppressed receptor activator of nuclear factor kappa-Β ligand (RANKL)-induced pregnane X receptor (PXR) ubiquitination and degradation, leading to increased binding of remaining PXR with P65. In vivo daily IPA administration or repeated C. spor. colonization protected against OVX-induced bone loss. To protect live bacteria from the harsh gastric environment and delay the emptying of orally administered C. spor. from the intestine, a C. spor.-encapsulated silk fibroin (SF) hydrogel system was developed, which achieved bone protection in OVX mice comparable to that achieved with repeated germ transplantation or daily IPA administration. Overall, we found that gut C. spor.-derived IPA was involved in estrogen deficiency-induced osteoclast overactivation by regulating the PXR/P65 complex. The C. spor.-encapsulated SF hydrogel system is a promising tool for combating postmenopausal osteoporosis without the disadvantages of repeated germ transplantation.
Collapse
Affiliation(s)
- Renpeng Peng
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Song
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuangquan Gou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Haiyang Liu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honglei Kang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimin Dong
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peixuan Hu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Hanfeng Guan
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Feng Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Attema B, Kummu O, Pitkänen S, Weisell J, Vuorio T, Pennanen E, Vorimo M, Rysä J, Kersten S, Levonen AL, Hakkola J. Metabolic effects of nuclear receptor activation in vivo after 28-day oral exposure to three endocrine-disrupting chemicals. Arch Toxicol 2024; 98:911-928. [PMID: 38182912 PMCID: PMC10861694 DOI: 10.1007/s00204-023-03658-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024]
Abstract
Environmental exposure to endocrine-disrupting chemicals (EDCs) can lead to metabolic disruption, resulting in metabolic complications including adiposity, dyslipidemia, hepatic lipid accumulation, and glucose intolerance. Hepatic nuclear receptor activation is one of the mechanisms mediating metabolic effects of EDCs. Here, we investigated the potential to use a repeated dose 28-day oral toxicity test for identification of EDCs with metabolic endpoints. Bisphenol A (BPA), pregnenolone-16α-carbonitrile (PCN), and perfluorooctanoic acid (PFOA) were used as reference compounds. Male and female wild-type C57BL/6 mice were orally exposed to 5, 50, and 500 μg/kg of BPA, 1000, 10 000, and 100 000 µg/kg of PCN and 50 and 300 μg/kg of PFOA for 28 days next to normal chow diet. Primary endpoints were glucose tolerance, hepatic lipid accumulation, and plasma lipids. After 28-day exposure, no changes in body weight and glucose tolerance were observed in BPA-, PCN-, or PFOA-treated males or females. PCN and PFOA at the highest dose in both sexes and BPA at the middle and high dose in males increased relative liver weight. PFOA reduced plasma triglycerides in males and females, and increased hepatic triglyceride content in males. PCN and PFOA induced hepatic expression of typical pregnane X receptor (PXR) and peroxisome proliferator-activated receptor (PPAR)α target genes, respectively. Exposure to BPA resulted in limited gene expression changes. In conclusion, the observed changes on metabolic health parameters were modest, suggesting that a standard repeated dose 28-day oral toxicity test is not a sensitive method for the detection of the metabolic effect of EDCs.
Collapse
Affiliation(s)
- Brecht Attema
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Sini Pitkänen
- A.I. Virtanen-Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jonna Weisell
- Finnish Institute of Occupational Health, Kuopio, Finland
| | - Taina Vuorio
- A.I. Virtanen-Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Pennanen
- A.I. Virtanen-Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maria Vorimo
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Anna-Liisa Levonen
- A.I. Virtanen-Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
6
|
Chenchula S, Sharma P, Ghanta MK, Amerneni KC, Rajakarunakaran P, Saggurthi P, Chandra MB, Gupta R, Chavan M. Association and Mechanisms of Proton Pump Inhibitors Use with Type-2Diabetes Mellitus Incidence in Adults: A Systemic Review andMeta-Analysis. Curr Diabetes Rev 2024; 20:e120124225581. [PMID: 38243950 DOI: 10.2174/0115733998254869231101095222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are an extensively prescribed class of anti-ulcer drugs. This systematic review aimed to investigate the association between PPI use and the risk of new-onset diabetes mellitus or type 2 diabetes (T2DM) incidence. METHODS A comprehensive literature search was conducted in PubMed, Scopus, Cochrane Library, and ClinicalTrials.gov using the search terms "proton pump inhibitor," "proton pump inhibitors," "PPIs," "diabetes mellitus," and "type 2 diabetes" from inception to February 2023. Statistical analyses were performed using the "Review Manager 5.4" version, and a statistically highly significant P-value <0.05 was set. RESULTS This systematic review identified 12 studies (8 cohort, 1 RCT, and 3 case-control) with a total of 12, 64, 816 population, and the median age ranged from ≥18 yrs to ≤ 75 yrs. The pooled relative risk (RR) observations of a random-effects meta-analysis model showed that chronic exposure to PPI use has a significant association with T2DM risk incidence (RR, 2.44; 95% confidence interval, 1.31-4.54; I2 = 99%, P < 0.00001). The systematic review findings of the three case-control studies also supported an association of dose-dependent and chronic use of PPIs with an incidence of T2DM among chronic users. CONCLUSION The systematic review concludes that chronic PPI exposure increases the risk of T2DM incidence. The authors recommend the shortest possible duration of PPI use and not prescribing PPIs to high-risk prediabetics and those without a compelling indication for PPI use. Regular education to patients regarding adverse reactions with prolonged use may decrease the risk of adverse effects associated with PPIs. The authors suggest that gut dysbiosis, hypergastrinemia, hypomagnesemia, decreased pancreatic secretions and IGF-1 levels, and PXR activation associated with chronic acid suppression among chronic PPI users and the potency of PPIs might explain the association between abnormal glucose metabolism and T2DM incidence. Finally, the authors recommend further randomized controlled trials to investigate the association between PPIs and the risk of new-onset T2DM incidence.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, India
| | - Phulen Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mohan Krishna Ghanta
- Department of Pharmacology, MVJ Medical College and Research Hospital, Bangalore, Karnataka, India
| | | | | | - Pavani Saggurthi
- Department of Pharmacology, Pharmacovigilance Associate, All India Institute of Medical Sciences, Mangalagiri, India
| | | | - Rupesh Gupta
- Department of Internal Medicine, Government Medical College, Shahdol, Madhya Pradesh, India
| | - Madhavrao Chavan
- Department of Pharmacology, Pharmacovigilance Associate, All India Institute of Medical Sciences, Mangalagiri, India
| |
Collapse
|
7
|
Amanat M, Ud Daula AFMS, Singh R. Potential Antidiabetic Activity of β-sitosterol from Zingiber roseum Rosc. via Modulation of Peroxisome Proliferator-activated Receptor Gamma (PPARγ). Comb Chem High Throughput Screen 2024; 27:1676-1699. [PMID: 38305397 DOI: 10.2174/0113862073260323231120134826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/09/2023] [Accepted: 10/02/2023] [Indexed: 02/03/2024]
Abstract
AIM To evaluate the antidiabetic potential of β-sitosterol from Zingiber roseum. BACKGROUND Diabetes mellitus is a cluster of metabolic disorders, and 90% of diabetic patients are affected with Type II diabetes (DM2). For the treatment of DM2, thiazolidinedione drugs (TZDs) were proposed, but recent studies have shown that TZDs have several detrimental effects, such as weight gain, kidney enlargement (hypertrophy), fluid retention, increased risk of bone fractures, and potential harm to the liver (hepatotoxicity). That is why a new molecule is needed to treat DM2. OBJECTIVE The current research aimed to assess the efficacy of β-Sitosterol from methanolic extract of Zingiber roseum in managing diabetes via PPARγ modulation. METHODS Zingiber roseum was extracted using methanol, and GC-MS was employed to analyze the extract. Through homology modeling, PPARγ structure was predicted. Molecular docking, MD simulation, free binding energies, QSAR, ADMET, and bioactivity and toxicity scores were all used during the in-depth computer-based research. RESULTS Clinically, agonists of synthetic thiazolidinedione (TZDs) have been used therapeutically to treat DM2, but these TZDs are associated with significant risks. Hence, GC-MS identified phytochemicals to search for a new PPAR-γ agonist. Based on the in-silico investigation, β-sitosterol was found to have a higher binding affinity (-8.9 kcal/mol) than standard drugs. MD simulations and MMGBSA analysis also demonstrated that β-sitosterol bound to the PPAR-γ active site stably. CONCLUSION It can be concluded that β-sitosterol from Z. roseum attenuates Type-II diabetes by modulating PPARγ activity.
