1
|
Terrell JA, Chen C. Extracellular Matrix Microstructures Modulate Hepatic Methionine Cycle and Methylations. Biomacromolecules 2025. [PMID: 40298277 DOI: 10.1021/acs.biomac.4c01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The field of mechanobiology has grown in the past decade, but limited studies investigate how the extracellular matrix affects the cell metabolome. The methionine cycle involves the catabolism and regeneration of methionine through the donation and recovery of a single methyl group; this methyl group can methylate DNA, RNA, and proteins to alter gene expression and protein-protein interactions. Through studying cells cultured on fibrous (mimicking healthy extracellular matrice (ECM)) and flat (mimicking severely fibrotic ECM) substrates, we observed an increase in methionine cycle enzyme expression in cells on the flat substrate. We also present how the methionine cycle is modulated by the ECM through transmembrane protein integrin β1. By inhibiting integrin activation through the ligand-mimicking peptide RGD, we observed that the methionine cycle was protected from alteration. The results presented provide insight into possible therapeutic targets for fibrotic diseases and knowledge of mechanisms by which the ECM alters cell processes.
Collapse
Affiliation(s)
- John A Terrell
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
2
|
Kotlyarov S, Oskin D. The Role of Inflammation in the Pathogenesis of Comorbidity of Chronic Obstructive Pulmonary Disease and Pulmonary Tuberculosis. Int J Mol Sci 2025; 26:2378. [PMID: 40141021 PMCID: PMC11942565 DOI: 10.3390/ijms26062378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The comorbid course of chronic obstructive pulmonary disease (COPD) and pulmonary tuberculosis is an important medical and social problem. Both diseases, although having different etiologies, have many overlapping relationships that mutually influence their course and prognosis. The aim of the current review is to discuss the role of different immune mechanisms underlying inflammation in COPD and pulmonary tuberculosis. These mechanisms are known to involve both the innate and adaptive immune system, including various cellular and intercellular interactions. There is growing evidence that immune mechanisms involved in the pathogenesis of both COPD and tuberculosis may jointly contribute to the tuberculosis-associated obstructive pulmonary disease (TOPD) phenotype. Several studies have reported prior tuberculosis as a risk factor for COPD. Therefore, the study of the mechanisms that link COPD and tuberculosis is of considerable clinical interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Dmitry Oskin
- Department of Infectious Diseases and Phthisiology, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
3
|
Farrokhnazar E, Moghbelinejad S, Najafipour R, Teimoori-Toolabi L. MiR-3664-3p through suppressing ABCG2, CYP3A4, MCL1, and MLH1 increases the sensitivity of colorectal cancer cells to irinotecan. Heliyon 2025; 11:e41933. [PMID: 39931465 PMCID: PMC11808512 DOI: 10.1016/j.heliyon.2025.e41933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 02/13/2025] Open
Abstract
Background Colorectal cancer (CRC) is the third most frequently diagnosed malignancy worldwide. Currently, irinotecan (CPT-11) is used alone or in combination with other drugs to treat patients with advanced CRC. However, the 5-year survival rate for metastatic CRC remains below 10 %, largely due to chemotherapy resistance. Several genes, including ABCG2, CYP3A4, MCL1, and MLH1 contribute to irinotecan resistance. This study aimed to identify microRNAs that simultaneously regulate the expression of these genes in irinotecan-resistant cell lines and study their effect on resistant colorectal cancer cells. Methods Irinotecan-resistant colorectal cancer cell lines were developed by intermittently exposing HCT116 and SW480 cell lines to gradually increasing doses of irinotecan over four generations. These resistant cell lines were designated HCT116-R1, HCT116-R2, HCT116-R3, HCT116-R4 and SW480-R1, SW480-R2, SW480-R3, SW480-R4. The induction of resistance was confirmed using MTT assays, by calculating IC50 values for each generation and comparing them to the parental cells. The expression levels of the ABCG2, CYP3A4, MCL1, and MLH1 genes, along with miR-3664-3p, were initially measured in all resistant and parental cell lines using quantitative real-time PCR. Following transfection of HCT116-R3 and SW480-R3 cells with pre-miR-3664-3p, the expression levels of ABCG2, CYP3A4, MCL1, MLH1, and miR-3664-3p were re-evaluated using real-time PCR. Results In resistant cell lines derived from HCT116 and SW480, increased expression of the ABCG2, CYP3A4, and MCL1 genes was observed. However, a reduction in CYP3A4 expression was noted in the final resistant lines from both cell lines. Additionally, while MLH1 expression increased in HCT116-derived cell lines, no significant increase was observed in SW480-derived lines. A consistent decrease in miR-3664-3p expression was found across all resistant cell lines. When we transfected HCT116-R3 and SW480-R3 cells with pre-miR-3664-3p, there was an increase in miR-3664-3p expression and a reduction in ABCG2, CYP3A4, MCL1, and MLH1 gene expression. This led to increased sensitivity to irinotecan. Conclusion It can be concluded that miR-3664-3p can be considered a regulator of resistance to irinotecan by modulating the expression of ABCG2, CYP3A4, MCL1, and MLH1 genes.
Collapse
Affiliation(s)
- Elham Farrokhnazar
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Iran
- Department of Molecular Medicine, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Moghbelinejad
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Reza Najafipour
- Genetics Research Center, University of Social Welfare and Rehabilitation Science, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Iran
| |
Collapse
|
4
|
Nazari A, Osati P, Seifollahy Fakhr S, Faghihkhorasani F, Ghanaatian M, Faghihkhorasani F, Rezaei-Tazangi F, Pazhouhesh Far N, Shourideh A, Ebrahimi N, Aref AR. New Emerging Therapeutic Strategies Based on Manipulation of the Redox Regulation Against Therapy Resistance in Cancer. Antioxid Redox Signal 2024. [PMID: 39506926 DOI: 10.1089/ars.2023.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Background: Resistance to standard therapeutic methods, including chemotherapy, immunotherapy, and targeted therapy, remains a critical challenge in effective cancer treatment. Redox homeostasis modification has emerged as a promising approach to address medication resistance. Objective: This review aims to explore the mechanisms of redox alterations and signaling pathways contributing to treatment resistance in cancer. Methods: In this study, a comprehensive review of the molecular mechanisms underlying drug resistance governed by redox signaling was conducted. Emphasis was placed on understanding how tumor cells manage increased reactive oxygen species (ROS) levels through upregulated antioxidant systems, enabling resistance across multiple therapeutic pathways. Results: Key mechanisms identified include alterations in drug efflux, target modifications, metabolic changes, enhanced DNA damage repair, stemness preservation, and tumor microenvironment remodeling. These pathways collectively facilitate tumor cells' adaptive response and resistance to various cancer treatments. Conclusion: Developing a detailed understanding of the interrelationships between these redox-regulated mechanisms and therapeutic resistance holds potential to improve treatment effectiveness, offering valuable insights for both fundamental and clinical cancer research. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Parisa Osati
- Department of Chemical Engineering, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Norway
| | - Ferdos Faghihkhorasani
- Department of Cardiology, Internal Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, 710061, China
| | - Masoud Ghanaatian
- Master 1 Bio-Santé-Parcours Toulouse Graduate School of Cancer, Ageing and Rejuvenation (CARe), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Fereshteh Faghihkhorasani
- General Physician in Medicine Program,General Doctorate Degree of Yazd Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Amir Shourideh
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA and Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Russell LE, Yadav J, Maldonato BJ, Chien HC, Zou L, Vergara AG, Villavicencio EG. Transporter-mediated drug-drug interactions: regulatory guidelines, in vitro and in vivo methodologies and translation, special populations, and the blood-brain barrier. Drug Metab Rev 2024:1-28. [PMID: 38967415 DOI: 10.1080/03602532.2024.2364591] [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: 02/13/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024]
Abstract
This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Quantitative, Translational, and ADME Sciences, AbbVie Inc, North Chicago, IL, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Huan-Chieh Chien
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ling Zou
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Rahway, NJ, USA
| | - Erick G Villavicencio
- Department of Biology-Discovery, Imaging and Functional Genomics, Merck & Co., Inc, Rahway, NJ, USA
| |
Collapse
|
6
|
Tomkiewicz C, Coumoul X, Nioche P, Barouki R, Blanc EB. Costs of molecular adaptation to the chemical exposome: a focus on xenobiotic metabolism pathways. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220510. [PMID: 38310928 PMCID: PMC10838638 DOI: 10.1098/rstb.2022.0510] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 12/04/2023] [Indexed: 02/06/2024] Open
Abstract
Organisms adapt to their environment through different pathways. In vertebrates, xenobiotics are detected, metabolized and eliminated through the inducible xenobiotic-metabolizing pathways (XMP) which can also generate reactive toxic intermediates. In this review, we will discuss the impacts of the chemical exposome complexity on the balance between detoxication and side effects. There is a large discrepancy between the limited number of proteins involved in these pathways (few dozens) and the diversity and complexity of the chemical exposome (tens of thousands of chemicals). Several XMP proteins have a low specificity which allows them to bind and/or metabolize a large number of chemicals. This leads to undesired consequences, such as cross-inhibition, inefficient metabolism, release of toxic intermediates, etc. Furthermore, several XMP proteins have endogenous functions that may be disrupted upon exposure to exogenous chemicals. The gut microbiome produces a very large number of metabolites that enter the body and are part of the chemical exposome. It can metabolize xenobiotics and either eliminate them or lead to toxic derivatives. The complex interactions between chemicals of different origins will be illustrated by the diverse roles of the aryl hydrocarbon receptor which binds and transduces the signals of a large number of xenobiotics, microbiome metabolites, dietary chemicals and endogenous compounds. This article is part of the theme issue 'Endocrine responses to environmental variation: conceptual approaches and recent developments'.
Collapse
Affiliation(s)
| | - Xavier Coumoul
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| | - Pierre Nioche
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| | - Robert Barouki
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
- Hôpital Necker Enfants malades, AP-HP, 75006 Paris, France
| | - Etienne B. Blanc
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| |
Collapse
|
7
|
Li D, Peng X, Hu Z, Li S, Chen J, Pan W. Small molecules targeting selected histone methyltransferases (HMTs) for cancer treatment: Current progress and novel strategies. Eur J Med Chem 2024; 264:115982. [PMID: 38056296 DOI: 10.1016/j.ejmech.2023.115982] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023]
Abstract
Histone methyltransferases (HMTs) play a critical role in gene post-translational regulation and diverse physiological processes, and are implicated in a plethora of human diseases, especially cancer. Increasing evidences demonstrate that HMTs may serve as a potential therapeutic target for cancer treatment. Thus, the development of HMTs inhibitor have been pursued with steadily increasing interest over the past decade. However, the disadvantages such as insufficient clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of conventional HMT inhibitors. New technologies and methods are imperative to enhance the anticancer activity of HMT inhibitors. In this review, we first review the structure and biological functions of the several essential HMTs, such as EZH2, G9a, PRMT5, and DOT1L. The internal relationship between these HMTs and cancer is also expounded. Next, we mainly focus on the latest progress in the development of HMT modulators encompassing dual-target inhibitors, targeted protein degraders and covalent inhibitors from perspectives such as rational design, pharmacodynamics, pharmacokinetics, and clinical status. Lastly, we also discuss the challenges and future directions for HMT-based drug discovery for cancer therapy.
