1
|
Shen Y, Liu J, Yao B, Zhang Y, Huang S, Liang C, Huang J, Tang Y, Wang X. Non-alcoholic fatty liver disease changes the expression and activity of hepatic drug-metabolizing enzymes and transporters in rats. Toxicol Lett 2024; 396:36-47. [PMID: 38663832 DOI: 10.1016/j.toxlet.2024.04.010] [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: 03/13/2024] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases, which can cause serious complications and gradually increase the mortality rate. However, the effects of NAFLD on drug-metabolizing enzymes and transporters remain unclear, which may cause some confusion regarding patient medication. In this study, a NAFLD rat model was constructed by feeding rats with methionine and choline deficiency diets for 6 weeks, and the mRNA and protein levels of drug-metabolizing enzymes and transporter were analyzed by real-time fluorescent quantitative PCR and Western blot, respectively. The activity of drug-metabolizing enzymes was detected by cocktail methods. In the NAFLD rat model, the mRNA expression of phase I enzymes, phase II enzymes, and transporters decreased. At the protein level, only CYP1A1, CYP1B1, CYP2C11, and CYP2J3 presented a decrease. In addition, the activities of CYP1A2, CYP2B1, CYP2C11, CYP2D1, CYP3A2, UGT1A1, UGT1A3, UGT1A6, and UGT1A9 decreased. These changes may be caused by the alteration of FXR, HNF4α, LXRα, LXRβ, PXR, and RXR. In conclusion, NAFLD changes the expression and activity of hepatic drug-metabolizing enzymes and transporters in rats, which may affect drug metabolism and pharmacokinetics. In clinical medication, drug monitoring should be strengthened to avoid potential risks.
Collapse
Affiliation(s)
- Yifei Shen
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yu Tang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
2
|
Acun A, Fan L, Oganesyan R, Uygun KM, Yeh H, Yarmush ML, Uygun BE. Effect of Donor Age and Liver Steatosis on Potential of Decellularized Liver Matrices to be used as a Platform for iPSC-Hepatocyte Culture. Adv Healthc Mater 2024; 13:e2302943. [PMID: 38266310 PMCID: PMC11102338 DOI: 10.1002/adhm.202302943] [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: 09/03/2023] [Revised: 11/13/2023] [Indexed: 01/26/2024]
Abstract
Decellularization of discarded whole livers and their recellularization with patient-specific induced pluripotent stem cells (iPSCs) to develop a functional organ is a promising approach to increasing the donor pool. The effect of extracellular matrix (ECM) of marginal livers on iPSC-hepatocyte differentiation and function has not been shown. To test the effect of donor liver ECM age and steatosis, young and old, as well as no, low, and high steatosis livers, are decellularized. All livers are decellularized successfully. High steatosis livers have fat remaining on the ECM after decellularization. Old donor liver ECM induces lower marker expression in early differentiation stages, compared to young liver ECM, while this difference is closed at later stages and do not affect iPSC-hepatocyte function significantly. High steatosis levels of liver ECM lead to higher albumin mRNA expression and secretion while at later stages of differentiation expression of major cytochrome (CYP) 450 enzymes is highest in low steatosis liver ECM. Both primary human hepatocytes and iPSC-hepatocytes show an increase in fat metabolism marker expression with increasing steatosis levels most likely induced by excess fat remaining on the ECM. Overall, removal of excess fat from liver ECM may be needed for inducing proper hepatic function after recellularization.
Collapse
Affiliation(s)
- Aylin Acun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
- Department of Biomedical Engineering, Widener University, Chester, PA, 19013, USA
| | - Letao Fan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
| | - Ruben Oganesyan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
| | - Korkut M. Uygun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
| | - Heidi Yeh
- Shriners Children’s, Boston, Boston, MA, 02114, USA
| | - Martin L. Yarmush
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Basak E. Uygun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Shriners Children’s, Boston, Boston, MA, 02114, USA
| |
Collapse
|
3
|
Odanga JJ, Anderson SM, Breathwaite EK, Presnell SC, LeCluyse EL, Chen J, Weaver JR. Characterization of diseased primary human hepatocytes in an all-human cell-based triculture system. Sci Rep 2024; 14:6772. [PMID: 38514705 PMCID: PMC10957907 DOI: 10.1038/s41598-024-57463-7] [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: 11/11/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Liver diseases, including NAFLD, are a growing worldwide health concern. Currently, there is a lack of suitable in vitro models that sustain basic primary human hepatocyte (PHH) morphology and functionality while supporting presentation of disease-associated phenotypic characteristics such as lipid accumulation and inflammasome activation. In TruVivo, an all-human triculture system (hTCS), basic metabolic functions were characterized in PHHs isolated from normal or diseased livers during two-weeks of culture. Decreases in albumin and urea levels and CYP3A4 activity were seen in diseased-origin PHHs compared to normal PHHs along with higher CYP2E1 expression. Positive expression of the macrophage markers CD68 and CD163 were seen in the diseased PHH preparations. Elevated levels of the pro-inflammatory cytokines IL-6 and MCP-1 and the fibrotic markers CK-18 and TGF-β were also measured. Gene expression of FASN, PCK1, and G6PC in the diseased PHHs was decreased compared to the normal PHHs. Further characterization revealed differences in lipogenesis and accumulation of intracellular lipids in normal and diseased PHHs when cultured with oleic acid and high glucose. TruVivo represents a promising new platform to study lipogenic mechanisms in normal and diseased populations due to the preservation of phenotypic differences over a prolonged culture period.
Collapse
Affiliation(s)
- Justin J Odanga
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Sharon M Anderson
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Erick K Breathwaite
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Sharon C Presnell
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Edward L LeCluyse
- Research and Development, LifeNet Health LifeSciences, 6 Davis Dr., Research Triangle Park, NC, USA
| | - Jingsong Chen
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Jessica R Weaver
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA.
- LifeSciences Product Development, LifeNet Health, 1864 Concert Drive, Virginia Beach, VA, 23453, USA.
| |
Collapse
|
4
|
Sanchez-Quant E, Richter ML, Colomé-Tatché M, Martinez-Jimenez CP. Single-cell metabolic profiling reveals subgroups of primary human hepatocytes with heterogeneous responses to drug challenge. Genome Biol 2023; 24:234. [PMID: 37848949 PMCID: PMC10583437 DOI: 10.1186/s13059-023-03075-9] [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: 06/08/2022] [Accepted: 09/26/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Xenobiotics are primarily metabolized by hepatocytes in the liver, and primary human hepatocytes are the gold standard model for the assessment of drug efficacy, safety, and toxicity in the early phases of drug development. Recent advances in single-cell genomics demonstrate liver zonation and ploidy as main drivers of cellular heterogeneity. However, little is known about the impact of hepatocyte specialization on liver function upon metabolic challenge, including hepatic metabolism, detoxification, and protein synthesis. RESULTS Here, we investigate the metabolic capacity of individual human hepatocytes in vitro. We assess how chronic accumulation of lipids enhances cellular heterogeneity and impairs the metabolisms of drugs. Using a phenotyping five-probe cocktail, we identify four functional subgroups of hepatocytes responding differently to drug challenge and fatty acid accumulation. These four subgroups display differential gene expression profiles upon cocktail treatment and xenobiotic metabolism-related specialization. Notably, intracellular fat accumulation leads to increased transcriptional variability and diminishes the drug-related metabolic capacity of hepatocytes. CONCLUSIONS Our results demonstrate that, upon a metabolic challenge such as exposure to drugs or intracellular fat accumulation, hepatocyte subgroups display different and heterogeneous transcriptional responses.
Collapse
Affiliation(s)
- Eva Sanchez-Quant
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Maria Lucia Richter
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Maria Colomé-Tatché
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich (TUM), 85354, Freising, Germany.
- Biomedical Center (BMC), Physiological Chemistry, Faculty of Medicine, Ludwig Maximilian University of Munich (LMU), 82152, Munich, Germany.
| | - Celia Pilar Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Zentrum München, 85764, Neuherberg, Germany.
- TUM School of Medicine, Technical University of Munich, Munich (TUM), 80333, Munich, Germany.
| |
Collapse
|
5
|
Basha A, May SC, Anderson RM, Samala N, Mirmira RG. Non-Alcoholic Fatty Liver Disease: Translating Disease Mechanisms into Therapeutics Using Animal Models. Int J Mol Sci 2023; 24:9996. [PMID: 37373143 PMCID: PMC10298283 DOI: 10.3390/ijms24129996] [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: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a range of pathologies arising from fat accumulation in the liver in the absence of excess alcohol use or other causes of liver disease. Its complications include cirrhosis and liver failure, hepatocellular carcinoma, and eventual death. NAFLD is the most common cause of liver disease globally and is estimated to affect nearly one-third of individuals in the United States. Despite knowledge that the incidence and prevalence of NAFLD are increasing, the pathophysiology of the disease and its progression to cirrhosis remain insufficiently understood. The molecular pathogenesis of NAFLD involves insulin resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. Better insight into these molecular pathways would allow for therapies that target specific stages of NAFLD. Preclinical animal models have aided in defining these mechanisms and have served as platforms for screening and testing of potential therapeutic approaches. In this review, we will discuss the cellular and molecular mechanisms thought to contribute to NAFLD, with a focus on the role of animal models in elucidating these mechanisms and in developing therapies.
Collapse
Affiliation(s)
- Amina Basha
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah C. May
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ryan M. Anderson
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Niharika Samala
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Le Moli R, Malandrino P, Russo M, Tumino D, Piticchio T, Naselli A, Rapicavoli V, Belfiore A, Frasca F. Levothyroxine therapy, calculated deiodinases activity and basal metabolic rate in obese or nonobese patients after total thyroidectomy for differentiated thyroid cancer, results of a retrospective observational study. Endocrinol Diabetes Metab 2023; 6:e406. [PMID: 36722311 PMCID: PMC10000637 DOI: 10.1002/edm2.406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Therapy for hypothyroid obese patients is still under definition since the thyrotropin-stimulating hormone (TSH) level is a less reliable marker of euthyroidism than nonobese patients. Indeed, TSH levels positively correlate with body mass index (BMI), and this increase may be a compensatory mechanism aimed at increasing energy expenditure in obese people. In contrast, the correlation of BMI with thyroid hormone levels is not completely clear, and conflicting results have been obtained by several studies. The L-T4 replacement dose is more variable in obese hypothyroid patients than in nonobese patients, and a recent study indicated that the L-T4 replacement dose is related to lean body mass in obese thyroidectomized patients. We aimed to study the correlations of L-T4-administered dose, thyroid hormone levels and TSH secretion with basal metabolic rate (BMR) and total calculated deiodinase activity (GD) in obese and nonobese athyreotic patients. We also looked for individualized L-T4 replacement dose set points to be used in clinical practice. METHODS We studied retrospectively 160 athyreotic patients, 120 nonobese and 40 obese. GD was calculated by SPINA Thyr 4.2, the responsiveness of the hypothalamic/pituitary thyrotrope by Jostel's thyrotropin (TSH) index and BMR by the Mifflin-St. Jeor formula, the interplay of GD and BMR with L-T4, thyroid hormones and TSH index (TSHI) was also evaluated. RESULTS In our study, the L-T4 dose was an independent predictor of GD, and approximately 30% of athyreotic patients under L-T4 therapy had a reduced GD; FT4 levels were higher and negatively modulated by BMR in obese athyreotic patients respect to nonobese, in these patients a T4 to T3 shunt, in terms of TSHI suppression is observed suggesting a defective hypothalamic pituitary T4 to T3 conversion and a resistance to L-T4 replacement therapy. CONCLUSIONS L-t4 dose is the most important predictor of GD, BMR modulates T4 levels in obese athyreotic patients that are resistant to L-T4 replacement therapy.
