1
|
Zimmer AA, Collier AC. Scaling factors to inform in vitro- in vivo extrapolation from preclinical and livestock animals: state of the field and recommendations for development of missing data. Drug Metab Rev 2025; 57:91-114. [PMID: 39898873 DOI: 10.1080/03602532.2025.2462527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
The use of in-vitro-in-vivo physiologically based pharmacokinetic (IVIVE-PBPK) modeling approaches assists for prediction of first-in animal or human trials. These approaches are underpinned by the scaling factors: microsomal protein per gram (MPPG) and cytosolic protein per gram (CPPG). In addition, IVIVE-PBPK has significant application in the reduction and refinement of live animal models in research. While human scaling factors are well-defined, many preclinical and livestock species remain poorly elucidated or uncharacterized. The MPPG parameter for liver (MPPGL) is the best characterized across all species and is well-defined for mouse, rat, and dog models. The MPPG parameters for Kidney (MPPGK) and intestine (MPPGI), are however; relatively indefinite for most species. Similarly, CPPG scaling factors for liver, kidney, and intestine (CPPGL/CPPGK/CPPGI) are generally sparse in all species. In addition to generation of mathematical values for scaling factors, methodological and animal-specific considerations, such as age, sex, and strain differences, have not yet been comprehensively described. Here, we review the current state-of-the-field for microsomal and cytosolic scaling factors, including highlighting areas that may need further description and development, with the intention of drawing attention to key knowledge gaps. The intention is to promote improved accuracy and precision in IVIVE-PBPK, concordance between laboratories, and stimulate work in underserved, but increasingly vital areas.
Collapse
Affiliation(s)
- Austin A Zimmer
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
- Prostate Cancer Foundation Canada, Surrey, Canada
| | - Abby C Collier
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
- Prostate Cancer Foundation Canada, Surrey, Canada
| |
Collapse
|
2
|
Ewert W, Bartens C, Ongouta J, Holmes M, Heutling A, Kishore A, Urbansky T, Zeilinger C, Preller M, Kirschning A. Structure and function of the geldanamycin amide synthase from Streptomyces hygroscopicus. Nat Commun 2025; 16:2464. [PMID: 40075103 PMCID: PMC11903869 DOI: 10.1038/s41467-025-57013-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Amide synthases catalyze the formation of macrolactam rings from aniline-containing polyketide-derived seco-acids as found in the important class of ansamycin antibiotics. One of these amide synthases is the geldanamycin amide synthase GdmF, which we recombinantly expressed, purified and studied in detail both functionally as well as structurally. Here we show that purified GdmF catalyzes the amide formation using synthetically derived substrates. The atomic structures of the ligand-free enzyme and in complex with simplified substrates reveal distinct structural features of the substrate binding site and a putative role of the flexible interdomain region for the catalysis reaction.
Collapse
Affiliation(s)
- Wiebke Ewert
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Christian Bartens
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Jekaterina Ongouta
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Monika Holmes
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anja Heutling
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anusha Kishore
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany
| | - Tim Urbansky
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany
| | - Carsten Zeilinger
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany.
| | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany.
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany.
- Uppsala Biomedical Center (BMC), University Uppsala, Uppsala, Sweden.
| |
Collapse
|
3
|
Zhang XT, Blacutt J, Lloyd T, Mencer M, Pratt V, Kotha J, Sheeran L, Adcock S. Enhancing clinical research with pharmacogenomics: a practical perspective. Bioanalysis 2025; 17:399-411. [PMID: 40118816 PMCID: PMC11970788 DOI: 10.1080/17576180.2025.2481019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
Pharmacogenomics (PGx) is transforming therapeutic development by providing insights into how genetic variations influence drug response, safety, and efficacy. This review provides a structured analysis of PGx in clinical research, beginning with an overview of key genes involved in drug metabolism, transport, and targets. Following this, it examines strategies for identifying PGx-relevant genes, including phenotype-driven, hypothesis-driven, population-focused, and clinical-driven approaches. Technical platforms such as PCR, MassARRAY, and next-generation sequencing are analyzed for their suitability in PGx studies. The discussion then shifts to assay validation processes, covering both analytical and clinical validation, to ensure data reliability in clinical trials. Finally, regulatory expectations for PGx in clinical trials are discussed, focusing on key requirements across all phases of drug development. This review aims to provide a clear and practical framework for integrating PGx into clinical research to enhance drug safety, efficacy, and personalized medicine.
Collapse
Affiliation(s)
| | - Jacob Blacutt
- Early Phase Unit, Worldwide Clinical Trials, Austin, TX, USA
| | - Thomas Lloyd
- Early Phase Unit, Worldwide Clinical Trials, Austin, TX, USA
| | - Mike Mencer
- Early Phase Unit, Worldwide Clinical Trials, Austin, TX, USA
| | - Vicky Pratt
- Pharmacogenetics, Agena Bioscience, San Diego, CA, USA
| | | | - Lona Sheeran
- Early Phase Unit, Worldwide Clinical Trials, Austin, TX, USA
| | - Sherilyn Adcock
- Early Phase Unit, Worldwide Clinical Trials, Austin, TX, USA
| |
Collapse
|
4
|
Oladipupo I, Ali T, Hein DW, Pagidas K, Bohler H, Doll MA, Mann ML, Gentry A, Chiang JL, Pierson RC, Torres S, Reece E, Taylor KC. Association between cigarette smoking and ovarian reserve among women seeking fertility care. PLoS One 2022; 17:e0278998. [PMID: 36512605 PMCID: PMC9746951 DOI: 10.1371/journal.pone.0278998] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION This study examined the association of smoking with ovarian reserve in a cross-sectional study of 207 women enrolled in the Louisville Tobacco Smoke Exposure, Genetic Susceptibility, and Infertility (LOUSSI) Study and assessed effect modification by NAT2 acetylator phenotype. METHODS Information on current smoking status was collected using a structured questionnaire and confirmed by cotinine assay. Serum anti-Müllerian hormone (AMH) levels were used to assess ovarian reserve. Diminished ovarian reserve (DOR) was defined as AMH <1ng/mL. Single nucleotide polymorphisms in the NAT2 gene, which metabolizes toxins found in cigarette smoke, were analyzed to determine NAT2 acetylator status. Linear and logistic regression were used to determine the effects of smoking on ovarian reserve and evaluate effect modification by NAT2. Regression analyses were stratified by polycystic ovary syndrome (PCOS) status and adjusted for age. RESULTS Current smoking status, either passive or active as measured by urinary cotinine assay, was not significantly associated with DOR. For dose-response assessed using self-report, the odds of DOR increased significantly for every additional cigarette currently smoked (Odds ratio, OR:1.08; 95% confidence interval, 95%CI:1.01-1.15); additionally, every 1 pack-year increase in lifetime exposure was associated with an increased odds of DOR among women without PCOS (OR: 1.08 95%CI: 0.99-1.18). These trends appear to be driven by the heavy or long-term smokers. Effect modification by NAT2 genotype was not established. CONCLUSION A history of heavy smoking may indicate increased risk of diminished ovarian reserve.