Collapse
Affiliation(s)
- Deping Li
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, PR China
| | - Xiaopeng Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Zhihao Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Shuqing Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 516000, PR China.
| | - Wanyi Pan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
8
|
Taylor ZL, Poweleit EA, Paice K, Somers KM, Pavia K, Vinks AA, Punt N, Mizuno T, Girdwood ST. Tutorial on model selection and validation of model input into precision dosing software for model-informed precision dosing. CPT Pharmacometrics Syst Pharmacol 2023; 12:1827-1845. [PMID: 37771190 PMCID: PMC10725261 DOI: 10.1002/psp4.13056] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
There has been rising interest in using model-informed precision dosing to provide personalized medicine to patients at the bedside. This methodology utilizes population pharmacokinetic models, measured drug concentrations from individual patients, pharmacodynamic biomarkers, and Bayesian estimation to estimate pharmacokinetic parameters and predict concentration-time profiles in individual patients. Using these individualized parameter estimates and simulated drug exposure, dosing recommendations can be generated to maximize target attainment to improve beneficial effect and minimize toxicity. However, the accuracy of the output from this evaluation is highly dependent on the population pharmacokinetic model selected. This tutorial provides a comprehensive approach to evaluating, selecting, and validating a model for input and implementation into a model-informed precision dosing program. A step-by-step outline to validate successful implementation into a precision dosing tool is described using the clinical software platforms Edsim++ and MwPharm++ as examples.
Collapse
Affiliation(s)
- Zachary L. Taylor
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Ethan A. Poweleit
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of Biomedical InformaticsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Biomedical InformaticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Research in Patient ServicesCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Kelli Paice
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Katherine M. Somers
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Hematology and Oncology, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Kathryn Pavia
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Division of Critical Care Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Alexander A. Vinks
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Research in Patient ServicesCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Nieko Punt
- Department of Clinical Pharmacy and Pharmacology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
- MedimaticsMaastrichtThe Netherlands
| | - Tomoyuki Mizuno
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Sonya Tang Girdwood
- Division of Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Hospital Medicine, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
9
|
Jackson KD, Achour B, Lee J, Geffert RM, Beers JL, Latham BD. Novel Approaches to Characterize Individual Drug Metabolism and Advance Precision Medicine. Drug Metab Dispos 2023; 51:1238-1253. [PMID: 37419681 PMCID: PMC10506699 DOI: 10.1124/dmd.122.001066] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 07/09/2023] Open
Abstract
Interindividual variability in drug metabolism can significantly affect drug concentrations in the body and subsequent drug response. Understanding an individual's drug metabolism capacity is important for predicting drug exposure and developing precision medicine strategies. The goal of precision medicine is to individualize drug treatment for patients to maximize efficacy and minimize drug toxicity. While advances in pharmacogenomics have improved our understanding of how genetic variations in drug-metabolizing enzymes (DMEs) affect drug response, nongenetic factors are also known to influence drug metabolism phenotypes. This minireview discusses approaches beyond pharmacogenetic testing to phenotype DMEs-particularly the cytochrome P450 enzymes-in clinical settings. Several phenotyping approaches have been proposed: traditional approaches include phenotyping with exogenous probe substrates and the use of endogenous biomarkers; newer approaches include evaluating circulating noncoding RNAs and liquid biopsy-derived markers relevant to DME expression and function. The goals of this minireview are to 1) provide a high-level overview of traditional and novel approaches to phenotype individual drug metabolism capacity, 2) describe how these approaches are being applied or can be applied to pharmacokinetic studies, and 3) discuss perspectives on future opportunities to advance precision medicine in diverse populations. SIGNIFICANCE STATEMENT: This minireview provides an overview of recent advances in approaches to characterize individual drug metabolism phenotypes in clinical settings. It highlights the integration of existing pharmacokinetic biomarkers with novel approaches; also discussed are current challenges and existing knowledge gaps. The article concludes with perspectives on the future deployment of a liquid biopsy-informed physiologically based pharmacokinetic strategy for patient characterization and precision dosing.
Collapse
Affiliation(s)
- Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Brahim Achour
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Raeanne M Geffert
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Jessica L Beers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Bethany D Latham
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| |
Collapse
|
10
|
Bian J, Zhao J, Zhao Y, Hao X, He S, Li Y, Huang L. Impact of individual factors on DNA methylation of drug metabolism genes: A systematic review. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2023; 64:401-415. [PMID: 37522536 DOI: 10.1002/em.22567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023]
Abstract
Individual differences in drug response have always existed in clinical treatment. Many non-genetic factors show non-negligible impacts on personalized medicine. Emerging studies have demonstrated epigenetic could connect non-genetic factors and individual treatment differences. We used systematic retrieval methods and reviewed studies that showed individual factors' impact on DNA methylation of drug metabolism genes. In total, 68 studies were included, and half (n = 36) were cohort studies. Six aspects of individual factors were summarized from the perspective of personalized medicine: parental exposure, environmental pollutants exposure, obesity and diet, drugs, gender and others. The most research (n = 11) focused on ABCG1 methylation. The majority of studies showed non-genetic factors could result in a significant DNA methylation alteration in drug metabolism genes, which subsequently affects the pharmacokinetic processes. However, the underlying mechanism remained unknown. Finally, some viewpoints were presented for future research.
Collapse
Affiliation(s)
- Jialu Bian
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing, China
| | - Jinxia Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing, China
| | - Yinyu Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing, China
| | - Xu Hao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Shiyu He
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing, China
| | - Yuanyuan Li
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Lin Huang
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| |
Collapse
|
11
|
Leuschner M, Cromarty AD. Critical Assessment of Phenotyping Cocktails for Clinical Use in an African Context. J Pers Med 2023; 13:1098. [PMID: 37511712 PMCID: PMC10381848 DOI: 10.3390/jpm13071098] [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: 06/12/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Interethnic and interindividual variability in in vivo cytochrome P450 (CYP450)-dependent metabolism and altered drug absorption via expressed transport channels such as P-glycoprotein (P-gp) contribute to the adverse drug reactions, drug-drug interaction and therapeutic failure seen in clinical practice. A cost-effective phenotyping approach could be advantageous in providing real-time information on in vivo phenotypes to assist clinicians with individualized drug therapy, especially in resource-constrained countries such as South Africa. A number of phenotyping cocktails have been developed and the aim of this study was to critically assess the feasibility of their use in a South African context. A literature search on library databases (including AccessMedicine, BMJ, ClinicalKey, MEDLINE (Ovid), PubMed, Scopus and TOXLINE) was limited to in vivo cocktails used in the human population to phenotype phase I metabolism and/or P-gp transport. The study found that the implementation of phenotyping in clinical practice is currently limited by multiple administration routes, the varying availability of probe drugs, therapeutic doses eliciting side effects, the interaction between probe drugs and extensive sampling procedures. Analytical challenges include complicated sample workup or extraction assays and impractical analytical procedures with low detection limits, analyte sensitivity and specificity. It was concluded that a single time point, non-invasive capillary sampling, combined with a low-dose probe drug cocktail, to simultaneously quantify in vivo drug and metabolite concentrations, would enhance the feasibility and cost-effectiveness of routine phenotyping in clinical practice; however, future research is needed to establish whether the quantitative bioanalysis of drugs in a capillary whole-blood matrix correlates with that of the standard plasma/serum matrixes used as a reference in the current clinical environment.
Collapse
Affiliation(s)
- Machel Leuschner
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa
| | - Allan Duncan Cromarty
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa
| |
Collapse
|
12
|
Stern S, Wang H, Sadrieh N. Microphysiological Models for Mechanistic-Based Prediction of Idiosyncratic DILI. Cells 2023; 12:1476. [PMID: 37296597 PMCID: PMC10253021 DOI: 10.3390/cells12111476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Drug-induced liver injury (DILI) is a major contributor to high attrition rates among candidate and market drugs and a key regulatory, industry, and global health concern. While acute and dose-dependent DILI, namely, intrinsic DILI, is predictable and often reproducible in preclinical models, the nature of idiosyncratic DILI (iDILI) limits its mechanistic understanding due to the complex disease pathogenesis, and recapitulation using in vitro and in vivo models is extremely challenging. However, hepatic inflammation is a key feature of iDILI primarily orchestrated by the innate and adaptive immune system. This review summarizes the in vitro co-culture models that exploit the role of the immune system to investigate iDILI. Particularly, this review focuses on advancements in human-based 3D multicellular models attempting to supplement in vivo models that often lack predictability and display interspecies variations. Exploiting the immune-mediated mechanisms of iDILI, the inclusion of non-parenchymal cells in these hepatoxicity models, namely, Kupffer cells, stellate cells, dendritic cells, and liver sinusoidal endothelial cells, introduces heterotypic cell-cell interactions and mimics the hepatic microenvironment. Additionally, drugs recalled from the market in the US between 1996-2010 that were studies in these various models highlight the necessity for further harmonization and comparison of model characteristics. Challenges regarding disease-related endpoints, mimicking 3D architecture with different cell-cell contact, cell source, and the underlying multi-cellular and multi-stage mechanisms are described. It is our belief that progressing our understanding of the underlying pathogenesis of iDILI will provide mechanistic clues and a method for drug safety screening to better predict liver injury in clinical trials and post-marketing.
Collapse
Affiliation(s)
- Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Nakissa Sadrieh
- Office of New Drugs, Center of Drug Evaluation and Research, FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
13
|
Fast-Fed Variability: Insights into Drug Delivery, Molecular Manifestations, and Regulatory Aspects. Pharmaceutics 2022; 14:pharmaceutics14091807. [PMID: 36145555 PMCID: PMC9505616 DOI: 10.3390/pharmaceutics14091807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/26/2022] Open
Abstract
Among various drug administration routes, oral drug delivery is preferred and is considered patient-friendly; hence, most of the marketed drugs are available as conventional tablets or capsules. In such cases, the administration of drugs with or without food has tremendous importance on the bioavailability of the drugs. The presence of food may increase (positive effect) or decrease (negative effect) the bioavailability of the drug. Such a positive or negative effect is undesirable since it makes dosage estimation difficult in several diseases. This may lead to an increased propensity for adverse effects of drugs when a positive food effect is perceived. However, a negative food effect may lead to therapeutic insufficiency for patients suffering from life-threatening disorders. This review emphasizes the causes of food effects, formulation strategies to overcome the fast-fed variability, and the regulatory aspects of drugs with food effects, which may open new avenues for researchers to design products that may help to eliminate fast-fed variability.
