1
|
Parvez MM, Thakur A, Mehrotra A, Stancil S, Pearce RE, Basit A, Leeder JS, Prasad B. Age-Dependent Abundance of CYP450 Enzymes Involved in Metronidazole Metabolism: Application to Pediatric PBPK Modeling. Clin Pharmacol Ther 2024; 116:1090-1099. [PMID: 38955794 DOI: 10.1002/cpt.3354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
The expression of cytochrome P450 (CYP) enzymes is highly variable and associated with factors, such as age, genotype, sex, and disease states. In this study, quantification of metronidazole metabolizing CYP isoforms (CYP2A6, CYP2E1, CYP3A4, CYP3A5, and CYP3A7) in human liver microsomes from 115 children and 35 adults was performed using a quantitative proteomics method. The data confirmed age-dependent increase in CYP2A6, CYP2E1, and CYP3A4 abundance, whereas, as expected, CYP3A7 abundance showed postnatal decrease with age. In particular, the fold difference (neonatal to adulthood levels) in the protein abundance of CYP2A6, CYP2E1, and CYP3A4 was 14, 11, and 20, respectively. In contrast, protein abundance of CYP3A7 was > 125-fold higher in the liver microsomes of neonates than of adults. The abundance of CYP2A6 and CYP3A5 was associated with genotypes, rs4803381 and rs776746, respectively. A proteomics-informed physiologically based pharmacokinetic (PBPK) model was developed to describe the pharmacokinetics of metronidazole and its primary metabolite, 2-hydroxymethylmetronidazole. The model revealed an increase in the metabolite-to-parent ratio with age and showed a strong correlation between CYP2A6 abundance and metabolite formation (r 2 = 0.75). Notably, the estimated contribution of CYP3A7 was ~ 75% in metronidazole clearance in neonates. These data suggest that variability in CYP2A6 and CYP3A7 in younger children poses the risk of variable pharmacokinetics of metronidazole and its active metabolite with a potential impact on drug efficacy and safety. No sex-dependent difference was observed in the protein abundance of the studied CYPs. The successful integration of hepatic CYP ontogeny data derived from a large liver bank into the pediatric PBPK model of metronidazole can be extended to other drugs metabolized by the studied CYPs.
Collapse
Affiliation(s)
- Md Masud Parvez
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Aarzoo Thakur
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Aanchal Mehrotra
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Stephani Stancil
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy-Kansas City, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Robin E Pearce
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy-Kansas City, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Abdul Basit
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy-Kansas City, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| |
Collapse
|
2
|
Sakamuru S, Ma D, Pierro JD, Baker NC, Kleinstreuer N, Cali JJ, Knudsen TB, Xia M. Development and validation of CYP26A1 inhibition assay for high-throughput screening. Biotechnol J 2024; 19:e2300659. [PMID: 38863121 PMCID: PMC11338008 DOI: 10.1002/biot.202300659] [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: 11/22/2023] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 06/13/2024]
Abstract
All-trans retinoic acid (atRA) is an endogenous ligand of the retinoic acid receptors, which heterodimerize with retinoid X receptors. AtRA is generated in tissues from vitamin A (retinol) metabolism to form a paracrine signal and is locally degraded by cytochrome P450 family 26 (CYP26) enzymes. The CYP26 family consists of three subtypes: A1, B1, and C1, which are differentially expressed during development. This study aims to develop and validate a high throughput screening assay to identify CYP26A1 inhibitors in a cell-free system using a luminescent P450-Glo assay technology. The assay performed well with a signal to background ratio of 25.7, a coefficient of variation of 8.9%, and a Z-factor of 0.7. To validate the assay, we tested a subset of 39 compounds that included known CYP26 inhibitors and retinoids, as well as positive and negative control compounds selected from the literature and/or the ToxCast/Tox21 portfolio. Known CYP26A1 inhibitors were confirmed, and predicted CYP26A1 inhibitors, such as chlorothalonil, prochloraz, and SSR126768, were identified, demonstrating the reliability and robustness of the assay. Given the general importance of atRA as a morphogenetic signal and the localized expression of Cyp26a1 in embryonic tissues, a validated CYP26A1 assay has important implications for evaluating the potential developmental toxicity of chemicals.
Collapse
Affiliation(s)
- Srilatha Sakamuru
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Dongping Ma
- Promega Corporation, Madison, Wisconsin, USA
| | - Jocylin D. Pierro
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | | | - Nicole Kleinstreuer
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | | | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
3
|
Czuba LC, Isoherranen N. LX-2 Stellate Cells Are a Model System for Investigating the Regulation of Hepatic Vitamin A Metabolism and Respond to Tumor Necrosis Factor α and Interleukin 1 β. Drug Metab Dispos 2024; 52:442-454. [PMID: 38485281 PMCID: PMC11023816 DOI: 10.1124/dmd.124.001679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/05/2024] [Indexed: 04/18/2024] Open
Abstract
Hepatic stellate cells (HSCs) are the major site of vitamin A (retinol) esterification and subsequent storage as retinyl esters within lipid droplets. However, retinyl esters become depleted in many pathophysiological states, including acute and chronic liver injuries. Recently, using a liver slice culture system as a model of acute liver injury and fibrogenesis, a time-dependent increase and decrease in the apparent formation of the bioactive retinoid all-trans-retinoic acid (atRA) and retinyl palmitate was measured, respectively. This coincided with temporal changes in the gene expression of retinoid-metabolizing enzymes and binding proteins, that preceded HSC activation. However, the underlying mechanisms that promote early changes in retinoid metabolism remain unresolved. We hypothesized that LX-2 cells could be applied to investigate differences in quiescent and activated HSC retinoid metabolism. We demonstrate that the hypermetabolic state of activated stellate cells relative to quiescent stellate cells may be attributed to induction of STRA6, RBP4, and CYP26A1, thereby reducing intracellular concentrations of atRA. We further hypothesized that paracrine and autocrine cytokine signaling regulates HSC vitamin A metabolism in both quiescent and activated cells. In quiescent cells, tumor necrosis factor α dose-dependently downregulated LRAT and CRBP1 mRNA, with EC50 values of 30-50 pg/mL. Likewise, interleukin-1β decreased LRAT and CRBP1 gene expression but with less potency. In activated stellate cells, multiple enzymes were downregulated, suggesting that the full effects of altered hepatic vitamin A metabolism in chronic conditions require both paracrine and autocrine signaling events. Further, this study suggests the potential for cell type-specific autocrine effects in hepatic retinoid signaling. SIGNIFICANCE STATEMENT: HSCs are the major site of vitamin A storage and important determinants of retinol metabolism during liver fibrogenesis. Here, two LX-2 culture methods were applied as models of hepatic retinoid metabolism to demonstrate the effects of activation status and dose-dependent cytokine exposure on the expression of genes involved in retinoid metabolism. This study suggests that compared to quiescent cells, activated HSCs are hypermetabolic and have reduced apparent formation of retinoic acid, which may alter downstream retinoic acid signaling.
Collapse
Affiliation(s)
- Lindsay C Czuba
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, Washington (L.C.C., N.I.) and Department of Pharmaceutical Sciences, University of Kentucky, College of Pharmacy, Lexington, Kentucky (L.C.C.)
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, Washington (L.C.C., N.I.) and Department of Pharmaceutical Sciences, University of Kentucky, College of Pharmacy, Lexington, Kentucky (L.C.C.)
| |
Collapse
|
4
|
Goel H, Printz RL, Pannala VR, AbdulHameed MDM, Wallqvist A. Probing Liver Injuries Induced by Thioacetamide in Human In Vitro Pooled Hepatocyte Experiments. Int J Mol Sci 2024; 25:3265. [PMID: 38542239 PMCID: PMC10970511 DOI: 10.3390/ijms25063265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 11/03/2024] Open
Abstract
Animal studies are typically utilized to understand the complex mechanisms associated with toxicant-induced hepatotoxicity. Among the alternative approaches to animal studies, in vitro pooled human hepatocytes have the potential to capture population variability. Here, we examined the effect of the hepatotoxicant thioacetamide on pooled human hepatocytes, divided into five lots, obtained from forty diverse donors. For 24 h, pooled human hepatocytes were exposed to vehicle, 1.33 mM (low dose), and 12 mM (high dose) thioacetamide, followed by RNA-seq analysis. We assessed gene expression variability using heat maps, correlation plots, and statistical variance. We used KEGG pathways and co-expression modules to identify underlying physiological processes/pathways. The co-expression module analysis showed that the majority of the lots exhibited activation for the bile duct proliferation module. Despite lot-to-lot variability, we identified a set of common differentially expressed genes across the lots with similarities in their response to amino acid, lipid, and carbohydrate metabolism. We also examined efflux transporters and found larger lot-to-lot variability in their expression patterns, indicating a potential for alteration in toxicant bioavailability within the cells, which could in turn affect the gene expression patterns between the lots. Overall, our analysis highlights the challenges in using pooled hepatocytes to understand mechanisms of toxicity.
Collapse
Affiliation(s)
- Himanshu Goel
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA; (V.R.P.); (M.D.M.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Richard L. Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Venkat R. Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA; (V.R.P.); (M.D.M.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Mohamed Diwan M. AbdulHameed
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA; (V.R.P.); (M.D.M.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA; (V.R.P.); (M.D.M.A.)
| |
Collapse
|
5
|
Cao DL, Ma LJ, Jiang BC, Gu Q, Gao YJ. Cytochrome P450 26A1 Contributes to the Maintenance of Neuropathic Pain. Neurosci Bull 2024; 40:293-309. [PMID: 37639183 PMCID: PMC10912416 DOI: 10.1007/s12264-023-01101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/02/2023] [Indexed: 08/29/2023] Open
Abstract
The cytochrome P450 proteins (CYP450s) have been implicated in catalyzing numerous important biological reactions and contribute to a variety of diseases. CYP26A1, a member of the CYP450 family, carries out the oxidative metabolism of retinoic acid (RA), the active metabolite of vitamin A. Here we report that CYP26A1 was dramatically upregulated in the spinal cord after spinal nerve ligation (SNL). CYP26A1 was mainly expressed in spinal neurons and astrocytes. HPLC analysis displayed that the content of all-trans-RA (at-RA), the substrate of CYP26A1, was reduced in the spinal cord on day 7 after SNL. Inhibition of CYP26A1 by siRNA or inhibition of CYP26A1-mediated at-RA catabolism by talarozole relieved the SNL-induced mechanical allodynia during the maintenance phase of neuropathic pain. Talarozole also reduced SNL-induced glial activation and proinflammatory cytokine production but increased anti-inflammatory cytokine (IL-10) production. The RA receptors RARα, RXRβ, and RXRγ were expressed in spinal neurons and glial cells. The promoter of Il-10 has several binding sites for RA receptors, and at-RA directly increased Il-10 mRNA expression in vitro. Finally, intrathecal IL-10 attenuated SNL-induced neuropathic pain and reduced the activation of astrocytes and microglia. Collectively, the inhibition of CYP26A1-mediated at-RA catabolism alleviates SNL-induced neuropathic pain by promoting the expression of IL-10 and suppressing glial activation. CYP26A1 may be a potential therapeutic target for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- De-Li Cao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
- Nantong University Medical School, Nantong, 226001, China
| | - Ling-Jie Ma
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Bao-Chun Jiang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China
| | - Qiang Gu
- Department of Pain Management, The Affiliated Hospital of Nantong University, Nantong, 226001, China.
