1
|
Fuchs CD, Simbrunner B, Baumgartner M, Campbell C, Reiberger T, Trauner M. Bile acid metabolism and signalling in liver disease. J Hepatol 2025; 82:134-153. [PMID: 39349254 DOI: 10.1016/j.jhep.2024.09.032] [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: 03/07/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024]
Abstract
Bile acids (BAs) serve as signalling molecules, efficiently regulating their own metabolism and transport, as well as key aspects of lipid and glucose homeostasis. BAs shape the gut microbial flora and conversely are metabolised by microbiota. Disruption of BA transport, metabolism and physiological signalling functions contribute to the pathogenesis and progression of a wide range of liver diseases including cholestatic disorders and MASLD (metabolic dysfunction-associated steatotic liver disease), as well as hepatocellular and cholangiocellular carcinoma. Additionally, impaired BA signalling may also affect the intestine and kidney, thereby contributing to failure of gut integrity and driving the progression and complications of portal hypertension, cholemic nephropathy and the development of extrahepatic malignancies such as colorectal cancer. In this review, we will summarise recent advances in the understanding of BA signalling, metabolism and transport, focusing on transcriptional regulation and novel BA-focused therapeutic strategies for cholestatic and metabolic liver diseases.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maximillian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Clarissa Campbell
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Qu Q, Chen Y, Wang Y, Long S, Wang W, Yang HY, Li M, Tian X, Wei X, Liu YH, Xu S, Zhang C, Zhu M, Lam SM, Wu J, Yun C, Chen J, Xue S, Zhang B, Zheng ZZ, Piao HL, Jiang C, Guo H, Shui G, Deng X, Zhang CS, Lin SC. Lithocholic acid phenocopies anti-ageing effects of calorie restriction. Nature 2024:10.1038/s41586-024-08329-5. [PMID: 39695227 DOI: 10.1038/s41586-024-08329-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 10/31/2024] [Indexed: 12/20/2024]
Abstract
Calorie restriction (CR) is a dietary intervention used to promote health and longevity1,2. CR causes various metabolic changes in both the production and the circulation of metabolites1; however, it remains unclear which altered metabolites account for the physiological benefits of CR. Here we use metabolomics to analyse metabolites that exhibit changes in abundance during CR and perform subsequent functional validation. We show that lithocholic acid (LCA) is one of the metabolites that alone can recapitulate the effects of CR in mice. These effects include activation of AMP-activated protein kinase (AMPK), enhancement of muscle regeneration and rejuvenation of grip strength and running capacity. LCA also activates AMPK and induces life-extending and health-extending effects in Caenorhabditis elegans and Drosophila melanogaster. As C. elegans and D. melanogaster are not able to synthesize LCA, these results indicate that these animals are able to transmit the signalling effects of LCA once administered. Knockout of AMPK abrogates LCA-induced phenotypes in all the three animal models. Together, we identify that administration of the CR-mediated upregulated metabolite LCA alone can confer anti-ageing benefits to metazoans in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Qi Qu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Yan Chen
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Yu Wang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Shating Long
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Weiche Wang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Heng-Ye Yang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Mengqi Li
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Xiao Tian
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Xiaoyan Wei
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Yan-Hui Liu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Shengrong Xu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Cixiong Zhang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Mingxia Zhu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | | | - Jianfeng Wu
- Laboratory Animal Research Centre, Xiamen University, Fujian, China
| | - Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
| | - Junjie Chen
- Analysis and Measurement Centre, School of Pharmaceutical Sciences, Xiamen University, Fujian, China
| | - Shengye Xue
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Baoding Zhang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Zhong-Zheng Zheng
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Liaoning, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, Department of Immunology, School of Basic Medical Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodelling, Peking University, Beijing, China
| | - Hao Guo
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
- Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Guanghou Shui
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, China
| | - Xianming Deng
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Chen-Song Zhang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China.
| | - Sheng-Cai Lin
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China.
| |
Collapse
|
3
|
Omotoso AO, Reyer H, Oster M, Ponsuksili S, Metzler-Zebeli B, Wimmers K. Hepatic Transcriptomics of Broilers with Low and High Feed Conversion in Response to Caloric Restriction. Metabolites 2024; 14:625. [PMID: 39590861 PMCID: PMC11596519 DOI: 10.3390/metabo14110625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In broiler chickens, the efficient utilization of macro- and micronutrients is influenced by various metabolic pathways that are closely linked to feed efficiency (FE), a critical metric in poultry industry, with residual feed intake (RFI) as the preferred proxy. Feed restriction is considered an approach to address the underlying molecular mechanisms of feed conversion. We hypothesized that broiler chickens with divergent RFI subjected to quantitative feed restriction differ in their pattern of molecular pathways for efficient nutrient utilization in liver as post-absorptive tissue. METHODS Cobb 500FF broiler chickens divergent for RFI (n = 112) were feed-restricted from day 9 until market weight at day 33-37 post-hatch. Based on a previous trial, feed restriction levels were set at 92% (low-RFI birds) and 80% (high-RFI birds) relative to the control groups. Transcriptomic analyses of the liver were conducted. RESULTS Due to the interaction of the RFI group and feeding regimen, a total of 140 to 507 differentially expressed genes were identified for the respective contrasts, with implications for hepatic metabolism and cellular stress response. Although the broilers did not realize their full growth potential under restrictive feeding (12.4% reduced body weight vs. controls, p = 0.094), the gene expression patterns indicate a lower susceptibility to blood coagulation (KNG1, FGG, and FGB), suggesting that controlled and mild feed restriction could lead to health benefits in less feed-efficient broilers. Moreover, FE traits are shown to be linked to cellular detoxification processes (MGST3 and CYP2AC2) and triacylglycerol syntheses (MOGAT1 and LPIN1). CONCLUSIONS Divergent transcriptional profiles between broiler groups under varied caloric conditions indicate potential for optimizing nutritional management strategies.
Collapse
Affiliation(s)
- Adewunmi O. Omotoso
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (A.O.O.); (H.R.); (M.O.); (S.P.)
| | - Henry Reyer
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (A.O.O.); (H.R.); (M.O.); (S.P.)
| | - Michael Oster
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (A.O.O.); (H.R.); (M.O.); (S.P.)
| | - Siriluck Ponsuksili
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (A.O.O.); (H.R.); (M.O.); (S.P.)
| | - Barbara Metzler-Zebeli
- Centre for Veterinary Systems Transformation and Sustainability, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Klaus Wimmers
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (A.O.O.); (H.R.); (M.O.); (S.P.)
- Faculty of Agricultural and Environmental Sciences, University Rostock, 18059 Rostock, Germany
| |
Collapse
|
4
|
MacLeod AK, Coquelin KS, Huertas L, Simeons FRC, Riley J, Casado P, Guijarro L, Casanueva R, Frame L, Pinto EG, Ferguson L, Duncan C, Mutter N, Shishikura Y, Henderson CJ, Cebrian D, Wolf CR, Read KD. Acceleration of infectious disease drug discovery and development using a humanized model of drug metabolism. Proc Natl Acad Sci U S A 2024; 121:e2315069121. [PMID: 38315851 PMCID: PMC10873626 DOI: 10.1073/pnas.2315069121] [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: 09/07/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
A key step in drug discovery, common to many disease areas, is preclinical demonstration of efficacy in a mouse model of disease. However, this demonstration and its translation to the clinic can be impeded by mouse-specific pathways of drug metabolism. Here, we show that a mouse line extensively humanized for the cytochrome P450 gene superfamily ("8HUM") can circumvent these problems. The pharmacokinetics, metabolite profiles, and magnitude of drug-drug interactions of a test set of approved medicines were in much closer alignment with clinical observations than in wild-type mice. Infection with Mycobacterium tuberculosis, Leishmania donovani, and Trypanosoma cruzi was well tolerated in 8HUM, permitting efficacy assessment. During such assessments, mouse-specific metabolic liabilities were bypassed while the impact of clinically relevant active metabolites and DDI on efficacy were well captured. Removal of species differences in metabolism by replacement of wild-type mice with 8HUM therefore reduces compound attrition while improving clinical translation, accelerating drug discovery.
Collapse
Affiliation(s)
- A. Kenneth MacLeod
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Kevin-Sebastien Coquelin
- Division of Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Ninewells Hospital, DundeeDD2 4GD, United Kingdom
| | - Leticia Huertas
- Global Health Research & Development, GlaxoSmithKline, Tres Cantos, Madrid28760, Spain
| | - Frederick R. C. Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Patricia Casado
- Global Health Research & Development, GlaxoSmithKline, Tres Cantos, Madrid28760, Spain
| | - Laura Guijarro
- Global Health Research & Development, GlaxoSmithKline, Tres Cantos, Madrid28760, Spain
| | - Ruth Casanueva
- Global Health Research & Development, GlaxoSmithKline, Tres Cantos, Madrid28760, Spain
| | - Laura Frame
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Erika G. Pinto
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Liam Ferguson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Christina Duncan
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Nicole Mutter
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Yoko Shishikura
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| | - Colin J. Henderson
- Division of Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Ninewells Hospital, DundeeDD2 4GD, United Kingdom
| | - David Cebrian
- Global Health Research & Development, GlaxoSmithKline, Tres Cantos, Madrid28760, Spain
| | - C. Roland Wolf
- Division of Systems Medicine, Jacqui Wood Cancer Centre, School of Medicine, University of Dundee, Ninewells Hospital, DundeeDD2 4GD, United Kingdom
| | - Kevin D. Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, DundeeDD1 5EH, United Kingdom
| |
Collapse
|
5
|
Shinya S, Kawai Y, Kondo M, Nakayama SMM, Ishizuka M, Ikenaka Y. Characteristics of cytochrome P450-dependent metabolism against acetamiprid in the musk shrew (Suncus murinus). Comp Biochem Physiol C Toxicol Pharmacol 2023; 265:109537. [PMID: 36563948 DOI: 10.1016/j.cbpc.2022.109537] [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: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Soricidae spp. (shrews) play an essential role in soil ecosystems and, due to their habitat and behavior, are exposed to soil pollutants, such as pesticides. Still, toxicity risk in Soricidae spp. has not been appropriately assessed. In this study, the musk shrew (Suncus murinus) was used as a model organism for toxicity assessment in Soricidae. Considering their carnivorous diet, it is reasonable to assume that the musk shrew has unique metabolic traits that are different from those of other common experimental models. This study describes the cytochrome P450 (CYP)-dependent metabolism affected by acetamiprid (ACP), a neonicotinoid insecticide. Pharmacokinetics analysis, an in vitro metabolic assay, and genetic analysis of CYP were performed and compared with data from mice and rats. Through phylogenetic and syntenic analyses, three families of CYP were identified in the musk shrew. Pharmacokinetic analysis showed that the blood concentration of ACP decreased more quickly in musk shrews than in mice. Moreover, the in vitro metabolic assay suggested more efficient metabolic responses toward ACP in musk shrews than in mice or rats. One of the CYP2A isoforms in musk shrews might be linked to a better ACP metabolism. From the results above, we describe novel metabolic traits of the musk shrew. Future research on recombinant CYP enzymes is necessary to fully understand CYP-dependent metabolism of xenobiotics in musk shrews.