Collapse
Affiliation(s)
- Muhammed Amanat
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda-151401, India
| | - A F M Shahid Ud Daula
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Sonapur-3814, Bangladesh
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda-151401, India
| |
Collapse
|
8
|
Karpale M, Kummu O, Kärkkäinen O, Lehtonen M, Näpänkangas J, Herfurth UM, Braeuning A, Rysä J, Hakkola J. Pregnane X receptor activation remodels glucose metabolism to promote NAFLD development in obese mice. Mol Metab 2023; 76:101779. [PMID: 37467962 PMCID: PMC10415798 DOI: 10.1016/j.molmet.2023.101779] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023] Open
Abstract
OBJECTIVE Both obesity and exposure to chemicals may induce non-alcoholic fatty liver disease (NAFLD). Pregnane X Receptor (PXR) is a central target of metabolism disrupting chemicals and disturbs hepatic glucose and lipid metabolism. We hypothesized that the metabolic consequences of PXR activation may be modified by existing obesity and associated metabolic dysfunction. METHODS Wildtype and PXR knockout male mice were fed high-fat diet to induce obesity and metabolic dysfunction. PXR was activated with pregnenolone-16α-carbonitrile. Glucose metabolism, hepatosteatosis, insulin signaling, glucose uptake, liver glycogen, plasma and liver metabolomics, and liver, white adipose tissue, and muscle transcriptomics were investigated. RESULTS PXR activation aggravated obesity-induced liver steatosis by promoting lipogenesis and inhibiting fatty acid disposal. Accordingly, hepatic insulin sensitivity was impaired and circulating alanine aminotransferase level increased. Lipid synthesis was facilitated by increased liver glucose uptake and utilization of glycogen reserves resulting in dissociation of hepatosteatosis and hepatic insulin resistance from the systemic glucose tolerance and insulin sensitivity. Furthermore, glucagon-induced hepatic glucose production was impaired. PXR deficiency did not protect from the metabolic manifestations of obesity, but the liver transcriptomics and metabolomics profiling suggest diminished activation of inflammation and less prominent changes in the overall metabolite profile. CONCLUSIONS Obesity and PXR activation by chemical exposure have a synergistic effect on NAFLD development. To support liver fat accumulation the PXR activation reorganizes glucose metabolism that seemingly improves systemic glucose metabolism. This implies that obese individuals, already predisposed to metabolic diseases, may be more susceptible to harmful metabolic effects of PXR-activating drugs and environmental chemicals.
Collapse
Affiliation(s)
- Mikko Karpale
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Olli Kärkkäinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Juha Näpänkangas
- Department of Pathology, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Uta M Herfurth
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment (BfR), Department of Food Safety, Berlin, Germany
| | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
9
|
Ming WH, Luan ZL, Yao Y, Liu HC, Hu SY, Du CX, Zhang C, Zhao YH, Huang YZ, Sun XW, Qiao RF, Xu H, Guan YF, Zhang XY. Pregnane X receptor activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/β-catenin signaling. Acta Pharmacol Sin 2023; 44:2075-2090. [PMID: 37344564 PMCID: PMC10545797 DOI: 10.1038/s41401-023-01113-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/18/2023] [Indexed: 06/23/2023]
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) with various etiologies, which seriously affects the structure and function of the kidney. Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily and plays a critical role in regulating the genes related to xenobiotic and endobiotic metabolism in mammals. Previous studies show that PXR is expressed in the kidney and has protective effect against acute kidney injury (AKI). In this study, we investigated the role of PXR in CKD. Adenine diet-induced CKD (AD) model was established in wild-type and PXR humanized (hPXR) mice, respectively, which were treated with pregnenolone-16α-carbonitrile (PCN, 50 mg/kg, twice a week for 4 weeks) or rifampicin (RIF, 10 mg·kg-1·d-1, for 4 weeks). We showed that both PCN and RIF, which activated mouse and human PXR, respectively, improved renal function and attenuated renal fibrosis in the two types of AD mice. In addition, PCN treatment also alleviated renal fibrosis in unilateral ureter obstruction (UUO) mice. On the contrary, PXR gene deficiency exacerbated renal dysfunction and fibrosis in both adenine- and UUO-induced CKD mice. We found that PCN treatment suppressed the expression of the profibrotic Wnt7a and β-catenin in AD mice and in cultured mouse renal tubular epithelial cells treated with TGFβ1 in vitro. We demonstrated that PXR was colocalized and interacted with p53 in the nuclei of tubular epithelial cells. Overexpression of p53 increased the expression of Wnt7a, β-catenin and its downstream gene fibronectin. We further revealed that p53 bound to the promoter of Wnt7a gene to increase its transcription and β-catenin activation, leading to increased expression of the downstream profibrotic genes, which was inhibited by PXR. Taken together, PXR activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wen-Hua Ming
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China
| | - Yao Yao
- Department of nephrology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226006, China
| | - Hang-Chi Liu
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Shu-Yuan Hu
- Department of nephrology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226006, China
| | - Chun-Xiu Du
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, 241100, China
| | - Cong Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Hang Zhao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ying-Zhi Huang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xiao-Wan Sun
- Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Rong-Fang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Hu Xu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian, 116044, China.
| | - Xiao-Yan Zhang
- Health Science Center, East China Normal University, Shanghai, 200241, China.