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
| |
Collapse
|
6
|
Faris A, Ibrahim IM, Alnajjar R, Hadni H, Bhat MA, Yaseen M, Chakraborty S, Alsakhen N, Shamkh IM, Mabood F, M Naglah A, Ullah I, Ziedan N, Elhallaoui M. QSAR-driven screening uncovers and designs novel pyrimidine-4,6-diamine derivatives as potent JAK3 inhibitors. J Biomol Struct Dyn 2023:1-30. [PMID: 38059345 DOI: 10.1080/07391102.2023.2283168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
This study presents a robust and integrated methodology that harnesses a range of computational techniques to facilitate the design and prediction of new inhibitors targeting the JAK3/STAT pathway. This methodology encompasses several strategies, including QSAR analysis, pharmacophore modeling, ADMET prediction, covalent docking, molecular dynamics (MD) simulations, and the calculation of binding free energies (MM/GBSA). An efficacious QSAR model was meticulously crafted through the employment of multiple linear regression (MLR). The initial MLR model underwent further refinement employing an artificial neural network (ANN) methodology aimed at minimizing predictive errors. Notably, both MLR and ANN exhibited commendable performance, showcasing R2 values of 0.89 and 0.95, respectively. The model's precision was assessed via leave-one-out cross-validation (CV) yielding a Q2 value of 0.65, supplemented by rigorous Y-randomization. , The pharmacophore model effectively differentiated between active and inactive drugs, identifying potential JAK3 inhibitors, and demonstrated validity with an ROC value of 0.86. The newly discovered and designed inhibitors exhibited high inhibitory potency, ranging from 6 to 8, as accurately predicted by the QSAR models. Comparative analysis with FDA-approved Tofacitinib revealed that the new compounds exhibited promising ADMET properties and strong covalent docking (CovDock) interactions. The stability of the new discovered and designed inhibitors within the JAK3 binding site was confirmed through 500 ns MD simulations, while MM/GBSA calculations supported their binding affinity. Additionally, a retrosynthetic study was conducted to facilitate the synthesis of these potential JAK3/STAT inhibitors. The overall integrated approach demonstrates the feasibility of designing novel JAK3/STAT inhibitors with robust efficacy and excellent ADMET characteristics that surpass Tofacitinib by a significant margin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdelmoujoud Faris
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi, Benghazi, Libya
| | - Hanine Hadni
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Yaseen
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Souvik Chakraborty
- Department of Physiology, Bhairab Ganguly College, Belghoria, Kolkata, West Bengal, India
| | - Nada Alsakhen
- Department of Chemistry, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Israa M Shamkh
- Botany and Microbiology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Fazal Mabood
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Ahmed M Naglah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ihsan Ullah
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Noha Ziedan
- Department of Physical, Mathematical and Engineering Science, Faculty of Science, Business and Enterprise, University of Chester, Chester, UK
| | - Menana Elhallaoui
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
7
|
Glass SM, Tateishi Y, Guengerich FP, Wang HJ. 3,4-Desaturation of retinoic acid by cytochrome P450 27C1 prevents P450-mediated catabolism. Arch Biochem Biophys 2023; 743:109669. [PMID: 37356607 PMCID: PMC11500474 DOI: 10.1016/j.abb.2023.109669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/28/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
Cytochrome P450 (P450, CYP) 27C1 is expressed in human skin and catalyzes the 3,4-desaturation of retinoids. The enzyme has a relatively high specificity constant (kcat/Km), and ∼¼ of the retinoids in human skin are in the desaturated form but their function is unknown. 3,4-Dehydroretinoic acid (also didehydroretinoic acid, ddRA) has similar affinity as all-trans retinoic acid (atRA) for retinoid X and retinoic acid receptors (RXRs/RAR). The metabolism of ddRA is unknown, and we considered the hypothesis that desaturation might be a protective mechanism in maintaining active retinoid levels in the body. There are limited theoretical products that can result from ddRA oxidation. We optimized conditions for oxidation of atRA by human liver microsomes-a slow loss of atRA was seen due to 4-oxidation but no loss of ddRA was observed under the same conditions. We evaluated the HPLC peaks that were observed in microsomal incubations with ddRA using UV spectroscopy, NaBH4 and NaBD4 reduction, and mass spectrometry. None were potential ddRA oxidation products, and none were increased in the presence of the P450 cofactor NADPH. Known P450 inhibitors had no effects on the levels of these compounds. We conclude that ddRA is not readily oxidized by P450s and that one role of desaturation may be the maintenance of levels of functional retinoids.
Collapse
Affiliation(s)
- Sarah M Glass
- The Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - Yasuhiro Tateishi
- The Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - F Peter Guengerich
- The Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - Hong-Jaan Wang
- The Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
8
|
Ojiro R, Okano H, Ozawa S, Yamagata H, Zou X, Tang Q, Jin M, Sasaki K, Yoshida T, Yoshinari T, Shibutani M. Pharmacokinetics and 28-day repeated-dose toxicity of enniatin B after oral administration in mice. Food Chem Toxicol 2023; 177:113814. [PMID: 37179047 DOI: 10.1016/j.fct.2023.113814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Enniatins are emerging mycotoxins that contaminate foods. The present study investigated the oral pharmacokinetics and 28-day repeated-dose oral toxicity of enniatin B (ENNB) in CD1 (ICR) mice. In the pharmacokinetic study, male mice received a single oral or intravenous dose of ENNB [30 mg/kg body weight (BW) and 1 mg/kg BW, respectively]. After oral dosing, ENNB exhibited 139.9% bioavailability, a 5.1-hr elimination half-life, 5.26% fecal excretion from 4 to 24 h post-dose, and upregulation of Cyp7a1, Cyp2a12, Cyp2b10, and Cyp26a1 in the liver 2 h post-dosing. In the 28-day toxicity study, ENNB was administered to male and female mice by oral gavage at 0, 7.5, 15, and 30 mg/kg BW/day. Females (7.5 and 30 mg/kg) showed dose-unrelated decreased food consumption without accompanying changes in clinical parameters. Males (30 mg/kg) showed low red blood cell counts and high blood urea nitrogen levels and absolute kidney weights; however, other related parameters including the histopathology of systemic organs/tissues were unchanged. These results suggest that ENNB may not induce toxicity after 28 days of oral administration in mice, despite high absorption. The no-observed-adverse-effect level of ENNB after 28 days of repeated oral doses was 30 mg/kg BW/day for both sexes of mice.
Collapse
Affiliation(s)
- Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Hiromu Okano
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Hiroshi Yamagata
- Toxicology Division, Gotemba Laboratory, BoZo Research Center Inc., 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan.
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Qian Tang
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing, 400715, PR China.
| | - Kazuaki Sasaki
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Laboratory of Veterinary Pharmacology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Tomoya Yoshinari
- Division of Microbiology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-9501, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
9
|
Villéger R, Chulkina M, Mifflin RC, Powell DW, Pinchuk IV. Disruption of retinol-mediated IL-6 expression in colon cancer-associated fibroblasts: new perspectives on the role of vitamin A metabolism. Oncotarget 2023; 14:377-381. [PMID: 37185128 PMCID: PMC10132993 DOI: 10.18632/oncotarget.28399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Stromal myo-/fibroblasts (MFs) account for up to 30% of lamina propria cells in the normal human colon and their number is dramatically increased in colon cancer (CRC). Fibroblasts from cancers, also known as cancer-associated fibroblasts (CAFs), differ from normal colonic MF (N-MFs) and support tumor-promoting inflammation, in part due to increased IL-6 secretion. In this editorial, we highlight recent data obtained regarding IL-6 regulation in colorectal cancer CAFs through vitamin A (retinol) metabolism, discuss current limitations in our understanding of the mechanisms leading to the CAF pro-inflammatory phenotype, and discuss potential approaches to target CAF retinoid metabolism during CRC treatment.
Collapse
Affiliation(s)
- Romain Villéger
- Université de Poitiers, UMR CNRS 7267, Ecologie et Biologie des Interactions, France
| | - Marina Chulkina
- Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Randy C Mifflin
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UTMB, Galveston, TX 77555, USA
| | - Don W Powell
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UTMB, Galveston, TX 77555, USA
- Institute for Translational Sciences, UTMB, Galveston, TX 77555, USA
- Department of Neuroscience and Cell Biology, UTMB, Galveston, TX 77555, USA
| | - Irina V Pinchuk
- Department of Medicine at PennState Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| |
Collapse
|
10
|
Jeong H, Armstrong AT, Isoherranen N, Czuba L, Yang A, Zumpf K, Ciolino J, Torres E, Stika CS, Wisner KL. Temporal changes in the systemic concentrations of retinoids in pregnant and postpartum women. PLoS One 2023; 18:e0280424. [PMID: 36795769 PMCID: PMC9934425 DOI: 10.1371/journal.pone.0280424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/02/2023] [Indexed: 02/17/2023] Open
Abstract
Retinoids and vitamin A are essential for multiple biological functions, including vision and immune responses, as well as the development of an embryo during pregnancy. Despite its importance, alterations in retinoid homeostasis during normal human pregnancy are incompletely understood. We aimed to characterize the temporal changes in the systemic retinoid concentrations across pregnancy and postpartum period. Monthly blood samples were collected from twenty healthy pregnant women, and plasma concentrations of retinol, all-trans-retinoic acid (atRA), 13-cis-retinoic acid (13cisRA), and 4-oxo-retinoic acids were measured using liquid chromatography-tandem mass spectrometry. Significant decreases in 13cisRA concentrations over the pregnancy were observed, with rebound increases in retinol and 13cisRA levels after delivery. Of note, atRA concentrations exhibited a unique temporal pattern with levels peaking at mid-pregnancy. While the 4-oxo-atRA concentration was below the limit of quantification, 4-oxo-13cisRA was readily detectable, and its temporal change mimicked that of 13cisRA. The time profiles of atRA and 13cisRA remained similar after correction by albumin levels for plasma volume expansion adjustment. Together, the comprehensive profiling of systemic retinoid concentrations over the course of pregnancy provides insights into pregnancy-mediated changes in retinoid disposition to maintain its homeostasis.
Collapse
Affiliation(s)
- Hyunyoung Jeong
- Department of Industrial and Physical Pharmacy and Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, IN, United States of America
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, IN, United States of America
| | - Abigail T. Armstrong
- Department of Industrial and Physical Pharmacy and Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, IN, United States of America
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Lindsay Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States of America
| | - Amy Yang
- Department of Preventive Medicine (Biostatistics), Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Katelynn Zumpf
- Department of Preventive Medicine (Biostatistics), Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Jody Ciolino
- Department of Preventive Medicine (Biostatistics), Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Elizabeth Torres
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Catherine S. Stika
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Katherine L. Wisner
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| |
Collapse
|
11
|
Abstract
Vitamin A (retinol) is a critical micronutrient required for the control of stem cell functions, cell differentiation, and cell metabolism in many different cell types, both during embryogenesis and in the adult organism. However, we must obtain vitamin A from food sources. Thus, the uptake and metabolism of vitamin A by intestinal epithelial cells, the storage of vitamin A in the liver, and the metabolism of vitamin A in target cells to more biologically active metabolites, such as retinoic acid (RA) and 4-oxo-RA, must be precisely regulated. Here, I will discuss the enzymes that metabolize vitamin A to RA and the cytochrome P450 Cyp26 family of enzymes that further oxidize RA. Because much progress has been made in understanding the regulation of ALDH1a2 (RALDH2) actions in the intestine, one focus of this review is on the metabolism of vitamin A in intestinal epithelial cells and dendritic cells. Another focus is on recent data that 4-oxo-RA is a ligand required for the maintenance of hematopoietic stem cell dormancy and the important role of RARβ (RARB) in these stem cells. Despite this progress, many questions remain in this research area, which links vitamin A metabolism to nutrition, immune functions, developmental biology, and nuclear receptor pharmacology.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, and Revlon Pharmaceutical Professor of Pharmacology and Toxicology, Pharmacology Department, and the Meyer Cancer Center of Weill Cornell Medicine of Cornell University, 1300 York Ave, New York, NY 10065
| |
Collapse
|
12
|
Hanna C, Boily M, Jumarie C. Pesticides Inhibit Retinoic Acid Catabolism in PLHC-1 and ZFL Fish Hepatic Cell Lines. Chem Res Toxicol 2022; 35:1045-1058. [PMID: 35608517 PMCID: PMC9214766 DOI: 10.1021/acs.chemrestox.2c00050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The population of yellow perch (Perca flavescens) in lake Saint-Pierre (QC, Canada) has been dramatically declining since 1995 without any sign of recovery. Previous studies have shown disrupted retinoid (vitamin A) metabolic pathways in these fish, possibly due to the influence of pesticides. Our study aimed to evaluate the impact of some herbicides and neonicotinoids on retinoic acid catabolism in the fish hepatic cell lines PLHC-1 and ZFL. We hypothesized that pesticides accelerate the catabolism of retinoic acid through oxidative stress that exacerbates the oxidation of retinoic acid. Results obtained with talarozole, a specific CYP26A1 inhibitor, and ketoconazole, a generalist inhibitor of cytochrome-P450 enzymes, revealed that CYP26A1 is mainly responsible for retinoic acid catabolism in ZFL but not PLHC-1 cells. The impacts of pesticides on retinoic acid catabolism were evaluated by incubating the cells with all-trans-retinoic acid and two herbicides, atrazine and glyphosate, or three neonicotinoids, clothianidin, imidacloprid, and thiamethoxam. Intracellular thiols and lipid peroxidation were measured following pesticide exposure. The possible causal relation between oxidative stress and the perturbation of retinoic acid catabolism was investigated using the antioxidant N-acetylcysteine. The data revealed that pesticides inhibit retinoic acid catabolism, with the involvement of oxidative stress in the case of atrazine, imidacloprid, and thiamethoxam but not with clothianidin and glyphosate. Pesticides also affected the isomerization of all-trans-retinoic acid over time, leading to an increased proportion of active isomers. These results hint at a possible perturbation of retinoic acid catabolism in fish living in pesticide-contaminated waters, as suggested by several in vivo studies. Such a disruption of retinoid metabolism is worrying, given the numerous physiological pathways driven by retinoids.