Collapse
Affiliation(s)
- So Shinya
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Yusuke Kawai
- Laboratory of Toxicology, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inaba-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Mitsuki Kondo
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Shouta M M Nakayama
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan; Biomedical Science Department, School of Veterinary Medicine, The University of Zambia, PO Box 32379, Lusaka, Zambia
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan; Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan; One Health Research Center, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan; Water Research Group, Unit for Environmental Sciences and Management, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa.
| |
Collapse
|
6
|
Kobayashi K, Minegishi G, Kuriyama N, Miyajima A, Abe S, Kazuki K, Kazuki Y. Metabolic Disposition of Triazolam and Clobazam in Humanized CYP3A Mice with a Double-Knockout Background of Mouse Cyp2c and Cyp3a Genes. Drug Metab Dispos 2023; 51:174-182. [PMID: 36379710 DOI: 10.1124/dmd.122.001087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/16/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022] Open
Abstract
Knockout (KO) of mouse Cyp3a genes increases the expression of hepatic CYP2C enzymes, which can metabolize triazolam, a typical substrate of human CYP3A. There is still marked formation of 1'-hydroxytriazolam in Cyp3a-KO (3aKO) mice after triazolam dosing. Here, we generated a new model of humanized CYP3A (hCYP3A) mice with a double-KO background of Cyp3a and Cyp2c genes (2c3aKO), and we examined the metabolic profiles of triazolam in wild-type (WT), 2c3aKO, and hCYP3A/2c3aKO mice in vitro and in vivo In vitro studies using liver microsomes showed that the formation of 1'-hydroxytriazolam in 2c3aKO mice was less than 8% of that in WT mice. The formation rate of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was eightfold higher than that in 2c3aKO mice. In vivo studies showed that area under the curve (AUC) of 1'-hydroxytriazolam in 2c3aKO mice was less than 3% of that in WT mice. The AUC of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was sixfold higher than that in 2c3aKO mice. These results showed that formation of 1'-hydroxytriazolam was significantly decreased in 2c3aKO mice. Metabolic functions of human CYP3A enzymes were distinctly found in hCYP3A mice with the 2c3aKO background. Moreover, hCYP3A/2c3aKO mice treated with clobazam showed human CYP3A-mediated formation of desmethylclobazam and prolonged elimination of desmethylclobazam, which is found in poor metabolizers of CYP2C19. The novel hCYP3A mouse model without mouse Cyp2c and Cyp3a genes (hCYP3A/2c3aKO) is expected to be useful to evaluate human CYP3A-mediated metabolism in vivo SIGNIFICANT STATEMENT: Humanized CYP3A (hCYP3A/2c3aKO) mice with a background of double knockout (KO) for mouse Cyp2c and Cyp3a genes were generated. Although CYP2C enzymes played a compensatory role in the metabolism of triazolam to 1'-hydroxytriazolam in the previous hCYP3A/3aKO mice with Cyp2c genes, the novel hCYP3A/2c3aKO mice clearly showed functions of human CYP3A enzymes introduced by chromosome engineering technology.
Collapse
Affiliation(s)
- Kaoru Kobayashi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Genki Minegishi
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Nina Kuriyama
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Atsushi Miyajima
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Satoshi Abe
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Kanako Kazuki
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yasuhiro Kazuki
- Department of Biopharmaceutics, Graduate School of Clinical Pharmacy, Meiji Pharmaceutical University, Kiyose, Japan (K.Ko., G.M., N.K., A.M.) and Chromosome Engineering Research Center (CERC) (S.A., K.Ka., Y.K.) and Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| |
Collapse
|
7
|
Dornburg A, Mallik R, Wang Z, Bernal MA, Thompson B, Bruford EA, Nebert DW, Vasiliou V, Yohe LR, Yoder JA, Townsend JP. Placing human gene families into their evolutionary context. Hum Genomics 2022; 16:56. [PMID: 36369063 PMCID: PMC9652883 DOI: 10.1186/s40246-022-00429-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Following the draft sequence of the first human genome over 20 years ago, we have achieved unprecedented insights into the rules governing its evolution, often with direct translational relevance to specific diseases. However, staggering sequence complexity has also challenged the development of a more comprehensive understanding of human genome biology. In this context, interspecific genomic studies between humans and other animals have played a critical role in our efforts to decode human gene families. In this review, we focus on how the rapid surge of genome sequencing of both model and non-model organisms now provides a broader comparative framework poised to empower novel discoveries. We begin with a general overview of how comparative approaches are essential for understanding gene family evolution in the human genome, followed by a discussion of analyses of gene expression. We show how homology can provide insights into the genes and gene families associated with immune response, cancer biology, vision, chemosensation, and metabolism, by revealing similarity in processes among distant species. We then explain methodological tools that provide critical advances and show the limitations of common approaches. We conclude with a discussion of how these investigations position us to gain fundamental insights into the evolution of gene families among living organisms in general. We hope that our review catalyzes additional excitement and research on the emerging field of comparative genomics, while aiding the placement of the human genome into its existentially evolutionary context.
Collapse
Affiliation(s)
- Alex Dornburg
- Department of Bioinformatics and Genomics, UNC-Charlotte, Charlotte, NC, USA.
| | - Rittika Mallik
- Department of Bioinformatics and Genomics, UNC-Charlotte, Charlotte, NC, USA
| | - Zheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Moisés A Bernal
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, USA
| | - Brian Thompson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Elspeth A Bruford
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Daniel W Nebert
- Department of Environmental Health, Center for Environmental Genetics, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati, OH, 45267, USA
- Department of Pediatrics and Molecular Developmental Biology, Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, OH, 45229, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Laurel R Yohe
- Department of Bioinformatics and Genomics, UNC-Charlotte, Charlotte, NC, USA
| | - Jeffrey A Yoder
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Jeffrey P Townsend
- Department of Bioinformatics and Genomics, UNC-Charlotte, Charlotte, NC, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
8
|
Hannon SL, Ding X. Assessing cytochrome P450 function using genetically engineered mouse models. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:253-284. [PMID: 35953157 PMCID: PMC10544722 DOI: 10.1016/bs.apha.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The ability to knock out and/or humanize different genes in experimental animals, globally or in cell- and tissue-specific patterns, has revolutionized scientific research in many areas. Genetically engineered mouse models, including knockout models, transgenic models, and humanized models, have played important roles in revealing the in vivo functions of various cytochrome P450 (CYP) enzymes. These functions are very diverse, ranging from the biotransformation of drugs and other xenobiotics, events that often dictate their pharmacokinetic or toxicokinetic properties and the associated therapeutic or adverse actions, to the metabolism of endogenous compounds, such as steroid hormones and other bioactive substances, that may determine susceptibility to many diseases, such as cancer and metabolic diseases. In this review, we provide a comprehensive list of Cyp-knockout, human CYP-transgenic, and CYP-humanized mouse models that target genes in the CYP1-4 gene families, and highlight their utility in assessing the in vivo metabolism, bioactivation, and toxicity of various xenobiotic compounds, including therapeutic agents and chemical carcinogens. We aim to showcase the advantages of utilizing these mouse models for in vivo drug metabolism and toxicology studies, and to encourage and facilitate greater utility of engineered mouse models to further improve our knowledge of the in vivo functions of various P450 enzymes, which is integral to our ability to develop safer and more effective therapeutics and to identify individuals predisposed to adverse drug reactions or environmental diseases.