Collapse
Affiliation(s)
- Islamiat Oladipupo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States of America
| | - T’shura Ali
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States of America
| | - David W. Hein
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Kelly Pagidas
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Henry Bohler
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Mark A. Doll
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Merry Lynn Mann
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Adrienne Gentry
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Jasmine L. Chiang
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Rebecca C. Pierson
- Department of Obstetrics, Gynecology, and Women’s Health, Division of Reproductive Endocrinology, and Infertility, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Sashia Torres
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States of America
| | - Emily Reece
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States of America
| | - Kira C. Taylor
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, United States of America
| |
Collapse
|
5
|
Habil MR, Doll MA, Hein DW. Acetyl coenzyme A kinetic studies on N-acetylation of environmental carcinogens by human N-acetyltransferase 1 and its NAT1*14B variant. Front Pharmacol 2022; 13:931323. [PMID: 36386142 PMCID: PMC9650386 DOI: 10.3389/fphar.2022.931323] [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: 04/28/2022] [Accepted: 09/30/2022] [Indexed: 01/11/2023] Open
Abstract
N-acetyltransferase 1 (NAT1) is a xenobiotic metabolizing enzyme that uses acetyl coenzyme A (AcCoA) as a cofactor for N-acetylation of many carcinogens including aromatic amines and alkylanilines. NAT1 is characterized by single nucleotide polymorphisms (SNPs) that may modulate affinity towards AcCoA. In the current study, we used Chinese hamster ovary (CHO) cells stably transfected with human NAT1*4 (reference allele) or NAT1*14B (variant allele) to measure AcCoA kinetic parameters for N-acetyltransferase activity measurements towards p-aminobenzoic acid (PABA), 4-aminobiphenyl (4-ABP), β-naphthylamine (BNA), benzidine and 3,4-dimethylaniline (3,4-DMA). Our results showed higher N-acetylation rates for each substrate catalyzed by NAT1*4 compared to NAT1*14B. NAT1*4 exhibited higher affinity to AcCoA when catalyzing the N-acetylation of BNA and benzidine compared to NAT1*14B. The results of the current study provide further insights into differences in carcinogen metabolism among individuals possessing the NAT1*14B haplotype.
Collapse
|
6
|
Yu L, Zhai J, Wang Y, Geng Y, Chen X, Wen Y, Tang H, Yu R, Zhang Y, Liu X. Exposure to N-monoacetyl-p-phenylenediamine impaired ovarian function in mice. J Appl Toxicol 2021; 41:2031-2041. [PMID: 34014586 DOI: 10.1002/jat.4183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/14/2021] [Accepted: 04/29/2021] [Indexed: 01/14/2023]
Abstract
p-Phenylenediamine (PPD) is the main constituent of permanent hair dye and is also widely used in the photographic and rubber industries. PPD and its metabolites have been shown to increase the risk of cancer (especially ovarian cancer); however, their effect on female reproduction is unclear. We investigated the effects of the PPD metabolite N-monoacetyl-PPD (MAPPD) on mouse blastocyst development and ovarian function. Sixty 8-week-old female Kunming mice were administered at 0-, 100-, and 300-mg/kg/day MPPD by gavage for 28 days. KGN (human ovarian granulosa cells) were treated with MAPPD at concentrations of 0, 50, 100, and 300 μg/ml for 48 h. The number of abnormal blastocysts increased on gestation day 3.5 in all treatment groups. Compared with the control group, in MAPPD exposed group, the number of antral follicles decreased, the levels of E2 and P4 decreased in ovarian tissue, the serum levels of E2 , P4 , luteinizing hormone (LH), and T decreased, and follicle-stimulating hormone (FSH) increased. The expression of FSH receptor (FSHR) and LH receptor (LHR) was significantly downregulated, and the level of oxidative stress was significantly increased. In KGN cells, the level of reactive oxygen species increased in a dose-dependent manner, and the mRNA levels of FSHR, LHR, and aromatase increased. These results suggest that MAPPD inhibits FSH- and LH-induced aromatase activity by causing oxidative stress, which decrease hormone levels, leading to abnormal follicle development. Meanwhile, MAPPD exposure could affect early embryonic development abnormalities by affecting the quality of ovum.
Collapse
Affiliation(s)
- Liliang Yu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jingwei Zhai
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yixian Wen
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Hongyu Tang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Rao Yu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yue Zhang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xueqing Liu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Liu X, Liu Y, Zhao G, Zhang Y, Liu L, Wang J, Wang Y, Zhang S, Li X, Guo D, Wang P, Xu X. Biochemical Characterization of Arylamine N-acetyltransferases From Vibrio vulnificus. Front Microbiol 2021; 11:595083. [PMID: 33537010 PMCID: PMC7847940 DOI: 10.3389/fmicb.2020.595083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/09/2020] [Indexed: 12/03/2022] Open
Abstract
Vibrio vulnificus is a zoonotic bacterium that is capable of causing highly lethal diseases in humans; this pathogen is responsible for 95% of all seafood-related deaths in the United States. Arylamine N-acetyltransferases (NAT, E.C. 2.3.1.5) is a major family of xenobiotic-metabolizing enzymes that can biotransform aromatic amine chemicals. In this research, to evaluate the effect of NAT on acetyl group transformation in arylamine antibiotics, we first used sequence alignment to study the structure of V. vulnificus NAT [(VIBVN)NAT]. The nat gene encodes a protein of 260 amino acids, which has an approximate molecular mass of 30 kDa. Then we purified recombinant (VIBVN)NAT and determined the enzyme activity by PNPA and DTNB methods. The DTNB method indicates that this prokaryotic NAT has a particular substrate specificity towards aromatic substrates. However, (VIBVN)NAT lost most of its activity after treatment with high concentrations of urea and H2O2. In addition, we also explored the stability of the enzyme at different temperatures and pH values. In analyzing the influence of metal ions, the enzyme activity was significantly inhibited by Zn2+ and Cu2+. The kinetic parameters Km and Vmax were determined using hydralazine, isoniazid, 4-amino salicylic acid, and 4-chloro-3-methylaniline as substrates, and the Tm, Tagg and size distribution of (VIBVN)NAT were observed. In particular, a molecular docking study on the structure of (VIBVN)NAT was conducted to understand its biochemical traits. These results showed that (VIBVN)NAT could acetylate various aromatic amine substrates and contribute to arylamine antibiotic resistance in V. vulnificus.
Collapse
Affiliation(s)
- Xinning Liu
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.,Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, China
| | - Yuanchang Liu
- Quality Control Department, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Guangjian Zhao
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yidan Zhang
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lu Liu
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Juan Wang
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yifan Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Siyu Zhang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Xin Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Dongliang Guo
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Peng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Ximing Xu
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.,Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, China
| |
Collapse
|
8
|
Abstract
Drug metabolizing enzymes catalyze the biotransformation of many of drugs and chemicals. The drug metabolizing enzymes are distributed among several evolutionary families and catalyze a range of detoxication reactions, including oxidation/reduction, conjugative, and hydrolytic reactions that serve to detoxify potentially toxic compounds. This detoxication function requires that drug metabolizing enzymes exhibit substrate promiscuity. In addition to their catalytic functions, many drug metabolizing enzymes possess functions unrelated to or in addition to catalysis. Such proteins are termed 'moonlighting proteins' and are defined as proteins with multiple biochemical or biophysical functions that reside in a single protein. This review discusses the diverse moonlighting functions of drug metabolizing enzymes and the roles they play in physiological functions relating to reproduction, vision, cell signaling, cancer, and transport. Further research will likely reveal new examples of moonlighting functions of drug metabolizing enzymes.