Collapse
Affiliation(s)
- Rosario Le Moli
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Pasqualino Malandrino
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Marco Russo
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Dario Tumino
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Tommaso Piticchio
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Adriano Naselli
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Valentina Rapicavoli
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| | - Francesco Frasca
- Endocrinology Unit, Department of Clinical and Experimental MedicineUniversity of Catania, Garibaldi Nesima HospitalCataniaItaly
| |
Collapse
|
7
|
Di Ciaula A, Shanmugam H, Ribeiro R, Pina A, Andrade R, Bonfrate L, Raposo JF, Macedo MP, Portincasa P. Liver fat accumulation more than fibrosis causes early liver dynamic dysfunction in patients with non-alcoholic fatty liver disease. Eur J Intern Med 2023; 107:52-59. [PMID: 36344354 DOI: 10.1016/j.ejim.2022.10.024] [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: 08/19/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION In Non-Alcoholic Fatty Liver Disease (NAFLD), events driving early hepatic dysfunction with respect to specific metabolic pathways are still poorly known. METHODS We enrolled 84 subjects with obesity and/or type 2 diabetes (T2D). FibroScan® served to assess NAFLD by controlled attenuation parameter (CAP), and fibrosis by liver stiffness (LS). Patients with LS above 7 kPa were excluded. APRI and FIB-4 were used as additional serum biomarkers of fibrosis. The stable-isotope dynamic breath test was used to assess the hepatic efficiency of portal extraction (as DOB15) and microsomal metabolization (as cPDR30) of orally-administered (13C)-methacetin. RESULTS NAFLD occurred in 45%, 65.9%, and 91.3% of normal weight, overweight, and obese subjects, respectively. Biomarkers of liver fibrosis were comparable across subgroups, and LS was higher in obese, than in normal weight subjects. DOB15 was 23.2 ± 1.5‰ in normal weight subjects, tended to decrease in overweight (19.9 ± 1.0‰) and decreased significantly in obese subjects (16.9 ± 1.3, P = 0.008 vs. normal weight). Subjects with NAFLD had lower DOB15 (18.7 ± 0.9 vs. 22.1 ± 1.2, P = 0.03) but higher LS (4.7 ± 0.1 vs. 4.0 ± 0.2 kPa, P = 0.0003) than subjects without NAFLD, irrespective of fibrosis. DOB15 (but not cPDR30) decreased with increasing degree of NAFLD (R = -0.26; P = 0.01) and LS (R = -0.23, P = 0.03). Patients with T2D showed increased rate of NAFLD than those without T2D but similar LS, DOB15 and cPDR30. CONCLUSIONS Overweight, obesity and liver fat accumulation manifest with deranged portal extraction efficiency of methacetin into the steatotic hepatocyte. This functional alteration occurs early, and irrespective of significant fibrosis and presence of T2D.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Medical School, Piazza Giulio Cesare 11, Bari 70124, Italy
| | - Harshitha Shanmugam
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Medical School, Piazza Giulio Cesare 11, Bari 70124, Italy
| | - Rogério Ribeiro
- Portuguese Diabetes Association-Education and Research Center (APDP-ERC), Lisbon 1150-082, Portugal
| | - Ana Pina
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal
| | - Rita Andrade
- Portuguese Diabetes Association-Education and Research Center (APDP-ERC), Lisbon 1150-082, Portugal
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Medical School, Piazza Giulio Cesare 11, Bari 70124, Italy.
| | - João F Raposo
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal; Portuguese Diabetes Association-Education and Research Center (APDP-ERC), Lisbon 1150-082, Portugal
| | - M Paula Macedo
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal; Portuguese Diabetes Association-Education and Research Center (APDP-ERC), Lisbon 1150-082, Portugal
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Medical School, Piazza Giulio Cesare 11, Bari 70124, Italy.
| |
Collapse
|
8
|
Sun F, Piao M, Zhang X, Zhang S, Wei Z, Liu L, Bu Y, Xu S, Zhao X, Meng X, Yue M. Multi-Omics Analysis of Transcriptomic and Metabolomics Profiles Reveal the Molecular Regulatory Network of Marbling in Early Castrated Holstein Steers. Animals (Basel) 2022; 12:3398. [PMID: 36496924 PMCID: PMC9736081 DOI: 10.3390/ani12233398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
The intramuscular fat (IMF), or so-called marbling, is known as potential determinant of the high quality beef in China, Korea, and Japan. Of the methods that affect IMF content in cattle, castration is markedly regarded as an effective and economical way to improve the deposition of IMF but with little attention to its multi-omics in early-castrated cattle. The aim of this study was to investigate the liver transcriptome and metabolome of early-castrated Holstein cattle and conduct a comprehensive analysis of two omics associated with the IMF deposition using transcriptomics and untargeted metabolomics under different treatments: non−castrated and slaughtered at 16 months of age (GL16), castrated at birth and slaughtered at 16 months of age (YL16), and castrated at birth and slaughtered at 26 months of age (YL26). The untargeted metabolome was analyzed using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The transcriptome of the hepatic genes was analyzed to identify marbling-related genes. Using untargeted metabolomics, the main altered metabolic pathways in the liver of cattle, including those for lipid and amino acid metabolism, were detected in the YL16 group relative to the GL16 and YL26 groups. Significant increases in the presence of betaine, alanine, and glycerol 3-phosphate were observed in the YL16 group (p < 0.05), which might have contributed to the improved beef-marbling production. Compared to the GL16 and YL26 groups, significant increases in the presence of glutathione, acetylcarnitine, and riboflavin but decreases in diethanolamine and 2-hydroxyglutarate were identified in YL16 group (p < 0.05), which might have been beneficial to the beef’s enhanced functional quality. The gene expressions of GLI1 and NUF2 were downregulated and that of CYP3A4 was upregulated in the YL16 group; these results were strongly correlated with the alanine, betaine, and leucine, respectively, in the liver of the cattle. In conclusion, implementation of early castration modified the hepatic metabolites and the related biological pathways by regulating the relevant gene expressions, which could represent a better rearing method for production of high marbled and healthier beef products.
Collapse
Affiliation(s)
- Fang Sun
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Minyu Piao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xinyue Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siqi Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ziheng Wei
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Li Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Ye Bu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Shanshan Xu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiaochuan Zhao
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiangren Meng
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Mengmeng Yue
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| |
Collapse
|
9
|
Nguyen HD, Kim MS. Effects of chemical mixtures on liver function biomarkers in the Korean adult population: thresholds and molecular mechanisms for non-alcoholic fatty liver disease involved. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:78555-78587. [PMID: 35696061 DOI: 10.1007/s11356-022-21090-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/21/2022] [Indexed: 06/15/2023]
Abstract
There is a scarcity of research on the effects of a mixture of chemicals on liver function biomarkers and non-alcoholic fatty liver disease (NAFLD) indices, including FSI, HIS, and FBI-4. Thus, we aimed to explore whether there is an association between chemical mixtures, including 26 chemicals found in blood and urine, liver function biomarkers, and non-alcoholic fatty liver disease (NAFLD) indices in Korean adults. The effects of exposure to chemical mixtures on liver function biomarkers and NAFLD indices were investigated using linear regression models, weighted quantile sum (WQS) regression, quantile g-computation (qgcomp), and Bayesian kernel machine regression (BKMR) among 3669 adults. In silico toxicogenomic data-mining, we evaluated molecular mechanisms associated with NAFLD, including pathways, diseases, genes, miRNAs, and biological processes. The linear regression models showed blood or urine Hg levels were the most important factors associated with AST, ALT, GGT, FSI, and HSI levels, and significant trends were observed for these chemical quartiles (p < 0.01). The WQS index was significantly associated with ALT, GGT, FSI, and HSI. The qgcomp index also found an association between chemicals and AST, ALT, GGT, and FSI. In the BKMR model, the overall effect of the mixture was significantly related to ALT, GGT, FSI, and HSI. In silico analysis, we found mixed chemicals interacted with the CYP1A2 gene and were associated with NAFLD. Seventy-eight percent of interactions were identified as physical interactions in the CYP1A2 gene related to NAFLD. Transcription factor regulation in adipogenesis and lipid metabolic processes are fundamental molecular mechanisms that could be influenced by NAFLD-related mixed chemicals. Cutoff thresholds for chemical exposure levels associated with liver function indicators and NAFLD indices were also reported. The strongest interactions and expression of miRNAs involved in NAFLD development were also identified.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, 57922, Republic of Korea
| | - Min-Sun Kim
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon, Jeonnam, 57922, Republic of Korea.
| |
Collapse
|
10
|
Klyushova LS, Perepechaeva ML, Grishanova AY. The Role of CYP3A in Health and Disease. Biomedicines 2022; 10:2686. [PMID: 36359206 PMCID: PMC9687714 DOI: 10.3390/biomedicines10112686] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
CYP3A is an enzyme subfamily in the cytochrome P450 (CYP) superfamily and includes isoforms CYP3A4, CYP3A5, CYP3A7, and CYP3A43. CYP3A enzymes are indiscriminate toward substrates and are unique in that these enzymes metabolize both endogenous compounds and diverse xenobiotics (including drugs); almost the only common characteristic of these compounds is lipophilicity and a relatively large molecular weight. CYP3A enzymes are widely expressed in human organs and tissues, and consequences of these enzymes' activities play a major role both in normal regulation of physiological levels of endogenous compounds and in various pathological conditions. This review addresses these aspects of regulation of CYP3A enzymes under physiological conditions and their involvement in the initiation and progression of diseases.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, 630117 Novosibirsk, Russia
| | | |
Collapse
|
11
|
Zhang T, Krekels EHJ, Smit C, Knibbe CAJ. Drug pharmacokinetics in the obese population: challenging common assumptions on predictors of obesity-related parameter changes. Expert Opin Drug Metab Toxicol 2022; 18:657-674. [PMID: 36217846 DOI: 10.1080/17425255.2022.2132931] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Obesity is associated with many physiological changes. We review available evidence regarding five commonly accepted assumptions to a priori predict the impact of obesity on drug pharmacokinetics (PK). AREAS COVERED The investigated assumptions are: 1) lean body weight is the preferred descriptor of clearance and dose adjustments; 2) volume of distribution increases for lipophilic, but not for hydrophilic drugs; 3) CYP-3A4 activity is suppressed and UGT activity is increased, implying decreased and increased dose requirements for substrates of these enzyme systems, respectively; 4) glomerular filtration rate is enhanced, necessitating higher doses for drugs cleared through glomerular filtration; 5) drug dosing information from obese adults can be extrapolated to obese adolescents. EXPERT OPINION Available literature contradicts, or at least limits the generalizability, of all five assumptions. Clinical studies should focus on quantifying the impact of duration and severity of obesity on drug PK in adults and adolescents, and also include oral bioavailability and pharmacodynamics in these studies. Physiologically-based PK approaches can be used to predict PK changes for individual drugs, but can also be used to define in general terms based on patient characteristics and drug properties, when certain assumptions can or cannot be expected to be systematically accurate.
Collapse
Affiliation(s)
- Tan Zhang
- Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elke H J Krekels
- Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Cornelis Smit
- Department of Clinical Pharmacy, Antonius Hospital Sneek, The Netherlands
| | - Catherijne A J Knibbe
- Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital Nieuwegein, The Netherlands
| |
Collapse
|
12
|
A Physiologically Based Pharmacokinetic Model to Predict the Impact of Metabolic Changes Associated with Metabolic Associated Fatty Liver Disease on Drug Exposure. Int J Mol Sci 2022; 23:ijms231911751. [PMID: 36233052 PMCID: PMC9570165 DOI: 10.3390/ijms231911751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic associated fatty liver disease (MAFLD) is the most common chronic liver disease, with an estimated prevalence of between 20 and 30% worldwide. Observational data supported by in vitro and pre-clinical animal models of MAFLD suggest meaningful differences in drug disposition in MAFLD patients. This study aimed to build a physiologically based pharmacokinetic (PBPK) model reflecting observed changes in physiological and molecular parameters relevant to drug disposition that are associated with MAFLD. A comprehensive literature review and meta-analysis was conducted to identify all studies describing in vivo physiological changes along with in vitro and pre-clinical model changes in CYP 1A2, 2C9, 2C19, 2D6 and 3A4 protein abundance associated with MAFLD. A MAFLD population profile was constructed in Simcyp (version 19.1) by adapting demographic and physiological covariates from the Sim-Healthy population profile based on a meta-analysis of observed data from the published literature. Simulations demonstrated that single dose and steady state area under the plasma concentration time curve (AUC) for caffeine, clozapine, omeprazole, metoprolol, dextromethorphan and midazolam, but not s-warfarin or rosiglitazone, were increased by >20% in the MAFLD population compared to the healthy control population. These findings indicate that MAFLD patients are likely to be experience meaningfully higher exposure to drugs that are primarily metabolized by CYP 1A2, 2C19, 2D6 and 3A4, but not CYP2C9. Closer monitoring of MAFLD patients using drugs primarily cleared by CYP 1A2, 2C19 and 3A4 is warranted as reduced metabolic activity and increased drug exposure are likely to result in an increased incidence of toxicity in this population.
Collapse
|
13
|
Brecklinghaus T, Albrecht W, Duda J, Kappenberg F, Gründler L, Edlund K, Marchan R, Ghallab A, Cadenas C, Rieck A, Vartak N, Tolosa L, Castell JV, Gardner I, Halilbasic E, Trauner M, Ullrich A, Zeigerer A, Demirci Turgunbayer Ö, Damm G, Seehofer D, Rahnenführer J, Hengstler JG. In vitro/in silico prediction of drug induced steatosis in relation to oral doses and blood concentrations by the Nile Red assay. Toxicol Lett 2022; 368:33-46. [PMID: 35963427 DOI: 10.1016/j.toxlet.2022.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022]
Abstract
The accumulation of lipid droplets in hepatocytes is a key feature of drug-induced liver injury (DILI) and can be induced by a subset of hepatotoxic compounds. In the present study, we optimized and evaluated an in vitro technique based on the fluorescent dye Nile Red, further named Nile Red assay to quantify lipid droplets induced by the exposure to chemicals. The Nile Red assay and a cytotoxicity test (CTB assay) were then performed on cells exposed concentration-dependently to 60 different compounds. Of these, 31 were known to induce hepatotoxicity in humans, and 13 were reported to also cause steatosis. In order to compare in vivo relevant blood concentrations, pharmacokinetic models were established for all compounds to simulate the maximal blood concentrations (Cmax) at therapeutic doses. The results showed that several hepatotoxic compounds induced an increase in lipid droplets at sub-cytotoxic concentrations. To compare how well (1) the cytotoxicity test alone, (2) the Nile Red assay alone, and (3) the combination of the cytotoxicity test and the Nile Red assay (based on the lower EC10 of both assays) allow the differentiation between hepatotoxic and non-hepatotoxic compounds, a previously established performance metric, the Toxicity Separation Index (TSI) was calculated. In addition, the Toxicity Estimation Index (TEI) was calculated to determine how well blood concentrations that cause an increased DILI risk can be estimated for hepatotoxic compounds. Our findings indicate that the combination of both assays improved the TSI and TEI compared to each assay alone. In conclusion, the study demonstrates that inclusion of the Nile Red assay into in vitro test batteries may improve the prediction of DILI compounds.