Collapse
Affiliation(s)
- Sarrah L Hannon
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, Ken R. Coit College of Pharmacy, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
9
|
Oteng AB, Higuchi S, Banks AS, Haeusler RA. Cyp2c-deficiency depletes muricholic acids and protects against high-fat diet-induced obesity in male mice but promotes liver damage. Mol Metab 2021; 53:101326. [PMID: 34438105 PMCID: PMC8449133 DOI: 10.1016/j.molmet.2021.101326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Murine-specific muricholic acids (MCAs) are reported to protect against obesity and associated metabolic disorders. However, the response of mice with genetic depletion of MCA to an obesogenic diet has not been evaluated. We used Cyp2c-deficient (Cyp2c−/−) mice, which lack MCAs and thus have a human-like bile acid (BA) profile, to directly investigate the potential role of MCAs in diet-induced obesity. Methods Male and female Cyp2c−/− mice and wild-type (WT) littermate controls were fed a standard chow diet or a high-fat diet (HFD) for 18 weeks. We measured BA composition from a pool of liver, gallbladder, and intestine, as well as weekly body weight, food intake, lean and fat mass, systemic glucose homeostasis, energy expenditure, intestinal lipid absorption, fecal lipid, and energy content. Results Cyp2c-deficiency depleted MCAs and caused other changes in BA composition, namely a decrease in the ratio of 12α-hydroxylated (12α-OH) BAs to non-12α-OH BAs, without altering the total BA levels. While WT male mice became obese after HFD feeding, Cyp2c−/− male mice were protected from obesity and associated metabolic dysfunctions. Cyp2c−/− male mice also showed reduced intestinal lipid absorption and increased lipid excretion, which was reversed by oral gavage with the 12α-OH BA and taurocholic acid (TCA). Cyp2c−/− mice also showed increased liver damage, which appeared stronger in females. Conclusions MCA does not protect against diet-induced obesity but may protect against liver injury. Reduced lipid absorption in Cyp2c-deficient male mice is potentially due to a reduced ratio of 12α-OH/non-12α-OH BAs. Presence of MCA does not necessarily protect against diet-induced obesity. Cyp2c deficiency promotes resistance to diet-induced obesity in males. Cyp2c-knockout mice have decreased the ratio of 12α-OH/non-12α-OH BAs that promotes decreased intestinal lipid absorption. Cyp2c-knockout mice have improved glucose homeostasis. Cyp2c-deficiency promotes mild and severe liver injury in male and female mice, respectively.
Collapse
Affiliation(s)
- Antwi-Boasiako Oteng
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sei Higuchi
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Alexander S Banks
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
10
|
de Boer JF, de Vries HD, Palmiotti A, Li R, Doestzada M, Hoogerland JA, Fu J, La Rose AM, Westerterp M, Mulder NL, Hovingh MV, Koehorst M, Kloosterhuis NJ, Wolters JC, Bloks VW, Haas JT, Dombrowicz D, Staels B, van de Sluis B, Kuipers F. Cholangiopathy and Biliary Fibrosis in Cyp2c70-Deficient Mice Are Fully Reversed by Ursodeoxycholic Acid. Cell Mol Gastroenterol Hepatol 2020; 11:1045-1069. [PMID: 33309945 PMCID: PMC7898074 DOI: 10.1016/j.jcmgh.2020.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Bile acids (BAs) aid intestinal fat absorption and exert systemic actions by receptor-mediated signaling. BA receptors have been identified as drug targets for liver diseases. Yet, differences in BA metabolism between humans and mice hamper translation of pre-clinical outcomes. Cyp2c70-ablation in mice prevents synthesis of mouse/rat-specific muricholic acids (MCAs), but potential (patho)physiological consequences of their absence are unknown. We therefore assessed age- and gender-dependent effects of Cyp2c70-deficiency in mice. METHODS The consequences of Cyp2c70-deficiency were assessed in male and female mice at different ages. RESULTS Cyp2c70-/- mice were devoid of MCAs and showed high abundances of chenodeoxycholic and lithocholic acids. Cyp2c70-deficiency profoundly impacted microbiome composition. Bile flow and biliary BA secretion were normal in Cyp2c70-/- mice of both sexes. Yet, the pathophysiological consequences of Cyp2c70-deficiency differed considerably between sexes. Three-week old male Cyp2c70-/- mice showed high plasma BAs and transaminases, which spontaneously decreased thereafter to near-normal levels. Only mild ductular reactions were observed in male Cyp2c70-/- mice up to 8 months of age. In female Cyp2c70-/- mice, plasma BAs and transaminases remained substantially elevated with age, gut barrier function was impaired and bridging fibrosis was observed at advanced age. Addition of 0.1% ursodeoxycholic acid to the diet fully normalized hepatic and intestinal functions in female Cyp2c70-/- mice. CONCLUSION Cyp2c70-/- mice show transient neonatal cholestasis and develop cholangiopathic features that progress to bridging fibrosis in females only. These consequences of Cyp2c70-deficiency are restored by treatment with UDCA, indicating a role of BA hydrophobicity in disease development.
Collapse
Affiliation(s)
- Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Hilde D de Vries
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; University of Groningen, Campus Fryslân, Leeuwarden, the Netherlands
| | - Anna Palmiotti
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Rumei Li
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Marwah Doestzada
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Genetics University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Joanne A Hoogerland
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1011-EGID, F-59000 Lille, France
| | - Jingyuan Fu
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Genetics University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anouk M La Rose
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Niels L Mulder
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Milaine V Hovingh
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Joel T Haas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1011-EGID, F-59000 Lille, France
| | - David Dombrowicz
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1011-EGID, F-59000 Lille, France
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands; iPSC/CRISPR Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
11
|
Liang RJ, Shih YN, Chen YL, Liu WY, Yang WL, Lee SY, Wang HJ. A dual system platform for drug metabolism: Nalbuphine as a model compound. Eur J Pharm Sci 2020; 141:105093. [DOI: 10.1016/j.ejps.2019.105093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/26/2019] [Accepted: 09/28/2019] [Indexed: 01/26/2023]
|
12
|
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, Wang X, Lyu Y, Chen X, Liu K, Yu AM, Zuo Z, Bi H. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144. [PMID: 31867160 PMCID: PMC6900561 DOI: 10.1016/j.apsb.2019.10.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Pharmacokinetics (PK) is the study of the absorption, distribution, metabolism, and excretion (ADME) processes of a drug. Understanding PK properties is essential for drug development and precision medication. In this review we provided an overview of recent research on PK with focus on the following aspects: (1) an update on drug-metabolizing enzymes and transporters in the determination of PK, as well as advances in xenobiotic receptors and noncoding RNAs (ncRNAs) in the modulation of PK, providing new understanding of the transcriptional and posttranscriptional regulatory mechanisms that result in inter-individual variations in pharmacotherapy; (2) current status and trends in assessing drug-drug interactions, especially interactions between drugs and herbs, between drugs and therapeutic biologics, and microbiota-mediated interactions; (3) advances in understanding the effects of diseases on PK, particularly changes in metabolizing enzymes and transporters with disease progression; (4) trends in mathematical modeling including physiologically-based PK modeling and novel animal models such as CRISPR/Cas9-based animal models for DMPK studies; (5) emerging non-classical xenobiotic metabolic pathways and the involvement of novel metabolic enzymes, especially non-P450s. Existing challenges and perspectives on future directions are discussed, and may stimulate the development of new research models, technologies, and strategies towards the development of better drugs and improved clinical practice.
Collapse
Affiliation(s)
- Yuhua Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
- The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Mengbi Yang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Hou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Tang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanfeng Lyu
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ai-Ming Yu
- UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Zhong Zuo
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
13
|
Honda A, Miyazaki T, Iwamoto J, Hirayama T, Morishita Y, Monma T, Ueda H, Mizuno S, Sugiyama F, Takahashi S, Ikegami T. Regulation of bile acid metabolism in mouse models with hydrophobic bile acid composition. J Lipid Res 2019; 61:54-69. [PMID: 31645370 DOI: 10.1194/jlr.ra119000395] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
The bile acid (BA) composition in mice is substantially different from that in humans. Chenodeoxycholic acid (CDCA) is an end product in the human liver; however, mouse Cyp2c70 metabolizes CDCA to hydrophilic muricholic acids (MCAs). Moreover, in humans, the gut microbiota converts the primary BAs, cholic acid and CDCA, into deoxycholic acid (DCA) and lithocholic acid (LCA), respectively. In contrast, the mouse Cyp2a12 reverts this action and converts these secondary BAs to primary BAs. Here, we generated Cyp2a12 KO, Cyp2c70 KO, and Cyp2a12/Cyp2c70 double KO (DKO) mice using the CRISPR-Cas9 system to study the regulation of BA metabolism under hydrophobic BA composition. Cyp2a12 KO mice showed the accumulation of DCAs, whereas Cyp2c70 KO mice lacked MCAs and exhibited markedly increased hepatobiliary proportions of CDCA. In DKO mice, not only DCAs or CDCAs but also DCAs, CDCAs, and LCAs were all elevated. In Cyp2c70 KO and DKO mice, chronic liver inflammation was observed depending on the hepatic unconjugated CDCA concentrations. The BA pool was markedly reduced in Cyp2c70 KO and DKO mice, but the FXR was not activated. It was suggested that the cytokine/c-Jun N-terminal kinase signaling pathway and the pregnane X receptor-mediated pathway are the predominant mechanisms, preferred over the FXR/small heterodimer partner and FXR/fibroblast growth factor 15 pathways, for controlling BA synthesis under hydrophobic BA composition. From our results, we hypothesize that these KO mice can be novel and useful models for investigating the roles of hydrophobic BAs in various human diseases.