Collapse
Affiliation(s)
- Philip G Board
- John Curtin School of Medical Research, ANU College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, New York, NY, USA
| |
Collapse
|
9
|
Pandi S, Chinniah R, Sevak V, Ravi PM, Vijayan M, Vellaiappan NA, Karuppiah B. Association of slow acetylator genotype of N-acetyltransferase 2 with Parkinson's disease in south Indian population. Neurosci Lett 2020; 735:135260. [PMID: 32682841 DOI: 10.1016/j.neulet.2020.135260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
AIM Parkinson's Disease (PD) is a neurodegenerative disorder with predisposing genetic and environmental factors. The present study was undertaken to elucidate the possible association of NAT2 gene polymorphism in PD patients from south India. METHODS Using previously validated PCR-RFLP assays, we genotyped 105 PD subjects and 101 healthy controls for N-acetyl transferase (NAT2) gene polymorphism. RESULTS We observed a significantly elevated frequencies of NAT2 *5/6 (OR = 4.21; p < 0.029) and *5/7 (OR = 2.73; p < 0.025) genotypes and NAT2*5 (OR = 1.83; p < 0.039) allele among PD cases showing susceptible associations. The age at onset analysis revealed a significant association of NAT2 *4/6 (OR = 4.62; p < 0.05) genotype with early onset PD (EOPD). A positive association with early onset disease was observed for *5/7 (OR = 3.88; p < 0.075) genotype, however without statistical significance. Whereas, in late onset PD (LOPD) cases, significant susceptible association was observed for NAT2 *5/7 (OR = 5.27; p < 0.029) genotype. We observed a highly significant protective association of NAT2 *4/6 (OR = 0.27; p < 0.012) genotype and NAT2 *4 (OR = 0.52; p < 0.027) allele with LOPD. The acetylator status phenotype analysis have revealed a higher risk for, 'NAT2 slow acetylator' in both overall PD (OR = 2.39; p < 0.002) and LOPD (OR = 2.88; p < 0.007). However, 'NAT2 intermediate acetylator' with a lower risk in both overall PD (OR = 0.47; p < 0.011) and LOPD (OR = 0.36; p < 0.007) cases revealed protective associations. CONCLUSIONS Thus, our results revealed the possible susceptible association of NAT2 slow acetylator in PD pathogenesis in south Indian population.
Collapse
Affiliation(s)
- Sasiharan Pandi
- Department of Immunology, School of Biological Sciences, Madurai, 625021, Tamil Nadu, India
| | - Rathika Chinniah
- Department of Immunology, School of Biological Sciences, Madurai, 625021, Tamil Nadu, India
| | - Vandit Sevak
- Department of Immunology, School of Biological Sciences, Madurai, 625021, Tamil Nadu, India
| | - Padma Malini Ravi
- Department of Immunology, School of Biological Sciences, Madurai, 625021, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | | | - Balakrishnan Karuppiah
- Department of Immunology, School of Biological Sciences, Madurai, 625021, Tamil Nadu, India.
| |
Collapse
|
10
|
Utility of Common Marmoset ( Callithrix jacchus) Embryonic Stem Cells in Liver Disease Modeling, Tissue Engineering and Drug Metabolism. Genes (Basel) 2020; 11:genes11070729. [PMID: 32630053 PMCID: PMC7397002 DOI: 10.3390/genes11070729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
The incidence of liver disease is increasing significantly worldwide and, as a result, there is a pressing need to develop new technologies and applications for end-stage liver diseases. For many of them, orthotopic liver transplantation is the only viable therapeutic option. Stem cells that are capable of differentiating into all liver cell types and could closely mimic human liver disease are extremely valuable for disease modeling, tissue regeneration and repair, and for drug metabolism studies to develop novel therapeutic treatments. Despite the extensive research efforts, positive results from rodent models have not translated meaningfully into realistic preclinical models and therapies. The common marmoset Callithrix jacchus has emerged as a viable non-human primate model to study various human diseases because of its distinct features and close physiologic, genetic and metabolic similarities to humans. C. jacchus embryonic stem cells (cjESC) and recently generated cjESC-derived hepatocyte-like cells (cjESC-HLCs) could fill the gaps in disease modeling, liver regeneration and metabolic studies. They are extremely useful for cell therapy to regenerate and repair damaged liver tissues in vivo as they could efficiently engraft into the liver parenchyma. For in vitro studies, they would be advantageous for drug design and metabolism in developing novel drugs and cell-based therapies. Specifically, they express both phase I and II metabolic enzymes that share similar substrate specificities, inhibition and induction characteristics, and drug metabolism as their human counterparts. In addition, cjESCs and cjESC-HLCs are advantageous for investigations on emerging research areas, including blastocyst complementation to generate entire livers, and bioengineering of discarded livers to regenerate whole livers for transplantation.
Collapse
|
11
|
Xu X, Zhang W, Berthelet J, Liu R, Michail C, Chaffotte AF, Dupret JM, Rodrigues-Lima F. From transglutaminases (TGs) to arylamine N-acetyltransferases (NATs): Insight into the role of a spatially conserved aromatic amino acid position in the active site of these two families of enzymes. Biochem Biophys Res Commun 2020; 525:308-312. [PMID: 32089267 DOI: 10.1016/j.bbrc.2020.02.082] [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: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 11/29/2022]
Abstract
Transglutaminases (TG) and arylamine N-acetyltransferases (NAT) are important family of enzymes. Although they catalyze different reactions and have distinct structures, these two families of enzymes share a spatially conserved catalytic triad (Cys, His, Asp residues). In active TGs, a conserved Trp residue located close to the triad cysteine is crucial for catalysis through stabilization of transition states. Here, we show that in addition to sharing a similar catalytic triad with TGs, functional NAT enzymes also possess in their active site an aromatic residue (Phe, Tyr or Trp) occupying a structural position similar to the Trp residue of active TGs. More importantly, as observed in active TGs, our data indicates that in functional NAT enzymes this conserved aromatic residue is also involved in stabilization of transition states. These results thus indicate that in addition to the three triad residues, these two families of enzymes also share a spatially conserved aromatic amino acid position important for catalysis. Identification of residues involved in the stabilization of transition states is important to develop potent inhibitors. Interestingly, NAT enzymes have been shown as potential targets of clinical interest.
Collapse
Affiliation(s)
- Ximing Xu
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France; Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Wenchao Zhang
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | | | - Rongxing Liu
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | | | - Alain F Chaffotte
- Institut Pasteur, Unité de Résonance Magnétique Nucléaire des Biomolécules, 75015, Paris, France
| | | | | |
Collapse
|
12
|
Rehman AU, Rahman MU, Lu S, Liu H, Li JY, Arshad T, Wadood A, Ng HL, Chen HF. Decoding allosteric communication pathways in protein lysine acetyltransferase. Int J Biol Macromol 2020; 149:70-80. [DOI: 10.1016/j.ijbiomac.2020.01.213] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
|
13
|
Zhu X, Liu Y, Chen G, Guo Q, Zhang Z, Zhao L, Wei R, Yin X, Zhang Y, Wang B, Li X. Association between NAT2 polymorphisms and acute leukemia risk: A meta-analysis. Medicine (Baltimore) 2019; 98:e14942. [PMID: 30896661 PMCID: PMC6709067 DOI: 10.1097/md.0000000000014942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND N-acetyl-transferase 2 (NAT2) polymorphisms have been demonstrated to be associated with acute leukemia (AL); however, the results remain controversial. The present meta-analysis was performed to provide more precise results. METHODS Pubmed, Embase, Cochrane Library, China National Knowledge Infrastructure, and Wanfang databases were used to identify eligible studies. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between NAT2 polymorphisms and AL risk. RESULTS Increased risk was found under both heterozygous (OR 1.24, 95% CI 1.02-1.51) and recessive model (OR 1.28, 95% CI 1.06-1.55) for rs1801280. The slow acetylator phenotype (OR 1.22, 95% CI 1.07-1.40) also increased AL risk. Subgroup analysis demonstrated that rs1801280 increased AL risk under the recessive model (OR 1.14, 95% CI 0.93-1.41) in Caucasian population and the co-dominant (OR 1.77, 95% CI 1.40-2.23), homozygous (OR 3.06, 95% CI 1.88-4.99), dominant (OR 2.22, 95% CI 1.56-3.17), recessive model (OR 2.06, 95% CI 1.35-3.16) in the Mixed populations. Association between rs1799929 and decreased AL risk was found in the co-dominant (OR 0.82, 95% CI 0.70-0.97), homozygous (OR 0.65, 95% CI 0.46-0.93), heterozygous (OR 0.71, 95% CI 0.51-1.00), and the recessive model (OR 0.68, 95% CI 0.49-0.94) in the Caucasian group. As for rs1799931, the same effects were found in the co-dominant (OR 0.68, 95% CI 0.49-0.94) and the dominant model (OR 0.68, 95% CI 0.48-0.97) in the mixed group. CONCLUSION rs1801280 and the slow acetylator phenotype are risk factors for AL.