Collapse
|
14
|
Gene Expression and Protein Abundance of Nuclear Receptors in Human Intestine and Liver: A New Application for Mass Spectrometry-Based Targeted Proteomics. Molecules 2022; 27:molecules27144629. [PMID: 35889510 PMCID: PMC9318449 DOI: 10.3390/molecules27144629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Unwanted drug-drug interactions (DDIs), as caused by the upregulation of clinically relevant drug metabolizing enzymes and transporter proteins in intestine and liver, have the potential to threaten the therapeutic efficacy and safety of drugs. The molecular mechanism of this undesired but frequently occurring scenario of polypharmacy is based on the activation of nuclear receptors such as the pregnane X receptor (PXR) or the constitutive androstane receptor (CAR) by perpetrator agents such as rifampin, phenytoin or St. John’s wort. However, the expression pattern of nuclear receptors in human intestine and liver remains uncertain, which makes it difficult to predict the extent of potential DDIs. Thus, it was the aim of this study to characterize the gene expression and protein abundance of clinically relevant nuclear receptors, i.e., the aryl hydrocarbon receptor (AhR), CAR, farnesoid X receptor (FXR), glucocorticoid receptor (GR), hepatocyte nuclear factor 4 alpha (HNF4α), PXR and small heterodimer partner (SHP), in the aforementioned organs. Methods: Gene expression analysis was performed by quantitative real-time PCR of jejunal, ileal, colonic and liver samples from eight human subjects. In parallel, a targeted proteomic method was developed and validated in order to determine the respective protein amounts of nuclear receptors in human intestinal and liver samples. The LC-MS/MS method was validated according to the current bioanalytical guidelines and met the criteria regarding linearity (0.1–50 nmol/L), within-day and between-day accuracy and precision, as well as the stability criteria. Results: The developed method was successfully validated and applied to determine the abundance of nuclear receptors in human intestinal and liver samples. Gene expression and protein abundance data demonstrated marked differences in human intestine and liver. On the protein level, only AhR and HNF4α could be detected in gut and liver, which corresponds to their highest gene expression. In transfected cell lines, PXR and CAR could be quantified. Conclusions: The substantially different expression pattern of nuclear receptors in human intestinal and liver tissue may explain the different extent of unwanted DDIs in the dependence on the administration route of drugs.
Collapse
|
15
|
Adhikari S, Bhattacharya A, Adhikary S, Singh V, Gadad S, Roy S, Das C. The paradigm of drug resistance in cancer: an epigenetic perspective. Biosci Rep 2022; 42:BSR20211812. [PMID: 35438143 PMCID: PMC9069444 DOI: 10.1042/bsr20211812] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Innate and acquired resistance towards the conventional therapeutic regimen imposes a significant challenge for the successful management of cancer for decades. In patients with advanced carcinomas, acquisition of drug resistance often leads to tumor recurrence and poor prognosis after the first therapeutic cycle. In this context, cancer stem cells (CSCs) are considered as the prime drivers of therapy resistance in cancer due to their 'non-targetable' nature. Drug resistance in cancer is immensely influenced by different properties of CSCs such as epithelial-to-mesenchymal transition (EMT), a profound expression of drug efflux pump genes, detoxification genes, quiescence, and evasion of apoptosis, has been highlighted in this review article. The crucial epigenetic alterations that are intricately associated with regulating different mechanisms of drug resistance, have been discussed thoroughly. Additionally, special attention is drawn towards the epigenetic mechanisms behind the interaction between the cancer cells and their microenvironment which assists in tumor progression and therapy resistance. Finally, we have provided a cumulative overview of the alternative treatment strategies and epigenome-modifying therapies that show the potential of sensitizing the resistant cells towards the conventional treatment strategies. Thus, this review summarizes the epigenetic and molecular background behind therapy resistance, the prime hindrance of present day anti-cancer therapies, and provides an account of the novel complementary epi-drug-based therapeutic strategies to combat drug resistance.
Collapse
Affiliation(s)
- Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Shrikanth S. Gadad
- Department of Molecular and Translational Medicine, Center of Emphasis in Cancer, Texas Tech University Health Sciences Center El Paso, El Paso, TX, U.S.A
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX 78229, U.S.A
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| |
Collapse
|
16
|
Lee YT, Tan YJ, Mok PY, Kaur G, Sreenivasan S, Falasca M, Oon CE. Sex-divergent expression of cytochrome P450 and SIRTUIN 1-7 proteins in toxicity evaluation of a benzimidazole-derived epigenetic modulator in mice. Toxicol Appl Pharmacol 2022; 445:116039. [PMID: 35489524 DOI: 10.1016/j.taap.2022.116039] [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: 01/08/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 10/18/2022]
Abstract
Efforts in precision medicine to combat aberrant epigenome have led to the development of epigenetic targeting drugs. We have previously reported the capability of the BZD9L1 epigenetic modulator to impede colorectal tumour growth in vitro and in vivo through sirtuin (SIRT) inhibition. Although most benzimidazole derivatives are commonly less toxic, their effects on SIRTs and cytochrome P450 (CYP) regulations have not been explored alongside toxicity assessments. SIRTs are histone deacetylases that are crucial in maintaining metabolic homeostasis, whereas CYP is essential in drug metabolism. This study aims to determine the toxicology profile of BZD9L1 through oral acute and repeated dose toxicity evaluations, along with molecular analyses of SIRT, CYP and relevant toxicity markers through western blot and quantitative polymerase chain reaction (qPCR). BZD9L1 demonstrated no sign of acute toxicity at the limit dose (2000 mg/kg). The 28-day toxicity study highlighted the tolerability of repeated dose administration without adverse effects. BZD9L1 showed a sex-divergent regulation of hepatic SIRT1-7, CYP2A5 and CYP2D proteins. Furthermore, BZD9L1 did not induce the expression of organ injury proteins or alter the gene expression of cellular function indicators in mouse liver and kidneys, hence demonstrating, at least in part, the safety of BZD9L1 in short-term evaluations. The present study cautions for personalised strategies when employing benzimidazole-derived epigenetic therapeutics.
Collapse
Affiliation(s)
- Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Pei Yi Mok
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Sasidharan Sreenivasan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Marco Falasca
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
17
|
Hao X, Li Y, Bian J, Zhang Y, He S, Yu F, Feng Y, Huang L. Impact of DNA methylation on ADME gene expression, drug disposition and efficacy. Drug Metab Rev 2022; 54:194-206. [PMID: 35412942 DOI: 10.1080/03602532.2022.2064488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Interindividual differences in drug response have always existed in clinical treatment. Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) play an important role in the process of pharmacokinetics. The effects of genetic polymorphism and nuclear receptors on the expression of drug metabolism enzymes and transporters can only explain some individual differences in clinical treatment. Several key ADME genes have been demonstrated to be regulated by epigenetic mechanisms that can potentially affect interindividual variability in medical treatment. Emerging studies have focused on the importance of DNA methylation for ADME gene expression and for drug response. Among them, the most studied is anti-tumor drugs, and followed by anti-tuberculous and anti-platelet drugs. Therefore, we provide an epigenetics perspective on variability in drug response. The review summarizes the correlation between ADME gene expression and DNA methylation, including the exact methylation locations, and focuses on the corresponding drug disposition and effects to illuminate interindividual differences in clinical medication.
Collapse
Affiliation(s)
- Xu Hao
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuanyuan Li
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jialu Bian
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China
| | - Ying Zhang
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China
| | - Shiyu He
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China
| | - Feng Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yufei Feng
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China
| | - Lin Huang
- Department of Pharmacy, Peking University People's Hospital, Beijing, 100044 China
| |
Collapse
|
18
|
Saiyed AN, Vasavada AR, Johar SRK. Recent trends in miRNA therapeutics and the application of plant miRNA for prevention and treatment of human diseases. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022; 8:24. [PMID: 35382490 PMCID: PMC8972743 DOI: 10.1186/s43094-022-00413-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/21/2022] [Indexed: 02/17/2023] Open
Abstract
Background Researchers now have a new avenue to investigate when it comes to miRNA-based therapeutics. miRNAs have the potential to be valuable biomarkers for disease detection. Variations in miRNA levels may be able to predict changes in normal physiological processes. At the epigenetic level, miRNA has been identified as a promising candidate for distinguishing and treating various diseases and defects. Main body In recent pharmacology, plants miRNA-based drugs have demonstrated a potential role in drug therapeutics. The purpose of this review paper is to discuss miRNA-based therapeutics, the role of miRNA in pharmacoepigenetics modulations, plant miRNA inter-kingdom regulation, and the therapeutic value and application of plant miRNA for cross-kingdom approaches. Target prediction and complementarity with host genes, as well as cross-kingdom gene interactions with plant miRNAs, are also revealed by bioinformatics research. We also show how plant miRNA can be transmitted from one species to another by crossing kingdom boundaries in this review. Despite several unidentified barriers to plant miRNA cross-transfer, plant miRNA-based gene regulation in trans-kingdom gene regulation may soon be valued as a possible approach in plant-based drug therapeutics. Conclusion This review summarised the biochemical synthesis of miRNAs, pharmacoepigenetics, drug therapeutics and miRNA transkingdom transfer.
Collapse
Affiliation(s)
- Atiyabanu N. Saiyed
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat India
- Ph.D. scholar of Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Abhay R. Vasavada
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat India
| | - S. R. Kaid Johar
- Department of Zoology, BMTC, Human Genetics, USSC, Gujarat University, Ahmedabad, Gujarat India
| |
Collapse
|
19
|
Li D, Chen M, Hong H, Tong W, Ning B. Integrative approaches for studying the role of noncoding RNAs in influencing drug efficacy and toxicity. Expert Opin Drug Metab Toxicol 2022; 18:151-163. [PMID: 35296201 PMCID: PMC9117541 DOI: 10.1080/17425255.2022.2054802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/14/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Drug efficacy and toxicity are important factors for evaluation in drug development. Drug metabolizing enzymes and transporters (DMETs) play an essential role in drug efficacy and toxicity. Noncoding RNAs (ncRNAs) have been implicated to influence inter-individual variations in drug efficacy and safety by regulating DMETs. An efficient strategy is urgently needed to identify and functionally characterize ncRNAs that mediate drug efficacy and toxicity through regulating DMETs. AREAS COVERED We outline an integrative strategy to identify ncRNAs that modulate DMETs. We include reliable tools and databases for computational prediction of ncRNA targets with regard to their advantages and limitations. Various biochemical, molecular, and cellular assays are discussed for in vitro experimental verification of the regulatory function of ncRNAs. In vivo approaches for association of ncRNAs with drug treatment and toxicity are also reviewed. EXPERT OPINION A streamlined integration of computational prediction and wet-lab validation is important to elucidate mechanisms of ncRNAs in the regulation of DMETs related to drug efficacy and safety. Bioinformatic analyses using open-access tools and databases serve as a powerful booster for ncRNA Research in toxicology. Further refinement of computational algorithms and experimental technologies is needed to improve accuracy and efficiency in ncRNA target identification and characterization.