Collapse
Affiliation(s)
- Akira Honda
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan; Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan.
| | - Teruo Miyazaki
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Junichi Iwamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Takeshi Hirayama
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Yukio Morishita
- Diagnostic Pathology Division, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Tadakuni Monma
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Hajime Ueda
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Tadashi Ikegami
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
14
|
Kapelyukh Y, Henderson CJ, Scheer N, Rode A, Wolf CR. Defining the Contribution of CYP1A1 and CYP1A2 to Drug Metabolism Using Humanized CYP1A1/1A2 and Cyp1a1/Cyp1a2 Knockout Mice. Drug Metab Dispos 2019; 47:907-918. [PMID: 31147315 PMCID: PMC6657216 DOI: 10.1124/dmd.119.087718] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/28/2019] [Indexed: 12/16/2022] Open
Abstract
Cytochrome P450s CYP1A1 and CYP1A2 can metabolize a broad range of foreign compounds and drugs. However, these enzymes have significantly overlapping substrate specificities. To establish their relative contribution to drug metabolism in vivo, we used a combination of mice humanized for CYP1A1 and CYP1A2 together with mice nulled at the Cyp1a1 and Cyp1a2 gene loci. CYP1A2 was constitutively expressed in the liver, and both proteins were highly inducible by 2,3,7,8-tetrachlorodibenzodioxin (TCDD) in a number of tissues, including the liver, lung, kidney, and small intestine. Using the differential inhibition of the human enzymes by quinidine, we developed a method to distinguish the relative contribution of CYP1A1 or CYP1A2 in the metabolism of drugs and foreign compounds. Both enzymes made a significant contribution to the hepatic metabolism of the probe compounds 7-methoxy and 7-ehthoxyresorufin in microsomal fractions from animals treated with TCDD. This enzyme kinetic approach allows modeling of the CYP1A1, CYP1A2, and non-CYP1A contribution to the metabolism of any substrate at any substrate, inhibitor, or enzyme concentration and, as a consequence, can be integrated into a physiologically based pharmacokinetics model. The validity of the model can then be tested in humanized mice in vivo. SIGNIFICANCE STATEMENT: Human CYP1A1 and CYP1A2 are important in defining the efficacy and toxicity/carcinogenicity of drugs and foreign compounds. In light of differences in substrate specificity and sensitivity to inhibitors, it is of central importance to understand their relative role in foreign compound metabolism. To address this issue, we have generated mice humanized or nulled at the Cyp1a gene locus and, through the use of these mouse lines and selective inhibitors, developed an enzyme kinetic-based model to enable more accurate prediction of the fate of new chemicals in humans and which can be validated in vivo using mice humanized for cytochrome P450-mediated metabolism.
Collapse
Affiliation(s)
- Y Kapelyukh
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (Y.K., C.J.H., C.R.W.) and Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.)
| | - C J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (Y.K., C.J.H., C.R.W.) and Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.)
| | - N Scheer
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (Y.K., C.J.H., C.R.W.) and Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.)
| | - A Rode
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (Y.K., C.J.H., C.R.W.) and Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.)
| | - C R Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (Y.K., C.J.H., C.R.W.) and Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.)
| |
Collapse
|
15
|
Li Y, Xu Y, Jadhav K, Zhu Y, Yin L, Zhang Y. Hepatic Forkhead Box Protein A3 Regulates ApoA-I (Apolipoprotein A-I) Expression, Cholesterol Efflux, and Atherogenesis. Arterioscler Thromb Vasc Biol 2019; 39:1574-1587. [PMID: 31291759 DOI: 10.1161/atvbaha.119.312610] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine the role of hepatic FOXA3 (forkhead box A3) in lipid metabolism and atherosclerosis. Approach and Results: Hepatic FOXA3 expression was reduced in diabetic or high fat diet-fed mice or patients with nonalcoholic steatohepatitis. We then used adenoviruses to overexpress or knock down hepatic FOXA3 expression. Overexpression of FOXA3 in the liver increased hepatic ApoA-I (apolipoprotein A-I) expression, plasma HDL-C (high-density lipoprotein cholesterol) level, macrophage cholesterol efflux, and macrophage reverse cholesterol transport. In contrast, knockdown of hepatic FOXA3 expression had opposite effects. We further showed that FOXA3 directly bound to the promoter of the Apoa1 gene to regulate its transcription. Finally, AAV8 (adeno-associated virus serotype 8)-mediated overexpression of human FOXA3 in the hepatocytes of Apoe-/- (apolipoprotein E-deficient) mice raised plasma HDL-C levels and significantly reduced atherosclerotic lesions. CONCLUSIONS Hepatocyte FOXA3 protects against atherosclerosis by inducing ApoA-I and macrophage reverse cholesterol transport.
Collapse
Affiliation(s)
- Yuanyuan Li
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yanyong Xu
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Kavita Jadhav
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yingdong Zhu
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Liya Yin
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| | - Yanqiao Zhang
- From the Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown
| |
Collapse
|
16
|
Henderson CJ, Kapelyukh Y, Scheer N, Rode A, McLaren AW, MacLeod AK, Lin D, Wright J, Stanley LA, Wolf CR. An Extensively Humanized Mouse Model to Predict Pathways of Drug Disposition and Drug/Drug Interactions, and to Facilitate Design of Clinical Trials. Drug Metab Dispos 2019; 47:601-615. [PMID: 30910785 PMCID: PMC6505380 DOI: 10.1124/dmd.119.086397] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Species differences in drug metabolism and disposition can confound the extrapolation of in vivo PK data to man and also profoundly compromise drug efficacy studies owing to differences in pharmacokinetics, in metabolites produced (which are often pharmacologically active), and in differential activation of the transcription factors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), which regulate the expression of such enzymes as P450s and drug transporters. These differences have gained additional importance as a consequence of the use of genetically modified mouse models for drug-efficacy testing and also patient-derived xenografts to predict individual patient responses to anticancer drugs. A number of humanized mouse models for cytochrome P450s, CAR, and PXR have been reported. However, the utility of these models has been compromised by the redundancy in P450 reactions across gene families, whereby the remaining murine P450s can metabolize the compounds being tested. To remove this confounding factor and create a mouse model that more closely reflects human pathways of drug disposition, we substituted 33 murine P450s from the major gene families involved in drug disposition, together with Car and Pxr, for human CAR, PXR, CYP1A1, CYP1A2, CYP2C9, CYP2D6, CYP3A4, and CYP3A7. We also created a mouse line in which 34 P450s were deleted from the mouse genome. Using model compounds and anticancer drugs, we demonstrated how these mouse lines can be applied to predict drug-drug interactions in patients and discuss here their potential application in the more informed design of clinical trials and the personalized treatment of cancer.
Collapse
Affiliation(s)
- C J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - Y Kapelyukh
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - N Scheer
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A Rode
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A W McLaren
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A K MacLeod
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - D Lin
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - J Wright
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - L A Stanley
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - C R Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| |
Collapse
|
17
|
Wang W, Yang J, Edin ML, Wang Y, Luo Y, Wan D, Yang H, Song CQ, Xue W, Sanidad KZ, Song M, Bisbee HA, Bradbury JA, Nan G, Zhang J, Shih PAB, Lee KSS, Minter LM, Kim D, Xiao H, Liu JY, Hammock BD, Zeldin DC, Zhang G. Targeted Metabolomics Identifies the Cytochrome P450 Monooxygenase Eicosanoid Pathway as a Novel Therapeutic Target of Colon Tumorigenesis. Cancer Res 2019; 79:1822-1830. [PMID: 30803995 PMCID: PMC6467714 DOI: 10.1158/0008-5472.can-18-3221] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/08/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
Abstract
Colon cancer is the third most common cancer and the second leading cause of cancer-related death in the United States, emphasizing the need for the discovery of new cellular targets. Using a metabolomics approach, we report here that epoxygenated fatty acids (EpFA), which are eicosanoid metabolites produced by cytochrome P450 (CYP) monooxygenases, were increased in both the plasma and colon of azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon cancer mice. CYP monooxygenases were overexpressed in colon tumor tissues and colon cancer cells. Pharmacologic inhibition or genetic ablation of CYP monooxygenases suppressed AOM/DSS-induced colon tumorigenesis in vivo. In addition, treatment with 12,13-epoxyoctadecenoic acid (EpOME), which is a metabolite of CYP monooxygenase produced from linoleic acid, increased cytokine production and JNK phosphorylation in vitro and exacerbated AOM/DSS-induced colon tumorigenesis in vivo. Together, these results demonstrate that the previously unappreciated CYP monooxygenase pathway is upregulated in colon cancer, contributes to its pathogenesis, and could be therapeutically explored for preventing or treating colon cancer. SIGNIFICANCE: This study finds that the previously unappreciated CYP monooxygenase eicosanoid pathway is deregulated in colon cancer and contributes to colon tumorigenesis.
Collapse
Affiliation(s)
- Weicang Wang
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
| | - Jun Yang
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California
| | - Matthew L Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Yuxin Wang
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Ying Luo
- Center for Nephrology and Metabolomics and Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Debin Wan
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California
| | - Haixia Yang
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
| | - Chun-Qing Song
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts
- Program in Molecular Medicine, Department of Molecular, Cell and Cancer Biology, and Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Katherine Z Sanidad
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
| | - Mingyue Song
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Heather A Bisbee
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
| | - Jennifer A Bradbury
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Guanjun Nan
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianan Zhang
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
| | - Pei-An Betty Shih
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Lisa M Minter
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Daeyoung Kim
- Department of Mathematics & Statistics, University of Massachusetts, Amherst, Massachusetts
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
| | - Jun-Yan Liu
- Center for Nephrology and Metabolomics and Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bruce D Hammock
- Department of Entomology and Comprehensive Cancer Center, University of California, Davis, California.
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina.
| | - Guodong Zhang
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts.
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
18
|
Ye H, Sui D, Liu W, Yuan Y, Ouyang Z, Wei Y. Effects of CYP2C11 gene knockout on the pharmacokinetics and pharmacodynamics of warfarin in rats. Xenobiotica 2019; 49:1478-1484. [PMID: 30724651 DOI: 10.1080/00498254.2019.1579006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
1. CYP2C11 is the most abundant isoform of cytochrome P450s (CYPs) in male rats and is considered the main enzyme for warfarin metabolism. 2. To further access the in vivo function of CYP2C11 in warfarin metabolism and efficacy, a CYP2C11-null rat model was used to study warfarin metabolism with both in vitro and in vivo approaches. Prothrombin time (PT) of warfarin was also determined. 3. The maximum rate of metabolism (Vmax) and intrinsic clearance (CLint) of liver microsomes from CYP2C11-null males were reduced by 37 and 64%, respectively, compared to those in Sprague Dawley (S-D) rats. The Km of liver microsomes from CYP2C11-null males was increased by 73% compared to that of S-D rats. The time to reach the maximum plasma concentration (Tmax) of warfarin in CYP2C11-null males was significantly delayed compared to that in S-D males, and the CL rate was also reduced. The PT of CYP2C11-null rats was moderately longer than that of S-D rats. 4. In conclusion, the clearance rate of warfarin was mildly decreased and its anticoagulant effect was moderately increased in male rats following CYP2C11 gene knockout. CYP2C11 played a certain role in the clearance and efficacy of warfarin, while it did not seem to be essential.