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, 241100, China.
| |
Collapse
|
10
|
Liang Y, Gong Y, Jiang Q, Yu Y, Zhang J. Environmental endocrine disruptors and pregnane X receptor action: A review. Food Chem Toxicol 2023; 179:113976. [PMID: 37532173 DOI: 10.1016/j.fct.2023.113976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/04/2023]
Abstract
The pregnane X receptor (PXR) is a kind of orphan nuclear receptor activated by a series of ligands. Environmental endocrine disruptors (EEDs) are a wide class of molecules present in the environment that are suspected to have adverse effects on the endocrine system by interfering with the synthesis, transport, degradation, or action of endogenous hormones. Since EEDs may modulate human/rodent PXR, this review aims to summarize EEDs as PXR modulators, including agonists and antagonists. The modular structure of PXR is also described, interestingly, the pharmacology of PXR have been confirmed to vary among different species. Furthermore, PXR play a key role in the regulation of endocrine function. Endocrine disruption of EEDs via PXR and its related pathways are systematically summarized. In brief, this review may provide a way to understand the roles of EEDs in interaction with the nuclear receptors (such as PXR) and the related pathways.
Collapse
Affiliation(s)
- Yuan Liang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Yiyao Gong
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Qiuyan Jiang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Yifan Yu
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
11
|
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
|
12
|
Yang H, Su M, Liu M, Sheng Y, Zhu L, Yang L, Mu R, Zou J, Liu X, Liu L. Hepatic retinaldehyde deficiency is involved in diabetes deterioration by enhancing PCK1- and G6PC-mediated gluconeogenesis. Acta Pharm Sin B 2023; 13:3728-3743. [PMID: 37719384 PMCID: PMC10501888 DOI: 10.1016/j.apsb.2023.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/14/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
Type 2 diabetes (T2D) is often accompanied with an induction of retinaldehyde dehydrogenase 1 (RALDH1 or ALDH1A1) expression and a consequent decrease in hepatic retinaldehyde (Rald) levels. However, the role of hepatic Rald deficiency in T2D progression remains unclear. In this study, we demonstrated that reversing T2D-mediated hepatic Rald deficiency by Rald or citral treatments, or liver-specific Raldh1 silencing substantially lowered fasting glycemia levels, inhibited hepatic glucogenesis, and downregulated phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6-phosphatase (G6PC) expression in diabetic db/db mice. Fasting glycemia and Pck1/G6pc mRNA expression levels were strongly negatively correlated with hepatic Rald levels, indicating the involvement of hepatic Rald depletion in T2D deterioration. A similar result that liver-specific Raldh1 silencing improved glucose metabolism was also observed in high-fat diet-fed mice. In primary human hepatocytes and oleic acid-treated HepG2 cells, Rald or Rald + RALDH1 silencing resulted in decreased glucose production and downregulated PCK1/G6PC mRNA and protein expression. Mechanistically, Rald downregulated direct repeat 1-mediated PCK1 and G6PC expression by antagonizing retinoid X receptor α, as confirmed by luciferase reporter assays and molecular docking. These results highlight the link between hepatic Rald deficiency, glucose dyshomeostasis, and the progression of T2D, whilst also suggesting RALDH1 as a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Hanyu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengxiang Su
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Sheng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ruijing Mu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jianjun Zou
- Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaodong Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
13
|
Zhao Y, Xie M, Wang C, Wang Y, Peng Y, Nie X. Effects of atorvastatin on the Sirtuin/PXR signaling pathway in Mugilogobius chulae. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:60009-60022. [PMID: 37016258 DOI: 10.1007/s11356-023-26736-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 03/27/2023] [Indexed: 05/10/2023]
Abstract
Atorvastatin (ATV) is a hypolipidemic drug widely detected in the aquatic environment. Nevertheless, limited information is provided about the toxic effects of ATV on estuary or coastal species and the underlying mechanisms. In the present study, the responses of genes expression in pregnane X receptor (PXR) signaling pathway and enzymatic activities in the liver of the estuarine benthic fish (Mugilogobius chulae) were investigated under acute and sub-chronic ATV exposure. Results showed that PXR was significantly inhibited in the highest exposure concentration of ATV for a shorter time (24 h, 500 μg L-1) but induced in a lower concentration (72 h, 5 μg L-1). The downstream genes in PXR signaling pathway such as CYP3A, SULT, UGT, and GST showed similar trends to PXR. P-gp and MRP1 were repressed in most treatments. GCLC associated with GSH synthesis was mostly induced under ATV exposure for a long time (168 h), suggesting that reactive oxygen species (ROS) were generated under ATV exposure. Similarly, GST and SOD enzymatic activities significantly increased in most exposure treatments. Under ATV exposure, SIRT1 and SIRT2 displayed induction to some extent in most treatments, suggesting that SIRTs may affect PXR expression by regulating the acetylation levels of PXR. The investigation demonstrated that ATV exposure affected the expression of the Sirtuin/PXR signaling pathway, thus further interfered adaption of M. chulae to the environment.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Meinan Xie
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Chao Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Ying Peng
- Research and Development Center for Watershed Environmental Eco-Engineering, Beijing Normal University, Zhuhai, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
14
|
Liang R, Feng X, Shi D, Wang B, Zhang Y, Liu W, Yu L, Ye Z, Zhou M, Chen W. Obesity modifies the association of environmental pyrethroid exposure with glucose homeostasis in the US general adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 328:121671. [PMID: 37080515 DOI: 10.1016/j.envpol.2023.121671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/02/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Environmental pyrethroids are concerning due to their widespread residues and potential implications on human health. We aimed to assess the association of pyrethroid exposure with glucose homeostasis and examine the interaction between obesity and pyrethroid exposure. A total of 4233 US general adults from the National Health and Nutrition Examination Survey with measured urinary pyrethroid metabolites, fasting plasma glucose (FPG), fasting insulin (FINS), and glycated hemoglobin A1c (HbA1c) were included in the study. The homeostasis model assessment (HOMA2) calculator was utilized to assess insulin resistance (HOMA2-IR), insulin sensitivity (HOMA2-IS), and beta-cell function (HOMA2-β). We estimated the associations of pyrethroid metabolites with glucose homeostasis parameters (FPG, FINS, HbA1c, HOMA2-IR, HOMA2-IS, and HOMA2-β) using multivariate linear regression models and restricted cubic spline models and further assessed the interaction between obesity and pyrethroid metabolites on glucose dyshomeostasis. Urinary 3-phenoxybenzoic acid (3-PBA) was the most detected pyrethroid metabolite (81%) with a median concentration of 0.43 (interquartile range 0.20-1.01) μg/g urinary creatinine. Compared with the participants in the lowest quartile, those in the highest quartile of 3-PBA had a 1.93% (95% confidence interval: 0.46%, 3.42%), 6.69% (1.96%, 11.64%), 1.60% (0.64%, 2.57%), 7.06% (2.33%, 12.01%), -6.59% (-10.72%, -2.28%), and 1.10% (-2.69%, 5.04%) alteration in FPG, FINS, HbA1c, HOMA2-IR, HOMA2-IS, and HOMA2-β, respectively. The restricted cubic spline model displayed a linear positive association between 3-PBA and FPG, FINS, HbA1c, and HOMA2-IR, and a negative association with HOMA2-IS (all P for overall <0.05 and P for non-linear >0.05). Additionally, the association between urinary 3-PBA and FPG was modified by general obesity (P for interaction <0.05), with a more pronounced association observed in obese participants than in non-obese participants. Our findings suggested that pyrethroid exposure was associated with glucose dyshomeostasis. General obesity significantly heightened the association between pyrethroid exposure and increased FPG level.