Collapse
Affiliation(s)
- Charbel Hanna
- Département des sciences biologiques, Groupe TOXEN, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Quebec H3C 3P8, Canada
| | - Monique Boily
- Département des sciences biologiques, Groupe TOXEN, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Quebec H3C 3P8, Canada
| | - Catherine Jumarie
- Département des sciences biologiques, Groupe TOXEN, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Quebec H3C 3P8, Canada
| |
Collapse
|
13
|
Yabut KCB, Isoherranen N. CRABPs Alter all-trans-Retinoic Acid Metabolism by CYP26A1 via Protein-Protein Interactions. Nutrients 2022; 14:nu14091784. [PMID: 35565751 PMCID: PMC9105409 DOI: 10.3390/nu14091784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Cellular retinoic acid binding proteins (CRABP1 and CRABP2) bind all-trans-retinoic acid (atRA), the active metabolite of vitamin A, with high affinity. CRABP1 and CRABP2 have been shown to interact with the atRA-clearing cytochrome P450 enzymes CYP26B1 and CYP26C1 and with nuclear retinoic acid receptors (RARs). We hypothesized that CRABP1 and CRABP2 also alter atRA metabolism and clearance by CYP26A1, the third key atRA-metabolizing enzyme in the CYP26 family. Based on stopped-flow experiments, atRA bound CRABP1 and CRABP2 with Kd values of 4.7 nM and 7.6 nM, respectively. The unbound atRA Km values for 4-OH-atRA formation by CYP26A1 were 4.7 ± 0.8 nM with atRA, 6.8 ± 1.7 nM with holo-CRABP1 and 6.1 ± 2.7 nM with holo-CRABP2 as a substrate. In comparison, the apparent kcat value was about 30% lower (0.71 ± 0.07 min−1 for holo-CRABP1 and 0.75 ± 0.09 min−1 for holo-CRABP2) in the presence of CRABPs than with free atRA (1.07 ± 0.08 min−1). In addition, increasing concentrations in apo-CRABPs decreased the 4-OH-atRA formation rates by CYP26A1. Kinetic analyses suggest that apo-CRABP1 and apo-CRABP2 inhibit CYP26A1 (Ki = 0.39 nM and 0.53 nM, respectively) and holo-CRABPs channel atRA for metabolism by CYP26A1. These data suggest that CRABPs play a critical role in modulating atRA metabolism and cellular atRA concentrations.
Collapse
|
14
|
O’Connor C, Varshosaz P, Moise AR. Mechanisms of Feedback Regulation of Vitamin A Metabolism. Nutrients 2022; 14:nu14061312. [PMID: 35334970 PMCID: PMC8950952 DOI: 10.3390/nu14061312] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Vitamin A is an essential nutrient required throughout life. Through its various metabolites, vitamin A sustains fetal development, immunity, vision, and the maintenance, regulation, and repair of adult tissues. Abnormal tissue levels of the vitamin A metabolite, retinoic acid, can result in detrimental effects which can include congenital defects, immune deficiencies, proliferative defects, and toxicity. For this reason, intricate feedback mechanisms have evolved to allow tissues to generate appropriate levels of active retinoid metabolites despite variations in the level and format, or in the absorption and conversion efficiency of dietary vitamin A precursors. Here, we review basic mechanisms that govern vitamin A signaling and metabolism, and we focus on retinoic acid-controlled feedback mechanisms that contribute to vitamin A homeostasis. Several approaches to investigate mechanistic details of the vitamin A homeostatic regulation using genomic, gene editing, and chromatin capture technologies are also discussed.
Collapse
Affiliation(s)
- Catherine O’Connor
- MD Program, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada;
| | - Parisa Varshosaz
- Biology and Biomolecular Sciences Ph.D. Program, Northern Ontario School of Medicine, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- Department of Chemistry and Biochemistry, Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7253
| |
Collapse
|
15
|
Variability of CYP2C8 Polymorphisms in Three Jordanian Populations: Circassians, Chechens and Jordanian-Arabs. J Immigr Minor Health 2021; 24:1167-1176. [PMID: 34448113 DOI: 10.1007/s10903-021-01264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
CYP2C8 is a member of Cytochrome P450 enzymes system. It plays an important role in metabolizing a wide range of exogenous and endogenous compounds. CYP2C8 is involved in the metabolism of more than 100 drugs, typical substrates include: anticancer agents, antidiabetic agents, antimalarial agents, lipid lowering drugs and many others that constitute 20% of clinically prescribed drugs. Genetic variations of CYP2C8 have been reported with different frequencies in different populations. These genetic polymorphisms can lead to differences in the efficacy and safety of different types of medications metabolized by CYP2C8. The aim of this study was to investigate the allele frequencies of CYP2C8*3 (rs10509681 and rs11572080) and CYP2C8*4 (rs1058930) polymorphisms in three populations living in Jordan; Circassians and Chechens and Jordanian-Arabs and compare those frequencies with other populations. A total of 200 healthy Jordanians, 93 Circassians and 88 Chechens were included in this study. Genotyping of CYP2C8*3 and CYP2C8*4 polymorphisms was done by using polymerase chain reaction (PCR) followed by Restriction Fragment Length Polymorphism (RFLP). Using the Chi-square test, we found that the prevalence of CYP2C8*3 and *4 among the three populations were significantly different. Moreover, the mutant allele CYP2C8*3 (416A) was only detected in the Jordanian-Arab population with an allele frequency of 0.082, while the mutant allele CYP2C8*4 (792G) was detected with frequencies of 0.065, 0.122, 0.017 in Jordanian-Arabs, Circassians and Chechens, respectively. As our results show, CYP2C8*3 was undetectable in our Circassians and Chechens samples, on the other hand, Circassians had the highest allele frequency of CYP2C8*4 compared to Chechens and Jordanian-Arabs. These genetic variations of the gene encoding the CYP2C8 drug metabolizing enzymes can lead to clinical differences in drug metabolism and ultimately variations in drug effectiveness and toxicities. This study provides evidence for the importance of personalized medicine in these populations and can be the foundation for future clinical studies.
Collapse
|
16
|
Wang Y, Wang S, Che Y, Chen D, Liu Y, Shi Y. Exploring new targets for the treatment of hepatitis-B virus and hepatitis-B virus-associated hepatocellular carcinoma: A new perspective in bioinformatics. Medicine (Baltimore) 2021; 100:e26917. [PMID: 34414947 PMCID: PMC8376394 DOI: 10.1097/md.0000000000026917] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/24/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Hepatitis B Virus (HBV) infection is a global public health problem. After infection, patients experience a natural course from chronic hepatitis to cirrhosis and even Hepatitis B associated Hepatocellular Carcinoma (HBV-HCC). With the multi-omics research, many differentially expressed genes from chronic hepatitis to HCC stages have been discovered. All these provide important clues for new biomarkers and therapeutic targets. The purpose of this study is to explore the differential gene expression of HBV and HBV-related liver cancer, and analyze their enrichments and significance of related pathways. METHODS In this study, we downloaded four microarray datasets GSE121248, GSE67764, GSE55092, GSE55092 and GSE83148 from the Gene Expression Omnibus (GEO) database. Using these four datasets, patients with chronic hepatitis B (CHB) differentially expressed genes (CHB DEGs) and patients with HBV-related HCC differentially expressed genes (HBV-HCC DEGs) were identified. Then Protein-protein Interaction (PPI) network analysis, Gene Ontology (GO) functional analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed to excavate the functional interaction of these two groups of DEGs and the common DEGs. Finally, the Kaplan website was used to analyze the role of these genes in HCC prognostic. RESULTS A total of 241 CHB DEGs, 276 HBV-HCC DEGs, and 4 common DEGs (cytochrome P450 family 26 subfamily A member 1 (CYP26A1), family with sequence similarity 110 member C(FAM110C), SET and MYND domain containing 3(SMYD3) and zymogen granule protein 16(ZG16)) were identified. CYP26A1, FAM110C, SMYD3 and ZG16 exist in 4 models and interact with 33 genes in the PPI network of CHB and HBV-HCC DEGs,. GO function analysis showed that: CYP26A1, FAM110C, SMYD3, ZG16, and the 33 genes in their models mainly affect the regulation of synaptic vesicle transport, tangential migration from the subventricular zone to the olfactory bulb, cellular response to manganese ion, protein localization to mitochondrion, cellular response to dopamine, negative regulation of neuron death in the biological process of CHB. In the biological process of HBV-HCC, they mainly affect tryptophan catabolic process, ethanol oxidation, drug metabolic process, tryptophan catabolic process to kynurenine, xenobiotic metabolic process, retinoic acid metabolic process, steroid metabolic process, retinoid metabolic process, steroid catabolic process, retinal metabolic process, and rogen metabolic process. The analysis of the 4 common DEGs related to the prognosis of liver cancer showed that: CYP26A1, FAM110C, SMYD3 and ZG16 are closely related to the development of liver cancer and patient survival. Besides, further investigation of the research status of the four genes showed that CYP26A1 and SMYD3 could also affect HBV replication and the prognosis of liver cancer. CONCLUSION CYP26A1, FAM110C, SMYD3 and ZG16 are unique genes to differentiate HBV infection and HBV-related HCC, and expected to be novel targets for HBV-related HCC occurrence and prognostic judgement.
Collapse
|
17
|
Czuba LC, Wu X, Huang W, Hollingshead N, Roberto JB, Kenerson HL, Yeung RS, Crispe IN, Isoherranen N. Altered vitamin A metabolism in human liver slices corresponds to fibrogenesis. Clin Transl Sci 2021; 14:976-989. [PMID: 33382909 PMCID: PMC8212748 DOI: 10.1111/cts.12962] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
All-trans-retinoic acid (atRA), the active metabolite of vitamin A, has antifibrogenic properties in vitro and in animal models. Liver vitamin A homeostasis is maintained by cell-specific enzymatic activities including storage in hepatic stellate cells (HSCs), secretion into circulation from hepatocytes, and formation and clearance of atRA. During chronic liver injury, HSC activation is associated with a decrease in liver retinyl esters and retinol concentrations. atRA is synthesized through two enzymatic steps from retinol, but it is unknown if the loss of retinoid stores is associated with changes in atRA formation and which cell types contribute to the metabolic changes. The aim of this study was to determine if the vitamin A metabolic flux is perturbed in acute liver injury, and if changes in atRA concentrations are associated with HSC activation and collagen expression. At basal levels, HSC and Kupffer cells expressed key genes involved in vitamin A metabolism, whereas after acute liver injury, complex changes to the metabolic flux were observed in liver slices. These changes include a reproducible spike in atRA tissue concentrations, decreased retinyl ester and atRA formation rate, and time-dependent changes to the expression of metabolizing enzymes. Kinetic simulations suggested that oxidoreductases are important in determining retinoid metabolic flux after liver injury. These early changes precede HSC activation and upregulation of profibrogenic gene expression, which were inversely correlated with atRA tissue concentrations, suggesting that HSC and Kupffer cells are key cells involved in changes to vitamin A metabolic flux and signaling after liver injury. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? Vitamin A is metabolized in the liver for storage as retinyl esters in hepatic stellate cell (HSCs) or to all-trans-retinoic acid (atRA), an active metabolite with antifibrogenic properties. Following chronic liver injury, vitamin A metabolic flux is perturbed, and HSC activation leads to diminished retinoid stores. WHAT QUESTION DID THIS STUDY ADDRESS? Do changes in the expression of vitamin A metabolizing enzymes explain changes in atRA concentrations and the regulation of fibrosis following acute liver injury? WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? In healthy liver, both HSC and Kupffer cells may mediate vitamin A homeostasis. Following acute liver injury, complex changes in metabolizing enzyme expression/activity alter the metabolic flux of retinoids, resulting in a transient peak in atRA concentrations. The atRA concentrations are inversely correlated with profibrogenic gene expression, HSC activation, and collagen deposition. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? Improved understanding of altered vitamin A metabolic flux in acute liver injury may provide insight into cell-specific contributions to vitamin A loss and lead to novel interventions in liver fibrosis.
Collapse
Affiliation(s)
- Lindsay C. Czuba
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Xia Wu
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Weize Huang
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Nicole Hollingshead
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jessica B. Roberto
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | | | - Raymond S. Yeung
- Department of SurgeryUniversity of WashingtonSeattleWashingtonUSA
| | - Ian N. Crispe
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Nina Isoherranen
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
18
|
Kuhn MJ, Mavangira V, Sordillo LM. Invited review: Cytochrome P450 enzyme involvement in health and inflammatory-based diseases of dairy cattle. J Dairy Sci 2020; 104:1276-1290. [PMID: 33358163 DOI: 10.3168/jds.2020-18997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Dairy cattle are at the greatest risk of developing diseases around the time of calving because of compromised immune responses and the occurrence of oxidative stress. Both the development of compromised immunity and oxidative stress are influenced directly or indirectly by the metabolism of polyunsaturated fatty acids (PUFA) and fat-soluble vitamins. The cytochrome P450 (CYP450) family of enzymes is central to the metabolism of both classes of these compounds, but to date, the importance of CYP450 in the health of dairy cattle is underappreciated. As certain CYP450 isoforms metabolize both PUFA and fat-soluble vitamins, potential interactions may occur between PUFA and fat-soluble vitamins that are largely unexplored. For example, one CYP450 that generates anti-inflammatory oxylipids from arachidonic acid additionally contributes to the activation of vitamin D. Other potential substrate interactions between PUFA and vitamins A and E may exist as well. The intersection of PUFA and fat-soluble vitamin metabolism by CYP450 suggest that this enzyme system could provide an understanding of how immune function and oxidant status interconnect, resulting in increased postpartum disease occurrence. This review will detail the known contributions of bovine CYP450 to the regulation of oxylipids with a focus on enzymes that may also be involved in the metabolism of fat-soluble vitamins A, D, and E that contribute to antioxidant defenses. Although the activity of specific CYP450 is generally conserved among mammals, important differences exist in cattle, such as the isoforms primarily responsible for activation of vitamin D that makes their specific study in cattle of great importance. Additionally, a CYP450-driven inflammatory positive feedback loop is proposed, which may contribute to the dysfunctional inflammatory responses commonly found during the transition period. Establishing the individual enzyme isoform contributions to oxylipid biosynthesis and the regulation of vitamins A, D, and E may reveal how the CYP450 family of enzymes can affect inflammatory responses during times of increased susceptibility to disease. Determining the potential effect of each CYP450 on disease susceptibility or pathogenesis may allow for the targeted manipulation of the CYP450 pathways to influence specific immune responses and antioxidant defenses during times of increased risk for health disorders.