Collapse
Affiliation(s)
- Dongying Li
- National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR, USA
| | - Minjun Chen
- National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR, USA
| | - Huixiao Hong
- National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR, USA
| | - Weida Tong
- National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR, USA
| | - Baitang Ning
- National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, AR, USA
| |
Collapse
|
20
|
Scionti F, Di Martino MT, Caracciolo D, Pensabene L, Tagliaferri P, Arbitrio M. Tools in Pharmacogenomics Biomarker Identification for Cancer Patients. Methods Mol Biol 2022; 2401:1-12. [PMID: 34902118 DOI: 10.1007/978-1-0716-1839-4_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] [Indexed: 06/14/2023]
Abstract
The understanding of the biological differences which underlie the inter-individual variability in drug response improved the efficacy of cancer therapy in the era of precision medicine. In fact molecularly targeted drugs and immunotherapy represent a revolution in cancer treatment. The identification of genetic predictive and/or prognostic biomarkers linked to drug pharmacokinetics (PK) and pharmacodynamics (PD) is allowed by the development of high-throughput omics tools for detecting and understanding biological differences among individuals, in order to improve drug efficacy and minimize risk of toxicity. Personalized medicine in cancer treatment reduces costs of the healthcare system. Unfortunately, pharmacogenomics biomarkers discovery is influenced by complexity, need of high-quality evidence, and a validation process for regulatory purposes. This chapter is focused on the critic analysis of presently available pharmacogenomics tools for discovering or testing genetic polymorphic variants in drug metabolizing enzyme to be introduced in clinical practice for the prospective stratification of cancer patients.
Collapse
Affiliation(s)
- Francesca Scionti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Messina, Italy
| | | | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Licia Pensabene
- Department of Medical and Surgical Sciences, Pediatric Unit, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), National Research Council (CNR), Catanzaro, Italy.
| |
Collapse
|
21
|
Yin J, Li F, Li Z, Yu L, Zhu F, Zeng S. Feature, Function, and Information of Drug Transporter-Related Databases. Drug Metab Dispos 2022; 50:76-85. [PMID: 34426411 DOI: 10.1124/dmd.121.000419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/20/2021] [Indexed: 11/22/2022] Open
Abstract
With the rapid progress in pharmaceutical experiments and clinical investigations, extensive knowledge of drug transporters (DTs) has accumulated, which is valuable data for the understanding of drug metabolism and disposition. However, such data are largely dispersed in the literature, which hampers its utility and significantly limits its possibility for comprehensive analysis. A variety of databases have, therefore, been constructed to provide DT-related data, and they were reviewed in this study. First, several knowledge bases providing data regarding clinically important drugs and their corresponding transporters were discussed, which constituted the most important resources of DT-centered data. Second, some databases describing the general transporters and their functional families were reviewed. Third, various databases offering transporter information as part of their entire data collection were described. Finally, customized database functions that are available to facilitate DT-related research were discussed. This review provided an overview of the whole collection of DT-related databases, which might facilitate research on precision medicine and rational drug use. SIGNIFICANCE STATEMENT: A collection of well established databases related to drug transporters were comprehensively reviewed, which were organized according to their importance in drug absorption, distribution, metabolism, and excretion research. These databases could collectively contribute to the research on rational drug use.
Collapse
Affiliation(s)
- Jiayi Yin
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| | - Fengcheng Li
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| | - Zhaorong Li
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| | - Feng Zhu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China (J.Y., F.L., L.Y., F.Z., S.Z.); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China (Z.L., F.Z.); Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China (F.Z.); and Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Cancer Center of Zhejiang University, Hangzhou 310058, China (S.Z.)
| |
Collapse
|
22
|
Genetic variations and epigenetic modulations in CYP genes: Implications in NSAID-treatment of arthritis patients. THE NUCLEUS 2021. [DOI: 10.1007/s13237-021-00373-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
23
|
Xu Q. Human Three-Dimensional Hepatic Models: Cell Type Variety and Corresponding Applications. Front Bioeng Biotechnol 2021; 9:730008. [PMID: 34631680 PMCID: PMC8497968 DOI: 10.3389/fbioe.2021.730008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
Owing to retained hepatic phenotypes and functions, human three-dimensional (3D) hepatic models established with diverse hepatic cell types are thought to recoup the gaps in drug development and disease modeling limited by a conventional two-dimensional (2D) cell culture system and species-specific variability in drug metabolizing enzymes and transporters. Primary human hepatocytes, human hepatic cancer cell lines, and human stem cell-derived hepatocyte-like cells are three main hepatic cell types used in current models and exhibit divergent hepatic phenotypes. Primary human hepatocytes derived from healthy hepatic parenchyma resemble in vivo-like genetic and metabolic profiling. Human hepatic cancer cell lines are unlimitedly reproducible and tumorigenic. Stem cell-derived hepatocyte-like cells derived from patients are promising to retain the donor's genetic background. It has been suggested in some studies that unique properties of cell types endue them with benefits in different research fields of in vitro 3D modeling paradigm. For instance, the primary human hepatocyte was thought to be the gold standard for hepatotoxicity study, and stem cell-derived hepatocyte-like cells have taken a main role in personalized medicine and regenerative medicine. However, the comprehensive review focuses on the hepatic cell type variety, and corresponding applications in 3D models are sparse. Therefore, this review summarizes the characteristics of different cell types and discusses opportunities of different cell types in drug development, liver disease modeling, and liver transplantation.
Collapse
Affiliation(s)
- Qianqian Xu
- School of Chinese Medicine, and Centre for Cancer and Inflammation Research, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
24
|
Mora Y, Reyes ME, Zanella L, Mora B, Buchegger K, Ili C, Brebi P. Resistance to platinum-based cancer drugs: a special focus on epigenetic mechanisms. Pharmacogenomics 2021; 22:777-790. [PMID: 34281355 DOI: 10.2217/pgs-2021-0020] [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] [Indexed: 12/18/2022] Open
Abstract
Chemoresistance is a significant clinical challenge, limiting the drug response in cancer. Several mechanisms associated with drug resistance have been characterized, and the role of epigenetics in generating resistance to platinum-based drugs has been clarified. Epigenetic mechanisms such as DNA methylation, histone modification, long noncoding RNA, and microRNA affect the expression of genes implicated in absorption, distribution, metabolism and excretion (ADME) of drugs, and other non-ADME genes that encode enzymes involved in the processes of cell proliferation, DNA repair, apoptosis and signal transduction key in the development of chemoresistance in cancer, specifically in platinum-based drugs. This review summarizes current discoveries in epigenetic regulation implicated in platinum drug resistance in cancer and the main clinical trials based on epigenetic therapy, evaluating their potential synergy with platinum-based drugs.
Collapse
Affiliation(s)
- Yuselin Mora
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile
| | - María Elena Reyes
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile.,Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago, 8370003, Chile
| | - Louise Zanella
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile
| | - Bárbara Mora
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, 4810101, Chile
| | - Kurt Buchegger
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile.,Departamento Ciencias Básicas, Facultad de Medicina, Universidad de La Frontera, Temuco, 4811230, Chile
| | - Carmen Ili
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile
| | - Priscilla Brebi
- Laboratory of Integrative Biology (LIBi), Scientific & Technological Bioresource Nucleus- Center for Excellence in Translational Medicine (BIOREN-CEMT), Universidad de La Frontera, Temuco, 4810296, Chile
| |
Collapse
|
25
|
Kringel D, Malkusch S, Lötsch J. Drugs and Epigenetic Molecular Functions. A Pharmacological Data Scientometric Analysis. Int J Mol Sci 2021; 22:7250. [PMID: 34298869 PMCID: PMC8311652 DOI: 10.3390/ijms22147250] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Interactions of drugs with the classical epigenetic mechanism of DNA methylation or histone modification are increasingly being elucidated mechanistically and used to develop novel classes of epigenetic therapeutics. A data science approach is used to synthesize current knowledge on the pharmacological implications of epigenetic regulation of gene expression. Computer-aided knowledge discovery for epigenetic implications of current approved or investigational drugs was performed by querying information from multiple publicly available gold-standard sources to (i) identify enzymes involved in classical epigenetic processes, (ii) screen original biomedical scientific publications including bibliometric analyses, (iii) identify drugs that interact with epigenetic enzymes, including their additional non-epigenetic targets, and (iv) analyze computational functional genomics of drugs with epigenetic interactions. PubMed database search yielded 3051 hits on epigenetics and drugs, starting in 1992 and peaking in 2016. Annual citations increased to a plateau in 2000 and show a downward trend since 2008. Approved and investigational drugs in the DrugBank database included 122 compounds that interacted with 68 unique epigenetic enzymes. Additional molecular functions modulated by these drugs included other enzyme interactions, whereas modulation of ion channels or G-protein-coupled receptors were underrepresented. Epigenetic interactions included (i) drug-induced modulation of DNA methylation, (ii) drug-induced modulation of histone conformations, and (iii) epigenetic modulation of drug effects by interference with pharmacokinetics or pharmacodynamics. Interactions of epigenetic molecular functions and drugs are mutual. Recent research activities on the discovery and development of novel epigenetic therapeutics have passed successfully, whereas epigenetic effects of non-epigenetic drugs or epigenetically induced changes in the targets of common drugs have not yet received the necessary systematic attention in the context of pharmacological plasticity.