Collapse
Affiliation(s)
- Ruyi Liang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaobing Feng
- Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da Shi
- Food and Human Nutritional Science, Faculty of Agriculture and Food Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yongfang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Linling Yu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zi Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
15
|
Guo YR, Liu XM, Wang GX. Exposure to proton pump inhibitors and risk of diabetes: A systematic review and meta-analysis. World J Diabetes 2023; 14:120-129. [PMID: 36926660 PMCID: PMC10011897 DOI: 10.4239/wjd.v14.i2.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Exposure to proton pump inhibitors (PPIs) has been reported to have a potential role in the development of diabetes.
AIM To determine the association between PPIs and diabetes.
METHODS This meta-analysis is registered on PROSPERO (CRD42022352704). In August 2022, eligible studies were identified through a comprehensive literature search. In this study, odds ratios were combined with 95% confidence intervals using a random-effects model. The source of heterogeneity was assessed using sensitivity analysis and subgroup analysis. The publication bias was evaluated using Egger’s test and Begg’s test.
RESULTS The meta-analysis included 9 studies with a total of 867185 participants. Results showed that the use of PPIs increased the risk of diabetes (odds ratio = 1.23, 95% confidence interval: 1.05-1.43, n = 9, I2 = 96.3%). Subgroup analysis showed that geographic location and study type had significant effects on the overall results. Both Egger’s and Begg’s tests showed no publication bias (P > 0.05). Sensitivity analysis also confirmed the stability of the results.
CONCLUSION The results of this study indicated that the use of PPIs was related to an increased risk of diabetes. However, more well-designed studies are needed to verify these results in the future.
Collapse
Affiliation(s)
- Yun-Ran Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Xin-Ming Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Gui-Xia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130000, Jilin Province, China
| |
Collapse
|
16
|
Zhang J, Jia Q, Li Y, He J. The Function of Xenobiotic Receptors in Metabolic Diseases. Drug Metab Dispos 2023; 51:237-248. [PMID: 36414407 DOI: 10.1124/dmd.122.000862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 09/01/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic diseases are a series of metabolic disorders that include obesity, diabetes, insulin resistance, hypertension, and hyperlipidemia. The increased prevalence of metabolic diseases has resulted in higher mortality and mobility rates over the past decades, and this has led to extensive research focusing on the underlying mechanisms. Xenobiotic receptors (XRs) are a series of xenobiotic-sensing nuclear receptors that regulate their downstream target genes expression, thus defending the body from xenobiotic and endotoxin attacks. XR activation is associated with the development of a number of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes, and cardiovascular diseases, thus suggesting an important role for XRs in modulating metabolic diseases. However, the regulatory mechanism of XRs in the context of metabolic disorders under different nutrient conditions is complex and remains controversial. This review summarizes the effects of XRs on different metabolic components (cholesterol, lipids, glucose, and bile acids) in different tissues during metabolic diseases. As chronic inflammation plays a critical role in the initiation and progression of metabolic diseases, we also discuss the impact of XRs on inflammation to comprehensively recognize the role of XRs in metabolic diseases. This will provide new ideas for treating metabolic diseases by targeting XRs. SIGNIFICANCE STATEMENT: This review outlines the current understanding of xenobiotic receptors on nutrient metabolism and inflammation during metabolic diseases. This work also highlights the gaps in this field, which can be used to direct the future investigations on metabolic diseases treatment by targeting xenobiotic receptors.