Collapse
Affiliation(s)
- Xiaoxiao Zhu
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| | - Yanbing Liu
- Breast Cancer Center, Shandong Cancer Hospital Affiliated to Shandong University
| | | | - Qiang Guo
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| | - Zhen Zhang
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| | - Lin Zhao
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| | - Ran Wei
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| | - Xunqiang Yin
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences
| | - Yunhong Zhang
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences
| | - Bin Wang
- Department of peripheral vascular disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xia Li
- Laboratory for molecular immunology, Institute of Basic Medicine, Shandong Academy of Medical Sciences
| |
Collapse
|
14
|
McGill MR, Jaeschke H. Biomarkers of drug-induced liver injury: progress and utility in research, medicine, and regulation. Expert Rev Mol Diagn 2018; 18:797-807. [PMID: 30080986 DOI: 10.1080/14737159.2018.1508998] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The difficulty of understanding and diagnosing drug-induced liver injury (DILI) has led to proliferation of serum and genetic biomarkers. Many applications of these biomarkers have been proposed, including investigation of mechanisms, prediction of DILI during early trials or before initiation of therapy in patients, and diagnosis of DILI during therapy. Areas covered: We review the definition and categories of DILI, describe recent developments in DILI biomarker development, and provide guidance for future directions in DILI biomarker research. Expert commentary: There are major obstacles to DILI biomarker development and implementation, including the low prevalence of idiosyncratic DILI (IDILI), weak associations of IDILI with genetic variants, and lack of specificity of many biomarkers for the liver. Certain serum biomarkers, like miR-122, may have clinical utility in early-presenting patients with either intrinsic or idiosyncratic DILI in the future, while others likely will not find use. Future research should focus on implementation of biomarkers to predict later injury and outcome in early presenters with intrinsic DILI, and on development of biomarkers of adaptation and repair in the liver that can be used to determine if a liver test abnormality is likely to be clinically significant in IDILI.
Collapse
Affiliation(s)
- Mitchell R McGill
- a Department of Environmental and Occupational Health , Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences , Little Rock , AR , USA.,b Department of Pharmacology and Toxicology , College of Medicine, University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Hartmut Jaeschke
- c Department of Pharmacology, Toxicology and Therapeutics , University of Kansas Medical Center , Kansas City , KS , USA
| |
Collapse
|
15
|
Gufford BT, Robarge JD, Eadon MT, Gao H, Lin H, Liu Y, Desta Z, Skaar TC. Rifampin modulation of xeno- and endobiotic conjugating enzyme mRNA expression and associated microRNAs in human hepatocytes. Pharmacol Res Perspect 2018; 6:e00386. [PMID: 29610665 PMCID: PMC5869567 DOI: 10.1002/prp2.386] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/12/2018] [Indexed: 01/06/2023] Open
Abstract
Rifampin is a pleiotropic inducer of multiple drug metabolizing enzymes and transporters. This work utilized a global approach to evaluate rifampin effects on conjugating enzyme gene expression with relevance to human xeno‐ and endo‐biotic metabolism. Primary human hepatocytes from 7 subjects were treated with rifampin (10 μmol/L, 24 hours). Standard methods for RNA‐seq library construction, EZBead preparation, and NextGen sequencing were used to measure UDP‐glucuronosyl transferase UGT, sulfonyltransferase SULT, N acetyltransferase NAT, and glutathione‐S‐transferase GST mRNA expression compared to vehicle control (0.01% MeOH). Rifampin‐induced (>1.25‐fold) mRNA expression of 13 clinically important phase II drug metabolizing genes and repressed (>1.25‐fold) the expression of 3 genes (P < .05). Rifampin‐induced miRNA expression changes correlated with mRNA changes and miRNAs were identified that may modulate conjugating enzyme expression. NAT2 gene expression was most strongly repressed (1.3‐fold) by rifampin while UGT1A4 and UGT1A1 genes were most strongly induced (7.9‐ and 4.8‐fold, respectively). Physiologically based pharmacokinetic modeling (PBPK) was used to simulate the clinical consequences of rifampin induction of CYP3A4‐ and UGT1A4‐mediated midazolam metabolism. Simulations evaluating isolated UGT1A4 induction predicted increased midazolam N‐glucuronide exposure (~4‐fold) with minimal reductions in parent midazolam exposure (~10%). Simulations accounting for simultaneous induction of both CYP3A4 and UGT1A4 predicted a ~10‐fold decrease in parent midazolam exposure with only a ~2‐fold decrease in midazolam N‐glucuronide metabolite exposure. These data reveal differential effects of rifampin on the human conjugating enzyme transcriptome and potential associations with miRNAs that form the basis for future mechanistic studies to elucidate the interplay of conjugating enzyme regulatory elements.
Collapse
Affiliation(s)
- Brandon T Gufford
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Jason D Robarge
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Michael T Eadon
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Hongyu Gao
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Hai Lin
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Yunlong Liu
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN
| | - Zeruesenay Desta
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| | - Todd C Skaar
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis IN
| |
Collapse
|
16
|
Alvarenga N, Birolli WG, Meira EB, Lucas SC, de Matos IL, Nitschke M, Romão LP, Porto AL. Biotransformation and biodegradation of methyl parathion by Brazilian bacterial strains isolated from mangrove peat. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2018. [DOI: 10.1016/j.bcab.2017.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Zhang QH, Yin RX, Huang F, Yang DZ, Lin WX, Pan SL. Association between the PINX1 and NAT2 polymorphisms and serum lipid levels. Oncotarget 2017; 8:114081-114094. [PMID: 29371971 PMCID: PMC5768388 DOI: 10.18632/oncotarget.23123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Jing nationality is a relatively conservative and isolated minority in China. Little is known about the association of the PIN2/TERF1-interacting telomerase inhibitor 1 (PINX1) and N-acetyltransferase 2 (NAT2) single nucleotide polymorphisms (SNPs) and serum lipid levels in the Chinese populations. This study aimed to clarify the association of 6 SNPs of the PINX1 and NAT2 and serum lipid levels in two Chinese populations. Genotyping of the SNPs was performed in 1236 Han subjects and 1248 Jing participants. Allelic and genotypic frequencies of these variants (except NAT2 rs1799931) were different between the two ethnic groups. The minor allele carriers had higher triglyceride (TG, rs11776767, rs1495743 and rs1799930), low-density lipoprotein cholesterol (rs6601530) levels and the apolipoprotein (Apo)A1/ApoB ratio (rs1495743) in Han nationality; and higher total cholesterol (rs1961456), TG (rs11776767, rs6601530 and rs1495743) and lower ApoA1 (rs6601530 and rs1799931) levels in Jing minority than the minor allele non-carriers. The SNPs were not statistically independent by the multiple-locus linkage disequilibrium analyses. The integrative haplotypes and gene-by-gene (G × G) interactions on serum lipid traits were also observed in the two populations. Association analysis based on haplotypes and G × G interactions might be powerful than single-locus tests. Differences in serum lipid profiles between the two populations might partially be attributed to these SNPs, their haplotypes and G × G interactions.