Collapse
Affiliation(s)
- Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (D.K.); (S.M.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
26
|
Loras A, Segovia C, Ruiz-Cerdá JL. Epigenomic and Metabolomic Integration Reveals Dynamic Metabolic Regulation in Bladder Cancer. Cancers (Basel) 2021; 13:2719. [PMID: 34072826 PMCID: PMC8198168 DOI: 10.3390/cancers13112719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer (BC) represents a clinical, social, and economic challenge due to tumor-intrinsic characteristics, limitations of diagnostic techniques and a lack of personalized treatments. In the last decade, the use of liquid biopsy has grown as a non-invasive approach to characterize tumors. Moreover, the emergence of omics has increased our knowledge of cancer biology and identified critical BC biomarkers. The rewiring between epigenetics and metabolism has been closely linked to tumor phenotype. Chromatin remodelers interact with each other to control gene silencing in BC, but also with stress-inducible factors or oncogenic signaling cascades to regulate metabolic reprogramming towards glycolysis, the pentose phosphate pathway, and lipogenesis. Concurrently, one-carbon metabolism supplies methyl groups to histone and DNA methyltransferases, leading to the hypermethylation and silencing of suppressor genes in BC. Conversely, α-KG and acetyl-CoA enhance the activity of histone demethylases and acetyl transferases, increasing gene expression, while succinate and fumarate have an inhibitory role. This review is the first to analyze the interplay between epigenome, metabolome and cell signaling pathways in BC, and shows how their regulation contributes to tumor development and progression. Moreover, it summarizes non-invasive biomarkers that could be applied in clinical practice to improve diagnosis, monitoring, prognosis and the therapeutic options in BC.
Collapse
Affiliation(s)
- Alba Loras
- Unidad Mixta de Investigación en TICs Aplicadas a la Reingeniería de Procesos Socio-Sanitarios (eRPSS), Universitat Politècnica de València-Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Cristina Segovia
- Epithelial Carcinogenesis Group, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - José Luis Ruiz-Cerdá
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València-Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain;
- Servicio de Urología, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Departamento de Cirugía, Facultad de Medicina y Odontología, Universitat de València, 46010 Valencia, Spain
| |
Collapse
|
27
|
Sun R, Du C, Li J, Zhou Y, Xiong W, Xiang J, Liu J, Xiao Z, Fang L, Li Z. Systematic Investigation of DNA Methylation Associated With Platinum Chemotherapy Resistance Across 13 Cancer Types. Front Pharmacol 2021; 12:616529. [PMID: 33995018 PMCID: PMC8117351 DOI: 10.3389/fphar.2021.616529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Platinum resistance poses a significant problem for oncology clinicians. As a result, the role of epigenetics and DNA methylation in platinum-based chemoresistance has gained increasing attention from researchers in recent years. A systematic investigation of aberrant methylation patterns related to platinum resistance across various cancer types is urgently needed. Methods: We analyzed the platinum chemotherapy response-related methylation patterns from different perspectives of 618 patients across 13 cancer types and integrated transcriptional and clinical data. Spearman’s test was used to evaluate the correlation between methylation and gene expression. Cox analysis, the Kaplan-Meier method, and log-rank tests were performed to identify potential risk biomarkers based on differentially methylated positions (DMPs) and compare survival based on DMP values. Support vector machines and receiver operating characteristic curves were used to identify the platinum-response predictive DMPs. Results: A total of 3,703 DMPs (p value < 0.001 and absolute delta beta >0.10) were identified, and the DMP numbers of each cancer type varied. A total of 39.83% of DMPs were hypermethylated and 60.17% were hypomethylated in platinum-resistant patients. Among them, 405 DMPs (Benjamini and Hochberg adjusted p value < 0.05) were found to be associated with prognosis in tumor patients treated with platinum-based regimens, and 664 DMPs displayed the potential to predict platinum chemotherapy response. In addition, we defined six DNA DMPs consisting of four gene members (mesothelin, protein kinase cAMP-dependent type II regulatory subunit beta, msh homeobox 1, and par-6 family cell polarity regulator alpha) that may have favorable prognostic and predictive values for platinum chemotherapy. Conclusion: The methylation-transcription axis exists and participates in the complex biological mechanism of platinum resistance in various cancers. Six DMPs and four associated genes may have the potential to serve as promising epigenetic biomarkers for platinum-based chemotherapy and guide clinical selection of optimal treatment.
Collapse
Affiliation(s)
- Ruizheng Sun
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Chao Du
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Jiaxin Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Juanjuan Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Jiheng Liu
- Department of Hematology and Oncology, The First Hospital of Changsha, Changsha, China
| | - Zhigang Xiao
- Department of General Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Li Fang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| |
Collapse
|
28
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 36:299-309. [PMID: 34773731 DOI: 10.1515/dmpt-2020-0186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- College of Pharmacy, Arab Academy for Science, Technology & Maritime Transport, Alexandria, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
29
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 0:dmdi-2020-0186. [PMID: 33818027 DOI: 10.1515/dmdi-2020-0186] [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: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
30
|
Wang X, Wei L, Yang J, Wang Y, Chen S, Yang K, Meng X, Zhang L. DNA methylation determines the regulation of pregnane X receptor on CYP3A4 expression. Clin Exp Pharmacol Physiol 2021; 48:250-259. [PMID: 33048369 DOI: 10.1111/1440-1681.13420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/22/2020] [Accepted: 10/07/2020] [Indexed: 11/30/2022]
Abstract
The expression and activity of CYP3A4 vary among individuals. With the development of epigenetics, it is now possible to elucidate interindividual differences in drug-metabolizing enzymes. Here, we aimed to explore the potential relationship between DNA methylation and CYP3A4 expression. We analyzed the effect of a DNA methylation inhibitor, 5-aza-2-deoxycytidine, on pregnane X receptor (PXR) and CYP3A4 expression in HepG2 cells. In addition, pCpGL-CYP3A4-promoter and pCpGL-CYP3A4-enhancer plus promoter plasmids were constructed, methylated, and transfected. We found that treatment with 5-aza-2-deoxycytidine significantly increased the expression of PXR and CYP3A4 in a concentration- and time-dependent manner. In addition, CYP3A4 expression was significantly enhanced by overexpressing PXR via transfection of pSG5-PXR plasmids. Methylation of CYP3A4 enhancer inhibited CYP3A4 transcriptional activity mediated through PXR and inhibited the binding of PXR and CYP3A4 promoter. We also observed that when the promoter and enhancer of CYP3A4 were methylated, CYP3A4 expression did not increase after treatment with rifampicin. In conclusion, the investigation demonstrates that DNA methylation of CYP3A4 enhancer significantly inhibits CYP3A4 expression, mediated through PXR, which is not influenced by rifampicin.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Luman Wei
- Department of Pharmacy, Zhengzhou People's Hospital, Zhengzhou, China
| | - Jingke Yang
- Laboratory of Cardiovascular Disease and Drug Research, The 7th People's Hospital of Zhengzhou, Zhengzhou, China
| | - Yiting Wang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Shitong Chen
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Kun Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Xiangguang Meng
- Laboratory of Cardiovascular Disease and Drug Research, The 7th People's Hospital of Zhengzhou, Zhengzhou, China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Yin J, Li F, Zhou Y, Mou M, Lu Y, Chen K, Xue J, Luo Y, Fu J, He X, Gao J, Zeng S, Yu L, Zhu F. INTEDE: interactome of drug-metabolizing enzymes. Nucleic Acids Res 2021; 49:D1233-D1243. [PMID: 33045737 PMCID: PMC7779056 DOI: 10.1093/nar/gkaa755] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Drug-metabolizing enzymes (DMEs) are critical determinant of drug safety and efficacy, and the interactome of DMEs has attracted extensive attention. There are 3 major interaction types in an interactome: microbiome-DME interaction (MICBIO), xenobiotics-DME interaction (XEOTIC) and host protein-DME interaction (HOSPPI). The interaction data of each type are essential for drug metabolism, and the collective consideration of multiple types has implication for the future practice of precision medicine. However, no database was designed to systematically provide the data of all types of DME interactions. Here, a database of the Interactome of Drug-Metabolizing Enzymes (INTEDE) was therefore constructed to offer these interaction data. First, 1047 unique DMEs (448 host and 599 microbial) were confirmed, for the first time, using their metabolizing drugs. Second, for these newly confirmed DMEs, all types of their interactions (3359 MICBIOs between 225 microbial species and 185 DMEs; 47 778 XEOTICs between 4150 xenobiotics and 501 DMEs; 7849 HOSPPIs between 565 human proteins and 566 DMEs) were comprehensively collected and then provided, which enabled the crosstalk analysis among multiple types. Because of the huge amount of accumulated data, the INTEDE made it possible to generalize key features for revealing disease etiology and optimizing clinical treatment. INTEDE is freely accessible at: https://idrblab.org/intede/.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kangli Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia Xue
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xu He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| |
Collapse
|
32
|
Okechukwu C. Deciphering and manipulating the epigenome for the treatment of Parkinson’s and Alzheimer’s disease. MGM JOURNAL OF MEDICAL SCIENCES 2021. [DOI: 10.4103/mgmj.mgmj_90_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
33
|
Huang L, Liao J, He J, Pan S, Zhang H, Yang X, Cheng J, Chen Y, Mo Z. Single-cell profiling reveals sex diversity in human renal proximal tubules. Gene 2020; 752:144790. [PMID: 32439376 DOI: 10.1016/j.gene.2020.144790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 01/11/2023]
Abstract
Many anatomical regions in the kidney, including proximal tubules, differ between males and females. While such differences in renal structures and functions under various physiological and pharmacological conditions have been identified, information relating to molecular mechanisms behind this gender disparity remain unknown. To understand gene expression differences in proximal tubules from human male and female kidneys, we reported on kidney cellular landscape using single-cell RNA sequencing. Differential gene expression profiles were observed in proximal tubules, between the sexes. Interestingly, the SLC22 family of anion transporters, including SLC22A6 and SLC22A8, had different expression profiles between male and female proximal tubule clusters but not sex-dependent abundance at the protein level. Moreover, in different species, we revealed a shared and species-specific differential gene expression between human and mouse kidney proximal tubules. Taken together, at single-cell resolution, this transcriptomic map represents a baseline description of gender biased genes in human kidney proximal tubules, which provide important insights for further studies of physiological differences in kidney.