Collapse
Affiliation(s)
- Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy (J.Z., Y.L., J.H.) and Department of Endocrinology and Metabolism (Q.J.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Wang J, Lu P, Xie W. Atypical functions of xenobiotic receptors in lipid and glucose metabolism. MEDICAL REVIEW (2021) 2022; 2:611-624. [PMID: 36785576 PMCID: PMC9912049 DOI: 10.1515/mr-2022-0032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/08/2022] [Indexed: 12/02/2022]
Abstract
Xenobiotic receptors are traditionally defined as xenobiotic chemical-sensing receptors, the activation of which transcriptionally regulates the expression of enzymes and transporters involved in the metabolism and disposition of xenobiotics. Emerging evidence suggests that "xenobiotic receptors" also have diverse endobiotic functions, including their effects on lipid metabolism and energy metabolism. Dyslipidemia is a major risk factor for cardiovascular disease, diabetes, obesity, metabolic syndrome, stroke, nonalcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH). Understanding the molecular mechanism by which transcriptional factors, including the xenobiotic receptors, regulate lipid homeostasis will help to develop preventive and therapeutic approaches. This review describes recent advances in our understanding the atypical roles of three xenobiotic receptors: aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR), and constitutive androstane receptor (CAR), in metabolic disorders, with a particular focus on their effects on lipid and glucose metabolism. Collectively, the literatures suggest the potential values of AhR, PXR and CAR as therapeutic targets for the treatment of NAFLD, NASH, obesity and diabetes, and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingyuan Wang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peipei Lu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Peng Q, Hao LY, Guo YL, Zhang ZQ, Ji JM, Xue Y, Liu YW, Lu JL, Li CG, Shi XL. Solute carrier family 2 members 1 and 2 as prognostic biomarkers in hepatocellular carcinoma associated with immune infiltration. World J Clin Cases 2022; 10:3989-4019. [PMID: 35665115 PMCID: PMC9131213 DOI: 10.12998/wjcc.v10.i13.3989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/17/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metabolic reprogramming has been identified as a core hallmark of cancer. Solute carrier family 2 is a major glucose carrier family. It consists of 14 members, and we mainly study solute carrier family 2 member 1 (SLC2A1) and solute carrier family 2 member 2 (SLC2A2) here. SLC2A1, mainly existing in human erythrocytes, brain endothelial cells, and normal placenta, was found to be increased in hepatocellular carcinoma (HCC), while SLC2A2, the major transporter of the normal liver, was decreased in HCC. AIM To identify if SLC2A1 and SLC2A2 were associated with immune infiltration in addition to participating in the metabolic reprogramming in HCC. METHODS The expression levels of SLC2A1 and SLC2A2 were tested in HepG2 cells, HepG215 cells, and multiple databases. The clinical characteristics and survival data of SLC2A1 and SLC2A2 were examined by multiple databases. The correlation between SLC2A1 and SLC2A2 was analyzed by multiple databases. The functions and pathways in which SLC2A1, SLC2A2, and frequently altered neighbor genes were involved were discussed in String. Immune infiltration levels and immune marker genes associated with SLC2A1 and SLC2A2 were discussed from multiple databases. RESULTS The expression level of SLC2A1 was up-regulated, but the expression level of SLC2A2 was down-regulated in HepG2 cells, HepG215 cells, and liver cancer patients. The expression levels of SLC2A1 and SLC2A2 were related to tumor volume, grade, and stage in HCC. Interestingly, the expression levels of SLC2A1 and SLC2A2 were negatively correlated. Further, high SLC2A1 expression and low SLC2A2 expression were linked to poor overall survival and relapse-free survival. SLC2A1, SLC2A2, and frequently altered neighbor genes played a major role in the occurrence and development of tumors. Notably, SLC2A1 was positively correlated with tumor immune infiltration, while SLC2A2 was negatively correlated with tumor immune infiltration. Particularly, SLC2A2 methylation was positively correlated with lymphocytes. CONCLUSION SLC2A1 and SLC2A2 are independent therapeutic targets for HCC, and they are quintessential marker molecules for predicting and regulating the number and status of immune cells in HCC.
Collapse
Affiliation(s)
- Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Li-Yuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Ying-Lin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Zhi-Qin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Jing-Min Ji
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Yu Xue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Yi-Wei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Jun-Lan Lu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Cai-Ge Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Xin-Li Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| |
Collapse
|
19
|
Liu P, Jiang L, Kong W, Xie Q, Li P, Liu X, Zhang J, Liu M, Wang Z, Zhu L, Yang H, Zhou Y, Zou J, Liu X, Liu L. PXR activation impairs hepatic glucose metabolism partly via inhibiting the HNF4 α-GLUT2 pathway. Acta Pharm Sin B 2022; 12:2391-2405. [PMID: 35646519 PMCID: PMC9136535 DOI: 10.1016/j.apsb.2021.09.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/05/2021] [Accepted: 09/16/2021] [Indexed: 01/20/2023] Open
Abstract
Drug-induced hyperglycemia/diabetes is a global issue. Some drugs induce hyperglycemia by activating the pregnane X receptor (PXR), but the mechanism is unclear. Here, we report that PXR activation induces hyperglycemia by impairing hepatic glucose metabolism due to inhibition of the hepatocyte nuclear factor 4-alpha (HNF4α)‒glucose transporter 2 (GLUT2) pathway. The PXR agonists atorvastatin and rifampicin significantly downregulated GLUT2 and HNF4α expression, and impaired glucose uptake and utilization in HepG2 cells. Overexpression of PXR downregulated GLUT2 and HNF4α expression, while silencing PXR upregulated HNF4α and GLUT2 expression. Silencing HNF4α decreased GLUT2 expression, while overexpressing HNF4α increased GLUT2 expression and glucose uptake. Silencing PXR or overexpressing HNF4α reversed the atorvastatin-induced decrease in GLUT2 expression and glucose uptake. In human primary hepatocytes, atorvastatin downregulated GLUT2 and HNF4α mRNA expression, which could be attenuated by silencing PXR. Silencing HNF4α downregulated GLUT2 mRNA expression. These findings were reproduced with mouse primary hepatocytes. Hnf4α plasmid increased Slc2a2 promoter activity. Hnf4α silencing or pregnenolone-16α-carbonitrile (PCN) suppressed the Slc2a2 promoter activity by decreasing HNF4α recruitment to the Slc2a2 promoter. Liver-specific Hnf4α deletion and PCN impaired glucose tolerance and hepatic glucose uptake, and decreased the expression of hepatic HNF4α and GLUT2. In conclusion, PXR activation impaired hepatic glucose metabolism partly by inhibiting the HNF4α‒GLUT2 pathway. These results highlight the molecular mechanisms by which PXR activators induce hyperglycemia/diabetes.
Collapse
|
20
|
Niu X, Wu T, Li G, Gu X, Tian Y, Cui H. Insights into the critical role of the PXR in preventing carcinogenesis and chemotherapeutic drug resistance. Int J Biol Sci 2022; 18:742-759. [PMID: 35002522 PMCID: PMC8741843 DOI: 10.7150/ijbs.68724] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Pregnane x receptor (PXR) as a nuclear receptor is well-established in drug metabolism, however, it has pleiotropic functions in regulating inflammatory responses, glucose metabolism, and protects normal cells against carcinogenesis. Most studies focus on its transcriptional regulation, however, PXR can regulate gene expression at the translational level. Emerging evidences have shown that PXR has a broad protein-protein interaction network, by which is implicated in the cross signaling pathways. Furthermore, the interactions between PXR and some critical proteins (e.g., p53, Tip60, p300/CBP-associated factor) in DNA damage pathway highlight its potential roles in this field. A thorough understanding of how PXR maintains genome stability and prevents carcinogenesis will help clinical diagnosis and finally benefit patients. Meanwhile, due to the regulation of CYP450 enzymes CYP3A4 and multidrug resistance protein 1 (MDR1), PXR contributes to chemotherapeutic drug resistance. It is worthy of note that the co-factor of PXR such as RXRα, also has contributions to this process, which makes the PXR-mediated drug resistance more complicated. Although single nucleotide polymorphisms (SNPs) vary between individuals, the amino acid substitution on exon of PXR finally affects PXR transcriptional activity. In this review, we have summarized the updated mechanisms that PXR protects the human body against carcinogenesis, and major contributions of PXR with its co-factors have made on multidrug resistance. Furthermore, we have also reviewed the current promising antagonist and their clinic applications in reversing chemoresistance. We believe our review will bring insight into PXR-targeted cancer therapy, enlighten the future study direction, and provide substantial evidence for the clinic in future.