Collapse
Affiliation(s)
- Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany.
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Julia Duda
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Franziska Kappenberg
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Lisa Gründler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Adrian Rieck
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Nachiket Vartak
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany
| | - Laia Tolosa
- Experimental Hepatology Unit, Health Research Institute La Fe, Valencia, Spain
| | - José V Castell
- Experimental Hepatology Unit, Health Research Institute La Fe, Valencia, Spain; Biochemistry Department, University of Valencia and CIBEREHD
| | | | - Emina Halilbasic
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Anett Ullrich
- Primacyt Cell Culture Technology GmbH, Schwerin, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
| | - Özlem Demirci Turgunbayer
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany; Department of Biology, Faculty of Science, Dicle University, 21280, Diyarbakır, Turkey
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Ardeystrasse 67, 44139 Dortmund, Germany.
| |
Collapse
|
14
|
Xiao J, Li X, Zhou Z, Guan S, Zhuo L, Gao B. Development of an in vitro insulin resistance dissociated model of hepatic steatosis by co-culture system. Biosci Trends 2022; 16:257-266. [PMID: 35965099 DOI: 10.5582/bst.2022.01242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The evidence shows that there is an associated relationship between hepatosteatosis and insulin resistance. While some existing genetic induction animal and patient models challenge this relationship, indicating that hepatosteatosis is dissociated from insulin resistance. However, the molecular mechanisms of this dissociation remain poorly understood due to a lack of available, reliable, and simplistic setup models. Currently, we used primary rat hepatocytes (rHPCs), co-cultured with rat hepatic stellate cells (HSC-T6) or human foreskin fibroblast cells (HFF-1) in stimulation with high insulin and glucose, to develop a model of steatosis charactered as dissociated lipid accumulation from insulin resistance. Oil-Red staining significantly showed intracellular lipid accumulated in the developed model. Gene expression of sterol regulatory element-binding protein 1c (SREBP1c) and elongase of very-long-chain fatty acids 6 (ELOVL6), key genes responsible for lipogenesis, were detected and obviously increased in this model. Inversely, the insulin resistance related genes expression included phosphoenolpyruvate carboxykinase 1 (PCK1), pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4), and glucose-6-phosphatase (G6pase) were decreased, suggesting a dissociation relationship between steatosis and insulin resistance in the developed model. As well, the drug metabolism of this developed model was investigated and showed up-regulation of cytochrome P450 3A (CYP3A) and down-regulation of cytochrome P450 2E1 (CYP2E1) and cytochrome P450 1A2 (CYP1A2). Taken together, those results demonstrate that the in vitro model of dissociated steatosis from insulin resistance was successfully created by our co-cultured cells in high insulin and glucose medium, which will be a potential model for investigating the mechanism of insulin resistance dissociated steatosis, and discovering a novel drug for its treatment.
Collapse
Affiliation(s)
- Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Xiang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zongbao Zhou
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Shuwen Guan
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Lingjian Zhuo
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| |
Collapse
|
15
|
Rey-Bedon C, Banik P, Gokaltun A, Hofheinz O, Yarmush ML, Uygun MK, Usta OB. CYP450 drug inducibility in NAFLD via an in vitro hepatic model: Understanding drug-drug interactions in the fatty liver. Biomed Pharmacother 2022; 146:112377. [PMID: 35062050 PMCID: PMC8792443 DOI: 10.1016/j.biopha.2021.112377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022] Open
Abstract
Drug-drug-interactions (DDIs) occur when a drug alters the metabolic rate, efficacy, and toxicity of concurrently used drugs. While almost 1 in 4 adults now use at least 3 concurrent prescription drugs in the United States, the Non-alcoholic fatty liver disease (NAFLD) prevalence has also risen over 25%. The effect of NALFD on DDIs is largely unknown. NAFLD is characterized by lipid vesicle accumulation in the liver, which can progress to severe steatohepatitis (NASH), fibrosis, cirrhosis, and hepatic carcinoma. The CYP450 enzyme family dysregulation in NAFLD, which might already alter the efficacy and toxicity of drugs, has been partially characterized. Nevertheless, the drug-induced dysregulation of CYP450 enzymes has not been studied in the fatty liver. These changes in enzymatic inducibility during NAFLD, when taking concurrent drugs, could cause unexpected fatalities through inadvertent DDIs. We have, thus, developed an in vitro model to investigate the CYP450 transcriptional regulation in NAFLD. Specifically, we cultured primary human hepatocytes in a medium containing free fatty acids, high glucose, and insulin for seven days. These cultures displayed intracellular macro-steatosis after 5 days and cytokine secretion resembling NAFLD patients. We further verified the model's dysregulation in the transcription of key CYP450 enzymes. We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. We, thus, conclude that the fatty liver could cause aggravated drug-drug interactions in NAFLD or NASH patients related to CYP2B6 and CYP2C9 enzymes.
Collapse
Affiliation(s)
- Camilo Rey-Bedon
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, United States
| | - Peony Banik
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - Aslihan Gokaltun
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Department of Chemical Engineering, Hacettepe University, 06532 Beytepe, Ankara, Turkey
| | - O Hofheinz
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - Martin L Yarmush
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, United States
| | - M Korkut Uygun
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States
| | - O Berk Usta
- Center for Engineering in Medicine and Surgery at Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States; Shriners Hospitals for Children, Boston, MA 02114, United States.
| |
Collapse
|
16
|
Zarezadeh M, Saedisomeolia A, Shekarabi M, Khorshidi M, Emami MR, Müller DJ. The effect of obesity, macronutrients, fasting and nutritional status on drug-metabolizing cytochrome P450s: a systematic review of current evidence on human studies. Eur J Nutr 2021; 60:2905-2921. [PMID: 33141242 DOI: 10.1007/s00394-020-02421-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 10/19/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cytochrome P450s (CYPs) are a class of hemoproteins involved in drug metabolism. It has been reported that body composition, proportion of dietary macronutrients, fasting and nutritional status can interfere with the activity of drug-metabolizing CYPs. OBJECTIVES The present systematic review was conducted to summarize the effect of obesity, weight reduction, macronutrients, fasting and malnutrition on the CYP-mediated drug metabolism. METHODS PubMed (Medline), Scopus, Embase and Cochrane Library databases and Google Scholar were searched up to June 2020 to obtain relevant studies. The PRISMA guidelines were employed during all steps. Two reviewers independently extracted the information from the included studies. Studies investigating CYPs activity directly or indirectly through pharmacokinetics of probe drugs, were included. Increase in clearance (CL) or decrease in elimination half-life (t½) and area under the curve (AUC) of probe drugs were considered as increase in CYPs activity. RESULTS A total of 6545 articles were obtained through searching databases among which 69 studies with 126 datasets fully met the inclusion criteria. The results indicated that obesity might decrease the activity of CYP3A4/5, CYP1A2 and CYP2C9 and increase the activity of CYP2E1. The effect of obesity on CYP2D6 is controversial. Also, weight loss increased CYP3A4 activity. Moreover, CYP1A2 activity was decreased by high carbohydrate diet, increased by high protein diet and fasting and unchanged by malnutrition. The activity of CYP2C19 was less susceptible to alterations compared to other CYPs. CONCLUSION The activity of drug-metabolizing CYPs are altered by body composition, dietary intake and nutritional status. This relationship might contribute to drug toxicity or reduce treatment efficacy and influence cost-effectiveness of medical care.
Collapse
Affiliation(s)
- Meysam Zarezadeh
- Department of Clinical Nutrition, Student Research Committee, Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Ahmad Saedisomeolia
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
- School of Medicine, Western Sydney University, Sydney, NSW, 2560, Australia.
| | - Mahoor Shekarabi
- Faculty of Medical Sciences and Technologies, Science and Research Branch, Azad University, Tehran, Iran
| | - Masoud Khorshidi
- Student's Research Committee, Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Emami
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Daniel J Müller
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
17
|
Zhang R, Xu D, Zhang Y, Wang R, Yang N, Lou Y, Zhao H, Aa J, Wang G, Xie Y. Silybin Restored CYP3A Expression through the Sirtuin 2/Nuclear Factor κ-B Pathway in Mouse Nonalcoholic Fatty Liver Disease. Drug Metab Dispos 2021; 49:770-779. [PMID: 34183378 DOI: 10.1124/dmd.121.000438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Silybin is widely used as a hepatoprotective agent in various liver disease therapies and has been previously identified as a CYP3A inhibitor. However, little is known about the effect of silybin on CYP3A and the regulatory mechanism during high-fat-diet (HFD)-induced liver inflammation. In our study, we found that silybin restored CYP3A expression and activity that were decreased by HFD and conditioned medium (CM) from palmitate-treated Kupffer cells. Moreover, silybin suppressed liver inflammation in HFD-fed mice and inhibited nuclear factor κ-B translocation into the nucleus through elevation of SIRT2 expression and promotion of p65 deacetylation. This effect was confirmed by overexpression of SIRT2, which suppressed p65 nuclear translocation and restored CYP3A transcription affected by CM. The hepatic NAD+ concentration markedly decreased in HFD-fed mice and CM-treated hepatocytes/HepG2 cells but increased after silybin treatment. Supplementing nicotinamide mononucleotide as an NAD+ donor inhibited p65 acetylation, decreased p65 nuclear translocation, and restored cyp3a transcription in both HepG2 cells and mouse hepatocytes. These results suggest that silybin regulates metabolic enzymes during liver inflammation by a mechanism related to the increase in NAD+ and SIRT2 levels. In addition, silybin enhanced the intracellular NAD+ concentration by decreasing poly-ADP ribosyl polymerase-1 expression. In summary, silybin increased NAD+ concentration, promoted SIRT2 expression, and lowered p65 acetylation both in vivo and in vitro, which supported the recovery of CYP3A expression. These findings indicate that the NAD+/SIRT2 pathway plays an important role in CYP3A regulation during nonalcoholic fatty liver disease. SIGNIFICANCE STATEMENT: This research revealed the differential regulation of CYP3A by silybin under physiological and fatty liver pathological conditions. In the treatment of nonalcoholic fatty liver disease, silybin restored, not inhibited, CYP3A expression and activity through the NAD+/ sirtuin 2 pathway in accordance with its anti-inflammatory effect.
Collapse
Affiliation(s)
- Ran Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Dan Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Yirui Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Rui Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Na Yang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Yunge Lou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Haokai Zhao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Jiye Aa
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| | - Yuan Xie
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China (R.Z., Y.Z., R.W., Y.L., H.Z., J.A., G.W., Y.X.); Research and Development Center, Nanjing Chia Tai Tianqing Pharmaceutical co., Ltd., Nanjing, China (D.X.); and Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China (N.Y.)
| |
Collapse
|
18
|
Kaempferol and Kaempferide Attenuate Oleic Acid-Induced Lipid Accumulation and Oxidative Stress in HepG2 Cells. Int J Mol Sci 2021; 22:ijms22168847. [PMID: 34445549 PMCID: PMC8396315 DOI: 10.3390/ijms22168847] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases which lacks ideal treatment options. Kaempferol and kaempferide, two natural flavonol compounds isolated from Hippophae rhamnoides L., were reported to exhibit a strong regulatory effect on lipid metabolism, for which the mechanism is largely unknown. In the present study, we investigated the effects of kaempferol and kaempferide on oleic acid (OA)-treated HepG2 cells, a widely used in vitro model of NAFLD. The results indicated an increased accumulation of lipid droplets and triacylglycerol (TG) by OA, which was attenuated by kaempferol and kaempferide (5, 10 and 20 μM). Western blot analysis demonstrated that kaempferol and kaempferide reduced expression of lipogenesis-related proteins, including sterol regulatory element-binding protein 1 (SREBP1), fatty acid synthase (FAS) and stearoyl-CoA desaturase 1 (SCD-1). Expression of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT enhancer binding proteins β (C/EBPβ), two adipogenic transcription factors, was also decreased by kaempferol and kaempferide treatment. In addition, western blot analysis also demonstrated that kaempferol and kaempferide reduced expression of heme oxygenase-1 (HO-1) and nuclear transcription factor-erythroid 2-related factor 2 (Nrf2). Molecular docking was performed to identify the direct molecular targets of kaempferol and kaempferide, and their binding to SCD-1, a critical regulator in lipid metabolism, was revealed. Taken together, our findings demonstrate that kaempferol and kaempferide could attenuate OA-induced lipid accumulation and oxidative stress in HepG2 cells, which might benefit the treatment of NAFLD.