Collapse
Affiliation(s)
- Lin Huang
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jinling Liao
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Juan He
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Siqiong Pan
- Department of Pathology, The Four Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiwen Cheng
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yang Chen
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Zengnan Mo
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
34
|
Yu D, Chen J, Chen S, Xu L, Wu L, Li D, Luo J, Jin Y, Zhao Y, Knox B, Tolleson WH, Wang X, Guo L, Tong W, Ning B. Coordinated Regulation of UGT2B15 Expression by Long Noncoding RNA LINC00574 and hsa-miR-129-5p in HepaRG Cells. Drug Metab Dispos 2020; 48:297-306. [PMID: 32086297 PMCID: PMC7153565 DOI: 10.1124/dmd.119.090043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that microRNAs and long noncoding RNAs (lncRNAs) regulate the expression of drug metabolizing enzymes (DMEs) in human hepatic cells and that a set of DMEs, including UDP glucuronosyltransferase (UGT) 2B15, is down-regulated dramatically in liver cells by toxic acetaminophen (APAP) concentrations. In this study we analyzed mRNA, microRNA, and lncRNA expression profiles in APAP-treated HepaRG cells to explore noncoding RNA-dependent regulation of DME expression. The expression of UGT2B15 and lncRNA LINC00574 was decreased in APAP-treated HepaRG cells. UGT2B15 levels were diminished by LINC00574 suppression using antisense oligonucleotides or small interfering RNA. Furthermore, we found that hsa-miR-129-5p suppressed LINC00574 and decreased UGT2B15 expression via LINC00574 in HepaRG cells. In conclusion, our results indicate that LINC00574 acts as an important regulator of UGT2B15 expression in human hepatic cells, providing experimental evidence and new clues to understand the role of cross-talk between noncoding RNAs. SIGNIFICANCE STATEMENT: We showed a molecular network that displays the cross-talk and consequences among mRNA, micro RNA, long noncoding RNA, and proteins in acetaminophen (APAP)-treated HepaRG cells. APAP treatment increased the level of hsa-miR-129-5p and decreased that of LINC00574, ultimately decreasing the production of UDP glucuronosyltransferase (UGT) 2B15. The proposed regulatory network suppresses UGT2B15 expression through interaction of hsa-miR-129-5p and LINC00574, which may be achieved potentially by recruiting RNA binding proteins.
Collapse
Affiliation(s)
- Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Jing Chen
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Si Chen
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Lin Xu
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Leihong Wu
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Dongying Li
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Yanjie Zhao
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Bridgett Knox
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - William H Tolleson
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Xubing Wang
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Lei Guo
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Weida Tong
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| | - Baitang Ning
- School of Public Health, Qingdao University, Qingdao, China; National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, Arkansas
| |
Collapse
|
35
|
Valodara AM, SR KJ. Sexual Dimorphism in Drug Metabolism and Pharmacokinetics. Curr Drug Metab 2020; 20:1154-1166. [DOI: 10.2174/1389200220666191021094906] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/27/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022]
Abstract
Background:Sex and gender-based differences are observed well beyond the sex organs and affect several physiological and biochemical processes involved in the metabolism of drug molecules. It is essential to understand not only the sex and gender-based differences in the metabolism of the drug but also the molecular mechanisms involved in the regulation of drug metabolism for avoiding sex-related adverse effects of drugs in the human.Method:The articles on the sex and gender-based differences in the metabolism of drug molecules were retrieved from the Pub Med database. The articles were classified into the metabolism of the drug molecule, gene expression regulation of drug-metabolizing enzymes, the effect of sex hormones on the metabolism of drug, expression of drugmetabolizing enzymes, etc.Result:Several drug molecules are known, which are metabolized differently in males and females. These differences in metabolism may be due to the genomic and non-genomic action of sex hormones. Several other drug molecules still require further evaluation at the molecular level regarding the sex and gender-based differences in their metabolism. Attention is also required at the effect of signaling cascades associated with the metabolism of drug molecules.Conclusion:Sex and gender-based differences in the metabolism of drugs exist at various levels and it may be due to the genomic and non-genomic action of sex hormones. Detailed understanding of the effect of sex and related condition on the metabolism of drug molecules will help clinicians to determine the effective therapeutic doses of drugs dependingon the condition of patient and disease.
Collapse
Affiliation(s)
- Askhi M. Valodara
- Department of Zoology, Biomedical Technology and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| | - Kaid Johar SR
- Department of Zoology, Biomedical Technology and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| |
Collapse
|
36
|
dos Santos PWDS, Machado ART, De Grandis RA, Ribeiro DL, Tuttis K, Morselli M, Aissa AF, Pellegrini M, Antunes LMG. Transcriptome and DNA methylation changes modulated by sulforaphane induce cell cycle arrest, apoptosis, DNA damage, and suppression of proliferation in human liver cancer cells. Food Chem Toxicol 2020; 136:111047. [DOI: 10.1016/j.fct.2019.111047] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/30/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
|
37
|
Sun J, Meng D, Yu T, Li F, Zhang G, Tian X, Zhao N, Li G, Li L, Wang H, Liu Y, Meng Y, Ma Y, Wan Z, Bao J, Piao H. N-terminal truncated carboxypeptidase E represses E-cadherin expression in lung cancer by stabilizing the Snail-HDAC complex. Am J Cancer Res 2020; 10:925-938. [PMID: 32266100 PMCID: PMC7136916 DOI: pmid/32266100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/16/2019] [Indexed: 02/05/2023] Open
Abstract
The N-terminal truncated carboxypeptidase E (CPEΔN) protein, an alternative splicing product of the carboxypeptidase E gene, has recently been recognized as an independent predictor for the recurrence and metastasis of lung adenocarcinoma. In this study, we showed that CPEΔN may accelerate lung cancer invasion via an E-cadherin-dependent mechanism. In vitro experiments and in vivo bioluminescence imaging assay revealed CPEΔN promoted the mobility and invasion of human lung cancer cells by suppressing endogenous expression of E-cadherin, a critical regulator for epithelial tissue homeostasis. Further mechanistic analyses revealed that CPEΔN directly interacted with and stabilized the Snail/HDAC1/HDAC3 complex within the promoter region of the E-cadherin-encoding CDH1 gene. CPEΔN overexpression led to a reduction of histone H3K9 acetylation and an increase of H3K9 and H3K27 trimethylation in the CHD1 gene promoter and ultimately inhibited E-cadherin transcription. In addition, correlations among CPEΔN, E-cadherin expression and tumor progression in 195 cases of lung adenocarcinoma patients were analyzed. Higher nuclear expression of CPEΔN was detected in patients with advanced stage of lung adenocarcinoma. Nuclear expression of CPEΔN was negatively correlated with the cell membrane expression of E-cadherin. Collectively, our findings illustrated that CPEΔN was involved in the transcriptional regulation of the epithelial-mesenchymal transition-related gene CDH1 and provide novel insights into CPEΔN-associated lung cancer metastasis.
Collapse
Affiliation(s)
- Jing Sun
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Dawei Meng
- Department of Otolaryngology Head and Neck Surgery, Liaoning Provincial Jinqiu HospitalShenyang 110016, Liaoning, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health of The PRC, China Medical UniversityShenyang 110122, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of The PRC, China Medical UniversityShenyang 110122, Liaoning, China
| | - Guirong Zhang
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Xin Tian
- Molecular Oncology Laboratory of Cancer Research Institute, China Medical University First HospitalShenyang 110001, Liaoning, China
| | - Nannan Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Guimin Li
- Department of Otolaryngology Head and Neck Surgery, Liaoning Provincial Jinqiu HospitalShenyang 110016, Liaoning, China
| | - Lu Li
- Research Center of Translational Medicine, The Second Affiliated Hospital of Shantou University Medical CollegeShantou 515000, Guangdong, China
| | - Hongyue Wang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Yeqiu Liu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Yiming Meng
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Yushu Ma
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Zhong Wan
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| | - Jimin Bao
- Department of Otolaryngology Head and Neck Surgery, Liaoning Provincial Jinqiu HospitalShenyang 110016, Liaoning, China
| | - Haozhe Piao
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & InstituteShenyang 110042, Liaoning, China
| |
Collapse
|
38
|
Protein Abundance of Clinically Relevant Drug Transporters in The Human Kidneys. Int J Mol Sci 2019; 20:ijms20215303. [PMID: 31653114 PMCID: PMC6862022 DOI: 10.3390/ijms20215303] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Renal drug transporters such as the organic cation transporters (OCTs), organic anion transporters (OATs) and multidrug resistance proteins (MRPs) play an important role in the tubular secretion of many drugs influencing their efficacy and safety. However, only little is known about the distinct protein abundance of these transporters in human kidneys, and about the impact of age and gender as potential factors of inter-subject variability in their expression and function. The aim of this study was to determine the protein abundance of MDR1, MRP1-4, BCRP, OAT1-3, OCT2-3, MATE1, PEPT1/2, and ORCTL2 by liquid chromatography-tandem mass spectrometry-based targeted proteomics in a set of 36 human cortex kidney samples (20 males, 16 females; median age 53 and 55 years, respectively). OAT1 and 3, OCT2 and ORCTL2 were found to be most abundant renal SLC transporters while MDR1, MRP1 and MRP4 were the dominating ABC transporters. Only the expression levels of MDR1 and ORCTL2 were significantly higher abundant in older donors. Moreover, we found several significant correlations between different transporters, which may indicate their functional interplay in renal vectorial transport processes. Our data may contribute to a better understanding of the molecular processes determining renal excretion of drugs.
Collapse
|
39
|
Transporters in the Mammary Gland-Contribution to Presence of Nutrients and Drugs into Milk. Nutrients 2019; 11:nu11102372. [PMID: 31590349 PMCID: PMC6836069 DOI: 10.3390/nu11102372] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
A large number of nutrients and bioactive ingredients found in milk play an important role in the nourishment of breast-fed infants and dairy consumers. Some of these ingredients include physiologically relevant compounds such as vitamins, peptides, neuroactive compounds and hormones. Conversely, milk may contain substances-drugs, pesticides, carcinogens, environmental pollutants-which have undesirable effects on health. The transfer of these compounds into milk is unavoidably linked to the function of transport proteins. Expression of transporters belonging to the ATP-binding cassette (ABC-) and Solute Carrier (SLC-) superfamilies varies with the lactation stages of the mammary gland. In particular, Organic Anion Transporting Polypeptides 1A2 (OATP1A2) and 2B1 (OATP2B1), Organic Cation Transporter 1 (OCT1), Novel Organic Cation Transporter 1 (OCTN1), Concentrative Nucleoside Transporters 1, 2 and 3 (CNT1, CNT2 and CNT3), Peptide Transporter 2 (PEPT2), Sodium-dependent Vitamin C Transporter 2 (SVCT2), Multidrug Resistance-associated Protein 5 (ABCC5) and Breast Cancer Resistance Protein (ABCG2) are highly induced during lactation. This review will focus on these transporters overexpressed during lactation and their role in the transfer of products into the milk, including both beneficial and harmful compounds. Furthermore, additional factors, such as regulation, polymorphisms or drug-drug interactions will be described.