Collapse
Affiliation(s)
- Qing-Hui Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Feng Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - De-Zhai Yang
- Department of Molecular Genetics, Medical Scientific Research Center, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Wei-Xiong Lin
- Department of Molecular Genetics, Medical Scientific Research Center, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Shang-Ling Pan
- Department of Pathophysiology, School of Premedical Sciences, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| |
Collapse
|
18
|
Homology modeling and prediction of the amino acid residues participating in the transfer of acetyl-CoA to arylalkylamine by the N-acetyltransferase from Chryseobacterium sp. Biotechnol Lett 2017; 39:1699-1707. [PMID: 28721586 DOI: 10.1007/s10529-017-2399-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVES To predict the amino acid residues playing important roles in acetyl-CoA and substrate binding and to study the acetyl group transfer mechanism of Chryseobacterium sp. 5-3B N-acetyltransferase (5-3B NatA). RESULTS A 3-dimensional homology model of 5-3B NatA was constructed to compare the theoretical structure of this compound with the structures of previously reported proteins belonging to the bacterial GCN5 N-acetyltransferase family. Homology modeling of the 5-3B NatA structure and a characterization of the enzyme's kinetic parameters identified the essential amino acid residues involved in binding and acetyl-group transfer. 126Leu, 132Leu, and 135Lys were implicated in the binding of phosphopantothenic acid, and 100Tyr and 131Lys in that of adenosyl biphosphate. The data supported the participation of 83Glu and 133Tyr in catalyzing acetyl-group transfer to L-2-phenylglycine. CONCLUSIONS 5-3B NatA catalyzes the enantioselective N-acetylation of L-2-phenylglycine via a ternary complex comprising the enzyme, acetyl-CoA, and the substrate.
Collapse
|
19
|
Association between NAT2 polymorphisms and the risk of schizophrenia in a Northern Chinese Han population. Psychiatr Genet 2017; 27:71-75. [PMID: 28187106 DOI: 10.1097/ypg.0000000000000164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The gene that encodes N-acetyltransferase 2 (NAT2), an enzyme that plays a crucial role in the metabolism of many drugs and xenobiotics, is located on chromosome 8p22, one of the most convictive susceptibility loci of schizophrenia. NAT2 genetic polymorphisms lead to various enzyme acetylation phenotypes. In the present study, six selected NAT2 exonic single nucleotide polymorphisms were genotyped in an independent case-control sample of a Northern Chinese Han population to verify the possible association between NAT2 and schizophrenia. Three (rs1801280T/341C, rs1799930/G590A, and rs1208/A803G) of the six single nucleotide polymorphisms showed significant allele frequency differences between the case and the control groups after rigorous Bonferroni correction. One protective fast-acetylation haplotype (NAT2*4) and two risk slow acetylation haplotypes (NAT2*5B and NAT2*6A) were discovered to be associated with schizophrenia. Our results indicate that NAT2 may be a susceptibility gene for schizophrenia in this Chinese Han population, and the risk haplotypes might cause the impairment of NAT2 in metabolizing neurotoxic substances.
Collapse
|
20
|
Begas E, Tsioutsiouliti A, Kouvaras E, Haroutounian SA, Kasiotis KM, Kouretas D, Asprodini E. Effects of peppermint tea consumption on the activities of CYP1A2, CYP2A6, Xanthine Oxidase, N-acetyltranferase-2 and UDP-glucuronosyltransferases-1A1/1A6 in healthy volunteers. Food Chem Toxicol 2017; 100:80-89. [DOI: 10.1016/j.fct.2016.12.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/18/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023]
|
21
|
Imran M, Usman HF, Shafi H, Sarwar M, Tahir MA. Development of Rapid and Economical Colorimetric Screening Method for p-Phenylenediamine in Variety of Biological Matrices and its Application to Eleven Fatal Cases of p-Phenylenediamine Poisoning. J Forensic Sci 2016; 62:483-487. [PMID: 27957731 DOI: 10.1111/1556-4029.13297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 05/03/2016] [Accepted: 05/15/2016] [Indexed: 12/01/2022]
Abstract
A rapid colorimetric method for detection of p-phenylenediamine (PPD) in various biological samples is developed. The o-cresol test for acetaminophen detection has been modified to detect PPD in blood, urine, gastric contents, and liver. After precipitating protein with trichloroacetic acid solution (2 mL, 10% w/v), biological specimens were required to convert PPD metabolites to PPD by acid hydrolysis. Finally, o-cresol solution (1 mL, 1% w/v), hydrogen peroxide (200 μL, 3%v/v), and concentrated ammonium hydroxide (0.5 mL) were added in the biological samples. The presence of PPD was indicated by formation of violet color which was turned to bluish green color within 10-15 min. The limit of detection was found to be 2 mg/L in blood, urine, and gastric contents and 2 mg/Kg in liver. This method is also free from any potential interference by p-aminophenol, acetaminophen, and other amine drugs under test conditions. This method was successfully employed to thirteen fatal cases of PPD poisoning.
Collapse
Affiliation(s)
- Muhammad Imran
- Forensic Toxicology department, Punjab Forensic Science Agency, Thokar Niaz Baig, Multan Road, Lahore, 53700, Pakistan
| | - Hafiz Faisal Usman
- Forensic Toxicology department, Punjab Forensic Science Agency, Thokar Niaz Baig, Multan Road, Lahore, 53700, Pakistan
| | - Humera Shafi
- Forensic Toxicology department, Punjab Forensic Science Agency, Thokar Niaz Baig, Multan Road, Lahore, 53700, Pakistan
| | - Muhammad Sarwar
- Forensic Toxicology department, Punjab Forensic Science Agency, Thokar Niaz Baig, Multan Road, Lahore, 53700, Pakistan
| | - Muhammad Ashraf Tahir
- Forensic Toxicology department, Punjab Forensic Science Agency, Thokar Niaz Baig, Multan Road, Lahore, 53700, Pakistan
| |
Collapse
|
22
|
Mycobacterium tuberculosis Arylamine N-Acetyltransferase Acetylates and Thus Inactivates para-Aminosalicylic Acid. Antimicrob Agents Chemother 2016; 60:7505-7508. [PMID: 27671064 DOI: 10.1128/aac.01312-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/19/2016] [Indexed: 12/31/2022] Open
Abstract
Mycobacterium tuberculosis arylamine N-acetyltransferase (TBNAT) is able to acetylate para-aminosalicylic acid (PAS) both in vitro and in vivo as determined by high-performance liquid chromatography (HPLC) and electrospray ionization-mass spectrometry (ESI-MS) techniques. The antituberculosis activity of the acetylated PAS is significantly reduced. As a result, overexpression of TBNAT in M. tuberculosis results in PAS resistance, as determined by MIC tests and drug exposure experiments. Taken together, our results suggest that TBNAT from M. tuberculosis is able to inactivate PAS by acetylating the compound.