Collapse
Affiliation(s)
- Huanying Ye
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Danjuan Sui
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Wei Liu
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Yuannan Yuan
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University , Zhenjiang , PR China
| |
Collapse
|
19
|
Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, Pankowicz FP, Zhang QY, Ding X. P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab Dispos 2018; 46:1734-1744. [PMID: 30093418 DOI: 10.1124/dmd.118.083303] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023] Open
Abstract
Preclinical evaluation of drug candidates in experimental animal models is an essential step in drug development. Humanized mouse models have emerged as a promising alternative to traditional animal models. The purpose of this mini-review is to provide a brief survey of currently available mouse models for studying human xenobiotic metabolism. Here, we describe both genetic humanization and human liver chimeric mouse models, focusing on the advantages and limitations while outlining their key features and applications. Although this field of biomedical science is relatively young, these humanized mouse models have the potential to transform preclinical drug testing and eventually lead to a more cost-effective and rapid development of new therapies.
Collapse
Affiliation(s)
- Karl-Dimiter Bissig
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Weiguo Han
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Mercedes Barzi
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Nataliia Kovalchuk
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Liang Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xiaoyu Fan
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Francis P Pankowicz
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Qing-Yu Zhang
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xinxin Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| |
Collapse
|
20
|
Li J, Dawson PA. Animal models to study bile acid metabolism. Biochim Biophys Acta Mol Basis Dis 2018; 1865:895-911. [PMID: 29782919 DOI: 10.1016/j.bbadis.2018.05.011] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022]
Abstract
The use of animal models, particularly genetically modified mice, continues to play a critical role in studying the relationship between bile acid metabolism and human liver disease. Over the past 20 years, these studies have been instrumental in elucidating the major pathways responsible for bile acid biosynthesis and enterohepatic cycling, and the molecular mechanisms regulating those pathways. This work also revealed bile acid differences between species, particularly in the composition, physicochemical properties, and signaling potential of the bile acid pool. These species differences may limit the ability to translate findings regarding bile acid-related disease processes from mice to humans. In this review, we focus primarily on mouse models and also briefly discuss dietary or surgical models commonly used to study the basic mechanisms underlying bile acid metabolism. Important phenotypic species differences in bile acid metabolism between mice and humans are highlighted.
Collapse
Affiliation(s)
- Jianing Li
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States
| | - Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
21
|
MacLeod AK, Lin D, Huang JTJ, McLaughlin LA, Henderson CJ, Wolf CR. Identification of Novel Pathways of Osimertinib Disposition and Potential Implications for the Outcome of Lung Cancer Therapy. Clin Cancer Res 2018; 24:2138-2147. [PMID: 29437786 DOI: 10.1158/1078-0432.ccr-17-3555] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/29/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Osimertinib is a third-generation inhibitor of the epidermal growth factor receptor used in treatment of non-small cell lung cancer. A full understanding of its disposition and capacity for interaction with other medications will facilitate its effective use as a single agent and in combination therapy.Experimental Design: Recombinant cytochrome P450s and liver microsomal preparations were used to identify novel pathways of osimertinib metabolism in vitro A panel of knockout and mouse lines humanized for pathways of drug metabolism were used to establish the relevance of these pathways in vivoResults: Although some osimertinib metabolites were similar in mouse and human liver samples there were several significant differences, in particular a marked species difference in the P450s involved. The murine Cyp2d gene cluster played a predominant role in mouse, whereas CYP3A4 was the major human enzyme responsible for osimertinib metabolism. Induction of this enzyme in CYP3A4 humanized mice substantially decreased circulating osimertinib exposure. Importantly, we discovered a further novel pathway of osimertinib disposition involving CPY1A1. Modulation of CYP1A1/CYP1A2 levels markedly reduced parent drug concentrations, significantly altering metabolite pharmacokinetics (PK) in humanized mice in vivoConclusions: We demonstrate that a P450 enzyme expressed in smokers' lungs and lung tumors has the capacity to metabolise osimertinib. This could be a significant factor in defining the outcome of osimertinib treatment. This work also illustrates how P450-humanized mice can be used to identify and mitigate species differences in drug metabolism and thereby model the in vivo effect of critical metabolic pathways on anti-tumor response. Clin Cancer Res; 24(9); 2138-47. ©2018 AACR.
Collapse
Affiliation(s)
- A Kenneth MacLeod
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - De Lin
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Jeffrey T-J Huang
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Lesley A McLaughlin
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Colin J Henderson
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - C Roland Wolf
- Division of Cancer Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom.
| |
Collapse
|
22
|
Graves JP, Gruzdev A, Bradbury JA, DeGraff LM, Edin ML, Zeldin DC. Characterization of the Tissue Distribution of the Mouse Cyp2c Subfamily by Quantitative PCR Analysis. Drug Metab Dispos 2017; 45:807-816. [PMID: 28450579 PMCID: PMC5478903 DOI: 10.1124/dmd.117.075697] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 01/08/2023] Open
Abstract
The CYP2C subfamily of the cytochrome P450 gene superfamily encodes heme-thiolate proteins that have a myriad of biologic functions. CYP2C proteins detoxify xenobiotics and metabolize endogenous lipids such as arachidonic acid to bioactive eicosanoids. We report new methods and results for the quantitative polymerase reaction (qPCR) analysis for the 15 members of the mouse Cyp2c subfamily (Cyp2c29, Cyp2c37, Cyp2c38, Cyp2c39, Cyp2c40, Cyp2c44, Cyp2c50, Cyp2c54, Cyp2c55, Cyp2c65, Cyp2c66, Cyp2c67, Cyp2c68, Cyp2c69, and Cyp2c70). Commercially available TaqMan primer/probe assays were compared with developed SYBR Green primer sets for specificity toward the mouse Cyp2c cDNAs and analysis of their tissue distribution. TaqMan primer/probe assays for 10 of the mouse Cyp2c isoforms were shown to be specific for their intended mouse Cyp2c cDNA; however, there were no TaqMan primer/probe assays specific for the mouse Cyp2c29, Cyp2c40, Cyp2c67, Cyp2c68, or Cyp2c69 transcripts. Each of the SYBR Green primer sets was specific for its intended mouse Cyp2c cDNA. The two qPCR methods confirmed similar patterns of Cyp2c tissue expression: Cyp2c37, Cyp2c38, Cyp2c39, Cyp2c44, Cyp2c50, Cyp2c54, and Cyp2c70 were most highly expressed in liver; Cyp2c55 was highly expressed in large intestine; Cyp2c65 was highly expressed in stomach, duodenum, and large intestine; and Cyp2c66 was highly expressed in both duodenum and jejunum. For isoforms without specific TaqMan primer/probe assays, the SYBR Green primer sets detected high level expression of Cyp2c29, Cyp2c40, Cyp2c67, Cyp2c68, and Cyp2c69 in the liver. Lower expression levels of the mouse Cyp2cs were also detected in other tissues.
Collapse
Affiliation(s)
- Joan P Graves
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Artiom Gruzdev
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - J Alyce Bradbury
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Laura M DeGraff
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Matthew L Edin
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Darryl C Zeldin
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
23
|
Chen X, DuBois DC, Almon RR, Jusko WJ. Characterization and Interspecies Scaling of rhTNF- α Pharmacokinetics with Minimal Physiologically Based Pharmacokinetic Models. Drug Metab Dispos 2017; 45:798-806. [PMID: 28411279 DOI: 10.1124/dmd.116.074799] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/12/2017] [Indexed: 01/26/2023] Open
Abstract
Tumor necrosis factor-α (TNF-α) is a soluble cytokine and target of specific monoclonal antibodies (mAbs) and other biologic agents used in the treatment of inflammatory diseases. These biologics exert their pharmacological effects through binding and neutralizing TNF-α, and thus they prevent TNF-α from interacting with its cell surface receptors. The magnitude of the pharmacological effects is governed not only by the pharmacokinetics (PK) of mAbs, but also by the kinetic fate of TNF-α We have examined the pharmacokinetics of recombinant human TNF-α (rhTNF-α) in rats at low doses and quantitatively characterized its pharmacokinetic features with a minimal physiologically based pharmacokinetic model. Our experimental and literature-digitalized PK data of rhTNF-α in rats across a wide range of doses were applied to global model fitting. rhTNF-α exhibits permeability rate-limited tissue distribution and its elimination is comprised of a saturable clearance pathway mediated by tumor necrosis factor receptor binding and disposition and renal filtration. The resulting model integrated with classic allometry was further used for interspecies PK scaling and resulted in model predictions that agreed well with experimental measurements in monkeys. In addition, a semimechanistic model was proposed and applied to explore the absorption kinetics of rhTNF-α following s.c. and other routes of administration. The model suggests substantial presystemic degradation of rhTNF-α for s.c. and i.m. routes and considerable lymph uptake contributing to the overall systemic absorption through the stomach wall and gastrointestinal wall routes of dosing. This report provides comprehensive modeling and key insights into the complexities of absorption and disposition of a major cytokine.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (X.C., D.C.D, R.R.A, W.J.J.), and Department of Biological Sciences (D.C.D, R.R.A), State University of New York at Buffalo, Buffalo, New York
| | - Debra C DuBois
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (X.C., D.C.D, R.R.A, W.J.J.), and Department of Biological Sciences (D.C.D, R.R.A), State University of New York at Buffalo, Buffalo, New York
| | - Richard R Almon
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (X.C., D.C.D, R.R.A, W.J.J.), and Department of Biological Sciences (D.C.D, R.R.A), State University of New York at Buffalo, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (X.C., D.C.D, R.R.A, W.J.J.), and Department of Biological Sciences (D.C.D, R.R.A), State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
24
|
Jilek JL, Tian Y, Yu AM. Effects of MicroRNA-34a on the Pharmacokinetics of Cytochrome P450 Probe Drugs in Mice. Drug Metab Dispos 2017; 45:512-522. [PMID: 28254952 DOI: 10.1124/dmd.116.074344] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 03/01/2017] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs or miRs), including miR-34a, have been shown to regulate nuclear receptor, drug-metabolizing enzyme, and transporter gene expression in various cell model systems. However, to what degree miRNAs affect pharmacokinetics (PK) at the systemic level remains unknown. In addition, miR-34a replacement therapy represents a new cancer treatment strategy, although it is unknown whether miR-34a therapeutic agents could elicit any drug-drug interactions. To address this question, we refined a practical single-mouse PK approach and investigated the effects of a bioengineered miR-34a agent on the PK of several cytochrome P450 probe drugs (midazolam, dextromethorphan, phenacetin, diclofenac, and chlorzoxazone) administered as a cocktail. This approach involves manual serial blood microsampling from a single mouse and requires a sensitive liquid chromatography-tandem mass spectrometry assay, which was able to illustrate the sharp changes in midazolam PK by ketoconazole and pregnenolone 16α-carbonitrile as well as phenacetin PK by α-naphthoflavone and 3-methylcholanthrene. Surprisingly, 3-methylcholanthrene also decreased systemic exposure to midazolam, whereas both pregnenolone 16α-carbonitrile and 3-methylcholanthrene largely reduced the exposure to dextromethorphan, diclofenac, and chlorzoxazone. Finally, the biologic miR-34a agent had no significant effects on the PK of cocktail drugs but caused a marginal (45%-48%) increase in systemic exposure to midazolam, phenacetin, and dextromethorphan in mice. In vitro validation of these data suggested that miR-34a slightly attenuated intrinsic clearance of dextromethorphan. These findings from single-mouse PK and corresponding mouse liver microsome models suggest that miR-34a might have minor or no effects on the PK of coadministered cytochrome P450-metabolized drugs.