Collapse
|
19
|
A network-based approach to identify protein kinases critical for regulating srebf1 in lipid deposition causing obesity. Funct Integr Genomics 2021; 21:557-570. [PMID: 34327622 DOI: 10.1007/s10142-021-00798-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
Obesity is a rapidly growing health pandemic, underlying a wide variety of disease conditions leading to increases in global mortality. It is known that the phosphorylation of various proteins regulates sterol regulatory element-binding transcription factors 1 (srebf1), a key lipogenic transcription factor, to cause the development of obesity. To detect the key protein kinases for regulating srebf1 in lipid deposition, we established the srebf1 knockout model in zebrafish (KO, srebf1-/-) by CRISPR/Cas9. The KO zebrafish exhibited a significant reduction of total free fatty acid content (fell 60.5%) and lipid deposition decrease compared with wild-type (WT) zebrafish. Meanwhile, srebf1 deletion in zebrafish eliminated lipid deposition induced by high-fat diet feeding. Compared with WT zebrafish, a total of 697 differentially expressed proteins and 316 differentially expressed phosphoproteins with 439 sites were identified in KO by differential proteomic and phosphoproteomic analyses. A significant number of proteins identified were involved in lipid and glucose metabolism. Moreover, some protein kinases critical for regulating srebf1 in lipid deposition, including Cdk2, Pkc, Prkceb, mTORC1, Mapk12, and Wnk1, were determined by network analyses. An in vitro study was performed to verify the network analysis results. Our findings provide potential targets (kinases) for human obesity treatments.
Collapse
|
20
|
Li Z, Lyu Y, Zhao J, Li D, Lin Z, To KKW, Yan X, Zuo Z. Disease Status-Dependent Drug-Herb Interactions: NASH Lowered the Risk of Hepatotoxicity in Rats Coadministered With Simvastatin and Gardenia jasminoides J. Ellis. Front Pharmacol 2021; 12:622040. [PMID: 33967756 PMCID: PMC8103205 DOI: 10.3389/fphar.2021.622040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
Concurrent use of simvastatin (SV) and Gardenia jasminoides J. Ellis (GJ) was adopted in patients with multi-morbidity, such as stroke rehabilitation patients with NASH. Although hepatotoxicity has been reported in both of them and NASH could alter the pharmacokinetics of drugs/herbs, the interaction between SV and GJ and the related hepatotoxicity remained uninvestigated under neither healthy nor NASH condition. The current study aimed to evaluate the potential hepatotoxicity resulted from the interactions between SV and GJ in both healthy and NASH rats. Both healthy and NASH rats received two-week SV (p. o., 8.66 mg/kg, once daily) and/or GJ (p.o., 325 mg/kg, twice daily). Pharmacokinetic profiles of SV, simvastatin acid (SVA, active metabolite of SV), and geniposide (major component in GJ); hepatic Cyp2c11/Oatp1b2/P-gp expression; and biomarker levels of liver function, lipid levels, and liver histology were compared to demonstrate the interactions in rats. To explore the mechanism of the interaction-mediated hepatotoxicity, hepatic genipin-protein adduct content and iNOS/COX-1/COX-2 expressions from related groups were compared. Moreover, liver histology of healthy/NASH rats at 90 days after discontinuation of two-week GJ in the absence and presence of SV was evaluated to estimate the long-term impact of the interactions. GJ reduced the systemic exposures of SV and SVA by up-regulating the hepatic P-gp expression in healthy but not NASH rats. Meanwhile, SV increased the systemic exposure of geniposide via inhibiting the activity of P-gp in both healthy and NASH rats. Although neither SV nor GJ induced hepatotoxicity in healthy rats, their co-treatment elevated serum ALT and AST levels, which may attribute to the aggravated genipin-protein adduct formation, inflammation infiltration, and iNOS/COX-1 expressions in the liver. In NASH rats, SV and/or GJ reduced serum ALT, AST, LDL/vLDL, and TC levels via alleviating hepatic inflammation infiltration and iNOS/COX-1 expressions. Moreover, in comparison to NASH rats, more severe fibrosis was observed in the livers of healthy rats at 90 days after discontinuation of two-week SV and GJ coadministration. Although interactions between SV and GJ induced short-term and long-term liver injuries in healthy rats, NASH condition in rats could lower such risk.
Collapse
Affiliation(s)
- Ziwei Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanfeng Lyu
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jiajia Zhao
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Dan Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhixiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiaoyu Yan
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
21
|
Garduno A, Wu T. Tobacco Smoke and CYP1A2 Activity in a US Population with Normal Liver Enzyme Levels. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2225. [PMID: 33668222 PMCID: PMC7956356 DOI: 10.3390/ijerph18052225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 11/18/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is common among 30% of American adults. Former and current smokers are at higher risk for NAFLD compared to never smokers. The ratio of urine caffeine metabolites to caffeine intake-namely, urine caffeine metabolite indices-has previously been used as a proxy for CYP1A2 activity, which is one of the main liver metabolizing enzymes. CYP1A2 activity is associated with NAFLD progression. No studies to our knowledge have examined the associations of liver enzymes, smoking intensity, and secondhand smoke (SES) with CYP1A2 activity (using caffeine metabolite indices) across smoking status. We analyzed national representative samples from the 2009-2010 National Health and Nutrition Examination Survey (NHANES). Interestingly, even within a normal range, several liver enzymes were associated with caffeine metabolite indices, and patterns of many of these associations varied by smoking status. For instance, within a normal range, aspartate aminotransferase (AST) in never smokers and bilirubin in current smokers were inversely associated with 1-methyluric acid and 5-acetylamino-6-amino-3-methyluracil (URXAMU). Furthermore, we observed a common pattern: across all smoking statuses, higher AST/alanine aminotransferase (AST/ALT) was associated with 1-methyluric acid and URXAMU. Moreover, in current smokers, increased lifelong smoking intensity was associated with reduced caffeine metabolite indices, but acute cigarette exposure as measured by SES levels was associated with increased caffeine metabolite indices among never smokers. In summary, commonly used liver enzyme tests can reflect the CYP1A2 activity even within a normal range, but the selection of these enzymes depends on the smoking status; the associations between smoking and the CYP1A2 activity not only depend on the intensity but also the duration of tobacco exposure.
Collapse
Affiliation(s)
- Alexis Garduno
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, USA;
- Division of Epidemiology and Biostatistics, School of Public Health, San Diego State University, San Diego, CA 92182, USA
| | - Tianying Wu
- Division of Epidemiology and Biostatistics, School of Public Health, San Diego State University, San Diego, CA 92182, USA
- Moores Cancer Center, School of Medicine, University of California, San Diego, CA 92037, USA
| |
Collapse
|
22
|
Lin YS, Thummel KE, Thompson BD, Totah RA, Cho CW. Sources of Interindividual Variability. Methods Mol Biol 2021; 2342:481-550. [PMID: 34272705 DOI: 10.1007/978-1-0716-1554-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The efficacy, safety, and tolerability of drugs are dependent on numerous factors that influence their disposition. A dose that is efficacious and safe for one individual may result in sub-therapeutic or toxic blood concentrations in others. A significant source of this variability in drug response is drug metabolism, where differences in presystemic and systemic biotransformation efficiency result in variable degrees of systemic exposure (e.g., AUC, Cmax, and/or Cmin) following administration of a fixed dose.Interindividual differences in drug biotransformation have been studied extensively. It is recognized that both intrinsic factors (e.g., genetics, age, sex, and disease states) and extrinsic factors (e.g., diet , chemical exposures from the environment, and the microbiome) play a significant role. For drug-metabolizing enzymes, genetic variation can result in the complete absence or enhanced expression of a functional enzyme. In addition, upregulation and downregulation of gene expression, in response to an altered cellular environment, can achieve the same range of metabolic function (phenotype), but often in a less predictable and time-dependent manner. Understanding the mechanistic basis for variability in drug disposition and response is essential if we are to move beyond the era of empirical, trial-and-error dose selection and into an age of personalized medicine that will improve outcomes in maintaining health and treating disease.
Collapse
Affiliation(s)
- Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Brice D Thompson
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Christi W Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Nonalcoholic Fatty Liver Disease (NAFLD) and Hepatic Cytochrome P450 (CYP) Enzymes. Pharmaceuticals (Basel) 2020; 13:ph13090222. [PMID: 32872474 PMCID: PMC7560175 DOI: 10.3390/ph13090222] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive fat in the liver. An international consensus panel has recently proposed to rename the disease to metabolic dysfunction associated with fatty liver disease (MAFLD). The disease can range from simple steatosis (fat accumulation) to nonalcoholic steatohepatitis (NASH) which represents a severe form of NAFLD and is accompanied by inflammation, fibrosis, and hepatocyte damage in addition to significant steatosis. This review collates current knowledge of changes in human hepatic cytochrome P450 enzymes in NAFLD. While the expression of these enzymes is well studied in healthy volunteers, our understanding of the alterations of these proteins in NAFLD is limited. Much of the existing knowledge on the subject is derived from preclinical studies, and clinical translation of these findings is poor. Wherever available, the effect of NAFLD on these proteins in humans is debatable and currently lacks a consensus among different reports. Protein expression is an important in vitro physiological parameter controlling the pharmacokinetics of drugs and the last decade has seen a rise in the accurate estimation of these proteins for use with physiologically based pharmacokinetic (PBPK) modeling to predict drug pharmacokinetics in special populations. The application of label-free, mass spectrometry-based quantitative proteomics as a promising tool to study NAFLD-associated changes has also been discussed.
Collapse
|
24
|
Mo J, Chen Z, Xu J, Wang A, Dai L, Cheng A, Meng X, Li H, Wang Y, Johnston SC, Wang Y, Zhao X, Wang Z, Xia H, Li B, Zhang G, Ren X, Ji C, Zhang G, Li J, Lu B, Wang L, Feng S, Wang D, Tang W, Li J, Zhang H, Li G, Wang B, Chen Y, Lian Y, Liu B, Teng J, Sui R, Li L, Yuan Z, Zang D, Lu Z, Sun L, Wang D, Hou L, Yuan D, Cao Y, Li H, Tan X, Wang H, Du H, Liu M, Wang S, Liu Q, Zhang Z, Cui Q, Wang R, Zhao J, Zhang J, Zhao J, Bi Q, Qi X, Liu J, Li C, Li L, Pan X, Zhang J, Jiao D, Han Z, Qian D, Xiao J, Xing Y, Du H, Huang G, Cui Y, Li Y, Feng L, Gao L, Xiao B, Cao Y, Wu Y, Liu J, Zhang Z, Dong Z, Wang L, He L, Wang X, Guo X, Wang M, Wang X, Jiang J, Zhao R, Zhou S, Hu H, He M, Yu F, Ouyang Q, Zhang J, Xu A, Qi X, Wang L, Shi F, Guo F, Wang J, et alMo J, Chen Z, Xu J, Wang A, Dai L, Cheng A, Meng X, Li H, Wang Y, Johnston SC, Wang Y, Zhao X, Wang Z, Xia H, Li B, Zhang G, Ren X, Ji C, Zhang G, Li J, Lu B, Wang L, Feng S, Wang D, Tang W, Li J, Zhang H, Li G, Wang B, Chen Y, Lian Y, Liu B, Teng J, Sui R, Li L, Yuan Z, Zang D, Lu Z, Sun L, Wang D, Hou L, Yuan D, Cao Y, Li H, Tan X, Wang H, Du H, Liu M, Wang S, Liu Q, Zhang Z, Cui Q, Wang R, Zhao J, Zhang J, Zhao J, Bi Q, Qi X, Liu J, Li C, Li L, Pan X, Zhang J, Jiao D, Han Z, Qian D, Xiao J, Xing Y, Du H, Huang G, Cui Y, Li Y, Feng L, Gao L, Xiao B, Cao Y, Wu Y, Liu J, Zhang Z, Dong Z, Wang L, He L, Wang X, Guo X, Wang M, Wang X, Jiang J, Zhao R, Zhou S, Hu H, He M, Yu F, Ouyang Q, Zhang J, Xu A, Qi X, Wang L, Shi F, Guo F, Wang J, Zhao F, Dou R, Wei D, Meng Q, Xia Y, Wang S, Xue Z, Xu Y, Ma L, Wang C, Wu J, Du Y, Wang Y, Xiao L, Song F, Hu W, Chen Z, Liu Q, Zhang J, Chen M, Yuan X, Liu Z, Li G, Li X, Tian T. Efficacy of Clopidogrel-Aspirin Therapy for Stroke Does Not Exist in
C
YP2C19 Loss-of-Function Allele Noncarriers With Overweight/Obesity. Stroke 2020; 51:224-231. [DOI: 10.1161/strokeaha.119.026845] [Show More Authors] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background and Purpose—
The role of dual-antiplatelet therapy with clopidogrel plus aspirin has been demonstrated to substantially decrease the risk of recurrent stroke among patients with minor stroke and transient ischemic attack. We aimed to determine whether the efficacy of clopidogrel-aspirin therapy among patients with minor stroke / transient ischemic attack was influenced by the stratification of
CYP2C19
genotype and body mass index (BMI).
Methods—
CYP2C19
loss-of-function allele (LoFA) carriers were defined as patients with either LoFA of *2 or *3. Low/normal weight and overweight/obesity was defined as BMI <25 and ≥25 kg/m
2
, respectively. Primary outcome was defined as stroke recurrence at 3 months.