Collapse
|
40
|
Yu L, Xie R, Tian T, Zheng L, Tang L, Cai S, Ma Z, Yang T, Han B, Yang Q. Suberoylanilide hydroxamic acid upregulates histone acetylation and activates endoplasmic reticulum stress to induce apoptosis in HepG2 liver cancer cells. Oncol Lett 2019; 18:3537-3544. [PMID: 31516571 PMCID: PMC6732958 DOI: 10.3892/ol.2019.10705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/26/2019] [Indexed: 01/07/2023] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor that has demonstrated clinical activity against various solid tumors. The aim of the present study was to explore the effects of SAHA on the apoptosis of HepG2 liver cancer cells, as well as the potential mechanisms involved in histone acetylation and endoplasmic reticulum (ER) stress. HepG2 cells were treated with various doses of SAHA (0, 1, 6 and 12 µM), and apoptosis was measured by flow cytometry. The levels of ER stress-associated molecules, including 78 kDa glucose-regulated protein (GRP78), PRKR-like endoplasmic reticulum kinase (PERK), phosphorylated (p)-PERK, activating transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP), were quantitated by western blot analysis and reverse transcription-quantitative PCR assay. The expression levels of acetylated histone H4 (acH4, acH4 lysine (K)5 and acH4K12) were detected by western blot analysis. The effects of SAHA on the acetylation of H4 in the promoter regions of GRP78, ATF4 and CHOP were evaluated by chromatin immunoprecipitation assays. Following treatment with higher doses of SAHA (6 and 12 µM) for 48 h, the proliferation of HepG2 cells was significantly suppressed. SAHA induced dose-dependent apoptosis and increased both protein and mRNA expression levels of GRP78, ATF4 and CHOP in HepG2 cells. The protein expression of PERK was markedly decreased by treatment with SAHA, whereas the p-PERK expression level was notably increased, which resulted in increased p-PERK/PERK ratio. Furthermore, the acetylation levels of H4 in the promoter regions of GRP78, ATF4 and CHOP were significantly increased in HepG2 cells exposed to 6 µM SAHA for 36 h. Thus, SAHA induces apoptosis in HepG2 cells by activating the ER stress-mediated apoptotic signaling pathway, at least partially by enhancing the acetylation of histone H4 on the promoter regions of ER-stress associated genes, including GRP78, ATF4 and CHOP.
Collapse
Affiliation(s)
- Lei Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathology, Hospital of Maternal and Childhood Health, Guiyang, Guizhou 550000, P.R. China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Tian Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lu Zheng
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lei Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Shuang Cai
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zihua Ma
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Qin Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
41
|
Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-Glycosyltransferase (UGT) Superfamily: New Members, New Functions, and Novel Paradigms. Physiol Rev 2019; 99:1153-1222. [DOI: 10.1152/physrev.00058.2017] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.
Collapse
Affiliation(s)
- Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A. McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Siti Nurul Mubarokah
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Alex Z. Haines
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Pramod C. Nair
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I. Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
42
|
Wang Y, Yan L, Liu J, Chen S, Liu G, Nie Y, Wang P, Yang W, Chen L, Zhong X, Han S, Zhang L. The HNF1 α-Regulated LncRNA HNF1 α-AS1 Is Involved in the Regulation of Cytochrome P450 Expression in Human Liver Tissues and Huh7 Cells. J Pharmacol Exp Ther 2019; 368:353-362. [PMID: 30602592 PMCID: PMC6367688 DOI: 10.1124/jpet.118.252940] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022] Open
Abstract
Expression of cytochrome P450s (P450s) is regulated by epigenetic factors, such as DNA methylation, histone modifications, and noncoding RNAs through different mechanisms. Among these factors, long noncoding RNAs (lncRNAs) have been shown to play important roles in the regulation of gene expression; however, little is known about the effects of lncRNAs on the regulation of P450 expression. The aim of this study was to explore the role of lncRNAs in the regulation of P450 expression by using human liver tissues and hepatoma Huh7 cells. Through lncRNA microarray analysis and quantitative polymerase chain reaction in human liver tissues, we found that the lncRNA hepatocyte nuclear factor 1 alpha antisense 1 (HNF1α-AS1), an antisense RNA of HNF1α, is positively correlated with the mRNA expression of CYP2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 as well as pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Gain- and loss-of-function studies in Huh7 cells transfected with small interfering RNAs or overexpression plasmids showed that HNF1α not only regulated the expression of HNF1α-AS1 and P450s, but also regulated the expression of CAR, PXR, and aryl hydrocarbon receptor (AhR). In turn, HNF1α-AS1 regulated the expression of PXR and most P450s without affecting the expression of HNF1α, AhR, and CAR. Moreover, the rifampicin-induced expression of P450s was also affected by HNF1α and HNF1α-AS1. In summary, the results of this study suggested that HNF1α-AS1 is involved in the HNF1α-mediated regulation of P450s in the liver at both basal and drug-induced levels.
Collapse
Affiliation(s)
- Yiting Wang
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Liang Yan
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Jingyang Liu
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Shitong Chen
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Guangming Liu
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Yali Nie
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Pei Wang
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Weihong Yang
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Liming Chen
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Xiaobo Zhong
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Shengna Han
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| | - Lirong Zhang
- Department of Pharmacology (Y.W., L.Y., J.L., S.C., G.L., Y.N., P.W., S.H., L.Z.) and Forensic Medicine (W.Y.), School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (L.Y.); and Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (L.C., X.Z.)
| |
Collapse
|
43
|
Ruoß M, Damm G, Vosough M, Ehret L, Grom-Baumgarten C, Petkov M, Naddalin S, Ladurner R, Seehofer D, Nussler A, Sajadian S. Epigenetic Modifications of the Liver Tumor Cell Line HepG2 Increase Their Drug Metabolic Capacity. Int J Mol Sci 2019; 20:347. [PMID: 30654452 PMCID: PMC6358789 DOI: 10.3390/ijms20020347] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 01/31/2023] Open
Abstract
Although human liver tumor cells have reduced metabolic functions as compared to primary human hepatocytes (PHH) they are widely used for pre-screening tests of drug metabolism and toxicity. The aim of the present study was to modify liver cancer cell lines in order to improve their drug-metabolizing activities towards PHH. It is well-known that epigenetics is strongly modified in tumor cells and that epigenetic regulators influence the expression and function of Cytochrome P450 (CYP) enzymes through altering crucial transcription factors responsible for drug-metabolizing enzymes. Therefore, we screened the epigenetic status of four different liver cancer cell lines (Huh7, HLE, HepG2 and AKN-1) which were reported to have metabolizing drug activities. Our results showed that HepG2 cells demonstrated the highest similarity compared to PHH. Thus, we modified the epigenetic status of HepG2 cells towards 'normal' liver cells by 5-Azacytidine (5-AZA) and Vitamin C exposure. Then, mRNA expression of Epithelial-mesenchymal transition (EMT) marker SNAIL and CYP enzymes were measured by PCR and determinate specific drug metabolites, associated with CYP enzymes by LC/MS. Our results demonstrated an epigenetic shift in HepG2 cells towards PHH after exposure to 5-AZA and Vitamin C which resulted in a higher expression and activity of specific drug metabolizing CYP enzymes. Finally, we observed that 5-AZA and Vitamin C led to an increased expression of Hepatocyte nuclear factor 4α (HNF4α) and E-Cadherin and a significant down regulation of Snail1 (SNAIL), the key transcriptional repressor of E-Cadherin. Our study shows, that certain phase I genes and their enzyme activities are increased by epigenetic modification in HepG2 cells with a concomitant reduction of EMT marker gene SNAIL. The enhancing of liver specific functions in hepatoma cells using epigenetic modifiers opens new opportunities for the usage of cell lines as a potential liver in vitro model for drug testing and development.
Collapse
Affiliation(s)
- Marc Ruoß
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany.
| | - Massoud Vosough
- Royan Institute for Stem Cell Biology and Technology, Department of Stem Cells and Developmental Biology, Tehran 16635-148, Iran.
| | - Lisa Ehret
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Carl Grom-Baumgarten
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Martin Petkov
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Silvio Naddalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany.
| | - Ruth Ladurner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany.
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, 04103 Leipzig, Germany.
| | - Andreas Nussler
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| | - Sahar Sajadian
- Siegfried Weller Institute, BG Trauma Clinic, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
44
|
Dietrich K, Baumgart J, Eshkind L, Reuter L, Gödtel-Armbrust U, Butt E, Musheev M, Marini F, More P, Grosser T, Niehrs C, Wojnowski L, Mathäs M. Health-Relevant Phenotypes in the Offspring of Mice Given CAR Activators Prior to Pregnancy. Drug Metab Dispos 2018; 46:1827-1835. [PMID: 30154105 DOI: 10.1124/dmd.118.082925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatic induction in response to drugs and environmental chemicals affects drug therapies and energy metabolism. We investigated whether the induction is transmitted to the offspring. We injected 3-day- and 6-week-old F0 female mice with TCPOBOP, an activator of the nuclear receptor constitutive androstane receptor (CAR, NR1I3), and mated them 1-6 weeks afterward. We detected in the offspring long-lasting alterations of CAR-mediated drug disposition, energy metabolism, and lipid profile. The transmission to the first filial generation (F1) was mediated by TCPOBOP transfer from the F0 adipose tissue via milk, as revealed by embryo transfer, crossfostering experiments, and liquid chromatography-mass spectrometry analyses. The important environmental pollutant PCB153 activated CAR in the F1 generation in a manner similar to TCPOBOP. Our findings indicate that chemicals accumulating and persisting in adipose tissue may exert liver-mediated, health-relevant effects on F1 offspring simply via physical transmission in milk. Such effects may occur even if treatment has been terminated far ahead of conception. This should be considered in assessing developmental toxicity and in the long-term follow-up of offspring of mothers exposed to both approved and investigational drugs, and to chemicals with known or suspected accumulation in adipose tissue.
Collapse
Affiliation(s)
- Karin Dietrich
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Jan Baumgart
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Leonid Eshkind
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Lea Reuter
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Ute Gödtel-Armbrust
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Elke Butt
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Michael Musheev
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Federico Marini
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Piyush More
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Tanja Grosser
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Christof Niehrs
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Leszek Wojnowski
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Marianne Mathäs
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| |
Collapse
|
45
|
Tian X, Wang C, Dong P, An Y, Zhao X, Jiang W, Wang G, Hou J, Feng L, Wang Y, Ge G, Huo X, Ning J, Ma X. Arenobufagin is a novel isoform-specific probe for sensing human sulfotransferase 2A1. Acta Pharm Sin B 2018; 8:784-794. [PMID: 30245965 PMCID: PMC6146385 DOI: 10.1016/j.apsb.2018.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/20/2018] [Accepted: 06/01/2018] [Indexed: 02/06/2023] Open
Abstract
Human cytosolic sulfotransferase 2A1 (SULT2A1) is an important phase II metabolic enzyme. The detection of SULT2A1 is helpful for the functional characterization of SULT2A1 and diagnosis of its related diseases. However, due to the overlapping substrate specificity among members of the sulfotransferase family, it is difficult to develop a probe substrate for selective detection of SULT2A1. In the present study, through characterization of the sulfation of series of bufadienolides, arenobufagin (AB) was proved as a potential probe substrate for SULT2A1 with high sensitivity and specificity. Subsequently, the sulfation of AB was characterized by experimental and molecular docking studies. The sulfate-conjugated metabolite was identified as AB-3-sulfate. The sulfation of AB displayed a high selectivity for SULT2A1 which was confirmed by in vitro reaction phenotyping assays. The sulfation of AB by human liver cytosols and recombinant SULT2A1 both obeyed Michaelis-Menten kinetics, with similar kinetic parameters. Molecular docking was performed to understand the interaction between AB and SULT2A1, in which the lack of interaction with Met-137 and Tyr-238 of SULT2A1 made it possible to eliminate substrate inhibition of AB sulfation. Finally, the probe was successfully used to determine the activity of SULT2A1 and its isoenzymes in tissue preparations of human and laboratory animals.