Collapse
Affiliation(s)
- Joseph L Jilek
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| | - Ye Tian
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California (J.L.J., Y.T., A.-M.Y.); and Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China (Y.T.)
| |
Collapse
|
25
|
Takahashi S, Fukami T, Masuo Y, Brocker CN, Xie C, Krausz KW, Wolf CR, Henderson CJ, Gonzalez FJ. Cyp2c70 is responsible for the species difference in bile acid metabolism between mice and humans. J Lipid Res 2016; 57:2130-2137. [PMID: 27638959 PMCID: PMC5321228 DOI: 10.1194/jlr.m071183] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/16/2016] [Indexed: 12/19/2022] Open
Abstract
Bile acids are synthesized from cholesterol in the liver and subjected to multiple metabolic biotransformations in hepatocytes, including oxidation by cytochromes P450 (CYPs) and conjugation with taurine, glycine, glucuronic acid, and sulfate. Mice and rats can hydroxylate chenodeoxycholic acid (CDCA) at the 6β-position to form α-muricholic acid (MCA) and ursodeoxycholic acid (UDCA) to form β-MCA. However, MCA is not formed in humans to any appreciable degree and the mechanism for this species difference is not known. Comparison of several Cyp-null mouse lines revealed that α-MCA and β-MCA were not detected in the liver samples from Cyp2c-cluster null (Cyp2c-null) mice. Global bile acid analysis further revealed the absence of MCAs and their conjugated derivatives, and high concentrations of CDCA and UDCA in Cyp2c-null mouse cecum and feces. Analysis of recombinant CYPs revealed that α-MCA and β-MCA were produced by oxidation of CDCA and UDCA by Cyp2c70, respectively. CYP2C9-humanized mice have similar bile acid metabolites as the Cyp2c-null mice, indicating that human CYP2C9 does not oxidize CDCA and UDCA, thus explaining the species differences in production of MCA. Because humans do not produce MCA, they lack tauro-β-MCA, a farnesoid X receptor antagonist in mouse that modulates obesity, insulin resistance, and hepatosteatosis.
Collapse
Affiliation(s)
- Shogo Takahashi
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tatsuki Fukami
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yusuke Masuo
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Chad N Brocker
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Cen Xie
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kristopher W Krausz
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - C Roland Wolf
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, United Kingdom
| | - Colin J Henderson
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, United Kingdom
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892.
| |
Collapse
|
26
|
Peters SA, Jones CR, Ungell AL, Hatley OJD. Predicting Drug Extraction in the Human Gut Wall: Assessing Contributions from Drug Metabolizing Enzymes and Transporter Proteins using Preclinical Models. Clin Pharmacokinet 2016; 55:673-96. [PMID: 26895020 PMCID: PMC4875961 DOI: 10.1007/s40262-015-0351-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Frankfurter Str. 250, F130/005, 64293, Darmstadt, Germany.
| | | | - Anna-Lena Ungell
- Investigative ADME, Non-Clinical Development, UCB New Medicines, BioPharma SPRL, Braine l'Alleud, Belgium
| | - Oliver J D Hatley
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
27
|
Wang X, Tang Y, Lu J, Shao Y, Qin X, Li Y, Wang L, Li D, Liu M. Characterization of novel cytochrome P450 2E1 knockout rat model generated by CRISPR/Cas9. Biochem Pharmacol 2016; 105:80-90. [DOI: 10.1016/j.bcp.2016.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
|
28
|
Scheer N, Kapelyukh Y, Rode A, Oswald S, Busch D, McLaughlin LA, Lin D, Henderson CJ, Wolf CR. Defining Human Pathways of Drug Metabolism In Vivo through the Development of a Multiple Humanized Mouse Model. Drug Metab Dispos 2015; 43:1679-90. [PMID: 26265742 DOI: 10.1124/dmd.115.065656] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 11/22/2022] Open
Abstract
Variability in drug pharmacokinetics is a major factor in defining drug efficacy and side effects. There remains an urgent need, particularly with the growing use of polypharmacy, to obtain more informative experimental data predicting clinical outcomes. Major species differences in multiplicity, substrate specificity, and regulation of enzymes from the cytochrome P450-dependent mono-oxygenase system play a critical role in drug metabolism. To develop an in vivo model for predicting human responses to drugs, we generated a mouse, where 31 P450 genes from the Cyp2c, Cyp2d, and Cyp3a gene families were exchanged for their relevant human counterparts. The model has been improved through additional humanization for the nuclear receptors constitutive androgen receptor and pregnane X receptor that control the expression of key drug metabolizing enzymes and transporters. In this most complex humanized mouse model reported to date, the cytochromes P450 function as predicted and we illustrate how these mice can be applied to predict drug-drug interactions in humans.
Collapse
Affiliation(s)
- Nico Scheer
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Yury Kapelyukh
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Anja Rode
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Stefan Oswald
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Diana Busch
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Lesley A McLaughlin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - De Lin
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - Colin J Henderson
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| | - C Roland Wolf
- Taconic Biosciences GmbH, Köln, Germany (N.S., A.R.); University Medicine of Greifswald, Center of Drug Absorption and Transport (C_DAT), Department of Clinical Pharmacology, Greifswald, Germany (S.O., D.B); and Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (Y.K., L.A.M., D.L., C.H., C.R.W)
| |
Collapse
|
29
|
Scheer N, Wilson ID. A comparison between genetically humanized and chimeric liver humanized mouse models for studies in drug metabolism and toxicity. Drug Discov Today 2015; 21:250-63. [PMID: 26360054 DOI: 10.1016/j.drudis.2015.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/07/2015] [Accepted: 09/01/2015] [Indexed: 12/12/2022]
Abstract
Mice that have been genetically humanized for proteins involved in drug metabolism and toxicity and mice engrafted with human hepatocytes are emerging and promising in vivo models for an improved prediction of the pharmacokinetic, drug-drug interaction and safety characteristics of compounds in humans. The specific advantages and disadvantages of these models should be carefully considered when using them for studies in drug discovery and development. Here, an overview on the corresponding genetically humanized and chimeric liver humanized mouse models described to date is provided and illustrated with examples of their utility in drug metabolism and toxicity studies. We compare the strength and weaknesses of the two different approaches, give guidance for the selection of the appropriate model for various applications and discuss future trends and perspectives.
Collapse
Affiliation(s)
| | - Ian D Wilson
- Imperial College London, South Kensington, London SW7 2AZ, UK.
| |
Collapse
|
30
|
Hough LB, Nalwalk JW, Ding X, Scheer N. Opioid Analgesia in P450 Gene Cluster Knockout Mice: A Search for Analgesia-Relevant Isoforms. Drug Metab Dispos 2015; 43:1326-30. [PMID: 26109562 DOI: 10.1124/dmd.115.065490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/24/2015] [Indexed: 01/10/2023] Open
Abstract
Cytochrome P450 monooxygenases (P450s), which are well-known drug-metabolizing enzymes, are thought to play a signal transduction role in µ opioid analgesia and may serve as high-affinity (3)H-cimetidine ((3)HCIM) binding sites in the brain. (3)HCIM binding sites may also be related to opioid or nonopioid analgesia. However, of the more than 100 murine P450 enzymes, the specific isoform(s) responsible for either function have not been identified. Presently, three lines of constitutive P450 gene cluster knockout (KO) mice with full-length deletions of 14 Cyp2c, 9 Cyp2d, and 7 Cyp3a genes were studied for deficiencies in (3)HCIM binding and for opioid analgesia. Liver and brain homogenates from all three genotypes showed normal (3)HCIM binding values, indicating that gene products of Cyp2d, Cyp3a, and Cyp2c are not (3)HCIM-binding proteins. Cyp2d KO and Cyp3a KO mice showed normal antinociceptive responses to a moderate systemic dose of morphine (20 mg/kg, s.c.), thereby excluding 16 P450 isoforms as mediators of opioid analgesia. In contrast, Cyp2c KO mice showed a 41% reduction in analgesic responses following systemically (s.c.) administered morphine. However, the significance of brain Cyp2c gene products in opioid analgesia is uncertain because little or no analgesic deficits were noted in Cyp2c KO mice following intracerebroventricular or intrathecalmorphine administration, respectively. These results show that the gene products of Cyp2d and Cyp3a do not contribute to µ opioid analgesia in the central nervous system. A possible role for Cyp2c gene products in opioid analgesia requires further consideration.