Results—
In a total of 2933 patients, there were 1726 (58.8%) LoFA carriers and 1275 (43.5%) patients with overweight/obesity (BMI ≥25 kg/m
2
). Stratified analyses by LoFA carrying status and BMI, hazard ratios (hazard ratios 95% CIs) of the clopidogrel-aspirin therapy for stroke recurrence were 0.90 (0.60–1.36), 0.87 (0.56–1.35), 0.65 (0.39–1.09), and 0.40 (0.22–0.71) among subgroups of LoFA carriers with overweight/obesity, LoFA carriers with low/normal weight, LoFA noncarriers with overweight/obesity, and LoFA noncarriers with low/normal weight, respectively, with
P
=0.049 for interaction.
Conclusions—
Efficacy of clopidogrel-aspirin therapy in reducing the risk of stroke recurrence is not present in
CYP2C19
LoFA noncarriers with overweight/obesity. Our study suggests that BMI significantly influences the correlation between
CYP2C19
genotype and efficacy of clopidogrel-aspirin therapy.
Clinical Trial Registration—
URL:
https://www.clinicaltrials.gov
. Unique identifier: NCT00979589.
Collapse
Affiliation(s)
- Jinglin Mo
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Zimo Chen
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Jie Xu
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Anxin Wang
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Liye Dai
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Aichun Cheng
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Xia Meng
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Hao Li
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | - Yongjun Wang
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China (J.M., Z.C., J.X., A.W., L.D., A.C., X.M., H.L., Y.W.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gabbia D, Roverso M, Guido M, Sacchi D, Scaffidi M, Carrara M, Orso G, Russo FP, Floreani A, Bogialli S, De Martin S. Western Diet-Induced Metabolic Alterations Affect Circulating Markers of Liver Function before the Development of Steatosis. Nutrients 2019; 11:1602. [PMID: 31311123 PMCID: PMC6683046 DOI: 10.3390/nu11071602] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/06/2019] [Accepted: 07/12/2019] [Indexed: 12/15/2022] Open
Abstract
Since nutrition might have a significant impact on liver function, we analyzed the early effect of Western-type diet on hepatic tissue and lipid and drug metabolism in Wistar-Kyoto rats (n = 8); eight rats fed with a standard diet were used as controls. Histological analysis of liver tissue was performed, and plasma biochemical parameters were measured. Plasma concentration of six bile acids was determined by ultra-liquid chromatography-tandem mass spectrometry UHPLC-MS/MS. Hepatic gene expressions of enzymes involved in drug and lipid metabolism were assessed by means of real-time reverse transcription (qRT)-PCR. Liver of rats fed with a Western diet did not show macroscopic histological alterations, but number and diameter of lipid droplets increased, as well as DGAT1, GPAT4, SCD, FASN and SREBP2 expression. Furthermore, Western diet-fed animals showed an increase in the activation of hepatic stellate cells and macrophage number in liver tissue, as well as a significant increase in AST and bilirubin levels (p < 0.01), and in the LDL:HDL cholesterol ratio (p < 0.001). Plasma chenodeoxycholic acid concentration increased significantly, whereas cholic acid decreased (p < 0.05), and cytochrome P450 genes were generally downregulated. Significant changes in hepatic lipid and drug metabolism are early induced by the Western diet, prior to steatosis development. Such changes are associated with a peculiar alteration in circulating bile acids, which could represent an early marker of non-alcoholic fatty liver disease (NAFLD) development.
Collapse
Affiliation(s)
- Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Marco Roverso
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| | - Maria Guido
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Diana Sacchi
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Michela Scaffidi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Maria Carrara
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Annarosa Floreani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Sara Bogialli
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, L.go Meneghetti 2, 35131 Padova, Italy.
| |
Collapse
|
26
|
Chen J, Jiang S, Wang J, Renukuntla J, Sirimulla S, Chen J. A comprehensive review of cytochrome P450 2E1 for xenobiotic metabolism. Drug Metab Rev 2019; 51:178-195. [PMID: 31203697 DOI: 10.1080/03602532.2019.1632889] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 2E1 (CYP2E1) plays a vital role in drug-induced hepatotoxicity and cancers (e.g. lung and bladder cancer), since it is responsible for metabolizing a number of medications and environmental toxins to reactive intermediate metabolites. CYP2E1 was recently found to be the highest expressed CYP enzyme in human livers using a proteomics approach, and CYP2E1-related toxicity is strongly associated with its protein level that shows significant inter-individual variability related to ethnicity, age, and sex. Furthermore, the expression of CYP2E1 demonstrates regulation by extensive genetic polymorphism, endogenous hormones, cytokines, xenobiotics, and varying pathological states. Over the past decade, the knowledge of pharmacology, toxicology, and biology about CYP2E1 has grown remarkably, but the research progress has yet to be summarized. This study presents a timely systematic review on CYP2E1's xenobiotic metabolism, genetic polymorphism, and inhibitors, with the focus on their clinical relevance for the efficacy and toxicity of various CYP2E1 substrates. Moreover, several knowledge gaps have been identified towards fully understanding the potential interactions among different CYP2E1 substrates in clinical settings. Through in-depth analyses of these knowns and unknowns, we expect this review will aid in future drug development and improve management of CYP2E1 related clinical toxicity.
Collapse
Affiliation(s)
- Jingxuan Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University , Guangzhou , China
| | - Sibo Jiang
- Department of Pharmaceutics, University of Florida , Orlando , FL , USA
| | - Jin Wang
- AbbVie Inc , North Chicago , IL , USA
| | - Jwala Renukuntla
- School of Pharmacy, The University of Texas at El Paso , El Paso , TX , USA
| | - Suman Sirimulla
- School of Pharmacy, The University of Texas at El Paso , El Paso , TX , USA
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University , Guangzhou , China
| |
Collapse
|
27
|
Huang Z, Wang M, Liu L, Peng J, Guo C, Chen X, Huang L, Tan J, Yang G. Transcriptional Repression of CYP3A4 by Increased miR-200a-3p and miR-150-5p Promotes Steatosis in vitro. Front Genet 2019; 10:484. [PMID: 31191607 PMCID: PMC6546834 DOI: 10.3389/fgene.2019.00484] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatic cytochrome P450 enzyme activities correlate with non-alcoholic fatty liver disease (NAFLD) and hepatic steatosis. The decreased activity of CYP3A4, an important drug-metabolizing enzyme, is associated with the progression of NAFLD. CYP3A4 is predicted as a target gene of miR-200a-3p and miR-150-5p by MicroInspector and TargetScan algorithms analyses. Here, we found decreased CYP3A4 and increased miR-200a-3p and miR-150-5p in LO2 cells with free fatty acid (FFA)-induced steatosis. Dual-luciferase assay confirmed that both miR-200a-3p and miR-150-5p targeted the 3'-untranslated region (3'-UTR) of CYP3A4 and that such interaction was abolished by miRNA binding site mutations in 3'-UTR of CYP3A4. Using miR-200a-3p and miR-150-5p mimics and inhibitors, we further confirmed that endogenous CYP3A4 was regulated posttranscriptionally by miR-200a-3p or miR-150-5p. Moreover, miR-200a-3p and miR-150-5p inhibitors attenuated FFA-induced steatosis in LO2 cells, and such effect was dependent on CYP3Y4 expression. These results suggest that miR-200a-3p and miR-150-5p, through directly targeting 3'-UTR of CYP3A4, contribute to the development of FFA-induced steatosis.
Collapse
Affiliation(s)
- Zhijun Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mengyao Wang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Li Liu
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jinfu Peng
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chengxian Guo
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Lu Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jieqiong Tan
- Center for Medical Genetics, Life Science School, Central South University, Changsha, China
| | - Guoping Yang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Grossmann M, Wierman ME, Angus P, Handelsman DJ. Reproductive Endocrinology of Nonalcoholic Fatty Liver Disease. Endocr Rev 2019; 40:417-446. [PMID: 30500887 DOI: 10.1210/er.2018-00158] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
The liver and the reproductive system interact in a multifaceted bidirectional fashion. Sex steroid signaling influences hepatic endobiotic and xenobiotic metabolism and contributes to the pathogenesis of functional and structural disorders of the liver. In turn, liver function affects the reproductive axis via modulating sex steroid metabolism and transport to tissues via sex hormone-binding globulin (SHBG). The liver senses the body's metabolic status and adapts its energy homeostasis in a sex-dependent fashion, a dimorphism signaled by the sex steroid milieu and possibly related to the metabolic costs of reproduction. Sex steroids impact the pathogenesis of nonalcoholic fatty liver disease, including development of hepatic steatosis, fibrosis, and carcinogenesis. Preclinical studies in male rodents demonstrate that androgens protect against hepatic steatosis and insulin resistance both via androgen receptor signaling and, following aromatization to estradiol, estrogen receptor signaling, through regulating genes involved in hepatic lipogenesis and glucose metabolism. In female rodents in contrast to males, androgens promote hepatic steatosis and dysglycemia, whereas estradiol is similarly protective against liver disease. In men, hepatic steatosis is associated with modest reductions in circulating testosterone, in part consequent to a reduction in circulating SHBG. Testosterone treatment has not been demonstrated to improve hepatic steatosis in randomized controlled clinical trials. Consistent with sex-dimorphic preclinical findings, androgens promote hepatic steatosis and dysglycemia in women, whereas endogenous estradiol appears protective in both men and women. In both sexes, androgens promote hepatic fibrosis and the development of hepatocellular carcinoma, whereas estradiol is protective.
Collapse
Affiliation(s)
- Mathis Grossmann
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Margaret E Wierman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Peter Angus
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Departments of Gastroenterology and Hepatology, Heidelberg, Victoria, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Xu YXZ, Mishra S. Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models. Cancers (Basel) 2018; 10:E523. [PMID: 30567335 PMCID: PMC6316427 DOI: 10.3390/cancers10120523] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/09/2018] [Accepted: 12/15/2018] [Indexed: 02/07/2023] Open
Abstract
The worldwide prevalence of obesity has doubled during the last 50 years, and according to the World Obesity Federation, one third of the people on Earth will be obese by the year 2025. Obesity is described as a chronic, relapsing and multifactorial disease that causes metabolic, biomechanical, and psychosocial health consequences. Growing evidence suggests that obesity is a risk factor for multiple cancer types and rivals smoking as the leading preventable cause for cancer incidence and mortality. The epidemic of obesity will likely generate a new wave of obesity-related cancers with high aggressiveness and shortened latency. Observational studies have shown that from cancer risk to disease prognosis, an individual with obesity is consistently ranked worse compared to their lean counterpart. Mechanistic studies identified similar sets of abnormalities under obesity that may lead to cancer development, including ectopic fat storage, altered adipokine profiles, hormone fluctuations and meta-inflammation, but could not explain how these common mechanisms produce over 13 different cancer types. A major hurdle in the mechanistic underpinning of obesity-related cancer is the lack of suitable pre-clinical models that spontaneously develop obesity-linked cancers like humans. Current approaches and animal models fall short when discerning the confounders that often coexist in obesity. In this mini-review, we will briefly survey advances in the different obesity-linked cancers and discuss the challenges and limitations in the rodent models employed to study their relationship. We will also provide our perspectives on the future of obesity-linked cancer research.
Collapse
Affiliation(s)
- Yang Xin Zi Xu
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Suresh Mishra
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
- Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
30
|
Tête A, Gallais I, Imran M, Chevanne M, Liamin M, Sparfel L, Bucher S, Burel A, Podechard N, Appenzeller BMR, Fromenty B, Grova N, Sergent O, Lagadic-Gossmann D. Mechanisms involved in the death of steatotic WIF-B9 hepatocytes co-exposed to benzo[a]pyrene and ethanol: a possible key role for xenobiotic metabolism and nitric oxide. Free Radic Biol Med 2018; 129:323-337. [PMID: 30268890 DOI: 10.1016/j.freeradbiomed.2018.09.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 12/15/2022]
Abstract
We previously demonstrated that co-exposing pre-steatotic hepatocytes to benzo[a]pyrene (B[a]P), a carcinogenic environmental pollutant, and ethanol, favored cell death. Here, the intracellular mechanisms underlying this toxicity were studied. Steatotic WIF-B9 hepatocytes, obtained by a 48h-supplementation with fatty acids, were then exposed to B[a]P/ethanol (10 nM/5 mM, respectively) for 5 days. Nitric oxide (NO) was demonstrated to be a pivotal player in the cell death caused by the co-exposure in steatotic hepatocytes. Indeed, by scavenging NO, CPTIO treatment of co-exposed steatotic cells prevented not only the increase in DNA damage and cell death, but also the decrease in the activity of CYP1, major cytochrome P450s of B[a]P metabolism. This would then lead to an elevation of B[a]P levels, thus possibly suggesting a long-lasting stimulation of the transcription factor AhR. Besides, as NO can react with superoxide anion to produce peroxynitrite, a highly oxidative compound, the use of FeTPPS to inhibit its formation indicated its participation in DNA damage and cell death, further highlighting the important role of NO. Finally, a possible key role for AhR was pointed out by using its antagonist, CH-223191. Indeed it prevented the elevation of ADH activity, known to participate to the ethanol production of ROS, notably superoxide anion. The transcription factor, NFκB, known to be activated by ROS, was shown to be involved in the increase in iNOS expression. Altogether, these data strongly suggested cooperative mechanistic interactions between B[a]P via AhR and ethanol via ROS production, to favor cell death in the context of prior steatosis.