Collapse
|
23
|
Kubiak X, Duval R, Pluvinage B, Chaffotte AF, Dupret JM, Rodrigues-Lima F. Xenobiotic-metabolizing enzymes in Bacillus anthracis: molecular and functional analysis of a truncated arylamine N-acetyltransferase isozyme. Br J Pharmacol 2016; 174:2174-2182. [PMID: 27846346 DOI: 10.1111/bph.13647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/01/2016] [Accepted: 09/26/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The arylamine N-acetyltransferases (NATs) are xenobiotic-metabolizing enzymes that play an important role in the detoxification and/or bioactivation of arylamine drugs and xenobiotics. In bacteria, NATs may contribute to the resistance against antibiotics such as isoniazid or sulfamides through their acetylation, which makes this enzyme family a possible drug target. Bacillus anthracis, a bacterial species of clinical significance, expresses three NAT isozymes with distinct structural and enzymatic properties, including an inactive isozyme ((BACAN)NAT3). (BACAN)NAT3 features both a non-canonical Glu residue in its catalytic triad and a truncated C-terminus domain. However, the role these unusual characteristics play in the lack of activity of the (BACAN)NAT3 isozyme remains unclear. EXPERIMENTAL APPROACH Protein engineering, recombinant expression, enzymatic analyses with aromatic amine substrates and phylogenetic analysis approaches were conducted. KEY RESULTS The deletion of guanine 580 (G580) in the nat3 gene was shown to be responsible for the expression of a truncated (BACAN)NAT3 isozyme. Artificial re-introduction of G580 in the nat3 gene led to a functional enzyme able to acetylate several arylamine drugs displaying structural characteristics comparable with its functional Bacillus cereus homologue ((BACCR)NAT3). Phylogenetic analysis of the nat3 gene in the B. cereus group further indicated that nat3 may constitute a pseudogene of the B. anthracis species. CONCLUSION AND IMPLICATIONS The existence of NATs with distinct properties and evolution in Bacillus species may account for their adaptation to their diverse chemical environments. A better understanding of these isozymes is of importance for their possible use as drug targets. LINKED ARTICLES This article is part of a themed section on Drug Metabolism and Antibiotic Resistance in Micro-organisms. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.14/issuetoc.
Collapse
Affiliation(s)
- Xavier Kubiak
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Romain Duval
- Sorbonne Paris Cité, Univ Paris Diderot, Unité BFA, CNRS UMR 8251, Paris, France
| | - Benjamin Pluvinage
- Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Alain F Chaffotte
- Unité de Résonance Magnétique Nucléaire des Biomolécules, Institut Pasteur, Paris, France
| | - Jean-Marie Dupret
- Sorbonne Paris Cité, Univ Paris Diderot, Unité BFA, CNRS UMR 8251, Paris, France
| | | |
Collapse
|
24
|
Matthaei J, Tzvetkov MV, Strube J, Sehrt D, Sachse-Seeboth C, Hjelmborg JB, Möller S, Halekoh U, Hofmann U, Schwab M, Kerb R, Brockmöller J. Heritability of Caffeine Metabolism: Environmental Effects Masking Genetic Effects on CYP1A2 Activity but Not on NAT2. Clin Pharmacol Ther 2016; 100:606-616. [DOI: 10.1002/cpt.444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 12/24/2022]
Affiliation(s)
- J Matthaei
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| | - MV Tzvetkov
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| | - J Strube
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| | - D Sehrt
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| | - C Sachse-Seeboth
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| | - JB Hjelmborg
- Department of Epidemiology, Biostatistics and Biodemography; University of Southern Denmark; Odense Denmark
| | - S Möller
- Department of Epidemiology, Biostatistics and Biodemography; University of Southern Denmark; Odense Denmark
| | - U Halekoh
- Department of Epidemiology, Biostatistics and Biodemography; University of Southern Denmark; Odense Denmark
| | - U Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen; Stuttgart Germany
| | - M Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen; Stuttgart Germany
- Department of Clinical Pharmacology; University Hospital Tübingen; Tübingen Germany
- Department of Pharmacy and Biochemistry; University of Tübingen; Tübingen Germany
| | - R Kerb
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen; Stuttgart Germany
| | - J Brockmöller
- Institute for Clinical Pharmacology, University Medical Center, Göttingen; Georg-August University; Göttingen Germany
| |
Collapse
|
25
|
Importance of the Evaluation of N-Acetyltransferase Enzyme Activity Prior to 5-Aminosalicylic Acid Medication for Ulcerative Colitis. Inflamm Bowel Dis 2016; 22:1793-802. [PMID: 27416043 PMCID: PMC4956520 DOI: 10.1097/mib.0000000000000823] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND 5-aminosalicylic acid (5-ASA) is a classic anti-inflammatory drug for the treatment of ulcerative colitis. N-acetyltransferase (NAT) enzymes convert 5-ASA to its metabolite N-acetyl-5-ASA, and it is unresolved whether 5-ASA or N-acetyl-5-ASA is the effective therapeutic molecule. We previously demonstrated that colonic production of N-acetyl-5-ASA (NAT activity) is decreased in dextran sulfate sodium-induced colitis. Our hypothesis is that 5-ASA is the therapeutic molecule to improve colitis, with the corollary that altered NAT activity affects drug efficacy. Since varying clinical effectiveness of 5-ASA has been reported, we also ask if NAT activity varies with inflammation in pediatric or adult patients. METHODS Acute colonic inflammation was induced in C57BL/6 NAT wild-type (WT) or knockout mice, using 3.5% dextran sulfate sodium (w/v) concurrent with 5-ASA treatment. Adult and pediatric rectosigmoid biopsies were collected from control or patients with ulcerative colitis. Tissue was analyzed for NAT and myeloperoxidase activity. RESULTS Dextran sulfate sodium-induced colitis was of similar severity in both NAT WT and knockout mice, and NAT activity was significantly decreased in NAT WT mice. In the setting of colitis, 5-ASA significantly restored colon length and decreased myeloperoxidase activity in NAT knockout but not in WT mice. Myeloperoxidase activity negatively correlated with NAT activity in pediatric patients, but correlation was not observed in adult patients. CONCLUSIONS Inflammation decreases NAT activity in the colon of mice and human pediatric patients. Decreased NAT activity enhances the therapeutic effect of 5-ASA in mice. A NAT activity assay could be useful to help predict the efficacy of 5-ASA therapy.
Collapse
|
26
|
Bohnert T, Patel A, Templeton I, Chen Y, Lu C, Lai G, Leung L, Tse S, Einolf HJ, Wang YH, Sinz M, Stearns R, Walsky R, Geng W, Sudsakorn S, Moore D, He L, Wahlstrom J, Keirns J, Narayanan R, Lang D, Yang X. Evaluation of a New Molecular Entity as a Victim of Metabolic Drug-Drug Interactions-an Industry Perspective. Drug Metab Dispos 2016; 44:1399-423. [PMID: 27052879 DOI: 10.1124/dmd.115.069096] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/31/2016] [Indexed: 12/15/2022] Open
Abstract
Under the guidance of the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), scientists from 20 pharmaceutical companies formed a Victim Drug-Drug Interactions Working Group. This working group has conducted a review of the literature and the practices of each company on the approaches to clearance pathway identification (fCL), estimation of fractional contribution of metabolizing enzyme toward metabolism (fm), along with modeling and simulation-aided strategy in predicting the victim drug-drug interaction (DDI) liability due to modulation of drug metabolizing enzymes. Presented in this perspective are the recommendations from this working group on: 1) strategic and experimental approaches to identify fCL and fm, 2) whether those assessments may be quantitative for certain enzymes (e.g., cytochrome P450, P450, and limited uridine diphosphoglucuronosyltransferase, UGT enzymes) or qualitative (for most of other drug metabolism enzymes), and the impact due to the lack of quantitative information on the latter. Multiple decision trees are presented with stepwise approaches to identify specific enzymes that are involved in the metabolism of a given drug and to aid the prediction and risk assessment of drug as a victim in DDI. Modeling and simulation approaches are also discussed to better predict DDI risk in humans. Variability and parameter sensitivity analysis were emphasized when applying modeling and simulation to capture the differences within the population used and to characterize the parameters that have the most influence on the prediction outcome.