Collapse
Affiliation(s)
- Lindsay B Hough
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York (L.B.H., J.W.N.); College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York (X.D.); and Taconic Biosciences GmbH, Cologne, Germany (N.S.)
| | - Julia W Nalwalk
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York (L.B.H., J.W.N.); College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York (X.D.); and Taconic Biosciences GmbH, Cologne, Germany (N.S.)
| | - Xinxin Ding
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York (L.B.H., J.W.N.); College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York (X.D.); and Taconic Biosciences GmbH, Cologne, Germany (N.S.)
| | - Nico Scheer
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York (L.B.H., J.W.N.); College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York (X.D.); and Taconic Biosciences GmbH, Cologne, Germany (N.S.)
| |
Collapse
|
31
|
Choo EF, Woolsey S, DeMent K, Ly J, Messick K, Qin A, Takahashi R. Use of Transgenic Mouse Models to Understand the Oral Disposition and Drug-Drug Interaction Potential of Cobimetinib, a MEK Inhibitor. Drug Metab Dispos 2015; 43:864-9. [DOI: 10.1124/dmd.115.063743] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 03/26/2015] [Indexed: 12/13/2022] Open
|
32
|
Henderson CJ, McLaughlin LA, Scheer N, Stanley LA, Wolf CR. Cytochrome b5 is a major determinant of human cytochrome P450 CYP2D6 and CYP3A4 activity in vivo. Mol Pharmacol 2015; 87:733-9. [PMID: 25657337 DOI: 10.1124/mol.114.097394] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The cytochrome P450-dependent mono-oxygenase system is responsible for the metabolism and disposition of chemopreventive agents, chemical toxins and carcinogens, and >80% of therapeutic drugs. Cytochrome P450 (P450) activity is regulated transcriptionally and by the rate of electron transfer from P450 reductase. In vitro studies have demonstrated that cytochrome b5 (Cyb5) also modulates P450 function. We recently showed that hepatic deletion of Cyb5 in the mouse (HBN) markedly alters in vivo drug pharmacokinetics; a key outstanding question is whether Cyb5 modulates the activity of the major human P450s in drug disposition in vivo. To address this, we crossed mice humanized for CYP2D6 or CYP3A4 with mice carrying a hepatic Cyb5 deletion. In vitro triazolam 4-hydroxylation (probe reaction for CYP3A4) was reduced by >50% in hepatic microsomes from CYP3A4-HBN mice compared with controls. Similar reductions in debrisoquine 4-hydroxylation and metoprolol α-hydroxylation were observed using CYP2D6-HBN microsomes, indicating a significant role for Cyb5 in the activity of both enzymes. This effect was confirmed by the concentration-dependent restoration of CYP3A4-mediated triazolam turnover and CYP2D6-mediated bufuralol and debrisoquine turnover on addition of Escherichia coli membranes containing recombinant Cyb5. In vivo, the peak plasma concentration and area under the concentration time curve from 0 to 8 hours (AUC0-8 h) of triazolam were increased 4- and 5.7-fold, respectively, in CYP3A4-HBN mice. Similarly, the pharmacokinetics of bufuralol and debrisoquine were significantly altered in CYP2D6-HBN mice, the AUC0-8 h being increased ∼1.5-fold and clearance decreased by 40-60%. These data demonstrate that Cyb5 can be a major determinant of CYP3A4 and CYP2D6 activity in vivo, with a potential impact on the metabolism, efficacy, and side effects of numerous therapeutic drugs.
Collapse
Affiliation(s)
- Colin J Henderson
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (C.J.H., L.A.M., C.R.W.), TaconicArtemis, Cologne, Germany (N.S.); and Consultant in Investigative Toxicology, Linlithgow, United Kingdom (L.A.S.)
| | - Lesley A McLaughlin
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (C.J.H., L.A.M., C.R.W.), TaconicArtemis, Cologne, Germany (N.S.); and Consultant in Investigative Toxicology, Linlithgow, United Kingdom (L.A.S.)
| | - Nico Scheer
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (C.J.H., L.A.M., C.R.W.), TaconicArtemis, Cologne, Germany (N.S.); and Consultant in Investigative Toxicology, Linlithgow, United Kingdom (L.A.S.)
| | - Lesley A Stanley
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (C.J.H., L.A.M., C.R.W.), TaconicArtemis, Cologne, Germany (N.S.); and Consultant in Investigative Toxicology, Linlithgow, United Kingdom (L.A.S.)
| | - C Roland Wolf
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (C.J.H., L.A.M., C.R.W.), TaconicArtemis, Cologne, Germany (N.S.); and Consultant in Investigative Toxicology, Linlithgow, United Kingdom (L.A.S.)
| |
Collapse
|
33
|
Jaiswal S, Sharma A, Shukla M, Vaghasiya K, Rangaraj N, Lal J. Novel pre-clinical methodologies for pharmacokinetic drug-drug interaction studies: spotlight on "humanized" animal models. Drug Metab Rev 2014; 46:475-93. [PMID: 25270219 DOI: 10.3109/03602532.2014.967866] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Poly-therapy is common due to co-occurrence of several ailments in patients, leading to the elevated possibility of drug-drug interactions (DDI). Pharmacokinetic DDI often accounts for severe adverse drug reactions in patients resulting in withdrawal of drug from the market. Hence, the prediction of DDI is necessary at pre-clinical stage of drug development. Several human tissue and cell line-based in vitro systems are routinely used for screening metabolic and transporter pathways of investigational drugs and for predicting their clinical DDI potentials. However, ample constraints are associated with the in vitro systems and sometimes in vitro-in vivo extrapolation (IVIVE) fail to assess the risk of DDI in clinic. In vitro-in vivo correlation model in animals combined with human in vitro studies may be helpful in better prediction of clinical outcome. Native animal models vary remarkably from humans in drug metabolizing enzymes and transporters, hence, the interpretation of results from animal DDI studies is difficult. With the advent of modern molecular biology and engineering tools, novel pre-clinical animal models, namely, knockout rat/mouse, transgenic rat/mouse with humanized drug metabolizing enzymes and/or transporters and chimeric rat/mouse with humanized liver are developed. These models nearly simulate human-like drug metabolism and help to validate the in vivo relevance of the in vitro human DDI data. This review briefly discusses the application of such novel pre-clinical models for screening various type of DDI along with their advantages and limitations.
Collapse
Affiliation(s)
- Swati Jaiswal
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute , Lucknow , India
| | | | | | | | | | | |
Collapse
|
34
|
Mitsui T, Nemoto T, Miyake T, Nagao S, Ogawa K, Kato M, Ishigai M, Yamada H. A Useful Model Capable of Predicting the Clearance of Cytochrome P450 3A4 (CYP3A4) Substrates in Humans: Validity of CYP3A4 Transgenic Mice Lacking Their Own Cyp3a Enzymes. Drug Metab Dispos 2014; 42:1540-7. [DOI: 10.1124/dmd.114.057935] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
35
|
Scheer N, McLaughlin LA, Rode A, Macleod AK, Henderson CJ, Wolf CR. Deletion of 30 murine cytochrome p450 genes results in viable mice with compromised drug metabolism. Drug Metab Dispos 2014; 42:1022-30. [PMID: 24671958 DOI: 10.1124/dmd.114.057885] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In humans, 75% of all drugs are metabolized by the cytochrome P450-dependent monooxygenase system. Enzymes encoded by the CYP2C, CYP2D, and CYP3A gene clusters account for ∼80% of this activity. There are profound species differences in the multiplicity of cytochrome P450 enzymes, and the use of mouse models to predict pathways of drug metabolism is further complicated by overlapping substrate specificity between enzymes from different gene families. To establish the role of the hepatic and extrahepatic P450 system in drug and foreign chemical disposition, drug efficacy, and toxicity, we created a unique mouse model in which 30 cytochrome P450 genes from the Cyp2c, Cyp2d, and Cyp3a gene clusters have been deleted. Remarkably, despite a wide range of putative important endogenous functions, Cyp2c/2d/3a KO mice were viable and fertile, demonstrating that these genes have evolved primarily as detoxification enzymes. Although there was no overt phenotype, detailed examination showed Cyp2c/2d/3a KO mice had a smaller body size (15%) and larger livers (20%). Changes in hepatic morphology and a decreased blood glucose (30%) were also noted. A five-drug cocktail of cytochrome P450 isozyme probe substrates were used to evaluate changes in drug pharmacokinetics; marked changes were observed in either the pharmacokinetics or metabolites formed from Cyp2c, Cyp2d, and Cyp3a substrates, whereas the metabolism of the Cyp1a substrate caffeine was unchanged. Thus, Cyp2c/2d/3a KO mice provide a powerful model to study the in vivo role of the P450 system in drug metabolism and efficacy, as well as in chemical toxicity.