Collapse
Affiliation(s)
- Xiaxia Niu
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Gege Li
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| | - Xinsheng Gu
- Department of Pharmacology, College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, USA
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, 730000, Lanzhou, China
| |
Collapse
|
21
|
Lv Y, Luo YY, Ren HW, Li CJ, Xiang ZX, Luan ZL. The role of pregnane X receptor (PXR) in substance metabolism. Front Endocrinol (Lausanne) 2022; 13:959902. [PMID: 36111293 PMCID: PMC9469194 DOI: 10.3389/fendo.2022.959902] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
As a member of the nuclear receptor (NR) superfamily, pregnane X receptor (PXR; NR1I2) is a ligand-activated transcription factor that plays a crucial role in the metabolism of xenobiotics and endobiotics in mammals. The tissue distribution of PXR is parallel to its function with high expression in the liver and small intestine and moderate expression in the kidney, stomach, skin, and blood-brain barrier, which are organs and tissues in frequent contact with xenobiotics. PXR was first recognized as an exogenous substance receptor regulating metabolizing enzymes and transporters and functioning in detoxification and drug metabolism in the liver. However, further research revealed that PXR acts as an equally important endogenous substance receptor in the metabolism and homeostasis of endogenous substances. In this review, we summarized the functions of PXR in metabolism of different substances such as glucose, lipid, bile acid, vitamin, minerals, and endocrines, and also included insights of the application of PXR ligands (drugs) in specific diseases.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian Medical University, Dalian, China
- *Correspondence: Zhi-Lin Luan,
| |
Collapse
|
22
|
Yu X, Xu M, Meng X, Li S, Liu Q, Bai M, You R, Huang S, Yang L, Zhang Y, Jia Z, Zhang A. Nuclear receptor PXR targets AKR1B7 to protect mitochondrial metabolism and renal function in AKI. Sci Transl Med 2021; 12:12/543/eaay7591. [PMID: 32404507 DOI: 10.1126/scitranslmed.aay7591] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 03/20/2020] [Indexed: 12/20/2022]
Abstract
Acute kidney injury (AKI) is a worldwide public health problem with no specific and satisfactory therapies in clinic. The nuclear pregnane X receptor (PXR) is involved in the progression of multiple diseases, including metabolic diseases, atherosclerosis, hypertension, liver injury, etc. However, its role in kidney injury remains to be understood. In this study, we have investigated the role of PXR in AKI and underlying mechanism(s) involved in its function. PXR was robustly down-regulated and negatively correlated with renal dysfunction in human and animal kidneys with AKI. Silencing PXR in rats enhanced cisplatin-induced AKI and induced severe mitochondrial abnormalities, whereas activating PXR protected against AKI. Using luciferase reporter assays, genomic manipulation, and proteomics data analysis on the kidneys of PXR-/- rats, we determined that PXR targeted Aldo-keto reductase family 1, member B7 (AKR1B7) to improve mitochondrial function, thereby ameliorating AKI. We confirmed the protective role of PXR against kidney injury using genomic and pharmacologic approaches in an ischemia/reperfusion model of AKI. These findings demonstrate that disabling the PXR/AKR1B7/mitochondrial metabolism axis is an important factor that can contribute to AKI, whereas reestablishing this axis can be useful for treating AKI.
Collapse
Affiliation(s)
- Xiaowen Yu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Man Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Xia Meng
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Shumin Li
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Ran You
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Li Yang
- Department of Nephrology, Peking University First Hospital, Beijing 100034, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China. .,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China. .,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China. .,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
23
|
Nabil H, Kummu O, Lehenkari P, Rysä J, Risteli J, Hakkola J, Hukkanen J. Rifampicin induces the bone form of alkaline phosphatase in humans. Basic Clin Pharmacol Toxicol 2021; 130 Suppl 1:81-94. [PMID: 33851518 DOI: 10.1111/bcpt.13586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022]
Abstract
Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor that regulates drug metabolism in the liver and intestine. In our clinical trials on healthy volunteers to discover novel metabolic functions of PXR activation, we observed that rifampicin, a well-established ligand for human PXR, 600 mg daily for a week, increased the plasma alkaline phosphatase (ALP) significantly compared with the placebo. Further analysis with lectin affinity electrophoresis revealed that especially the bone form of ALP was elevated. To investigate the mechanism(s) of bone ALP induction, we employed osteoblast lineage differentiated from human primary bone marrow-derived mesenchymal stromal cells. Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN and OPG). PXR expression was detected in the cells, but the expression was very low compared with the human liver. To further investigate the potential role of PXR in the ALP induction, we treated mice and rats with a rodent PXR ligand pregnenolone 16α-carbonitrile (PCN). However, PCN treatment did not increase plasma ALP activity or bone ALP mRNA expression. In conclusion, rifampicin treatment induces the bone form of ALP in the serum of healthy human volunteers. Further studies are required to establish the mechanism of this novel finding.
Collapse
Affiliation(s)
- Heba Nabil
- Research Unit of Biomedicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Petri Lehenkari
- Cancer Research and Translational Medicine Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Juha Risteli
- Cancer Research and Translational Medicine Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Nordlab, Oulu University Hospital, Oulu, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Janne Hukkanen
- Biocenter Oulu, University of Oulu, Oulu, Finland.,Research Unit of Internal Medicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
24
|
Cai X, Young GM, Xie W. The xenobiotic receptors PXR and CAR in liver physiology, an update. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166101. [PMID: 33600998 DOI: 10.1016/j.bbadis.2021.166101] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/25/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022]
Abstract
Pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are two nuclear receptors that are well-known for their roles in xenobiotic detoxification by regulating the expression of drug-metabolizing enzymes and transporters. In addition to metabolizing drugs and other xenobiotics, the same enzymes and transporters are also responsible for the production and elimination of numerous endogenous chemicals, or endobiotics. Moreover, both PXR and CAR are highly expressed in the liver. As such, it is conceivable that PXR and CAR have major potentials to affect the pathophysiology of the liver by regulating the homeostasis of endobiotics. In recent years, the physiological functions of PXR and CAR in the liver have been extensively studied. Emerging evidence has suggested the roles of PXR and CAR in energy metabolism, bile acid homeostasis, cell proliferation, to name a few. This review summarizes the recent progress in our understanding of the roles of PXR and CAR in liver physiology.