Collapse
Affiliation(s)
- M J Kuhn
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - V Mavangira
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824
| | - L M Sordillo
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing 48824.
| |
Collapse
|
19
|
Sharma S, Liu J, Zhang X, Sharma SS, Sorensen EJ, Bureik M. New luciferin-based probe substrates for human CYP26A1. Biochem Biophys Rep 2020; 24:100861. [PMID: 33294638 PMCID: PMC7695906 DOI: 10.1016/j.bbrep.2020.100861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 01/27/2023] Open
Abstract
Activity of human CYP26A1 towards six proluciferin probe substrates and their ester derivatives was monitored. These included three monofluorobenzyl ether isomers and three five-membered heterocycles. Overall, luciferin substrates with a free acid group gave higher activities than the ester compounds. Also, luciferin derivatives with six-ring structures were better metabolized than those with five-rings. The best substrates identified in this study are Luciferin 6′ 3-fluorobenzyl ether (Luciferin-3FBE) and its methyl ester (Luciferin-3FBEME). Taken together, we describe eleven new probe substrates for CYP26A1 and demonstrate for the first time that CYP26A1 does not only accept acid substrates but can also metabolize esters. Eleven new probe substrates for CYP26A1 were identified. CYP26A1 is shown to metabolize ester substrates. The best probe substrate identified is Luciferin 6′ 3-fluorobenzyl ether.
Collapse
Affiliation(s)
- Shishir Sharma
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Jingyao Liu
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Xue Zhang
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Sangeeta Shrestha Sharma
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
| | - Erik J. Sorensen
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Health Sciences Platform, Tianjin University, Tianjin, 300072, China
- Corresponding author.
| |
Collapse
|
20
|
Snyder JM, Zhong G, Hogarth C, Huang W, Topping T, LaFrance J, Palau L, Czuba LC, Griswold M, Ghiaur G, Isoherranen N. Knockout of Cyp26a1 and Cyp26b1 during postnatal life causes reduced lifespan, dermatitis, splenomegaly, and systemic inflammation in mice. FASEB J 2020; 34:15788-15804. [PMID: 33105029 DOI: 10.1096/fj.202001734r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
Abstract
All-trans-retinoic acid (atRA), the active metabolite of vitamin A, is an essential signaling molecule in all chordates. Global knockouts of the atRA clearing enzymes Cyp26a1 or Cyp26b1 are embryonic lethal. In adult rodents, inhibition of Cyp26a1 and Cyp26b1 increases atRA concentrations and signaling. However, postnatal knockout of Cyp26a1 does not cause a severe phenotype. We hypothesized that Cyp26b1 is the main atRA clearing Cyp in postnatal mammals. This hypothesis was tested by generating tamoxifen-inducible knockout mouse models of Cyp26b1 alone or with Cyp26a1. Both mouse models showed dermatitis, blepharitis, and splenomegaly. Histology showed infiltration of inflammatory cells including neutrophils and T lymphocytes into the skin and hyperkeratosis/hyperplasia of the nonglandular stomach. The mice lacking both Cyp26a1 and Cyp26b1 also had a reduced lifespan, failed to gain weight, and showed fat atrophy. There were significant changes in vitamin A homeostasis. Postnatal knockout of Cyp26b1 resulted in increased atRA concentrations in the skin while the postnatal knockout of both Cyp26a1 and Cyp26b1 resulted in increased atRA concentrations in the liver, serum, skin, spleen, and intestines. This study demonstrates the paramount role of Cyp26b1 in regulating retinoid homeostasis in postnatal life.
Collapse
Affiliation(s)
- Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Cathryn Hogarth
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA.,Department of Pharmacy and Biomedical Science, School of Molecular Science, La Trobe University, Wodonga, VIC, Australia
| | - Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Traci Topping
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| | - Jeffrey LaFrance
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Laura Palau
- School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Lindsay C Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Michael Griswold
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| | - Gabriel Ghiaur
- School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
21
|
Ambinder AJ, Norsworthy K, Hernandez D, Palau L, Paun B, Duffield A, Chandraratna R, Sanders M, Varadhan R, Jones RJ, Douglas Smith B, Ghiaur G. A Phase 1 Study of IRX195183, a RARα-Selective CYP26 Resistant Retinoid, in Patients With Relapsed or Refractory AML. Front Oncol 2020; 10:587062. [PMID: 33194741 PMCID: PMC7645224 DOI: 10.3389/fonc.2020.587062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Subsets of non-acute promyelocytic leukemia (APL) acute myelogenous leukemia (AML) exhibit aberrant retinoid signaling and demonstrate sensitivity to retinoids in vitro. We present the results of a phase 1 dose-escalation study that evaluated the safety, pharmacodynamics, and efficacy of IRX195183, a novel retinoic acid receptor α agonist, in patients with relapsed or refractory myelodysplastic syndrome (MDS) or AML. In this single center, single arm study, eleven patients with relapsed or refractory MDS/AML were enrolled and treated. Oral IRX195183 was administered at two dose levels: 50 mg daily or 75 mg daily for a total of two 28-day cycles. Patients with stable disease or better were allowed to continue on the drug for four additional 28-day cycles. Common adverse events included hypertriglyceridemia, fatigue, dyspnea, and edema. Three patients at the first dose level developed asymptomatic Grade 3 hypertriglyceridemia. The maximally tolerated dose was not reached. Four of the eleven patients had (36%) stable disease or better. One had a morphological complete remission with incomplete hematologic recovery while on the study drug. Two patients had evidence of in vivo leukemic blast maturation, as reflected by increased CD38 expression. In a pharmacodynamics study, plasma samples from four patients treated at the lowest dose level demonstrated the capacity to differentiate leukemic cells from the NB4 cell line in vitro. These results suggest that IRX195183 is safe, achieves biologically meaningful plasma concentrations and may be efficacious in a subset of patients with MDS/AML. Clinical Trial Registration: clinicaltrials.gov, identifier NCT02749708.
Collapse
Affiliation(s)
- Alexander J. Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kelly Norsworthy
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Hernandez
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura Palau
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bogdan Paun
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amy Duffield
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | | | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Richard J. Jones
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - B. Douglas Smith
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gabriel Ghiaur
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Bilip M, Shah S, Mathiyalakan M, Tagalakis AD, Hart SL, Maeshima R, Eaton S, Orford M, Irving E, Di Florio A, Simons C, Stoker AW. Liposomal delivery of hydrophobic RAMBAs provides good bioavailability and significant enhancement of retinoic acid signalling in neuroblastoma tumour cells. J Drug Target 2020; 28:643-654. [PMID: 31903789 PMCID: PMC7609071 DOI: 10.1080/1061186x.2019.1710157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/29/2019] [Accepted: 12/25/2019] [Indexed: 01/04/2023]
Abstract
Retinoid treatment is employed during residual disease treatment in neuroblastoma, where the aim is to induce neural differentiation or death in tumour cells. However, although therapeutically effective, retinoids have only modest benefits and suffer from poor pharmacokinetic properties. In vivo, retinoids induce CYP26 enzyme production in the liver, enhancing their own rapid metabolic clearance, while retinoid resistance in tumour cells themselves is considered to be due in part to increased CYP26 production. Retinoic acid metabolism blocking agents (RAMBAs), which inhibit CYP26 enzymes, can improve retinoic acid (RA) pharmacokinetics in pre-clinical neuroblastoma models. Here, we demonstrate that in cultured neuroblastoma tumour cells, RAMBAs enhance RA action as seen by morphological differentiation, AKT signalling and suppression of MYCN protein. Although active as retinoid enhancers, these RAMBAs are highly hydrophobic and their effective delivery in humans will be very challenging. Here, we demonstrate that such RAMBAs can be loaded efficiently into cationic liposomal particles, where the RAMBAs achieve good bioavailability and activity in cultured tumour cells. This demonstrates the efficacy of RAMBAs in enhancing retinoid signalling in neuroblastoma cells and shows for the first time that liposomal delivery of hydrophobic RAMBAs is a viable approach, providing novel opportunities for their delivery and application in humans.
Collapse
Affiliation(s)
- Maja Bilip
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | - Shreya Shah
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | | | | | - Stephen L. Hart
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | - Ruhina Maeshima
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | - Michael Orford
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | - Elsa Irving
- Great Ormond Street Institute of Child Health, UCL, London, UK
| | | | - Claire Simons
- Great Ormond Street Institute of Child Health, UCL, London, UK
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
23
|
Czuba LC, Zhong G, Yabut KC, Isoherranen N. Analysis of vitamin A and retinoids in biological matrices. Methods Enzymol 2020; 637:309-340. [PMID: 32359651 DOI: 10.1016/bs.mie.2020.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vitamin A signaling pathways are predominantly driven by the cellular concentrations of all-trans-retinoic acid (atRA), as the main mechanism of retinoid signaling is via activation of retinoic acid receptors. atRA concentrations are in turn controlled by the storage of vitamin A and enzymatic processes that synthesize and clear atRA. This has resulted in the need for robust and highly specific analytical methods to accurately quantify retinoids in diverse biological matrices. Tissue-specific differences in both the quantity of retinoids and background matrix interferences can confound the quantification of retinoids, and the bioanalysis requires high performance instrumentation, such as liquid chromatography mass-spectrometry (LC-MS). Successful bioanalysis of retinoids is further complicated by the innate structural instability of retinoids and their relatively high lipophilicity. Further, in vitro experiments with retinoids require attention to experimental design and interpretation to account for the instability of retinoids due to isomerization and degradation, sequential metabolism to numerous structurally similar metabolites, and substrate depletion during experiments. In addition, in vitro biological activity is often confounded by residual presence of retinoids in common biological reagents such as cell culture media. This chapter identifies common biological and analytical complexities in retinoid bioanalysis in diverse biological matrices, and in the use of retinoids in cell culture and metabolic incubations. In addition, this chapter highlights best practices for the successful detection and quantification of the vitamin A metabolome in a wide range of biological matrices.
Collapse
Affiliation(s)
- Lindsay C Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - King C Yabut
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States.
| |
Collapse
|
24
|
Roberts C. Regulating Retinoic Acid Availability during Development and Regeneration: The Role of the CYP26 Enzymes. J Dev Biol 2020; 8:jdb8010006. [PMID: 32151018 PMCID: PMC7151129 DOI: 10.3390/jdb8010006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022] Open
Abstract
This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Cyp26 enzymes are necessary to generate RA gradients and to protect specific tissues from RA signalling. Disruption of RA homeostasis leads to a wide variety of embryonic defects affecting many tissues. Here, the function of CYP26 enzymes is discussed in the context of the RA signalling pathway, enzymatic structure and biochemistry, human genetic disease, and function in development and regeneration as elucidated from animal model studies.
Collapse
Affiliation(s)
- Catherine Roberts
- Developmental Biology of Birth Defects, UCL-GOS Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK;
- Institute of Medical and Biomedical Education St George’s, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
| |
Collapse
|
25
|
Isoherranen N, Zhong G. Biochemical and physiological importance of the CYP26 retinoic acid hydroxylases. Pharmacol Ther 2019; 204:107400. [PMID: 31419517 PMCID: PMC6881548 DOI: 10.1016/j.pharmthera.2019.107400] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022]
Abstract
The Cytochrome P450 (CYP) family 26 enzymes contribute to retinoic acid (RA) metabolism and homeostasis in humans, mammals and other chordates. The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it's 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. While no crystal structures of CYP26 enzymes are available, the binding of various ligands has been extensively explored via homology modeling. All three CYP26 enzymes are inducible by treatment with atRA in various prenatal and postnatal tissues and cell types. However, current literature shows that in addition to regulation by atRA, CYP26 enzyme expression is also regulated by other endogenous processes and inflammatory cytokines. In humans and in animal models the expression patterns of CYP26 enzymes have been shown to be tissue and cell type specific, and the expression of the CYP26 enzymes is believed to regulate the formation of critical atRA concentration gradients in various tissue types. Yet, very little data exists on direct disease associations of altered CYP26 expression or activity. Nevertheless, data is emerging describing a variety of human genetic variations in the CYP26 enzymes that are associated with different pathologies. Interestingly, some of these genetic variants result in increased activity of the CYP26 enzymes potentially leading to complex gene-environment interactions due to variability in dietary intake of retinoids. This review highlights the current knowledge of structure-function of CYP26 enzymes and focuses on their role in human retinoid metabolism in different tissues.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA.
| | - Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Tuteja S, Qu L, Vujkovic M, Dunbar RL, Chen J, DerOhannessian S, Rader DJ. Genetic Variants Associated With Plasma Lipids Are Associated With the Lipid Response to Niacin. J Am Heart Assoc 2019; 7:e03488. [PMID: 30371334 PMCID: PMC6404865 DOI: 10.1161/jaha.117.008461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Background Niacin is a broad-spectrum lipid-modulating drug, but its mechanism of action is unclear. Genome-wide association studies have identified multiple loci associated with blood lipid levels and lipoprotein (a). It is unknown whether these loci modulate response to niacin. Methods and Results Using data from the AIM - HIGH (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL /High Triglycerides and Impact on Global Health Outcomes) trial (n=2054 genotyped participants), we determined whether genetic variations at validated loci were associated with a differential change in plasma lipids and lipoprotein (a) 1 year after randomization to either statin+placebo or statin+niacin in a variant-treatment interaction model. Nominally significant interactions ( P<0.05) were found for genetic variants in MVK , LIPC , PABPC 4, AMPD 3 with change in high-density lipoprotein cholesterol; SPTLC 3 with change in low-density lipoprotein cholesterol; TOM 1 with change in total cholesterol; PDXDC 1 and CYP 26A1 with change in triglycerides; and none for lipoprotein (a). We also investigated whether these loci were associated with cardiovascular events. The risk of coronary disease related death was higher in the minor allele carriers at the LIPC locus in the placebo group (odds ratio 2.08, 95% confidence interval 1.11-3.90, P=0.02) but not observed in the niacin group (odds ratio 0.89, 95% confidence interval 0.48-1.65, P=0.7); P-interaction =0.02. There was a greater risk for acute coronary syndrome (odds ratio 1.85, 95% confidence interval 1.16-2.77, P=0.02) and revascularization events (odds ratio 1.64, 95% confidence interval 1.2-2.22, P=0.002) in major allele carriers at the CYP 26A1 locus in the placebo group not seen in the niacin group. Conclusions Genetic variation at loci previously associated with steady-state lipid levels displays evidence for lipid response to niacin treatment. Clinical Trials Registration URL: https://www.clinicaltrials.gov . Unique identifier: NCT00120289.