Collapse
Affiliation(s)
- Arnaud Tête
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Muhammad Imran
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Marie Liamin
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Simon Bucher
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000 Rennes, France
| | - Agnès Burel
- Univ Rennes, Biosit - UMS 3480, US_S 018, F-35000 Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Brice M R Appenzeller
- HBRU, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg
| | - Bernard Fromenty
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000 Rennes, France
| | - Nathalie Grova
- HBRU, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
31
|
Bessone F, Dirchwolf M, Rodil MA, Razori MV, Roma MG. Review article: drug-induced liver injury in the context of nonalcoholic fatty liver disease - a physiopathological and clinical integrated view. Aliment Pharmacol Ther 2018; 48:892-913. [PMID: 30194708 DOI: 10.1111/apt.14952] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 03/25/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nonalcoholic fatty disease (NAFLD) is the most common liver disease, since it is strongly associated with obesity and metabolic syndrome pandemics. NAFLD may affect drug disposal and has common pathophysiological mechanisms with drug-induced liver injury (DILI); this may predispose to hepatoxicity induced by certain drugs that share these pathophysiological mechanisms. In addition, drugs may trigger fatty liver and inflammation per se by mimicking NAFLD pathophysiological mechanisms. AIMS To provide a comprehensive update on (a) potential mechanisms whereby certain drugs can be more hepatotoxic in NAFLD patients, (b) the steatogenic effects of drugs, and (c) the mechanism involved in drug-induced steatohepatitis (DISH). METHODS A language- and date-unrestricted Medline literature search was conducted to identify pertinent basic and clinical studies on the topic. RESULTS Drugs can induce macrovesicular steatosis by mimicking NAFLD pathogenic factors, including insulin resistance and imbalance between fat gain and loss. Other forms of hepatic fat accumulation exist, such as microvesicular steatosis and phospholipidosis, and are mostly associated with acute mitochondrial dysfunction and defective lipophagy, respectively. Drug-induced mitochondrial dysfunction is also commonly involved in DISH. Patients with pre-existing NAFLD may be at higher risk of DILI induced by certain drugs, and polypharmacy in obese individuals to treat their comorbidities may be a contributing factor. CONCLUSIONS The relationship between DILI and NAFLD may be reciprocal: drugs can cause NAFLD by acting as steatogenic factors, and pre-existing NAFLD could be a predisposing condition for certain drugs to cause DILI. Polypharmacy associated with obesity might potentiate the association between this condition and DILI.
Collapse
Affiliation(s)
- Fernando Bessone
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - Melisa Dirchwolf
- Unidad de Transplante Hepático, Servicio de Hepatología, Hospital Privado de Rosario, Rosario, Argentina
| | - María Agustina Rodil
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - María Valeria Razori
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
32
|
Skubic C, Drakulić Ž, Rozman D. Personalized therapy when tackling nonalcoholic fatty liver disease: a focus on sex, genes, and drugs. Expert Opin Drug Metab Toxicol 2018; 14:831-841. [PMID: 29969922 DOI: 10.1080/17425255.2018.1492552] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is the most frequent liver disease in the world. It describes a term for a group of hepatic diseases including steatosis, fibrosis, and cirrhosis that can finally lead to hepatocellular carcinoma. There are many factors influencing NAFLD initiation and progression, such as obesity, dyslipidemia, insulin resistance, genetic factors, and hormonal changes. However, there is also lean-NAFLD which is not associated with obesity. NAFLD is considered to be a sexually dimorphic disease. In most cases, men have a higher prevalence for the disease compared to premenopausal women. Areas covered: In this review, we first summarize the NAFLD disease epidemiology, pathology, and diagnosis. We describe NAFLD progression with the focus on sexual and genetic differences for disease development and pharmacological treatment. Personalized treatment for multifactorial NAFLD is discussed in consideration of different factors, including genetics, gender and sex. Expert opinion: The livers of female and male NAFLD patients have different metabolic capacities which influence the metabolism of all drugs applied to such patients. This aspect is not yet sufficiently taken into account. The liver computational models might quicken the pace toward assessing personalized disease progression and treatment options.
Collapse
Affiliation(s)
- Cene Skubic
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Živa Drakulić
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| | - Damjana Rozman
- a Centre for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|
33
|
Angrish MM, McQueen CA, Cohen-Hubal E, Bruno M, Ge Y, Chorley BN. Editor's Highlight: Mechanistic Toxicity Tests Based on an Adverse Outcome Pathway Network for Hepatic Steatosis. Toxicol Sci 2018; 159:159-169. [PMID: 28903485 DOI: 10.1093/toxsci/kfx121] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Risk assessors use liver endpoints in rodent toxicology studies to assess the safety of chemical exposures. Yet, rodent endpoints may not accurately reflect human responses. For this reason and others, human-based invitro models are being developed and anchored to adverse outcome pathways to better predict adverse human health outcomes. Here, a networked adverse outcome pathway-guided selection of biology-based assays for lipid uptake, lipid efflux, fatty acid oxidation, and lipid accumulation were developed. These assays were evaluated in a metabolically competent human hepatocyte cell model (HepaRG) exposed to compounds known to cause steatosis (amiodarone, cyclosporine A, and T0901317) or activate lipid metabolism pathways (troglitazone, Wyeth-14,643, and 22(R)-hydroxycholesterol). All of the chemicals activated at least one assay, however, only T0901317 and cyclosporin A dose-dependently increased lipid accumulation. T0901317 and cyclosporin A increased fatty acid uptake, decreased lipid efflux (inferred from apolipoprotein B100 levels), and increased fatty acid synthase protein levels. Using this biologically-based evaluation of key events regulating hepatic lipid levels, we demonstrated dysregulation of compensatory pathways that normally balance hepatic lipid levels. This approach may provide biological plausibility and data needed to increase confidence in linking invitro-based measurements to chemical effects on adverse human health outcomes.
Collapse
Affiliation(s)
- Michelle M Angrish
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Charlene A McQueen
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Elaine Cohen-Hubal
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Maribel Bruno
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Yue Ge
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Brian N Chorley
- National Health and Environmental Effects Research Laboratory , United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
34
|
Jamwal R, de la Monte SM, Ogasawara K, Adusumalli S, Barlock BB, Akhlaghi F. Nonalcoholic Fatty Liver Disease and Diabetes Are Associated with Decreased CYP3A4 Protein Expression and Activity in Human Liver. Mol Pharm 2018; 15:2621-2632. [PMID: 29792708 DOI: 10.1021/acs.molpharmaceut.8b00159] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major cause of chronic liver disease in the Western population. We investigated the association of nonalcoholic fatty liver disease (NAFLD) and diabetes mellitus on CYP3A4 activity in human liver tissue from brain dead donors ( n = 74). Histopathologically graded livers were grouped into normal ( n = 24), nonalcoholic fatty liver (NAFL, n = 26), and nonalcoholic steatohepatitis (NASH, n = 24) categories. The rate of conversion of midazolam to its 1-hydroxy metabolite was used to assess in vitro CYP3A4 activity in human liver microsomes (HLM). A proteomics approach was utilized to quantify the protein expression of CYP3A4 and related enzymes. Moreover, a physiologically based pharmacokinetic (PBPK) model was developed to allow prediction of midazolam concentration in NAFL and NASH livers. CYP3A4 activity in NAFL and NASH was 1.9- and 3.1-fold ( p < 0.05) lower than normal donors, respectively. Intrinsic clearance (CLint) was 2.7- ( p < 0.05) and 4.1-fold ( p < 0.01) lower in donors with NAFL and NASH, respectively. CYP3A4 protein expression was significantly lower in NAFL and NASH donors ( p < 0.05) and accounted for significant midazolam hydroxylation variability in a multiple linear regression analysis (β = 0.869, r2 = 0.762, P < 0.01). Diabetes was also associated with decreased CYP3A4 activity and protein expression. Both midazolam CLint and CYP3A4 protein abundance decreased significantly with increase in hepatic fat accumulation. Age and gender did not exhibit any significant association with the observed alterations. Predicted midazolam exposure was 1.7- and 2.3-fold higher for NAFL and NASH, respectively, which may result in a longer period of sedation in these disease-states. Data suggests that NAFLD and diabetes are associated with the decreased hepatic CYP3A4 activity. Thus, further evaluation of clinical consequences of these findings on the efficacy and safety of CYP3A4 substrates is warranted.
Collapse
Affiliation(s)
- Rohitash Jamwal
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Suzanne M de la Monte
- Departments of Medicine, Pathology, Neurology, and Neurosurgery , Rhode Island Hospital and the Warren Alpert Medical School of Brown University , Providence , Rhode Island 02903 , United States
| | - Ken Ogasawara
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Sravani Adusumalli
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Benjamin B Barlock
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Fatemeh Akhlaghi
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| |
Collapse
|
35
|
Bucher S, Tête A, Podechard N, Liamin M, Le Guillou D, Chevanne M, Coulouarn C, Imran M, Gallais I, Fernier M, Hamdaoui Q, Robin MA, Sergent O, Fromenty B, Lagadic-Gossmann D. Co-exposure to benzo[a]pyrene and ethanol induces a pathological progression of liver steatosis in vitro and in vivo. Sci Rep 2018; 8:5963. [PMID: 29654281 PMCID: PMC5899096 DOI: 10.1038/s41598-018-24403-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatic steatosis (i.e. lipid accumulation) and steatohepatitis have been related to diverse etiologic factors, including alcohol, obesity, environmental pollutants. However, no study has so far analyzed how these different factors might interplay regarding the progression of liver diseases. The impact of the co-exposure to the environmental carcinogen benzo[a]pyrene (B[a]P) and the lifestyle-related hepatotoxicant ethanol, was thus tested on in vitro models of steatosis (human HepaRG cell line; hybrid human/rat WIF-B9 cell line), and on an in vivo model (obese zebrafish larvae). Steatosis was induced prior to chronic treatments (14, 5 or 7 days for HepaRG, WIF-B9 or zebrafish, respectively). Toxicity and inflammation were analyzed in all models; the impact of steatosis and ethanol towards B[a]P metabolism was studied in HepaRG cells. Cytotoxicity and expression of inflammation markers upon co-exposure were increased in all steatotic models, compared to non steatotic counterparts. A change of B[a]P metabolism with a decrease in detoxification was detected in HepaRG cells under these conditions. A prior steatosis therefore enhanced the toxicity of B[a]P/ethanol co-exposure in vitro and in vivo; such a co-exposure might favor the appearance of a steatohepatitis-like state, with the development of inflammation. These deleterious effects could be partly explained by B[a]P metabolism alterations.
Collapse
Affiliation(s)
- Simon Bucher
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Arnaud Tête
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Marie Liamin
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Dounia Le Guillou
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Cédric Coulouarn
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Muhammad Imran
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Morgane Fernier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Quentin Hamdaoui
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Marie-Anne Robin
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France
| | - Bernard Fromenty
- Univ Rennes, Inserm, Inra, Institut NUMECAN (Nutrition Metabolisms and Cancer) - UMR_S 1241, UMR_A 1341, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
36
|
Tanner N, Kubik L, Luckert C, Thomas M, Hofmann U, Zanger UM, Böhmert L, Lampen A, Braeuning A. Regulation of Drug Metabolism by the Interplay of Inflammatory Signaling, Steatosis, and Xeno-Sensing Receptors in HepaRG Cells. Drug Metab Dispos 2018; 46:326-335. [PMID: 29330220 DOI: 10.1124/dmd.117.078675] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/11/2018] [Indexed: 02/13/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), which is characterized by triglyceride deposition in hepatocytes resulting from imbalanced lipid homeostasis, is of increasing concern in Western countries, along with progression to nonalcoholic steatohepatitis (NASH), liver fibrosis, and cirrhosis. Previous studies suggest a complex, mutual influence of hepatic fat accumulation, NASH-related inflammatory mediators, and drug-sensing receptors regulating xenobiotic metabolism. Here, we investigated the suitability of human HepaRG hepatocarcinoma cells as a model for NAFLD and NASH. Cells were incubated for up to 14 days with an oleate/palmitate mixture (125 µM each) and/or with 10 ng/ml of the inflammatory mediator interleukin-6 (IL-6). Effects of these conditions on the regulation of drug metabolism were studied using xenobiotic agonists of the aryl hydrocarbon receptor (AHR), pregnane X receptor (PXR), constitutive androstane receptor (CAR), nuclear factor (erythroid-derived 2)-like 2, and peroxisome proliferator-activated receptor α (PPARα). Results underpin the suitability of HepaRG cells for NAFLD- and NASH-related research and constitute a broad-based analysis of the impact of hepatic fatty acid accumulation and inflammation on drug metabolism and its inducibility by xenobiotics. IL-6 exerted pronounced negative regulatory effects on basal as well as on PXR-, CAR-, and PPARα-, but not AHR-dependent induction of drug-metabolizing enzymes. This inhibition was related to diminished transactivation potential of the respective receptors rather than to reduced transcription of nuclear receptor-encoding mRNAs. The most striking effects of IL-6 and/or fatty acid treatment were observed in HepaRG cells after 14 days of treatment, making these cultures appear a suitable model for studying the relationship of fatty acid accumulation, inflammation, and xenobiotic-induced drug metabolism.