Collapse
Affiliation(s)
- Tonika Bohnert
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Aarti Patel
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ian Templeton
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Yuan Chen
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Chuang Lu
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - George Lai
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Louis Leung
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Susanna Tse
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Heidi J Einolf
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ying-Hong Wang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Michael Sinz
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ralph Stearns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Robert Walsky
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Wanping Geng
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Sirimas Sudsakorn
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - David Moore
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ling He
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jan Wahlstrom
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jim Keirns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Rangaraj Narayanan
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Dieter Lang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Xiaoqing Yang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| |
Collapse
|
27
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Agúndez JAG. NAT2 polymorphisms and risk for Parkinson's disease: a systematic review and meta-analysis. Expert Opin Drug Metab Toxicol 2016; 12:937-46. [PMID: 27216438 DOI: 10.1080/17425255.2016.1192127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Several studies suggested a possible association between certain polymorphisms in the N-acetyl-transferase 2 (NAT2) gene (which encodes a very important enzyme involved in xenobiotic metabolism) and the risk for Parkinson's disease (PD). As the results of studies on this issue are controversial, we conducted a systematic review and a meta-analysis of eligible studies on this putative association. AREAS COVERED The authors revised the relationship between NAT2 polymorphisms and the risk of developing PD using several databases, and performed a meta-analysis using the software Meta-Disc1.1.1. In addition heterogeneity between studies was analyzed. A description of studies regarding gene-gene interactions and gene-environmental interactions involving NAT2 polymorphisms is also made. EXPERT OPINION Despite several recent meta-analyses showing an association between several polymorphisms in genes related with detoxification mechanisms such as cytochrome P4502D6 (CYP2D6), and glutathione transferases M1 and T1 (GSTM1, and GSTT1), data on NAT2 gene polymorphisms obtained from the current meta-analysis do not support a major association with PD risk, except in Asian populations. However, data from many studies are incomplete and therefore insufficient data exists to draw definitive conclusions. Several studies suggesting gene-gene and gene-environmental factors involving NAT2 gene in PD risk await confirmation.
Collapse
Affiliation(s)
- Félix Javier Jiménez-Jiménez
- a Section of Neurology , Hospital Universitario del Sureste , Arganda del Rey , Madrid , Spain.,b Department of Medicine-Neurology, Hospital 'Príncipe de Asturias' , Universidad de Alcalá , Alcalá de Henares , Madrid , Spain
| | | | | | - José A G Agúndez
- c Department of Pharmacology , University of Extremadura , Cáceres , Spain
| |
Collapse
|
28
|
Gómez-Gómez GJ, Masedo &A, Yela C, Martínez-Montiel MDP, Casís B. Current stage in inflammatory bowel disease: What is next? World J Gastroenterol 2015; 21:11282-11303. [PMID: 26525013 PMCID: PMC4616205 DOI: 10.3748/wjg.v21.i40.11282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/12/2015] [Accepted: 09/02/2015] [Indexed: 02/06/2023] Open
Abstract
In recent years, the incidence of inflammatory bowel disease (IBD) has been on the rise, extending to countries where it was infrequent in the past. As a result, the gap between high and low incidence countries is decreasing. The disease, therefore, has an important economic impact on the healthcare system. Advances in recent years in pharmacogenetics and clinical pharmacology have allowed for the development of treatment strategies adjusted to the patient profile. Concurrently, new drugs aimed at inflammatory targets have been developed that may expand future treatment options. This review examines advances in the optimization of existing drug treatments and the development of novel treatment options for IBD.
Collapse
|
29
|
Stepp MW, Mamaliga G, Doll MA, States JC, Hein DW. Folate-Dependent Hydrolysis of Acetyl-Coenzyme A by Recombinant Human and Rodent Arylamine N-Acetyltransferases. Biochem Biophys Rep 2015; 3:45-50. [PMID: 26309907 PMCID: PMC4545580 DOI: 10.1016/j.bbrep.2015.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Arylamine N-acetyltransferases (NATs) are drug and xenobiotic metabolizing enzymes that catalyze the N-acetylation of arylamines and hydrazines and the O-acetylation of N-hydroxy-arylamines. Recently, studies report that human NAT1 and mouse Nat2 hydrolyze acetyl-coenzyme A (AcCoA) into acetate and coenzyme A in a folate-dependent fashion, a previously unknown function. In this study, our goal was to confirm these findings and determine the apparent Michaelis-Menten kinetic constants (Vmax and Km) of the folate-dependent AcCoA hydrolysis for human NAT1/NAT2, and the rodent analogs rat Nat1/Nat2, mouse Nat1/Nat2, and hamster Nat1/Nat2. We also compared apparent Vmax values for AcCoA hydrolysis and N-acetylation of the substrate para-aminobenzoic acid (PABA). Human NAT1 and its rodent analogs rat Nat2, mouse Nat2 and hamster Nat2 catalyzed AcCoA hydrolysis in a folate-dependent manner. Rates of AcCoA hydrolysis were between 0.25 - 1% of the rates for N-acetylation of PABA catalyzed by human NAT1 and its rodent orthologs. In contrast to human NAT1, human NAT2 and its rodent analogs rat Nat1, mouse Nat1, and hamster Nat1 did not hydrolyze AcCoA in a folate-dependent manner. These results are consistent with the possibility that human NAT1 and its rodent analogs regulate endogenous AcCoA levels.
Collapse
Affiliation(s)
- Marcus W Stepp
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, School of Medicine, University of Louisville
| | - Galina Mamaliga
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, School of Medicine, University of Louisville
| | - Mark A Doll
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, School of Medicine, University of Louisville
| | - J Christopher States
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, School of Medicine, University of Louisville
| | - David W Hein
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, School of Medicine, University of Louisville
| |
Collapse
|
30
|
Abstract
Pharmacogenetic studies have been performed for almost all classes of drugs that have been used in IBD but very few have generated consistent findings or have been replicated. The genetic test that has been approved for clinical practice is TPMT testing prior to starting treatment with thiopurine drugs. Research in IBD pharmacogenetics has focused on prediction of drug efficacy and toxicity by identifying polymorphisms in the genes encoding enzymes that are involved in metabolic pathways. Recent research has mainly focused on therapeutic agents such as azathioprine, methotrexate, aminosalicylates, corticosteroids, infliximab and adalimumab. Future pharmaceutical trials should include pharmacogenetic research to test appropriate candidate genes in a prospective manner and correlate genetic associations with trial outcomes and relevant functional data.
Collapse
|
31
|
Milioli HH, Vimieiro R, Riveros C, Tishchenko I, Berretta R, Moscato P. The Discovery of Novel Biomarkers Improves Breast Cancer Intrinsic Subtype Prediction and Reconciles the Labels in the METABRIC Data Set. PLoS One 2015; 10:e0129711. [PMID: 26132585 PMCID: PMC4488510 DOI: 10.1371/journal.pone.0129711] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prediction of breast cancer intrinsic subtypes has been introduced as a valuable strategy to determine patient diagnosis and prognosis, and therapy response. The PAM50 method, based on the expression levels of 50 genes, uses a single sample predictor model to assign subtype labels to samples. Intrinsic errors reported within this assay demonstrate the challenge of identifying and understanding the breast cancer groups. In this study, we aim to: a) identify novel biomarkers for subtype individuation by exploring the competence of a newly proposed method named CM1 score, and b) apply an ensemble learning, as opposed to the use of a single classifier, for sample subtype assignment. The overarching objective is to improve class prediction. METHODS AND FINDINGS The microarray transcriptome data sets used in this study are: the METABRIC breast cancer data recorded for over 2000 patients, and the public integrated source from ROCK database with 1570 samples. We first computed the CM1 score to identify the probes with highly discriminative patterns of expression across samples of each intrinsic subtype. We further assessed the ability of 42 selected probes on assigning correct subtype labels using 24 different classifiers from the Weka software suite. For comparison, the same method was applied on the list of 50 genes from the PAM50 method. CONCLUSIONS The CM1 score portrayed 30 novel biomarkers for predicting breast cancer subtypes, with the confirmation of the role of 12 well-established genes. Intrinsic subtypes assigned using the CM1 list and the ensemble of classifiers are more consistent and homogeneous than the original PAM50 labels. The new subtypes show accurate distributions of current clinical markers ER, PR and HER2, and survival curves in the METABRIC and ROCK data sets. Remarkably, the paradoxical attribution of the original labels reinforces the limitations of employing a single sample classifiers to predict breast cancer intrinsic subtypes.