Collapse
Affiliation(s)
- Sony Tuteja
- 1 Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Liming Qu
- 1 Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Marijana Vujkovic
- 2 Department of Biostatistics and Epidemiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Richard L Dunbar
- 1 Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,4 Cardiometabolic and Lipid Clinic Corporal Michael J. Crescenz VA Medical Center Philadelphia PA.,5 ICON plc North Wales PA
| | - Jinbo Chen
- 2 Department of Biostatistics and Epidemiology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Stephanie DerOhannessian
- 1 Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| | - Daniel J Rader
- 1 Department of Medicine Perelman School of Medicine at the University of Pennsylvania Philadelphia PA.,3 Department of Genetics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA
| |
Collapse
|
27
|
Zhong G, Hogarth C, Snyder JM, Palau L, Topping T, Huang W, Czuba LC, LaFrance J, Ghiaur G, Isoherranen N. The retinoic acid hydroxylase Cyp26a1 has minor effects on postnatal vitamin A homeostasis, but is required for exogenous atRA clearance. J Biol Chem 2019; 294:11166-11179. [PMID: 31167781 DOI: 10.1074/jbc.ra119.009023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/01/2019] [Indexed: 12/20/2022] Open
Abstract
The all-trans-retinoic acid (atRA) hydroxylase Cyp26a1 is essential for embryonic development and may play a key role in regulating atRA clearance also in adults. We hypothesized that loss of Cyp26a1 activity via inducible knockout in juvenile or adult mice would result in decreased atRA clearance and increased tissue atRA concentrations and atRA-related adverse effects. To test these hypotheses, Cyp26a1 was knocked out in juvenile and adult male and female Cyp26a1 floxed mice using standard Cre-Lox technology and tamoxifen injections. Biochemical and histological methods were used to study the effects of global Cyp26a1 knockout. The Cyp26a1 knockout did not result in consistent histopathological changes in any major organs. Cyp26a1 -/- mice gained weight normally and exhibited no adverse phenotypes for up to 1 year after loss of Cyp26a1 expression. Similarly, atRA concentrations were not increased in the liver, testes, spleen, or serum of these mice, and the Cyp26a1 knockout did not cause compensatory induction of lecithin:retinol acetyltransferase (Lrat) or retinol dehydrogenase 11 (Rdh11) mRNA or a decrease in aldehyde dehydrogenase 1a1 (Aldh1a1) mRNA in the liver compared with tamoxifen-treated controls. However, the Cyp26a1 -/- mice showed increased bone marrow cellularity and decreased frequency of erythroid progenitor cells in the bone marrow consistent with a retinoid-induced myeloid skewing of hematopoiesis. In addition, the Cyp26a1 knockout decreased clearance of exogenous atRA by 70% and increased atRA half-life 6-fold. These findings demonstrate that despite lacking a major impact on endogenous atRA signaling, Cyp26a1 critically contributes as a barrier for exogenous atRA exposure.
Collapse
Affiliation(s)
- Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Cathryn Hogarth
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington 98195
| | - Laura Palau
- School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21231
| | - Traci Topping
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Lindsay C Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Jeffrey LaFrance
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Gabriel Ghiaur
- School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21231
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| |
Collapse
|
28
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
29
|
Grindheim JM, Nicetto D, Donahue G, Zaret KS. Polycomb Repressive Complex 2 Proteins EZH1 and EZH2 Regulate Timing of Postnatal Hepatocyte Maturation and Fibrosis by Repressing Genes With Euchromatic Promoters in Mice. Gastroenterology 2019; 156:1834-1848. [PMID: 30689973 PMCID: PMC6599454 DOI: 10.1053/j.gastro.2019.01.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/02/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Little is known about mechanisms that underlie postnatal hepatocyte maturation and fibrosis at the chromatin level. We investigated the transcription of genes involved in maturation and fibrosis in postnatal hepatocytes of mice, focusing on the chromatin compaction the roles of the Polycomb repressive complex 2 histone methyltransferases EZH1 and EZH2. METHODS Hepatocytes were isolated from mixed background C57BL/6J-C3H mice, as well as mice with liver-specific disruption of Ezh1 and/or Ezh2, at postnatal day 14 and 2 months after birth. Liver tissues were collected and analyzed by RNA sequencing, H3K27me3 chromatin immunoprecipitation sequencing, and sonication-resistant heterochromatin sequencing (a method to map heterochromatin and euchromatin). Liver damage was characterized by histologic analysis. RESULTS We found more than 3000 genes differentially expressed in hepatocytes during liver maturation from postnatal day 14 to month 2 after birth. Disruption of Ezh1 and Ezh2 in livers caused perinatal hepatocytes to differentiate prematurely and to express genes at postnatal day 14 that would normally be induced by month 2 and differentiate prematurely. Loss of Ezh1 and Ezh2 also resulted in liver fibrosis. Genes with H3K27me3-postive and H3K4me3-positive euchromatic promoters were prematurely induced in hepatocytes with loss of Ezh1 and Ezh2-these genes included those that regulate hepatocyte maturation, fibrosis, and genes not specifically associated with the liver lineage. CONCLUSIONS The Polycomb repressive complex 2 proteins EZH1 and EZH2 regulate genes that control hepatocyte maturation and fibrogenesis and genes not specifically associated with the liver lineage by acting at euchromatic promoter regions. EZH1 and EZH2 thereby promote liver homeostasis and prevent liver damage. Strategies to manipulate Polycomb proteins might be used to improve hepatocyte derivation protocols or developed for treatment of patients with liver fibrosis.
Collapse
Affiliation(s)
- Jessica Mae Grindheim
- Institute for Regenerative Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Penn Epigenetics Institute, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Dept. Cell and Developmental Biology, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Dept. of Cancer Biology, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Perelman School of Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA
| | - Dario Nicetto
- Institute for Regenerative Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Penn Epigenetics Institute, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Dept. Cell and Developmental Biology, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA
| | - Greg Donahue
- Institute for Regenerative Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Penn Epigenetics Institute, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Dept. Cell and Developmental Biology, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA.,Perelman School of Medicine, University of Pennsylvania, Smilow Center for Translational Research, 3400 Civic Center Blvd, Bldg. 421, Philadelphia, PA 19104-5157, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, University of Pennsylvania, Smilow Center for Translational Research, Philadelphia, Pennsylvania; Penn Epigenetics Institute, University of Pennsylvania, Smilow Center for Translational Research, Philadelphia, Pennsylvania; Department of Cell and Developmental Biology, University of Pennsylvania, Smilow Center for Translational Research, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Smilow Center for Translational Research, Philadelphia, Pennsylvania.
| |
Collapse
|
30
|
Scaling in vitro activity of CYP3A7 suggests human fetal livers do not clear retinoic acid entering from maternal circulation. Sci Rep 2019; 9:4620. [PMID: 30874620 PMCID: PMC6420499 DOI: 10.1038/s41598-019-40995-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/21/2019] [Indexed: 01/09/2023] Open
Abstract
All-trans-retinoic acid (atRA), the active metabolite of vitamin A, is a critical signaling molecule during embryonic and fetal development and is necessary for maternal health. Fetal exposure to endogenous atRA is tightly regulated during gestation in a tissue specific manner and maternal exposure to exogenous retinoids during pregnancy is teratogenic. The clearance of atRA is primarily mediated by the cytochrome P450 (CYP) 26 enzymes, which play an essential role in controlling retinoid gradients during organogenesis. We hypothesized that CYP26 enzymes in the human fetal liver also function as a protective barrier to prevent maternal atRA reaching fetal circulation. Using human fetal liver tissue, we found that the mRNA of CYP26A1 and CYP26B1 enzymes is expressed in the human fetal liver. However, based on inhibition studies, metabolite profiles and correlation of atRA metabolism with testosterone hydroxylation, clearance of atRA in the fetal livers was mediated by CYP3A7. Based on in vitro-to-in vivo scaling, atRA clearance in the fetal liver was quantitatively minimal, thus providing an insufficient maternal-fetal barrier for atRA exposure.
Collapse
|
31
|
Ghaffari H, Varner JD, Petzold LR. Analysis of the role of thrombomodulin in all-trans retinoic acid treatment of coagulation disorders in cancer patients. Theor Biol Med Model 2019; 16:3. [PMID: 30764845 PMCID: PMC6376718 DOI: 10.1186/s12976-019-0099-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clinical studies have shown that all-trans retinoic acid (RA), which is often used in treatment of cancer patients, improves hemostatic parameters and bleeding complications such as disseminated intravascular coagulation (DIC). However, the mechanisms underlying this improvement have yet to be elucidated. In vitro studies have reported that RA upregulates thrombomodulin (TM) expression on the endothelial cell surface. The objective of this study was to investigate how and to what extent the TM concentration changes after RA treatment in cancer patients, and how this variation influences the blood coagulation cascade. RESULTS In this study, we introduced an ordinary differential equation (ODE) model of gene expression for the RA-induced upregulation of TM concentration. Coupling the gene expression model with a two-compartment pharmacokinetic model of RA, we obtained the time-dependent changes in TM and thrombomodulin-mRNA (TMR) concentrations following oral administration of RA. Our results indicated that the TM concentration reached its peak level almost 14 h after taking a single oral dose (110 [Formula: see text]) of RA. Continuous treatment with RA resulted in oscillatory expression of TM on the endothelial cell surface. We then coupled the gene expression model with a mechanistic model of the coagulation cascade, and showed that the elevated levels of TM over the course of RA therapy with a single daily oral dose (110 [Formula: see text]) of RA, reduced the peak thrombin levels and endogenous thrombin potential (ETP) up to 50 and 49%, respectively. We showed that progressive reductions in plasma levels of RA, observed in continuous RA therapy with a once-daily oral dose (110 [Formula: see text]) of RA, did not affect TM-mediated reduction of thrombin generation significantly. This finding prompts the hypothesis that continuous RA treatment has more consistent therapeutic effects on coagulation disorders than on cancer. CONCLUSIONS Our results indicate that the oscillatory upregulation of TM expression on the endothelial cells over the course of RA therapy could potentially contribute to the treatment of coagulation abnormalities in cancer patients. Further studies on the impacts of RA therapy on the procoagulant activity of cancer cells are needed to better elucidate the mechanisms by which RA therapy improves hemostatic abnormalities in cancer.