Collapse
Affiliation(s)
- Norman Tanner
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Lisa Kubik
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Claudia Luckert
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Maria Thomas
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Ute Hofmann
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Ulrich M Zanger
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Linda Böhmert
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department Food Safety, Berlin (N.T., L.K., C.L., L.B., A.L., A.B.), and Dr. Margarete Fischer-Bosch-Institute for Clinical Pharmacology, Stuttgart (M.T., U.H., U.M.Z.), Germany
| |
Collapse
|
37
|
Strategies for Determining Correct Cytochrome P450 Contributions in Hepatic Clearance Predictions: In Vitro-In Vivo Extrapolation as Modelling Approach and Tramadol as Proof-of Concept Compound. Eur J Drug Metab Pharmacokinet 2018; 42:537-543. [PMID: 27317395 DOI: 10.1007/s13318-016-0355-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND OBJECTIVE Although the measurement of cytochrome P450 (CYP) contributions in metabolism assays is straightforward, determination of actual in vivo contributions might be challenging. How representative are in vitro for in vivo CYP contributions? This article proposes an improved strategy for the determination of in vivo CYP enzyme-specific metabolic contributions, based on in vitro data, using an in vitro-in vivo extrapolation (IVIVE) approach. Approaches are exemplified using tramadol as model compound, and CYP2D6 and CYP3A4 as involved enzymes. METHODS Metabolism data for tramadol and for the probe substrates midazolam (CYP3A4) and dextromethorphan (CYP2D6) were gathered in human liver microsomes (HLM) and recombinant human enzyme systems (rhCYP). From these probe substrates, an activity-adjustment factor (AAF) was calculated per CYP enzyme, for the determination of correct hepatic clearance contributions. As a reference, tramadol CYP contributions were scaled-back from in vivo data (retrograde approach) and were compared with the ones derived in vitro. In this view, the AAF is an enzyme-specific factor, calculated from reference probe activity measurements in vitro and in vivo, that allows appropriate scaling of a test drug's in vitro activity to the 'healthy volunteer' population level. Calculation of an AAF, thus accounts for any 'experimental' or 'batch-specific' activity difference between in vitro HLM and in vivo derived activity. RESULTS In this specific HLM batch, for CYP3A4 and CYP2D6, an AAF of 0.91 and 1.97 was calculated, respectively. This implies that, in this batch, the in vitro CYP3A4 activity is 1.10-fold higher and the CYP2D6 activity 1.97-fold lower, compared to in vivo derived CYP activities. CONCLUSION This study shows that, in cases where the HLM pool does not represent the typical mean population CYP activities, AAF correction of in vitro metabolism data, optimizes CYP contributions in the prediction of hepatic clearance. Therefore, in vitro parameters for any test compound, obtained in a particular batch, should be corrected with the AAF for the respective enzymes. In the current study, especially the CYP2D6 contribution was found, to better reflect the average in vivo situation. It is recommended that this novel approach is further evaluated using a broader range of compounds.
Collapse
|
38
|
Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A. Liver 'organ on a chip'. Exp Cell Res 2018; 363:15-25. [PMID: 29291400 PMCID: PMC5944300 DOI: 10.1016/j.yexcr.2017.12.023] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems.
Collapse
Affiliation(s)
- Colin H Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
39
|
Li H, Toth E, Cherrington NJ. Asking the Right Questions With Animal Models: Methionine- and Choline-Deficient Model in Predicting Adverse Drug Reactions in Human NASH. Toxicol Sci 2018; 161:23-33. [PMID: 29145614 PMCID: PMC6454421 DOI: 10.1093/toxsci/kfx253] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In the past few decades, great conceptual and technological advances have been made in the field of toxicology, but animal model-based research still remains one of the most widely used and readily available tools for furthering our current knowledge. However, animal models are not perfect in predicting all systemic toxicity in humans. Extrapolating animal data to accurately predict human toxicities remains a challenge, and researchers are obligated to question the appropriateness of their chosen animal model. This paper provides an assessment of the utility of the methionine- and choline-deficient (MCD) diet fed animal model in reflecting human nonalcoholic steatohepatitis (NASH) and the potential risks of adverse drug reactions and toxicities that are associated with the disease. As a commonly used NASH model, the MCD model fails to exhibit most metabolic abnormalities in a similar manner to the human disease. The MCD model, on the other hand, closely resembles human NASH histology and reflects signatures of drug transporter alterations in humans. Due to the nature of the MCD model, it should be avoided in studies of NASH pathogenesis, metabolic parameter evaluation, and biomarker identification. But it can be used to accurately predict altered drug disposition due to NASH-associated transporter alterations.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Erica Toth
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | | |
Collapse
|
40
|
Heath RD, Ertem F, Romana BS, Ibdah JA, Tahan V. Hepatocyte transplantation: Consider infusion before incision. World J Transplant 2017; 7:317-323. [PMID: 29312860 PMCID: PMC5743868 DOI: 10.5500/wjt.v7.i6.317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 11/27/2017] [Accepted: 12/05/2017] [Indexed: 02/05/2023] Open
Abstract
Human hepatocyte transplantation is undergoing study as a bridge, or even alternative, to orthotopic liver transplantation (OLT). This technique has undergone multiple developments over the past thirty years in terms of mode of delivery, source and preparation of cell cultures, monitoring of graft function, and use of immunosuppression. Further refinements and improvements in these techniques will likely allow improved graft survival and function, granting patients higher yield from this technique and potentially significantly delaying need for OLT.
Collapse
Affiliation(s)
- Ryan D Heath
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, United States
| | - Furkan Ertem
- Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States
| | - Bhupinder S Romana
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, United States
| | - Jamal A Ibdah
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, United States
| | - Veysel Tahan
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
41
|
Dietrich CG, Rau M, Jahn D, Geier A. Changes in drug transport and metabolism and their clinical implications in non-alcoholic fatty liver disease. Expert Opin Drug Metab Toxicol 2017; 13:625-640. [PMID: 28359183 DOI: 10.1080/17425255.2017.1314461] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The incidence of non-alcoholic fatty liver disease (NAFLD) is rising, especially in Western countries. Drug treatment in patients with NAFLD is common since it is linked to other conditions like diabetes, obesity, and cardiovascular disease. Consequently, changes in drug metabolism may have serious clinical implications. Areas covered: A literature search for studies in animal models or patients with obesity, fatty liver, non-alcoholic steatohepatitis (NASH) or NASH cirrhosis published before November 2016 was performed. After discussing epidemiology and animal models for NAFLD, we summarized both basic as well as clinical studies investigating changes in drug transport and metabolism in NAFLD. Important drug groups were assessed separately with emphasis on clinical implications for drug treatment in patients with NAFLD. Expert opinion: Given the frequency of NAFLD even today, a high degree of drug treatment in NAFLD patients appears safe and well-tolerated despite considerable changes in hepatic uptake, distribution, metabolism and transport of drugs in these patients. NASH causes changes in biliary excretion, systemic concentrations, and renal handling of drugs leading to alterations in drug efficacy or toxicity under specific circumstances. Future clinical drug studies should focus on this special patient population in order to avoid serious adverse events in NAFLD patients.
Collapse
Affiliation(s)
- Christoph G Dietrich
- a Bethlehem Center of Health , Department of Medicine , Stolberg/Rhineland , Germany
| | - Monika Rau
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Daniel Jahn
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Andreas Geier
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| |
Collapse
|
42
|
Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev 2017; 49:197-211. [PMID: 28303724 DOI: 10.1080/03602532.2017.1293683] [Citation(s) in RCA: 437] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of liver disorders. It is defined by the presence of steatosis in more than 5% of hepatocytes with little or no alcohol consumption. Insulin resistance, the metabolic syndrome or type 2 diabetes and genetic variants of PNPLA3 or TM6SF2 seem to play a role in the pathogenesis of NAFLD. The pathological progression of NAFLD follows tentatively a "three-hit" process namely steatosis, lipotoxicity and inflammation. The presence of steatosis, oxidative stress and inflammatory mediators like TNF-α and IL-6 has been implicated in the alterations of nuclear factors such as CAR, PXR, PPAR-α in NAFLD. These factors may result in altered expression and activity of drug metabolizing enzymes (DMEs) or transporters. Existing evidence suggests that the effect of NAFLD on CYP3A4, CYP2E1 and MRP3 is more consistent across rodent and human studies. CYP3A4 activity is down-regulated in NASH whereas the activity of CYP2E1 and the efflux transporter MRP3 is up-regulated. However, it is not clear how the majority of CYPs, UGTs, SULTs and transporters are influenced by NAFLD either in vivo or in vitro. The alterations associated with NAFLD could be a potential source of drug variability in patients and could have serious implications for the safety and efficacy of xenobiotics. In this review, we summarize the effects of NAFLD on the regulation, expression and activity of major DMEs and transporters. We also discuss the potential mechanisms underlying these alterations.
Collapse
Affiliation(s)
- Enoch Cobbina
- a Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences , University of Rhode Island , Kingston , RI , USA
| | - Fatemeh Akhlaghi
- a Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences , University of Rhode Island , Kingston , RI , USA
| |
Collapse
|
43
|
Kostrzewski T, Cornforth T, Snow SA, Ouro-Gnao L, Rowe C, Large EM, Hughes DJ. Three-dimensional perfused human in vitro model of non-alcoholic fatty liver disease. World J Gastroenterol 2017; 23:204-215. [PMID: 28127194 PMCID: PMC5236500 DOI: 10.3748/wjg.v23.i2.204] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/25/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
AIM To develop a human in vitro model of non-alcoholic fatty liver disease (NAFLD), utilising primary hepatocytes cultured in a three-dimensional (3D) perfused platform.
METHODS Fat and lean culture media were developed to directly investigate the effects of fat loading on primary hepatocytes cultured in a 3D perfused culture system. Oil Red O staining was used to measure fat loading in the hepatocytes and the consumption of free fatty acids (FFA) from culture medium was monitored. Hepatic functions, gene expression profiles and adipokine release were compared for cells cultured in fat and lean conditions. To determine if fat loading in the system could be modulated hepatocytes were treated with known anti-steatotic compounds.
RESULTS Hepatocytes cultured in fat medium were found to accumulate three times more fat than lean cells and fat uptake was continuous over a 14-d culture. Fat loading of hepatocytes did not cause any hepatotoxicity and significantly increased albumin production. Numerous adipokines were expressed by fatty cells and genes associated with NAFLD and liver disease were upregulated including: Insulin-like growth factor-binding protein 1, fatty acid-binding protein 3 and CYP7A1. The metabolic activity of hepatocytes cultured in fatty conditions was found to be impaired and the activities of CYP3A4 and CYP2C9 were significantly reduced, similar to observations made in NAFLD patients. The utility of the model for drug screening was demonstrated by measuring the effects of known anti-steatotic compounds. Hepatocytes, cultured under fatty conditions and treated with metformin, had a reduced cellular fat content compared to untreated controls and consumed less FFA from cell culture medium.
CONCLUSION The 3D in vitro NAFLD model recapitulates many features of clinical NAFLD and is an ideal tool for analysing the efficacy of anti-steatotic compounds.
Collapse
|
44
|
Cho SJ, Kim SB, Cho HJ, Chong S, Chung SJ, Kang IM, Lee JI, Yoon IS, Kim DD. Effects of Nonalcoholic Fatty Liver Disease on Hepatic CYP2B1 and in Vivo Bupropion Disposition in Rats Fed a High-Fat or Methionine/Choline-Deficient Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:5598-5606. [PMID: 27321734 DOI: 10.1021/acs.jafc.6b01663] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) refers to hepatic pathologies, including simple fatty liver (SFL), nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis, that may progress to hepatocellular carcinoma. These liver disease states may affect the activity and expression levels of drug-metabolizing enzymes, potentially resulting in an alteration in the pharmacokinetics, therapeutic efficacy, and safety of drugs. This study investigated the hepatic cytochrome P450 (CYP) 2B1-modulating effect of a specific NAFLD state in dietary rat models. Sprague-Dawley rats were given a methionine/choline-deficient (MCD) or high-fat (HF) diet to induce NASH and SFL, respectively. The induction of these disease states was confirmed by plasma chemistry and liver histological analysis. Both the protein and mRNA levels of hepatic CYP2B1 were considerably reduced in MCD diet-fed rats; however, they were similar between the HF diet-fed and control rats. Consistently, the enzyme-kinetic and pharmacokinetic parameters for CYP2B1-mediated bupropion metabolism were considerably reduced in MCD diet-fed rats; however, they were also similar between the HF diet-fed and control rats. These results may promote a better understanding of the influence of NAFLD on CYP2B1-mediated metabolism, which could have important implications for the safety and pharmacokinetics of drug substrates for the CYP2B subfamily in patients with NAFLD.