Collapse
Affiliation(s)
- Xiangge Tian
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
- Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Chao Wang
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Peipei Dong
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Yue An
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Xinyu Zhao
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Weiru Jiang
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Gang Wang
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Jie Hou
- Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Lei Feng
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
- Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Yan Wang
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Guangbo Ge
- Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China
| | - Xiaokui Huo
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Jing Ning
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
- Corresponding author at: College of Pharmacy, Research Institute of Integrated Traditional and Western Medicine, Dalian Medical University, Western 9 Lvshun South Road, Dalian 116044, China. Tel./fax: +86 411 86110419.
| | - Xiaochi Ma
- Academy of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Second Affliated Hospital, Dalian Medical University, Dalian 116044, China
- Institute of Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
- Corresponding author at: College of Pharmacy, Research Institute of Integrated Traditional and Western Medicine, Dalian Medical University, Western 9 Lvshun South Road, Dalian 116044, China. Tel./fax: +86 411 86110419.
| |
Collapse
|
46
|
Nie YL, Meng XG, Liu JY, Yan L, Wang P, Bi HZ, Kan QC, Zhang LR. Histone Modifications Regulate the Developmental Expression of Human Hepatic UDP-Glucuronosyltransferase 1A1. Drug Metab Dispos 2017; 45:1372-1378. [PMID: 29025858 DOI: 10.1124/dmd.117.076109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/29/2017] [Indexed: 12/23/2022] Open
Abstract
Human UDP-glucuronosyltransferase 1A1 (UGT1A1) is a unique enzyme involved in bilirubin conjugation. We previously characterized the hepatic expression of transcription factors affecting UGT1A1 expression during development. Accordingly, in this study, we characterized the ontogenetic expression of hepatic UGT1A1 from the perspective of epigenetic regulation. We observed significant histone-3-lysine-4 dimethylation (H3K4me2) enrichment in the adult liver and histone-3-lysine-27 trimethylation (H3K27me3) enrichment in the fetal liver, indicating that dynamic alterations of histone methylation were associated with ontogenetic UGT1A1 expression. We further showed that the transcription factor hepatocyte nuclear factor 1α (HNF1A) affects histone modifications around the UGT1A1 locus. In particular, we demonstrated that by recruiting HNF1A the cofactors mixed-lineage leukemia 1, the transcriptional coactivator p300, and nuclear receptor coactivator 6 aggregate at the UGT1A1 promoter, thereby regulating histone modifications and subsequent UGT1A1 expression. In this study, we proposed new ideas for the developmental regulation of metabolic enzymes via histone modifications, and our findings will potentially contribute to the development of age-specific therapies.
Collapse
Affiliation(s)
- Ya-Li Nie
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Xiang-Guang Meng
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Jing-Yang Liu
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Liang Yan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Pei Wang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Hong-Zheng Bi
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Quan-Cheng Kan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University (Y.-L.N., J.-Y.L., L.Y., P.W., H.-Z.B., L.-R.Z.); Laboratory of Cardiovascular Disease and Drug Research, Seventh People's Hospital of Zhengzhou (X.-G.M.); Department of Clinical Pharmacology, First Affiliated Hospital, Zhengzhou University (Q.-C.K.), Zhengzhou, People's Republic of China
| |
Collapse
|
47
|
Awortwe C, Kaehler M, Rosenkranz B, Cascorbi I, Bruckmueller H. MicroRNA-655-3p regulates Echinacea purpurea mediated activation of ABCG2. Xenobiotica 2017; 48:1050-1058. [PMID: 28990842 DOI: 10.1080/00498254.2017.1390624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
1. The aim of this study was to investigate the regulatory effect of Echinacea purpurea (EP) on efflux transporters ABCB1 and ABCG2 and to identify specific microRNAs contributing to their post-transcriptional regulation. 2. ABCB1 and ABCG2 levels were assessed in human hepatoblastoma HepG2 cells treated with 50 µg/mL methanolic extract of commercial EP capsules for different durations. The microRNA expression profile of HepG2 cells after EP treatment was evaluated and in silico target prediction was subsequently conducted to identify specific microRNAs with binding sites in the 3'-UTR of ABCB1 and ABCG2. Luciferase reporter gene assays and site-directed mutagenesis were used to confirm the binding site of identified microRNA within the 3'-UTR of the target gene. 3. EP increased ABCB1 (10-fold ± 3.4, p < 0.001) and ABCG2 (2.7-fold ± 0.5, p < 0.01) mRNA levels after 12 h exposure. Twenty-four microRNAs showed significant expression differences at all durations of exposure to EP. MiR-655-3p showed a 6.79-fold decrease in expression after 12 h exposure compared to 0 h, was predicted in silico to bind ABCG2 3'-UTR and showed a significant negative correlation (p = 0.01) to ABCG2 expression level. The binding of miR-655-3p to ABCG2 3'-UTR was confirmed by reporter gene assays (reduction of reporter gene activity to 60%; p = 0.0001). 4. These results suggest that EP regulates ABCG2 expression via downregulation of miR-655-3p in the liver cells. Thus, miR-655-3p downregulation could be applied to predict EP mediated drug interactions.
Collapse
Affiliation(s)
- Charles Awortwe
- a Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, University of Stellenbosch , Tygerberg , South Africa.,b Institute for Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein , Kiel , Germany , and.,c Biomedical Research and Innovation Platform, South African Medical Research Council , Tygerberg , South Africa
| | - Meike Kaehler
- b Institute for Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein , Kiel , Germany , and
| | - Bernd Rosenkranz
- a Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, University of Stellenbosch , Tygerberg , South Africa
| | - Ingolf Cascorbi
- b Institute for Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein , Kiel , Germany , and
| | - Henrike Bruckmueller
- b Institute for Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein , Kiel , Germany , and
| |
Collapse
|
48
|
Yan L, Wang Y, Liu J, Nie Y, Zhong XB, Kan Q, Zhang L. Alterations of Histone Modifications Contribute to Pregnane X Receptor-Mediated Induction of CYP3A4 by Rifampicin. Mol Pharmacol 2017; 92:113-123. [PMID: 28546420 PMCID: PMC5508193 DOI: 10.1124/mol.117.108225] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/22/2017] [Indexed: 01/28/2023] Open
Abstract
CYP3A4 is one of the major drug-metabolizing enzymes in human and is responsible for the metabolism of 60% of clinically used drugs. Many drugs are able to induce the expression of CYP3A4, which usually causes drug-drug interactions and adverse drug reactions. This study aims to explore the role of histone modifications in rifampicin-induced expression of CYP3A4 in LS174T cells. We found that the induction of CYP3A4 mRNA (4- to 15-fold) by rifampicin in LS174T cells was associated with increased levels of histone H3 lysine 4 trimethylation (H3K4me3, above 1.8-fold) and H3 acetylation (above 2-fold) and a decreased level of histone H3 lysine 27 trimethylation (H3K27me3, about 50%) in the CYP3A4 promoter. Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Silencing NCOA6 or p300 by short-hairpin RNAs resulted in inhibition of the CYP3A4 induction as well as altered levels of H3K4me3, H3K27me3, or H3 acetylation in the CYP3A4 promoter. Knockdown of pregnane X receptor (PXR) expression not only suppressed the recruitment of NCOA6 and p300 but also abolished the changes caused by rifampicin in H3K4me3, H3K27me3, and H3 acetylation levels in the CYP3A4 promoter. Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. In conclusion, we show that the alterations of histone modifications contribute to the PXR-mediated induction of CYP3A4 by rifampicin.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yiting Wang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Jingyang Liu
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Yali Nie
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Xiao-Bo Zhong
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Quancheng Kan
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| | - Lirong Zhang
- Department of Pharmacology (L.Y., J.L., Y.N, L.Z.) and Department of Forensic Medicine (Y.W.), School of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut (X.Z.); The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (Q.K.)
| |
Collapse
|
49
|
Yu AM, Ingelman-Sundberg M, Cherrington NJ, Aleksunes LM, Zanger UM, Xie W, Jeong H, Morgan ET, Turnbaugh PJ, Klaassen CD, Bhatt AP, Redinbo MR, Hao P, Waxman DJ, Wang L, Zhong XB. Regulation of drug metabolism and toxicity by multiple factors of genetics, epigenetics, lncRNAs, gut microbiota, and diseases: a meeting report of the 21 st International Symposium on Microsomes and Drug Oxidations (MDO). Acta Pharm Sin B 2017; 7:241-248. [PMID: 28388695 PMCID: PMC5343155 DOI: 10.1016/j.apsb.2016.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 01/16/2023] Open
Abstract
Variations in drug metabolism may alter drug efficacy and cause toxicity; better understanding of the mechanisms and risks shall help to practice precision medicine. At the 21st International Symposium on Microsomes and Drug Oxidations held in Davis, California, USA, in October 2-6, 2016, a number of speakers reported some new findings and ongoing studies on the regulation mechanisms behind variable drug metabolism and toxicity, and discussed potential implications to personalized medications. A considerably insightful overview was provided on genetic and epigenetic regulation of gene expression involved in drug absorption, distribution, metabolism, and excretion (ADME) and drug response. Altered drug metabolism and disposition as well as molecular mechanisms among diseased and special populations were presented. In addition, the roles of gut microbiota in drug metabolism and toxicology as well as long non-coding RNAs in liver functions and diseases were discussed. These findings may offer new insights into improved understanding of ADME regulatory mechanisms and advance drug metabolism research.
Collapse
|
50
|
Dixit VA, Deshpande S. Advances in Computational Prediction of Regioselective and Isoform-Specific Drug Metabolism Catalyzed by CYP450s. ChemistrySelect 2016. [DOI: 10.1002/slct.201601051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| | - Shirish Deshpande
- Department of Pharmaceutical Chemistry; School of Pharmacy and Technology Management (SPTM), Shri Vile Parle Kelavani Mandal's (SVKM's) Narsee Monjee Institute of Management Studies (NMIMS), Mukesh Patel Technology Park, Babulde, Bank of Tapi River; Mumbai-Agra Road Shirpur, Dist. Dhule−425405 India
| |
Collapse
|