Collapse
Affiliation(s)
- Hamed Ghaffari
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.
| | - Jeffrey D Varner
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Linda R Petzold
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| |
Collapse
|
32
|
Zhong G, Ortiz D, Zelter A, Nath A, Isoherranen N. CYP26C1 Is a Hydroxylase of Multiple Active Retinoids and Interacts with Cellular Retinoic Acid Binding Proteins. Mol Pharmacol 2018; 93:489-503. [PMID: 29476041 DOI: 10.1124/mol.117.111039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/22/2018] [Indexed: 01/10/2023] Open
Abstract
The clearance of retinoic acid (RA) and its metabolites is believed to be regulated by the CYP26 enzymes, but the specific roles of CYP26A1, CYP26B1, and CYP26C1 in clearing active vitamin A metabolites have not been defined. The goal of this study was to establish the substrate specificity of CYP26C1, and determine whether CYP26C1 interacts with cellular retinoic acid binding proteins (CRABPs). CYP26C1 was found to effectively metabolize all-trans retinoic acid (atRA), 9-cis-retinoic acid (9-cis-RA), 13-cis-retinoic acid, and 4-oxo-atRA with the highest intrinsic clearance toward 9-cis-RA. In comparison with CYP26A1 and CYP26B1, CYP26C1 resulted in a different metabolite profile for retinoids, suggesting differences in the active-site structure of CYP26C1 compared with other CYP26s. Homology modeling of CYP26C1 suggested that this is attributable to the distinct binding orientation of retinoids within the CYP26C1 active site. In comparison with other CYP26 family members, CYP26C1 was up to 10-fold more efficient in clearing 4-oxo-atRA (intrinsic clearance 153 μl/min/pmol) than CYP26A1 and CYP26B1, suggesting that CYP26C1 may be important in clearing this active retinoid. In support of this, CRABPs delivered 4-oxo-atRA and atRA for metabolism by CYP26C1. Despite the tight binding of 4-oxo-atRA and atRA with CRABPs, the apparent Michaelis-Menten constant in biological matrix (Km) value of these substrates with CYP26C1 was not increased when the substrates were bound with CRABPs, in contrast to what is predicted by free drug hypothesis. Together these findings suggest that CYP26C1 is a 4-oxo-atRA hydroxylase and may be important in regulating the concentrations of this active retinoid in human tissues.
Collapse
Affiliation(s)
- Guo Zhong
- Departments of Pharmaceutics (G.Z., N.I.) and Medicinal Chemistry (D.O., A.N.), School of Pharmacy, and Department of Biochemistry, School of Medicine (A.Z.), University of Washington, Seattle, Washington
| | - David Ortiz
- Departments of Pharmaceutics (G.Z., N.I.) and Medicinal Chemistry (D.O., A.N.), School of Pharmacy, and Department of Biochemistry, School of Medicine (A.Z.), University of Washington, Seattle, Washington
| | - Alex Zelter
- Departments of Pharmaceutics (G.Z., N.I.) and Medicinal Chemistry (D.O., A.N.), School of Pharmacy, and Department of Biochemistry, School of Medicine (A.Z.), University of Washington, Seattle, Washington
| | - Abhinav Nath
- Departments of Pharmaceutics (G.Z., N.I.) and Medicinal Chemistry (D.O., A.N.), School of Pharmacy, and Department of Biochemistry, School of Medicine (A.Z.), University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Departments of Pharmaceutics (G.Z., N.I.) and Medicinal Chemistry (D.O., A.N.), School of Pharmacy, and Department of Biochemistry, School of Medicine (A.Z.), University of Washington, Seattle, Washington
| |
Collapse
|
33
|
Dubey A, Rose RE, Jones DR, Saint-Jeannet JP. Generating retinoic acid gradients by local degradation during craniofacial development: One cell's cue is another cell's poison. Genesis 2018; 56:10.1002/dvg.23091. [PMID: 29330906 PMCID: PMC5818312 DOI: 10.1002/dvg.23091] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/02/2023]
Abstract
Retinoic acid (RA) is a vital morphogen for early patterning and organogenesis in the developing embryo. RA is a diffusible, lipophilic molecule that signals via nuclear RA receptor heterodimeric units that regulate gene expression by interacting with RA response elements in promoters of a significant number of genes. For precise RA signaling, a robust gradient of the morphogen is required. The developing embryo contains regions that produce RA, and specific intracellular concentrations of RA are created through local degradation mediated by Cyp26 enzymes. In order to elucidate the mechanisms by which RA executes precise developmental programs, the kinetics of RA metabolism must be clearly understood. Recent advances in techniques for endogenous RA detection and quantification have paved the way for mechanistic studies to shed light on downstream gene expression regulation coordinated by RA. It is increasingly coming to light that RA signaling operates not only at precise concentrations but also employs mechanisms of degradation and feedback inhibition to self-regulate its levels. A global gradient of RA throughout the embryo is often found concurrently with several local gradients, created by juxtaposed domains of RA synthesis and degradation. The existence of such local gradients has been found especially critical for the proper development of craniofacial structures that arise from the neural crest and the cranial placode populations. In this review, we summarize the current understanding of how local gradients of RA are established in the embryo and their impact on craniofacial development.
Collapse
Affiliation(s)
- Aditi Dubey
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry
| | - Rebecca E. Rose
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | - Drew R. Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | | |
Collapse
|
34
|
Jing J, Nelson C, Paik J, Shirasaka Y, Amory JK, Isoherranen N. Physiologically Based Pharmacokinetic Model of All- trans-Retinoic Acid with Application to Cancer Populations and Drug Interactions. J Pharmacol Exp Ther 2017; 361:246-258. [PMID: 28275201 PMCID: PMC5399637 DOI: 10.1124/jpet.117.240523] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/06/2017] [Indexed: 11/22/2022] Open
Abstract
All-trans retinoic acid (atRA) is a front-line treatment of acute promyelocytic leukemia (APL). Due to its activity in regulating the cell cycle, it has also been evaluated for the treatment of other cancers. However, the efficacy of atRA has been limited by atRA inducing its own metabolism during therapy, resulting in a decrease of atRA exposure during continuous dosing. Frequent relapse occurs in patients receiving atRA monotherapy. In an attempt to combat therapy resistance, inhibitors of atRA metabolism have been developed. Of these, ketoconazole and liarozole have shown some benefits, but their usage is limited by side effects and low potency toward the cytochrome P450 26A1 isoform (CYP26A1), the main atRA hydroxylase. We determined the pharmacokinetic basis of therapy resistance to atRA and tested whether the complex disposition kinetics of atRA could be predicted in healthy subjects and in cancer patients in the presence and absence of inhibitors of atRA metabolism using physiologically based pharmacokinetic (PBPK) modeling. A PBPK model of atRA disposition was developed and verified in healthy individuals and in cancer patients. The population-based PBPK model of atRA disposition incorporated saturable metabolic clearance of atRA, induction of CYP26A1 by atRA, and the absorption and distribution kinetics of atRA. It accurately predicted the changes in atRA exposure after continuous dosing and when coadministered with ketoconazole and liarozole. The developed model will be useful in interpretation of atRA disposition and efficacy, design of novel dosing strategies, and development of next-generation atRA metabolism inhibitors.
Collapse
Affiliation(s)
- Jing Jing
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| | - Cara Nelson
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| | - Jisun Paik
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| | - Yoshiyuki Shirasaka
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| | - John K Amory
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics (J.J., C.N., Y.S., N.I.), Department of Medicine (J.A.), and Department of Comparative Medicine (J.P.), University of Washington, Seattle, Washington
| |
Collapse
|
35
|
Stevison F, Hogarth C, Tripathy S, Kent T, Isoherranen N. Inhibition of the all-trans Retinoic Acid ( atRA) Hydroxylases CYP26A1 and CYP26B1 Results in Dynamic, Tissue-Specific Changes in Endogenous atRA Signaling. Drug Metab Dispos 2017; 45:846-854. [PMID: 28446509 DOI: 10.1124/dmd.117.075341] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022] Open
Abstract
All-trans retinoic acid (atRA), the active metabolite of vitamin A, is a ligand for several nuclear receptors and acts as a critical regulator of many physiologic processes. The cytochrome P450 family 26 (CYP26) enzymes are responsible for atRA clearance, and are potential drug targets to increase concentrations of endogenous atRA in a tissue-specific manner. Talarozole is a potent inhibitor of CYP26A1 and CYP26B1, and has shown some success in clinical trials. However, it is not known what magnitude of change is needed in tissue atRA concentrations to promote atRA signaling changes. The aim of this study was to quantify the increase in endogenous atRA concentrations necessary to alter atRA signaling in target organs, and to establish the relationship between CYP26 inhibition and altered atRA concentrations in tissues. Following a single 2.5-mg/kg dose of talarozole to mice, atRA concentrations increased up to 5.7-, 2.7-, and 2.5-fold in serum, liver, and testis, respectively, resulting in induction of Cyp26a1 in the liver and testis and Rar β and Pgc 1β in liver. The increase in atRA concentrations was well predicted from talarozole pharmacokinetics and in vitro data of CYP26 inhibition. After multiple doses of talarozole, a significant increase in atRA concentrations was observed in serum but not in liver or testis. This lack of increase in atRA concentrations correlated with an increase in CYP26A1 expression in the liver. The increased atRA concentrations in serum without a change in liver suggest that CYP26B1 in extrahepatic sites plays a key role in regulating systemic atRA exposure.
Collapse
Affiliation(s)
- Faith Stevison
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (F.S., S.T., N.I.); and School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington (C.H., T.K.)
| | - Cathryn Hogarth
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (F.S., S.T., N.I.); and School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington (C.H., T.K.)
| | - Sasmita Tripathy
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (F.S., S.T., N.I.); and School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington (C.H., T.K.)
| | - Travis Kent
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (F.S., S.T., N.I.); and School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington (C.H., T.K.)
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (F.S., S.T., N.I.); and School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington (C.H., T.K.)
| |
Collapse
|
36
|
Nilsson O, Isoherranen N, Guo MH, Lui JC, Jee YH, Guttmann-Bauman I, Acerini C, Lee W, Allikmets R, Yanovski JA, Dauber A, Baron J. Accelerated Skeletal Maturation in Disorders of Retinoic Acid Metabolism: A Case Report and Focused Review of the Literature. Horm Metab Res 2016; 48:737-744. [PMID: 27589347 PMCID: PMC5534175 DOI: 10.1055/s-0042-114038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nutritional excess of vitamin A, a precursor for retinoic acid (RA), causes premature epiphyseal fusion, craniosynostosis, and light-dependent retinopathy. Similarly, homozygous loss-of-function mutations in CYP26B1, one of the major RA-metabolizing enzymes, cause advanced bone age, premature epiphyseal fusion, and craniosynostosis. In this paper, a patient with markedly accelerated skeletal and dental development, retinal scarring, and autism-spectrum disease is presented and the role of retinoic acid in longitudinal bone growth and skeletal maturation is reviewed. Genetic studies were carried out using SNP array and exome sequencing. RA isomers were measured in the patient, family members, and in 18 age-matched healthy children using high-performance liquid chromatography coupled to tandem mass spectrometry. A genomic SNP array identified a novel 8.3 megabase microdeletion on chromosome 10q23.2-23.33. The 79 deleted genes included CYP26A1 and C1, both major RA-metabolizing enzymes. Exome sequencing did not detect any variants that were predicted to be deleterious in the remaining alleles of these genes or other known retinoic acid-metabolizing enzymes. The patient exhibited elevated plasma total RA (16.5 vs. 12.6±1.5 nM, mean±SD, subject vs. controls) and 13-cisRA (10.7 nM vs. 6.1±1.1). The findings support the hypothesis that elevated RA concentrations accelerate bone and dental maturation in humans. CYP26A1 and C1 haploinsufficiency may contribute to the elevated retinoic acid concentrations and clinical findings of the patient, although this phenotype has not been reported in other patients with similar deletions, suggesting that other unknown genetic or environmental factors may also contribute.
Collapse
Affiliation(s)
- Ola Nilsson
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Center for Molecular Medicine and Pediatric Endocrinology Unit, Department of Women’s and Children’s Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Nina Isoherranen
- Department of Pharmaceutics School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Michael H. Guo
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Julian C. Lui
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Youn Hee Jee
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Ines Guttmann-Bauman
- Harold Schnitzer Diabetes Health Center, Oregon Health and Science University, Portland, OR, USA
| | - Carlo Acerini
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Winston Lee
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Rando Allikmets
- Department of Ophthalmology, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jack A. Yanovski
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Dauber
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffrey Baron
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of Cytochrome P450 2C8 in Drug Metabolism and Interactions. Pharmacol Rev 2016; 68:168-241. [PMID: 26721703 DOI: 10.1124/pr.115.011411] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During the last 10-15 years, cytochrome P450 (CYP) 2C8 has emerged as an important drug-metabolizing enzyme. CYP2C8 is highly expressed in human liver and is known to metabolize more than 100 drugs. CYP2C8 substrate drugs include amodiaquine, cerivastatin, dasabuvir, enzalutamide, imatinib, loperamide, montelukast, paclitaxel, pioglitazone, repaglinide, and rosiglitazone, and the number is increasing. Similarly, many drugs have been identified as CYP2C8 inhibitors or inducers. In vivo, already a small dose of gemfibrozil, i.e., 10% of its therapeutic dose, is a strong, irreversible inhibitor of CYP2C8. Interestingly, recent findings indicate that the acyl-β-glucuronides of gemfibrozil and clopidogrel cause metabolism-dependent inactivation of CYP2C8, leading to a strong potential for drug interactions. Also several other glucuronide metabolites interact with CYP2C8 as substrates or inhibitors, suggesting that an interplay between CYP2C8 and glucuronides is common. Lack of fully selective and safe probe substrates, inhibitors, and inducers challenges execution and interpretation of drug-drug interaction studies in humans. Apart from drug-drug interactions, some CYP2C8 genetic variants are associated with altered CYP2C8 activity and exhibit significant interethnic frequency differences. Herein, we review the current knowledge on substrates, inhibitors, inducers, and pharmacogenetics of CYP2C8, as well as its role in clinically relevant drug interactions. In addition, implications for selection of CYP2C8 marker and perpetrator drugs to investigate CYP2C8-mediated drug metabolism and interactions in preclinical and clinical studies are discussed.