Collapse
Affiliation(s)
- Sung-Joon Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| | - Sang-Bum Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University , Gangwon 24341, Republic of Korea
| | - Saeho Chong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| | - Suk-Jae Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| | - Il-Mo Kang
- Advanced Geo-materials R&D Department, Korea Institute of Geoscience and Mineral Resources, Pohang Branch , Gyeongbuk 37559, Republic of Korea
| | - Jangik Ike Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| | - In-Soo Yoon
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University , Jeonnam 58554, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University , Seoul 08826, Republic of Korea
| |
Collapse
|
45
|
Davidson MD, Ballinger KR, Khetani SR. Long-term exposure to abnormal glucose levels alters drug metabolism pathways and insulin sensitivity in primary human hepatocytes. Sci Rep 2016; 6:28178. [PMID: 27312339 PMCID: PMC4911593 DOI: 10.1038/srep28178] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 05/31/2016] [Indexed: 12/13/2022] Open
Abstract
Hyperglycemia in type 2 diabetes mellitus has been linked to non-alcoholic fatty liver disease, which can progress to inflammation, fibrosis/cirrhosis, and hepatocellular carcinoma. Understanding how chronic hyperglycemia affects primary human hepatocytes (PHHs) can facilitate the development of therapeutics for these diseases. Conversely, elucidating the effects of hypoglycemia on PHHs may provide insights into how the liver adapts to fasting, adverse diabetes drug reactions, and cancer. In contrast to declining PHH monocultures, micropatterned co-cultures (MPCCs) of PHHs and 3T3-J2 murine embryonic fibroblasts maintain insulin-sensitive glucose metabolism for several weeks. Here, we exposed MPCCs to hypo-, normo- and hyperglycemic culture media for ~3 weeks. While albumin and urea secretion were not affected by glucose level, hypoglycemic MPCCs upregulated CYP3A4 enzyme activity as compared to other glycemic states. In contrast, hyperglycemic MPCCs displayed significant hepatic lipid accumulation in the presence of insulin, while also showing decreased sensitivity to insulin-mediated inhibition of glucose output relative to a normoglycemic control. In conclusion, we show for the first time that PHHs exposed to hypo- and hyperglycemia can remain highly functional, but display increased CYP3A4 activity and selective insulin resistance, respectively. In the future, MPCCs under glycemic states can aid in novel drug discovery and mechanistic investigations.
Collapse
Affiliation(s)
- Matthew D Davidson
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA.,Department of Bioengineering, University of Illinois, Chicago, IL 60607, USA
| | - Kimberly R Ballinger
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Salman R Khetani
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA.,Department of Bioengineering, University of Illinois, Chicago, IL 60607, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
46
|
Tolosa L, Gómez-Lechón MJ, López S, Guzmán C, Castell JV, Donato MT, Jover R. Human Upcyte Hepatocytes: Characterization of the Hepatic Phenotype and Evaluation for Acute and Long-Term Hepatotoxicity Routine Testing. Toxicol Sci 2016; 152:214-29. [PMID: 27208088 DOI: 10.1093/toxsci/kfw078] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The capacity of human hepatic cell-based models to predict hepatotoxicity depends on the functional performance of cells. The major limitations of human hepatocytes include the scarce availability and rapid loss of the hepatic phenotype. Hepatoma cells are readily available and easy to handle, but are metabolically poor compared with hepatocytes. Recently developed human upcyte hepatocytes offer the advantage of combining many features of primary hepatocytes with the unlimited availability of hepatoma cells. We analyzed the phenotype of upcyte hepatocytes comparatively with HepG2 cells and adult primary human hepatocytes to characterize their functional features as a differentiated hepatic cell model. The transcriptomic analysis of liver characteristic genes confirmed that the upcyte hepatocytes expression profile comes closer to human hepatocytes than HepG2 cells. CYP activities were measurable and showed a similar response to prototypical CYP inducers than primary human hepatocytes. Upcyte hepatocytes also retained conjugating activities and key hepatic functions, e.g. albumin, urea, lipid and glycogen synthesis, at levels close to hepatocytes. We also investigated the suitability of this cell model for preclinical hepatotoxicity risk assessments using multiparametric high-content screening, as well as transcriptomics and targeted metabolomic analysis. Compounds with well-documented in vivo hepatotoxicity were screened after acute and repeated doses up to 1 week. The evaluation of complex mechanisms of cell toxicity, drug-induced steatosis and oxidative stress biomarkers demonstrated that, by combining the phenotype of primary human hepatocytes and the ease of handling of HepG2 cells, upcyte hepatocytes offer suitable properties to be potentially used for toxicological assessments during drug development.
Collapse
Affiliation(s)
- Laia Tolosa
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain
| | - M José Gómez-Lechón
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain CIBEREHD, Madrid, Spain
| | - Silvia López
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain
| | - Carla Guzmán
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain
| | - José V Castell
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain CIBEREHD, Madrid, Spain Departamento de Bioquímica Y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Spain
| | - M Teresa Donato
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain CIBEREHD, Madrid, Spain Departamento de Bioquímica Y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Spain CIBEREHD, Madrid, Spain
| | - Ramiro Jover
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), Unidad de Hepatología Experimental, Avda. Fernando Abril Martorell, N° 106- Torre A, Valencia, 46026, Spain CIBEREHD, Madrid, Spain Departamento de Bioquímica Y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Spain
| |
Collapse
|
47
|
Michaut A, Le Guillou D, Moreau C, Bucher S, McGill MR, Martinais S, Gicquel T, Morel I, Robin MA, Jaeschke H, Fromenty B. A cellular model to study drug-induced liver injury in nonalcoholic fatty liver disease: Application to acetaminophen. Toxicol Appl Pharmacol 2015; 292:40-55. [PMID: 26739624 DOI: 10.1016/j.taap.2015.12.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Obesity and nonalcoholic fatty liver disease (NAFLD) can increase susceptibility to hepatotoxicity induced by some xenobiotics including drugs, but the involved mechanisms are poorly understood. For acetaminophen (APAP), a role of hepatic cytochrome P450 2E1 (CYP2E1) is suspected since the activity of this enzyme is consistently enhanced during NAFLD. The first aim of our study was to set up a cellular model of NAFLD characterized not only by triglyceride accumulation but also by higher CYP2E1 activity. To this end, human HepaRG cells were incubated for one week with stearic acid or oleic acid, in the presence of different concentrations of insulin. Although cellular triglycerides and the expression of lipid-responsive genes were similar with both fatty acids, CYP2E1 activity was significantly increased only by stearic acid. CYP2E1 activity was reduced by insulin and this effect was reproduced in cultured primary human hepatocytes. Next, APAP cytotoxicity was assessed in HepaRG cells with or without lipid accretion and CYP2E1 induction. Experiments with a large range of APAP concentrations showed that the loss of ATP and glutathione was almost always greater in the presence of stearic acid. In cells pretreated with the CYP2E1 inhibitor chlormethiazole, recovery of ATP was significantly higher in the presence of stearate with low (2.5mM) or high (20mM) concentrations of APAP. Levels of APAP-glucuronide were significantly enhanced by insulin. Hence, HepaRG cells can be used as a valuable model of NAFLD to unveil important metabolic and hormonal factors which can increase susceptibility to drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Anaïs Michaut
- INSERM, U991, Université de Rennes 1, Rennes, France
| | | | - Caroline Moreau
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Simon Bucher
- INSERM, U991, Université de Rennes 1, Rennes, France
| | - Mitchell R McGill
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Thomas Gicquel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Isabelle Morel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | | | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|
48
|
Aljomah G, Baker SS, Liu W, Kozielski R, Oluwole J, Lupu B, Baker RD, Zhu L. Induction of CYP2E1 in non-alcoholic fatty liver diseases. Exp Mol Pathol 2015; 99:677-81. [PMID: 26551085 DOI: 10.1016/j.yexmp.2015.11.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/04/2015] [Indexed: 01/01/2023]
Abstract
Mounting evidence supports a contribution of endogenous alcohol metabolism in the pathogenesis of non-alcoholic steatohepatitis (NASH). However, it is not known whether the expression of alcohol metabolism genes is altered in the livers of simple steatosis. There is also a current debate on whether fatty acids induce CYP2E1 in fatty livers. In this study, expression of alcohol metabolizing genes in the liver biopsies of simple steatosis patients was examined by quantitative real-time PCR (qRT-PCR), in comparison to biopsies of NASH livers and normal controls. Induction of alcohol metabolizing genes was also examined in cultured HepG2 cells treated with ethanol or oleic acid, by qRT-PCR and Western blots. We found that the mRNA expression of alcohol metabolizing genes including ADH1C, ADH4, ADH6, catalase and CYP2E1 was elevated in the livers of simple steatosis, to similar levels found in NASH livers. In cultured HepG2 cells, ethanol induced the expression of CYP2E1 mRNA and protein, but not ADH4 or ADH6; oleic acid did not induce any of these genes. These results suggest that elevated alcohol metabolism may contribute to the pathogenesis of NAFLD at the stage of simple steatosis as well as more severe stages. Our in vitro data support that CYP2E1 is induced by endogenous alcohol but not by fatty acids.
Collapse
Affiliation(s)
- Ghanim Aljomah
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States
| | - Susan S Baker
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States.
| | - Wensheng Liu
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States
| | - Rafal Kozielski
- Department of Pathology, SUNY at Buffalo, Women and Children's Hospital of Buffalo, Buffalo, NY 14214, United States
| | - Janet Oluwole
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States
| | - Benita Lupu
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States
| | - Robert D Baker
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States
| | - Lixin Zhu
- Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, Department of Pediatrics, SUNY at Buffalo, United States.
| |
Collapse
|
49
|
Woolsey SJ, Mansell SE, Kim RB, Tirona RG, Beaton MD. CYP3A Activity and Expression in Nonalcoholic Fatty Liver Disease. Drug Metab Dispos 2015; 43:1484-90. [PMID: 26231377 DOI: 10.1124/dmd.115.065979] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/29/2015] [Indexed: 02/13/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of liver disease in the Western world, given its association with obesity, type 2 diabetes, and dyslipidemia. Medications are widely used in NAFLD to manage comorbid conditions, and there is significant interest in developing new drug therapies to treat the disease. Despite this, little is known about the effects of NAFLD on drug metabolism. We examined the activity and expression of the major drug-metabolizing enzyme subfamily, CYP3A, in subjects with NAFLD as well as in mouse and cellular models. CYP3A activity was determined in healthy volunteers and subjects with biopsy-proven NAFLD by oral midazolam phenotyping and measurement of plasma 4β-hydroxycholesterol, an endogenous metabolic biomarker. CYP3A4 transcriptional activity, metabolic activity, and expression were also assessed in a mouse and cellular model of NAFLD. Subjects with nonalcoholic steatohepatitis (NASH) had 2.4-fold higher plasma midazolam levels compared with controls. Plasma 4β-hydroxycholesterol was 51% and 37% lower than controls in subjects with simple steatosis and NASH, respectively. Fibrosis was associated with 57% lower plasma 4β-hydroxycholesterol levels than controls. Furthermore, hepatic CYP3A4 mRNA expression in NASH was 69% lower than control livers. CYP3A4 gene luciferase activity in the livers of NAFLD mice was 38% lower than that of controls. Lipid-loaded Huh7 human hepatoma cells had a 38% reduction in CYP3A4 activity and 80% lower CYP3A4 mRNA expression compared with the control. CYP3A activity is reduced in human NAFLD in addition to mouse and in vitro cell models of the disease.
Collapse
Affiliation(s)
- Sarah J Woolsey
- Divisions of Clinical Pharmacology (S.J.W, S.E.M, R.B.K, R.G.T.) and Gastroenterology (M.D.B), Departments of Medicine (S.J.W, S.E.M, R.B.K, R.G.T., M.D.B.) and Physiology and Pharmacology (S.J.W, R.B.K, R.G.T.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sara E Mansell
- Divisions of Clinical Pharmacology (S.J.W, S.E.M, R.B.K, R.G.T.) and Gastroenterology (M.D.B), Departments of Medicine (S.J.W, S.E.M, R.B.K, R.G.T., M.D.B.) and Physiology and Pharmacology (S.J.W, R.B.K, R.G.T.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Richard B Kim
- Divisions of Clinical Pharmacology (S.J.W, S.E.M, R.B.K, R.G.T.) and Gastroenterology (M.D.B), Departments of Medicine (S.J.W, S.E.M, R.B.K, R.G.T., M.D.B.) and Physiology and Pharmacology (S.J.W, R.B.K, R.G.T.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Rommel G Tirona
- Divisions of Clinical Pharmacology (S.J.W, S.E.M, R.B.K, R.G.T.) and Gastroenterology (M.D.B), Departments of Medicine (S.J.W, S.E.M, R.B.K, R.G.T., M.D.B.) and Physiology and Pharmacology (S.J.W, R.B.K, R.G.T.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Melanie D Beaton
- Divisions of Clinical Pharmacology (S.J.W, S.E.M, R.B.K, R.G.T.) and Gastroenterology (M.D.B), Departments of Medicine (S.J.W, S.E.M, R.B.K, R.G.T., M.D.B.) and Physiology and Pharmacology (S.J.W, R.B.K, R.G.T.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
50
|
Effect of tetrahydrocurcumin on the profiles of drug-metabolizing enzymes induced by a high fat and high fructose diet in mice. Chem Biol Interact 2015; 239:67-75. [DOI: 10.1016/j.cbi.2015.06.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/12/2015] [Accepted: 06/18/2015] [Indexed: 12/26/2022]
|