Collapse
Affiliation(s)
- Nico Scheer
- TaconicArtemis, Köln, Germany (N.S., A.R.); and Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom (L.A.M., A.K.M., C.J.H., C.R.W.)
| | | | | | | | | | | |
Collapse
|
36
|
Henderson CJ, McLaughlin LA, Finn RD, Ronseaux S, Kapelyukh Y, Wolf CR. A role for cytochrome b5 in the In vivo disposition of anticancer and cytochrome P450 probe drugs in mice. Drug Metab Dispos 2013; 42:70-7. [PMID: 24115751 DOI: 10.1124/dmd.113.055277] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The role of microsomal cytochrome b5 (Cyb5) in defining the rate of drug metabolism and disposition has been intensely debated for several decades. Recently we described mouse models involving the hepatic or global deletion of Cyb5, demonstrating its central role in in vivo drug disposition. We have now used the cytochrome b5 complete null (BCN) model to determine the role of Cyb5 in the metabolism of ten pharmaceuticals metabolized by a range of cytochrome P450s, including five anticancer drugs, in vivo and in vitro. The extent to which metabolism was significantly affected by the absence of Cyb5 was substrate-dependent; AUC increased (75-245%) and clearance decreased (35-72%) for phenacetin, metoprolol, and chlorzoxazone. Tolbutamide disposition was not significantly altered by Cyb5 deletion, while for midazolam clearance was decreased by 66%. The absence of Cyb5 had no effect on gefitinib and paclitaxel disposition, while significant changes in the in vivo pharmacokinetics were measured for: cyclophosphamide [maximum plasma concentration (Cmax) and terminal half-life increased 55% and 40%, respectively], tamoxifen (AUClast and Cmax increased 370% and 233%, respectively), and anastrozole (AUC and terminal half-life increased 125% and 62%, respectively; clearance down 80%). These data provide strong evidence that both hepatic and extrahepatic Cyb5 levels are an important determinant of in vivo drug disposition catalyzed by a range of cytochrome P450s, including currently prescribed anticancer agents, and that individuality in Cyb5 expression could be a significant determinant in rates of drug disposition in man.
Collapse
Affiliation(s)
- Colin J Henderson
- Division of Cancer Research, Medical Research Institute, University of Dundee, and Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | | | | | | | | | |
Collapse
|
37
|
Divanovic S, Dalli J, Jorge-Nebert LF, Flick LM, Gálvez-Peralta M, Boespflug ND, Stankiewicz TE, Fitzgerald JM, Somarathna M, Karp CL, Serhan CN, Nebert DW. Contributions of the three CYP1 monooxygenases to pro-inflammatory and inflammation-resolution lipid mediator pathways. THE JOURNAL OF IMMUNOLOGY 2013; 191:3347-57. [PMID: 23956430 DOI: 10.4049/jimmunol.1300699] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
All three cytochrome P450 1 (CYP1) monooxygenases are believed to participate in lipid mediator biosynthesis and/or their local inactivation; however, distinct metabolic steps are unknown. We used multiple-reaction monitoring and liquid chromatography-UV coupled with tandem mass spectrometry-based lipid-mediator metabololipidomics to identify and quantify three lipid-mediator metabolomes in basal peritoneal and zymosan-stimulated inflammatory exudates, comparing Cyp1a1/1a2/1b1(⁻/⁻) C57BL/6J-background triple-knockout mice with C57BL/6J wild-type mice. Significant differences between untreated triple-knockout and wild-type mice were not found for peritoneal cell number or type or for basal CYP1 activities involving 11 identified metabolic steps. Following zymosan-initiated inflammation, 18 lipid mediators were identified, including members of the eicosanoids and specialized proresolving mediators (i.e., resolvins and protectins). Compared with wild-type mice, Cyp1 triple-knockout mice exhibited increased neutrophil recruitment in zymosan-treated peritoneal exudates. Zymosan stimulation was associated with eight statistically significantly altered metabolic steps: increased arachidonic acid-derived leukotriene B₄ (LTB₄) and decreased 5S-hydroxyeicosatetraenoic acid; decreased docosahexaenoic acid-derived neuroprotectin D1/protectin D1, 17S-hydroxydocosahexaenoic acid, and 14S-hydroxydocosahexaenoic acid; and decreased eicosapentaenoic acid-derived 18R-hydroxyeicosapentaenoic acid (HEPE), 15S-HEPE, and 12S-HEPE. In neutrophils analyzed ex vivo, elevated LTB₄ levels were shown to parallel increased neutrophil numbers, and 20-hydroxy-LTB₄ formation was found to be deficient in Cyp1 triple-knockout mice. Together, these results demonstrate novel contributions of CYP1 enzymes to the local metabolite profile of lipid mediators that regulate neutrophilic inflammation.
Collapse
Affiliation(s)
- Senad Divanovic
- Division of Cellular and Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati OH 45229
| | - Jesmond Dalli
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Lucia F Jorge-Nebert
- Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati OH 45267-0056
| | - Leah M Flick
- Division of Cellular and Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati OH 45229
| | - Marina Gálvez-Peralta
- Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati OH 45267-0056
| | - Nicholas D Boespflug
- Division of Cellular and Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati OH 45229
| | - Traci E Stankiewicz
- Division of Cellular and Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati OH 45229
| | - Jonathan M Fitzgerald
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Maheshika Somarathna
- Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati OH 45267-0056
| | - Christopher L Karp
- Division of Cellular and Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati OH 45229
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Daniel W Nebert
- Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati OH 45267-0056
| |
Collapse
|
38
|
Scheer N, Snaith M, Wolf CR, Seibler J. Generation and utility of genetically humanized mouse models. Drug Discov Today 2013; 18:1200-11. [PMID: 23872278 DOI: 10.1016/j.drudis.2013.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/20/2013] [Accepted: 07/11/2013] [Indexed: 01/15/2023]
Abstract
Identifying in vivo models that are naturally predictive for particular areas of study in humans can be challenging due to the divergence that has occurred during speciation. One solution to this challenge that is gaining increasing traction is the use of genetic engineering to introduce human genes into mice to generate superior models for predicting human responses. This review describes the state-of-the-art for generating such models, provides an overview of the types of genetically humanized mouse models described to date and their applications in basic research, drug discovery and development and to understand clinical drug toxicity. We discuss limitations and explore promising future directions for the use of genetically humanized mice to further improve translational research.
Collapse
Affiliation(s)
- Nico Scheer
- TaconicArtemis, Neurather Ring 1, Koeln 51063, Germany.
| | | | | | | |
Collapse
|
39
|
Scheer N, Wolf CR. Genetically humanized mouse models of drug metabolizing enzymes and transporters and their applications. Xenobiotica 2013; 44:96-108. [DOI: 10.3109/00498254.2013.815831] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
40
|
Grimsley A, Gallagher R, Hutchison M, Pickup K, Wilson ID, Samuelsson K. Drug-drug interactions and metabolism in cytochrome P450 2C knockout mice: application to troleandomycin and midazolam. Biochem Pharmacol 2013; 86:529-38. [PMID: 23732297 DOI: 10.1016/j.bcp.2013.05.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 12/11/2022]
Abstract
Drug-drug interactions (DDIs) may cause serious drug toxicity and delay development of candidate drugs. Screening using human liver microsomes and hepatocytes can help predict DDIs but do not always provide the degree of certainty required for confident progression of a candidate drug. Thus a suitable in vivo test system could be of great value. Here a Cyp2c knockout (KO) mouse was investigated for studying DDIs using midazolam (MDZ) a standard human CYP3A4 substrate and troleandomycin (TAO) a potent human CYP3A4 inhibitor. Pharmacokinetics (PK) and biotransformation of MDZ were investigated following dosing to Cyp2c KO and wild type mice before and after TAO treatment. The noteworthy differences in the metabolism of MDZ in Cyp2c KO compared to wild type mice confirms the important role that Cyp2c enzymes play in the murine metabolism of MDZ in vivo. The impact of Cyp3a inhibition produced a further increase in circulating MDZ concentrations in all individuals from both strains of mice though the impact of the elimination of the Cyp2c pathway in the KO mice on the AUC was less than perhaps expected. We have shown that TAO produces an increase in the MDZ concentration and a reduction in the 1'hydroxymidazolam/midazolam formation ratio but the expected difference in the magnitude of this effect between the wild type and the Cyp2c KO mice was not seen. The magnitude of the TAO effect was also smaller than is reported in humans. Hence further work is required before this animal model could be used to predict clinical interactions.
Collapse
Affiliation(s)
- Aidan Grimsley
- Global DMPK, AstraZeneca UK Ltd., Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | | | | | | |
Collapse
|
41
|
Nebert DW, Wikvall K, Miller WL. Human cytochromes P450 in health and disease. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120431. [PMID: 23297354 DOI: 10.1098/rstb.2012.0431] [Citation(s) in RCA: 343] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There are 18 mammalian cytochrome P450 (CYP) families, which encode 57 genes in the human genome. CYP2, CYP3 and CYP4 families contain far more genes than the other 15 families; these three families are also the ones that are dramatically larger in rodent genomes. Most (if not all) genes in the CYP1, CYP2, CYP3 and CYP4 families encode enzymes involved in eicosanoid metabolism and are inducible by various environmental stimuli (i.e. diet, chemical inducers, drugs, pheromones, etc.), whereas the other 14 gene families often have only a single member, and are rarely if ever inducible or redundant. Although the CYP2 and CYP3 families can be regarded as largely redundant and promiscuous, mutations or other defects in one or more genes of the remaining 16 gene families are primarily the ones responsible for P450-specific diseases-confirming these genes are not superfluous or promiscuous but rather are more directly involved in critical life functions. P450-mediated diseases comprise those caused by: aberrant steroidogenesis; defects in fatty acid, cholesterol and bile acid pathways; vitamin D dysregulation and retinoid (as well as putative eicosanoid) dysregulation during fertilization, implantation, embryogenesis, foetogenesis and neonatal development.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental Health, Center for Environmental Genetics, University of Cincinnati Medical Center, Cincinnati, OH 45267-0056, USA.
| | | | | |
Collapse
|