Collapse
Affiliation(s)
- Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki (J.T.B., A.M.F., M.N., P.J.N.), and Helsinki University Hospital, Helsinki, Finland (J.T.B., M.N., P.J.N.)
| |
Collapse
|
38
|
Nelson CH, Peng CC, Lutz JD, Yeung CK, Zelter A, Isoherranen N. Direct protein-protein interactions and substrate channeling between cellular retinoic acid binding proteins and CYP26B1. FEBS Lett 2016; 590:2527-35. [PMID: 27416800 DOI: 10.1002/1873-3468.12303] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/23/2016] [Accepted: 06/29/2016] [Indexed: 11/11/2022]
Abstract
Cellular retinoic acid binding proteins (CRABPs) bind all-trans-retinoic acid (atRA) tightly. This study aimed to determine whether atRA is channeled directly to cytochrome P450 (CYP) CYP26B1 by CRABPs, and whether CRABPs interact directly with CYP26B1. atRA bound to CRABPs (holo-CRABP) was efficiently metabolized by CYP26B1. Isotope dilution experiments showed that delivery of atRA to CYP26B1 in solution was similar with or without CRABP. Holo-CRABPs had higher affinity for CYP26B1 than free atRA, but both apo-CRABPs inhibited the formation of 4-OH-RA by CYP26B1. Similar protein-protein interactions between soluble binding proteins and CYPs may be important for other lipophilic CYP substrates.
Collapse
Affiliation(s)
- Cara H Nelson
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Chi-Chi Peng
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Justin D Lutz
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Catherine K Yeung
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Alex Zelter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| |
Collapse
|
39
|
Foti RS, Diaz P, Douguet D. Comparison of the ligand binding site of CYP2C8 with CYP26A1 and CYP26B1: a structural basis for the identification of new inhibitors of the retinoic acid hydroxylases. J Enzyme Inhib Med Chem 2016; 31:148-161. [PMID: 27424662 DOI: 10.1080/14756366.2016.1193734] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The CYP26s are responsible for metabolizing retinoic acid and play an important role in maintaining homeostatic levels of retinoic acid. Given the ability of CYP2C8 to metabolize retinoic acid, we evaluated the potential for CYP2C8 inhibitors to also inhibit CYP26. In vitro assays were used to evaluate the inhibition potencies of CYP2C8 inhibitors against CYP26A1 and CYP26B1. Using tazarotenic acid as a substrate for CYP26, IC50 values for 17 inhibitors of CYP2C8 were determined for CYP26A1 and CYP26B1, ranging from ∼20 nM to 100 μM, with a positive correlation observed between IC50s for CYP2C8 and CYP26A1. An evaluation of IC50's versus in vivo Cmax values suggests that inhibitors such as clotrimazole or fluconazole may interact with CYP26 at clinically relevant concentrations and may alter levels of retinoic acid. These findings provide insight into drug interactions resulting in elevated retinoic acid concentrations and expand upon the pharmacophore of CYP26 inhibition.
Collapse
Affiliation(s)
- Robert S Foti
- a Amgen Pharmacokinetics and Drug Metabolism , Cambridge , MA , USA
| | - Philippe Diaz
- b Department of Biomedical and Pharmaceutical Sciences , Core Laboratory for Neuromolecular Production, University of Montana , Missoula , MT , USA.,c Dermaxon , Missoula , MT , USA , and
| | - Dominique Douguet
- d CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Nice Sophia Antipolis , Valbonne , France
| |
Collapse
|
40
|
Centritto F, Paroni G, Bolis M, Garattini SK, Kurosaki M, Barzago MM, Zanetti A, Fisher JN, Scott MF, Pattini L, Lupi M, Ubezio P, Piccotti F, Zambelli A, Rizzo P, Gianni' M, Fratelli M, Terao M, Garattini E. Cellular and molecular determinants of all-trans retinoic acid sensitivity in breast cancer: Luminal phenotype and RARα expression. EMBO Mol Med 2016; 7:950-72. [PMID: 25888236 PMCID: PMC4520659 DOI: 10.15252/emmm.201404670] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Forty-two cell lines recapitulating mammary carcinoma heterogeneity were profiled for all-trans retinoic acid (ATRA) sensitivity. Luminal and ER+ (estrogen-receptor-positive) cell lines are generally sensitive to ATRA, while refractoriness/low sensitivity is associated with a Basal phenotype and HER2 positivity. Indeed, only 2 Basal cell lines (MDA-MB157 and HCC-1599) are highly sensitive to the retinoid. Sensitivity of HCC-1599 cells is confirmed in xenotransplanted mice. Short-term tissue-slice cultures of surgical samples validate the cell-line results and support the concept that a high proportion of Luminal/ER+ carcinomas are ATRA sensitive, while triple-negative (Basal) and HER2-positive tumors tend to be retinoid resistant. Pathway-oriented analysis of the constitutive gene-expression profiles in the cell lines identifies RARα as the member of the retinoid pathway directly associated with a Luminal phenotype, estrogen positivity and ATRA sensitivity. RARα3 is the major transcript in ATRA-sensitive cells and tumors. Studies in selected cell lines with agonists/antagonists confirm that RARα is the principal mediator of ATRA responsiveness. RARα over-expression sensitizes retinoid-resistant MDA-MB453 cells to ATRA anti-proliferative action. Conversely, silencing of RARα in retinoid-sensitive SKBR3 cells abrogates ATRA responsiveness. All this is paralleled by similar effects on ATRA-dependent inhibition of cell motility, indicating that RARα may mediate also ATRA anti-metastatic effects. We define gene sets of predictive potential which are associated with ATRA sensitivity in breast cancer cell lines and validate them in short-term tissue cultures of Luminal/ER+ and triple-negative tumors. In these last models, we determine the perturbations in the transcriptomic profiles afforded by ATRA. The study provides fundamental information for the development of retinoid-based therapeutic strategies aimed at the stratified treatment of breast cancer subtypes.
Collapse
Affiliation(s)
- Floriana Centritto
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Gabriela Paroni
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Marco Bolis
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Silvio Ken Garattini
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Mami Kurosaki
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Maria Monica Barzago
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Adriana Zanetti
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - James Neil Fisher
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Mark Francis Scott
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Linda Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Monica Lupi
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Paolo Ubezio
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | | | | | - Paola Rizzo
- Gene Therapy and Cellular Reprogramming, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Bergamo, Italy
| | - Maurizio Gianni'
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Maddalena Fratelli
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Mineko Terao
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milano, Italy
| |
Collapse
|
41
|
Diaz P, Huang W, Keyari CM, Buttrick B, Price L, Guilloteau N, Tripathy S, Sperandio VG, Fronczek FR, Astruc-Diaz F, Isoherranen N. Development and Characterization of Novel and Selective Inhibitors of Cytochrome P450 CYP26A1, the Human Liver Retinoic Acid Hydroxylase. J Med Chem 2016; 59:2579-95. [PMID: 26918322 DOI: 10.1021/acs.jmedchem.5b01780] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytochrome P450 CYP26 enzymes are responsible for all-trans-retinoic acid (atRA) clearance. Inhibition of CYP26 enzymes will increase endogenous atRA concentrations and is an attractive therapeutic target. However, the selectivity and potency of the existing atRA metabolism inhibitors toward CYP26A1 and CYP26B1 is unknown, and no selective CYP26A1 or CYP26B1 inhibitors have been developed. Here the synthesis and potent inhibitory activity of the first CYP26A1 selective inhibitors is reported. A series of nonazole CYP26A1 selective inhibitors was identified with low nM potency. The lead compound 3-{4-[2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1,3-dioxolan-2-yl] phenyl}4-propanoic acid (24) had 43-fold selectivity toward CYP26A1 with an IC50 of 340 nM. Compound 24 and its two structural analogues also inhibited atRA metabolism in HepG2 cells, resulting in increased potency of atRA toward RAR activation. The identified compounds have potential to become novel treatments aiming to elevate endogenous atRA concentrations and may be useful as cotreatment with atRA to combat therapy resistance.
Collapse
Affiliation(s)
- Philippe Diaz
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana , 32 Campus Drive, Missoula, Montana 59812, United States.,DermaXon LLC , 32 Campus Drive, Missoula, Montana 59812, United States
| | - Weize Huang
- Department of Pharmaceutics, University of Washington , 1959 NE Pacific Street, Health Sciences Building, Box 357610, Seattle, Washington 98195, United States
| | - Charles M Keyari
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana , 32 Campus Drive, Missoula, Montana 59812, United States
| | - Brian Buttrick
- Department of Pharmaceutics, University of Washington , 1959 NE Pacific Street, Health Sciences Building, Box 357610, Seattle, Washington 98195, United States
| | - Lauren Price
- Department of Pharmaceutics, University of Washington , 1959 NE Pacific Street, Health Sciences Building, Box 357610, Seattle, Washington 98195, United States
| | | | - Sasmita Tripathy
- Department of Pharmaceutics, University of Washington , 1959 NE Pacific Street, Health Sciences Building, Box 357610, Seattle, Washington 98195, United States
| | - Vanessa G Sperandio
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana , 32 Campus Drive, Missoula, Montana 59812, United States
| | - Frank R Fronczek
- Chemistry Department, Louisiana State University , 232 Choppin Hall, Baton Rouge, Louisiana 70803, United States
| | - Fanny Astruc-Diaz
- DermaXon LLC , 32 Campus Drive, Missoula, Montana 59812, United States
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington , 1959 NE Pacific Street, Health Sciences Building, Box 357610, Seattle, Washington 98195, United States
| |
Collapse
|
42
|
Foti RS, Isoherranen N, Zelter A, Dickmann LJ, Buttrick BR, Diaz P, Douguet D. Identification of Tazarotenic Acid as the First Xenobiotic Substrate of Human Retinoic Acid Hydroxylase CYP26A1 and CYP26B1. J Pharmacol Exp Ther 2016; 357:281-92. [PMID: 26937021 DOI: 10.1124/jpet.116.232637] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/26/2016] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 (CYP) 26A1 and 26B1 are heme-containing enzymes responsible for metabolizing all-trans retinoic acid (at-RA). No crystal structures have been solved, and therefore homology models that provide structural information are extremely valuable for the development of inhibitors of cytochrome P450 family 26 (CYP26). The objectives of this study were to use homology models of CYP26A1 and CYP26B1 to characterize substrate binding characteristics, to compare structural aspects of their active sites, and to support the role of CYP26 in the metabolism of xenobiotics. Each model was verified by dockingat-RA in the active site and comparing the results to known metabolic profiles ofat-RA. The models were then used to predict the metabolic sites of tazarotenic acid with results verified by in vitro metabolite identification experiments. The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. Metabolite identification experiments indicated that CYP26A1 and CYP26B1 oxidatively metabolized tazarotenic acid on the predicted moiety, with in vitro rates of metabolite formation by CYP26A1 and CYP26B1 being the highest across a panel of enzymes. Molecular analysis of the active sites estimated the active-site volumes of CYP26A1 and CYP26B1 to be 918 Å(3)and 977 Å(3), respectively. Overall, the homology models presented herein describe the enzyme characteristics leading to the metabolism of tazarotenic acid by CYP26A1 and CYP26B1 and support a potential role for the CYP26 enzymes in the metabolism of xenobiotics.
Collapse
Affiliation(s)
- Robert S Foti
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Nina Isoherranen
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Alex Zelter
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Leslie J Dickmann
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Brian R Buttrick
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Philippe Diaz
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| | - Dominique Douguet
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F.); Department of Pharmaceutics, University of Washington, Seattle, Washington (N.I., A.Z., L.J.D., B.R.B.); Core Laboratory for Neuromolecular Production, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana (P.D.); CNRS, Université Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Valbonne, France (D.D.)
| |
Collapse
|
43
|
Mwanza-Lisulo M, Kelly P. Potential for use of retinoic acid as an oral vaccine adjuvant. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0145. [PMID: 25964457 DOI: 10.1098/rstb.2014.0145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite the heavy burden of diarrhoeal disease across much of the tropical world, only two diarrhoea-causing pathogens, cholera and rotavirus, are the target of commercially available vaccines. Oral vaccines are generally less immunogenic than the best parenteral vaccines, but the reasons for this are still debated. Over the past decade, several lines of evidence from work in experimental animals have suggested that all-trans retinoic acid (ATRA), a form of vitamin A which is highly transcriptionally active, can alter the homing receptor expression of T lymphocytes. Increased expression of α4β7 integrin and the chemokine receptor CCR9 following exposure to ATRA can be used to redirect T cells to the gut. Early work in human volunteers suggests that oral ATRA administration 1 h prior to dosing with oral typhoid vaccine can augment secretion of specific IgA against vaccine-derived lipopolysaccharide into gut secretions. In this review, we set out the rationale for using ATRA in this way and assess its likely applicability to vaccination programmes for protection of children in low-income countries from the considerable mortality caused by diarrhoeal disease. Comparison of recent work in experimental animals, non-human primates and men suggests that a more detailed understanding of ATRA dosage and kinetics will be important to taking forward translational work into human vaccinology.