Collapse
Affiliation(s)
- Xinran Cai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gregory M Young
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
25
|
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: 11] [Impact Index Per Article: 2.2] [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
|
26
|
Negishi M, Kobayashi K, Sakuma T, Sueyoshi T. Nuclear receptor phosphorylation in xenobiotic signal transduction. J Biol Chem 2020; 295:15210-15225. [PMID: 32788213 DOI: 10.1074/jbc.rev120.007933] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
Nuclear pregnane X receptor (PXR, NR1I2) and constitutive active/androstane receptor (CAR, NR1I3) are nuclear receptors characterized in 1998 by their capability to respond to xenobiotics and activate cytochrome P450 (CYP) genes. An anti-epileptic drug, phenobarbital (PB), activates CAR and its target CYP2B genes, whereas PXR is activated by drugs such as rifampicin and statins for the CYP3A genes. Inevitably, both nuclear receptors have been investigated as ligand-activated nuclear receptors by identifying and characterizing xenobiotics and therapeutics that directly bind CAR and/or PXR to activate them. However, PB, which does not bind CAR directly, presented an alternative research avenue for an indirect ligand-mediated nuclear receptor activation mechanism: phosphorylation-mediated signal regulation. This review summarizes phosphorylation-based mechanisms utilized by xenobiotics to elicit cell signaling. First, the review presents how PB activates CAR (and other nuclear receptors) through a conserved phosphorylation motif located between two zinc fingers within its DNA-binding domain. PB-regulated phosphorylation at this motif enables nuclear receptors to form communication networks, integrating their functions. Next, the review discusses xenobiotic-induced PXR activation in the absence of the conserved DNA-binding domain phosphorylation motif. In this case, phosphorylation occurs at a motif located within the ligand-binding domain to transduce cell signaling that regulates hepatic energy metabolism. Finally, the review delves into the implications of xenobiotic-induced signaling through phosphorylation in disease development and progression.
Collapse
Affiliation(s)
- Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | - Kaoru Kobayashi
- Department of Biopharmaceutics, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Tsutomu Sakuma
- School of Pharmaceutical Sciences, Ohu University, Koriyama, Fukushima, Japan
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
27
|
Torres-Vergara P, Ho YS, Espinoza F, Nualart F, Escudero C, Penny J. The constitutive androstane receptor and pregnane X receptor in the brain. Br J Pharmacol 2020; 177:2666-2682. [PMID: 32201941 DOI: 10.1111/bph.15055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/16/2022] Open
Abstract
Since their discovery, the orphan nuclear receptors constitutive androstane receptor (CAR;NR1I3) and pregnane X receptor (PXR;NR1I2) have been regarded as master regulators of drug disposition and detoxification mechanisms. They regulate the metabolism and transport of endogenous mediators and xenobiotics in organs including the liver, intestine and brain. However, with proposals of new physiological functions for NR1I3 and NR1I2, there is increasing interest in the role of these receptors in influencing brain function. This review will summarise key findings regarding the expression and function of NR1I3 and NR1I2 in the brain, hereby highlighting the need for further research in this field.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.,Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Universidad del Bío Bío, Chillán, Chile
| | - Yu Siong Ho
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| | - Francisca Espinoza
- Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Francisco Nualart
- Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Laboratorio de FisiologíaVascular, Departamento de Ciencias Básicas, Facultad de Ciencias Básicas, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Universidad del Bío Bío, Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| |
Collapse
|
28
|
Xing Y, Yan J, Niu Y. PXR: a center of transcriptional regulation in cancer. Acta Pharm Sin B 2020; 10:197-206. [PMID: 32082968 PMCID: PMC7016272 DOI: 10.1016/j.apsb.2019.06.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/10/2019] [Accepted: 06/20/2019] [Indexed: 02/05/2023] Open
Abstract
Pregnane X receptor (PXR, NR1I2) is a prototypical member of the nuclear receptor superfamily. PXR can be activated by both endobiotics and xenobiotics. As a key xenobiotic receptor, the cellular function of PXR is mostly exerted by its binding to the regulatory gene sequences in a ligand-dependent manner. Classical downstream target genes of PXR participate in xenobiotic responses, such as detoxification, metabolism and inflammation. Emerging evidence also implicates PXR signaling in the processes of apoptosis, cell cycle arrest, proliferation, angiogenesis and oxidative stress, which are closely related to cancer. Here, we discussed, in addition to the characterization of PXR per se, the biological function and regulatory mechanism of PXR signaling in cancer, and its potential for the targeted prevention and therapeutics.
Collapse
Affiliation(s)
- Yaqi Xing
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Jiong Yan
- Center for Pharmacogenetics, University of Pittsburgh, PA 15261, USA
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
- Corresponding author.
| |
Collapse
|
29
|
Tu J, Yang Y, Zhang J, Lu G, Ke L, Tong Z, Kasimu M, Hu D, Xu Q, Li W. Regulatory effect of chemerin and therapeutic efficacy of chemerin‑9 in pancreatogenic diabetes mellitus. Mol Med Rep 2020; 21:981-988. [PMID: 31922216 PMCID: PMC7002998 DOI: 10.3892/mmr.2020.10915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022] Open
Abstract
Chemerin is a novel adipokine that regulates immune responses, adipocyte differentiation, and glucose metabolism. However, the role of chemerin in pancreatogenic diabetes mellitus (PDM) remains unknown. PDM is recognized as DM occurring secondary to chronic pancreatitis or pancreatic resection due to the loss of the loss of islet cell mass. The aim of the present study was to investigate the role of chemerin in PDM by collecting blooding samples from DM patients and establishing in vivo PDM model. The present study demonstrated that chemerin levels are decreased in the serum of patients with PDM and are negatively associated with the insulin resistance (IR) status. Chemerin levels also decreased during the development of PDM in C57BL/6 mice, together with increasing serum levels of interleukin-1 and tumor necrosis factor-α and decreasing mRNA expression levels of glucose transporter 2 (GLUT2) and pancreatic and duodenal homeobox 1 (PDX1). Treatment of PDM model mice with chemerin chemokine-like receptor 1 (CMKLR1) agonist, chemerin-9, elevated the serum levels of chemerin and mRNA expression levels of GLUT2 and PDX1, leading to the alleviation of glucose intolerance and IR in these animals. Together, the accumulated data indicated that chemerin may exert a protective function in PDM, perhaps by regulating perhaps by regulating GLUT2 and PDX1 expression, and that the restoration of the chemerin/CMKLR1 pathway may represent a novel therapeutic strategy for PDM.
Collapse
Affiliation(s)
- Jianfeng Tu
- Department of General Surgery, Akesu First People's Hospital, Akesu, Xinjiang 843000, P.R. China
| | - Yue Yang
- Hangzhou Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Jingzhu Zhang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Guotao Lu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Lu Ke
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Zhihui Tong
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Maimaitijiang Kasimu
- Department of General Surgery, Akesu First People's Hospital, Akesu, Xinjiang 843000, P.R. China
| | - Dejun Hu
- Department of General Surgery, Akesu First People's Hospital, Akesu, Xinjiang 843000, P.R. China
| | - Qiuran Xu
- Department of Emergency Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Weiqin Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
30
|
Hassani-Nezhad-Gashti F, Kummu O, Karpale M, Rysä J, Hakkola J. Nutritional status modifies pregnane X receptor regulated transcriptome. Sci Rep 2019; 9:16728. [PMID: 31723190 PMCID: PMC6853963 DOI: 10.1038/s41598-019-53101-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 10/24/2019] [Indexed: 12/25/2022] Open
Abstract
Pregnane X receptor (PXR) regulates glucose and lipid metabolism, but little is known of the nutritional regulation of PXR function. We investigated the genome wide effects of the nutritional status on the PXR mediated gene regulation in the liver. Mice were treated with a PXR ligand pregnenolone 16α-carbonitrile (PCN) for 4 days and subsequently either fasted for 5 hours or after 4-hour fast treated with intragastric glucose 1 hour before sample collection. Gene expression microarray study indicated that PCN both induced and repressed much higher number of genes in the glucose fed mice and the induction of multiple well-established PXR target genes was potentiated by glucose. A subset of genes, including bile acid synthesis gene Cyp8b1, responded in an opposite direction during fasting and after glucose feeding. PXR knockout abolished these effects. In agreement with the Cyp8b1 regulation, PCN also modified the bile acid composition in the glucose fed mice. Contribution of glucose, insulin and glucagon on the observed nutritional effects was investigated in primary hepatocytes. However, only mild impact on PXR function was observed. These results show that nutritional status modifies the PXR regulated transcriptome both qualitatively and quantitatively and reveal a complex crosstalk between PXR and energy homeostasis.
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
| | - 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
| | - 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
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, 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
|
31
|
Pu S, Wu X, Yang X, Zhang Y, Dai Y, Zhang Y, Wu X, Liu Y, Cui X, Jin H, Cao J, Li R, Cai J, Cao Q, Hu L, Gao Y. The Therapeutic Role of Xenobiotic Nuclear Receptors Against Metabolic Syndrome. Curr Drug Metab 2019; 20:15-22. [PMID: 29886826 DOI: 10.2174/1389200219666180611083155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/05/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diabetes, with an increased prevalence and various progressive complications, has become a significant global health challenge. The concrete mechanisms responsible for the development of diabetes still remain incompletely unknown, although substantial researches have been conducted to search for the effective therapeutic targets. This review aims to reveal the novel roles of Xenobiotic Nuclear Receptors (XNRs), including the Peroxisome Proliferator-Activated Receptor (PPAR), the Farnesoid X Receptor (FXR), the Liver X Receptor (LXR), the Pregnane X Receptor (PXR) and the Constitutive Androstane Receptor (CAR), in the development of diabetes and provide potential strategies for research and treatment of metabolic diseases. METHODS We retrieved a large number of original data about these five XNRs and organized to focus on their recently discovered functions in diabetes and its complications. RESULTS Increasing evidences have suggested that PPAR, FXR, LXR ,PXR and CAR are involved in the development of diabetes and its complications through different mechanisms, including the regulation of glucose and lipid metabolism, insulin and inflammation response and related others. CONCLUSION PPAR, FXR, LXR, PXR, and CAR, as the receptors for numerous natural or synthetic compounds, may be the most effective therapeutic targets in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Shuqi Pu
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojie Wu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology and Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Yunzhan Zhang
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunkai Dai
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yueling Zhang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, China
| | - Xiaoting Wu
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, China
| | - Yan Liu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xiaona Cui
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Haiyong Jin
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianhong Cao
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruliu Li
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiazhong Cai
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qizhi Cao
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Ling Hu
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Gao
- PI-WEI Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Lynch C, Mackowiak B, Huang R, Li L, Heyward S, Sakamuru S, Wang H, Xia M. Identification of Modulators That Activate the Constitutive Androstane Receptor From the Tox21 10K Compound Library. Toxicol Sci 2019; 167:282-292. [PMID: 30247703 PMCID: PMC6657574 DOI: 10.1093/toxsci/kfy242] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The constitutive androstane receptor (CAR; NR1I3) is a nuclear receptor involved in all phases of drug metabolism and disposition. However, recently it's been implicated in energy metabolism, tumor progression, and cancer therapy as well. It is, therefore, important to identify compounds that induce human CAR (hCAR) activation to predict drug-drug interactions and potential therapeutic usage. In this study, we screen the Tox21 10,000 compound collection to characterize hCAR activators. A potential novel structural cluster of compounds was identified, which included nitazoxanide and tenonitrozole, whereas known structural clusters, such as flavones and prazoles, were also detected. Four compounds, neticonazole, diphenamid, phenothrin, and rimcazole, have been identified as novel hCAR activators, one of which, rimcazole, shows potential selectivity toward hCAR over its sister receptor, the pregnane X receptor (PXR). All 4 compounds translocated hCAR from the cytoplasm into the nucleus demonstrating the first step to CAR activation. Profiling these compounds as hCAR activators would enable an estimation of drug-drug interactions, as well as identify prospective therapeutically beneficial drugs.
Collapse
Affiliation(s)
- Caitlin Lynch
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Ruili Huang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | | | - Srilatha Sakamuru
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Menghang Xia
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
33
|
Mackowiak B, Hodge J, Stern S, Wang H. The Roles of Xenobiotic Receptors: Beyond Chemical Disposition. Drug Metab Dispos 2018; 46:1361-1371. [PMID: 29759961 PMCID: PMC6124513 DOI: 10.1124/dmd.118.081042] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/07/2018] [Indexed: 02/06/2023] Open
Abstract
Over the past 20 years, the ability of the xenobiotic receptors to coordinate an array of drug-metabolizing enzymes and transporters in response to endogenous and exogenous stimuli has been extensively characterized and well documented. The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are the xenobiotic receptors that have received the most attention since they regulate the expression of numerous proteins important to drug metabolism and clearance and formulate a central defensive mechanism to protect the body against xenobiotic challenges. However, accumulating evidence has shown that these xenobiotic sensors also control many cellular processes outside of their traditional realms of xenobiotic metabolism and disposition, including physiologic and/or pathophysiologic responses in energy homeostasis, cell proliferation, inflammation, tissue injury and repair, immune response, and cancer development. This review will highlight recent advances in studying the noncanonical functions of xenobiotic receptors with a particular focus placed on the roles of CAR and PXR in energy homeostasis and cancer development.
Collapse
Affiliation(s)
- Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Jessica Hodge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| |
Collapse
|
34
|
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
|