Collapse
Affiliation(s)
- Mpala Mwanza-Lisulo
- Tropical Gastroenterology and Nutrition group, Department of Internal Medicine, University of Zambia School of Medicine, Lusaka, Zambia
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition group, Department of Internal Medicine, University of Zambia School of Medicine, Lusaka, Zambia
| |
Collapse
|
44
|
Brown HR, Castellino S, Groseclose MR, Elangbam CS, Mellon-Kusibab K, Yoon LW, Gates LD, Krull DL, Cariello NF, Arrington-Brown L, Tillman T, Fowler S, Shah V, Bailey D, Miller RT. Drug-induced Liver Fibrosis. Toxicol Pathol 2016; 44:112-31. [DOI: 10.1177/0192623315617033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Nevirapine (NVP) is associated with hepatotoxicity in 1–5% of patients. In rodent studies, NVP has been shown to cause hepatic enzyme induction, centrilobular hypertrophy, and skin rash in various rat strains but not liver toxicity. In an effort to understand whether NVP is metabolized differently in a transiently inflamed liver and whether a heightened immune response alters NVP-induced hepatic responses, female brown Norway rats were dosed with either vehicle or NVP alone (75 mg/kg/day for 15 days) or galactosamine alone (single intraperitoneal [ip] injection on day 7 to mimic viral hepatitis) or a combination of NVP (75/100/150 mg/kg/day for 15 days) and galactosamine (single 750 mg/kg ip on day 7). Livers were collected at necropsy for histopathology, matrix-assisted laser desorption/ionization imaging mass spectrometry and gene expression. Eight days after galactosamine, hepatic fibrosis was noted in rats dosed with the combination of NVP and galactosamine. No fibrosis occurred with NVP alone or galactosamine alone. Gene expression data suggested a viral-like response initiated by galactosamine via RNA sensors leading to apoptosis, toll-like receptor, and dendritic cell responses. These were exacerbated by NVP-induced growth factor, retinol, apoptosis, and periostin effects. This finding supports clinical reports warning against exacerbation of fibrosis by NVP in patients with hepatitis C.
Collapse
Affiliation(s)
- H. Roger Brown
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Stephen Castellino
- Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - M. Reid Groseclose
- Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Chandikumar S. Elangbam
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Kathryn Mellon-Kusibab
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Lawrence W. Yoon
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Lisa D. Gates
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - David L. Krull
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Neal F. Cariello
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Leigh Arrington-Brown
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Tony Tillman
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Serita Fowler
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Vishal Shah
- Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - David Bailey
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Richard T. Miller
- Department of Safety Assessment, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| |
Collapse
|
45
|
Abstract
Retinoic acid (RA) was identified as the biologically active form of vitamin A almost 70 years ago and work on its function and mechanism of action is still of major interest both from a scientific and a clinical perspective. The currently accepted model postulates that RA is produced in two sequential oxidative steps: first, retinol is oxidized reversibly to retinaldehyde, and then retinaldehyde is oxidized irreversibly to RA. Excess RA is inactivated by conversion to hydroxylated derivatives. Much is left to learn, especially about retinoid binding proteins and the trafficking of the hydrophobic retinoid substrates between membrane bound and cytosolic enzymes. Here, background on development of the field and an update on recent advances in our understanding of the enzymatic pathways and mechanisms that control the rate of RA production and degradation are presented with a focus on the many questions that remain unanswered.
Collapse
|
46
|
Shmarakov IO. Retinoid-xenobiotic interactions: the Ying and the Yang. Hepatobiliary Surg Nutr 2015; 4:243-67. [PMID: 26311625 DOI: 10.3978/j.issn.2304-3881.2015.05.05] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/13/2015] [Indexed: 12/20/2022]
Abstract
The literature provides compelling evidence pointing to tight metabolic interactions between retinoids and xenobiotics. These are extensive and important for understanding xenobiotic actions in the body. Within the body, retinoids affect xenobiotic metabolism and actions and conversely, xenobiotics affect retinoid metabolism and actions. This article summarizes data that establish the importance of retinoid-dependent metabolic pathways for sustaining the body's responses to xenobiotic exposure, including the roles of all-trans- and 9-cis-retinoic acid for protecting mammals from harmful xenobiotic effects and for ensuring xenobiotic elimination from the body. This review will also consider molecular mechanisms underlying xenobiotic toxicity focusing on how this may contribute to retinoid deficiency and disruption of normal retinoid homeostasis. Special attention is paid to xenobiotic molecular targets (nuclear receptors, regulatory proteins, enzymes, and transporters) which affect retinoid metabolism and signaling.
Collapse
Affiliation(s)
- Igor O Shmarakov
- Department of Biochemistry and Biotechnology, Chernivtsi National University, Chernivtsi, Ukraine
| |
Collapse
|
47
|
Arnold SLM, Kent T, Hogarth CA, Griswold MD, Amory JK, Isoherranen N. Pharmacological inhibition of ALDH1A in mice decreases all-trans retinoic acid concentrations in a tissue specific manner. Biochem Pharmacol 2015; 95:177-92. [PMID: 25764981 PMCID: PMC4420653 DOI: 10.1016/j.bcp.2015.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/03/2015] [Indexed: 12/27/2022]
Abstract
all-trans retinoic acid (atRA), the active metabolite of vitamin A, is an essential signaling molecule. Specifically the concentrations of atRA are spatiotemporally controlled in target tissues such as the liver and the testes. While the enzymes of the aldehyde dehydrogenase 1A family (ALDH1A) are believed to control the synthesis of atRA, a direct relationship between altered ALDH1A activity and tissue atRA concentrations has never been shown. To test whether inhibition of ALDH1A enzymes decreases atRA concentrations in a tissue specific manner, the potent ALDH1A inhibitor WIN 18,446 was used to inhibit ALDH1A activity in mice. The ALDH1A expression, atRA formation kinetics, ALDH1A inhibition by WIN 18,446 and WIN 18,446 disposition were used to predict the time course and extent of inhibition of atRA formation in the testis and liver. The effect of WIN 18,446 on atRA concentrations in testis, liver and serum were measured following single and multiple doses of WIN 18,446. ALDH1A1 and ALDH1A2 were responsible for the majority of atRA formation in the testis while ALDH1A1 and aldehyde oxidase contributed to atRA formation in the liver. Due to the different complement of enzymes contributing to atRA formation in different tissues and different inhibition of ALDH1A1 and ALDH1A2 by WIN 18,446, WIN 18,446 caused only a 50% decrease in liver atRA but testicular atRA decreased over 90%. Serum atRA concentrations were also reduced. These data demonstrate that inhibition of ALDH1A enzymes will decrease atRA concentrations in a tissue specific manner and selective ALDH1A inhibition could be used to alter atRA concentrations in select target tissues.
Collapse
Affiliation(s)
- Samuel L M Arnold
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Travis Kent
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Cathryn A Hogarth
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Michael D Griswold
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - John K Amory
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
48
|
Role of Retinoic Acid-Metabolizing Cytochrome P450s, CYP26, in Inflammation and Cancer. ADVANCES IN PHARMACOLOGY 2015; 74:373-412. [PMID: 26233912 DOI: 10.1016/bs.apha.2015.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vitamin A (retinol) and its active metabolite, all-trans-retinoic acid (atRA), play critical roles in regulating the differentiation, growth, and migration of immune cells. Similarly, as critical signaling molecules in the regulation of the cell cycle, retinoids are important in cancers. Concentrations of atRA are tightly regulated in tissues, predominantly by the availability of retinol, synthesis of atRA by ALDH1A enzymes and metabolism and clearance of atRA by CYP26 enzymes. The ALDH1A and CYP26 enzymes are expressed in several cell types in the immune system and in cancer cells. In the immune system, the ALDH1A and CYP26 enzymes appear to modulate RA concentrations. Consequently, alterations in the activity of ALDH1A and CYP26 enzymes are expected to change disease outcomes in inflammation. There is increasing evidence from various disease models of intestinal and skin inflammation that treatment with atRA has a positive effect on disease markers. However, whether aberrant atRA concentrations or atRA synthesis and metabolism play a role in inflammatory disease development and progression is not well understood. In cancers, especially in acute promyelocytic leukemia and neuroblastoma, increasing intracellular concentrations of atRA appears to provide clinical benefit. Inhibition of the CYP26 enzymes to increase atRA concentrations and combat therapy resistance has been pursued as a drug target in these cancers. This chapter covers the current knowledge of how atRA and retinol regulate the immune system and inflammation, how retinol and atRA metabolism is altered in inflammation and cancer, and what roles atRA-metabolizing enzymes have in immune responses and cancers.
Collapse
|
49
|
Arnold SL, Kent T, Hogarth CA, Schlatt S, Prasad B, Haenisch M, Walsh T, Muller CH, Griswold MD, Amory JK, Isoherranen N. Importance of ALDH1A enzymes in determining human testicular retinoic acid concentrations. J Lipid Res 2014; 56:342-57. [PMID: 25502770 DOI: 10.1194/jlr.m054718] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Retinoic acid (RA), the active metabolite of vitamin A, is required for spermatogenesis and many other biological processes. RA formation requires irreversible oxidation of retinal to RA by aldehyde dehydrogenase enzymes of the 1A family (ALDH1A). While ALDH1A1, ALDH1A2, and ALDH1A3 all form RA, the expression pattern and relative contribution of these enzymes to RA formation in the testis is unknown. In this study, novel methods to measure ALDH1A protein levels and intrinsic RA formation were used to accurately predict RA formation velocities in individual human testis samples and an association between RA formation and intratesticular RA concentrations was observed. The distinct localization of ALDH1A in the testis suggests a specific role for each enzyme in controlling RA formation. ALDH1A1 was found in Sertoli cells, while only ALDH1A2 was found in spermatogonia, spermatids, and spermatocytes. In the absence of cellular retinol binding protein (CRBP)1, ALDH1A1 was predicted to be the main contributor to intratesticular RA formation, but when CRBP1 was present, ALDH1A2 was predicted to be equally important in RA formation as ALDH1A1. This study provides a comprehensive novel methodology to evaluate RA homeostasis in human tissues and provides insight to how the individual ALDH1A enzymes mediate RA concentrations in specific cell types.
Collapse
Affiliation(s)
- Samuel L Arnold
- Department of Pharmaceutics, School of Pharmacy, School of Medicine, University of Washington, Seattle, WA 98195
| | - Travis Kent
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - Cathryn A Hogarth
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - Stefan Schlatt
- Center for Reproductive Medicine and Andrology, Munster, Germany
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, School of Medicine, University of Washington, Seattle, WA 98195
| | - Michael Haenisch
- Departments of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195
| | - Thomas Walsh
- Urology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Charles H Muller
- Urology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Michael D Griswold
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - John K Amory
- Medicine, School of Medicine, University of Washington, Seattle, WA 98195
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, School of Medicine, University of Washington, Seattle, WA 98195
| |
Collapse
|
50
|
Topletz AR, Tripathy S, Foti RS, Shimshoni JA, Nelson WL, Isoherranen N. Induction of CYP26A1 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Mol Pharmacol 2014; 87:430-41. [PMID: 25492813 DOI: 10.1124/mol.114.096784] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
All-trans-retinoic acid (atRA), the active metabolite of vitamin A, induces gene transcription via binding to nuclear retinoic acid receptors (RARs). The primary hydroxylated metabolites formed from atRA by CYP26A1, and the subsequent metabolite 4-oxo-atRA, bind to RARs and potentially have biologic activity. Hence, CYP26A1, the main atRA hydroxylase, may function either to deplete bioactive retinoids or to form active metabolites. This study aimed to determine the role of CYP26A1 in modulating RAR activation via formation and elimination of active retinoids. After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARβ were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. However, >60% of the 4-OH-atRA enantiomers were converted to 4-oxo-atRA in the first 12 hours of treatment, suggesting that the activity of the 4-OH-atRA was due to 4-oxo-atRA. In human hepatocytes, atRA, 4-OH-atRA, and 4-oxo-atRA induced CYP26A1 and 4-oxo-atRA formation was observed from 4-OH-atRA. In HepG2 cells, 4-oxo-atRA formation was observed even in the absence of CYP26A1 activity and this formation was not inhibited by ketoconazole. In human liver microsomes, 4-oxo-atRA formation was supported by NAD(+), suggesting that 4-oxo-atRA formation is mediated by a microsomal alcohol dehydrogenase. Although 4-oxo-atRA was not formed by CYP26A1, it was depleted by CYP26A1 (Km = 63 nM and intrinsic clearance = 90 μl/min per pmol). Similarly, CYP26A1 depleted 18-OH-atRA and the 4-OH-atRA enantiomers. These data support the role of CYP26A1 to clear bioactive retinoids, and suggest that the enzyme forming active 4-oxo-atRA may be important in modulating retinoid action.
Collapse
Affiliation(s)
- Ariel R Topletz
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| | - Sasmita Tripathy
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| | - Robert S Foti
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| | - Jakob A Shimshoni
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| | - Wendel L Nelson
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| | - Nina Isoherranen
- Departments of Pharmaceutics (A.R.T., S.T., J.A.S., N.I.) and Medicinal Chemistry (W.L.N.), University of Washington, Seattle, Washington; and Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (R.S.F.)
| |
Collapse
|