Collapse
Affiliation(s)
- Heloisa Helena Milioli
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Renato Vimieiro
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centro de Informática, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Carlos Riveros
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Inna Tishchenko
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Regina Berretta
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Pablo Moscato
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
32
|
Valente C, Alvarez L, Marks SJ, Lopez-Parra AM, Parson W, Oosthuizen O, Oosthuizen E, Amorim A, Capelli C, Arroyo-Pardo E, Gusmão L, Prata MJ. Exploring the relationship between lifestyles, diets and genetic adaptations in humans. BMC Genet 2015; 16:55. [PMID: 26018448 PMCID: PMC4445807 DOI: 10.1186/s12863-015-0212-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 04/30/2015] [Indexed: 12/05/2022] Open
Abstract
Background One of the most important dietary shifts underwent by human populations began to occur in the Neolithic, during which new modes of subsistence emerged and new nutrients were introduced in diets. This change might have worked as a selective pressure over the metabolic pathways involved in the breakdown of substances extracted from food. Here we applied a candidate gene approach to investigate whether in populations with different modes of subsistence, diet-related genetic adaptations could be identified in the genes AGXT, PLRP2, MTRR, NAT2 and CYP3A5. Results At CYP3A5, strong signatures of positive selection were detected, though not connected to any dietary variable, but instead to an environmental factor associated with the Tropic of Cancer. Suggestive signals of adaptions that could indeed be connected with differences in dietary habits of populations were only found for PLRP2 and NAT2. Contrarily, the demographic history of human populations seemed enough to explain patterns of diversity at AGXT and MTRR, once both conformed the evolutionary expectations under selective neutrality. Conclusions Accumulated evidence indicates that CYP3A5 has been under adaptive evolution during the history of human populations. PLRP2 and NAT2 also appear to have been modelled by some selective constrains, although clear support for that did not resist to a genome wide perspective. It is still necessary to clarify which were the biological mechanisms and the environmental factors involved as well as their interactions, to understand the nature and strength of the selective pressures that contributed to shape current patterns of genetic diversity at those loci. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0212-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cristina Valente
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal. .,Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Luis Alvarez
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.
| | - Sarah J Marks
- Department of Zoology, University of Oxford, Oxford, UK.
| | - Ana M Lopez-Parra
- Departamento de Toxicología y Legislación Sanitaria, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| | - Walther Parson
- Institute of Legal Medicine, Innsbruck Medical University, Innsbruck, Austria. .,Penn State Eberly College of Science, University Park, Pennsylvania, USA.
| | | | | | - António Amorim
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal. .,Faculty of Sciences, University of Porto, Porto, Portugal.
| | | | - Eduardo Arroyo-Pardo
- Departamento de Toxicología y Legislación Sanitaria, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| | - Leonor Gusmão
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal. .,DNA Diagnostic Laboratory (LDD), State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil.
| | - Maria J Prata
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal. .,Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
33
|
Geller F, Soborg B, Koch A, Michelsen SW, Bjorn-Mortensen K, Carstensen L, Birch E, Nordholm AC, Johansen MMB, Børresen ML, Feenstra B, Melbye M. Determination of NAT2 acetylation status in the Greenlandic population. Arch Toxicol 2015; 90:883-9. [PMID: 25794903 DOI: 10.1007/s00204-015-1501-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/05/2015] [Indexed: 11/26/2022]
Abstract
N-acetyltransferase 2 (NAT2) is a well-studied phase II xenobiotic metabolizing enzyme relevant in drug metabolism and cancerogenesis. NAT2 activity is largely determined by genetic polymorphisms in the coding region of the corresponding gene. We investigated NAT2 acetylation status in 1556 individuals from Greenland based on four different single nucleotide polymorphism (SNP) panels and the tagging SNP rs1495741. There was good concordance between the NAT2 status inferred by the different SNP combinations. Overall, the fraction of slow acetylators was low with 17.5 % and varied depending on the degree of Inuit ancestry; in individuals with <50 % Inuit ancestry, we observed more than 25 % slow acetylators reflecting European ancestry. Greenland has a high incidence of tuberculosis, and individual dosing of isoniazid according to NAT2 status has been shown to improve treatment and reduce side effects. Our findings could be a first step in pharmacogenetics-based tuberculosis therapy in Greenland.
Collapse
Affiliation(s)
- Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark.
| | - Bolette Soborg
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Anders Koch
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Sascha Wilk Michelsen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Karen Bjorn-Mortensen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Lisbeth Carstensen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Emilie Birch
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | | | | | - Malene Landbo Børresen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen S, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200, Copenhagen N, Denmark
- Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
34
|
Xu X, Li de la Sierra-Gallay I, Kubiak X, Duval R, Chaffotte AF, Dupret JM, Haouz A, Rodrigues-Lima F. Insight into cofactor recognition in arylamine N-acetyltransferase enzymes: structure of Mesorhizobium loti arylamine N-acetyltransferase in complex with coenzyme A. ACTA ACUST UNITED AC 2015; 71:266-73. [PMID: 25664736 DOI: 10.1107/s139900471402522x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/17/2014] [Indexed: 01/05/2023]
Abstract
Arylamine N-acetyltransferases (NATs) are xenobiotic metabolizing enzymes that catalyze the acetyl-CoA-dependent acetylation of arylamines. To better understand the mode of binding of the cofactor by this family of enzymes, the structure of Mesorhizobium loti NAT1 [(RHILO)NAT1] was determined in complex with CoA. The F42W mutant of (RHILO)NAT1 was used as it is well expressed in Escherichia coli and displays enzymatic properties similar to those of the wild type. The apo and holo structures of (RHILO)NAT1 F42W were solved at 1.8 and 2 Å resolution, respectively. As observed in the Mycobacterium marinum NAT1-CoA complex, in (RHILO)NAT1 CoA binding induces slight structural rearrangements that are mostly confined to certain residues of its `P-loop'. Importantly, it was found that the mode of binding of CoA is highly similar to that of M. marinum NAT1 but different from the modes reported for Bacillus anthracis NAT1 and Homo sapiens NAT2. Therefore, in contrast to previous data, this study shows that different orthologous NATs can bind their cofactors in a similar way, suggesting that the mode of binding CoA in this family of enzymes is less diverse than previously thought. Moreover, it supports the notion that the presence of the `mammalian/eukaryotic insertion loop' in certain NAT enzymes impacts the mode of binding CoA by imposing structural constraints.
Collapse
Affiliation(s)
- Ximing Xu
- Université Paris Diderot, Sorbonne Paris Cité, Unité BFA, CNRS UMR 8251, 75013 Paris, France
| | - Inés Li de la Sierra-Gallay
- Université Paris-Sud, Institut de Biochimie et Biophysique Moléculaire et Cellulaire, CNRS UMR 8619, 91405 Orsay, France
| | - Xavier Kubiak
- Université Paris Diderot, Sorbonne Paris Cité, Unité BFA, CNRS UMR 8251, 75013 Paris, France
| | - Romain Duval
- Université Paris Diderot, Sorbonne Paris Cité, Unité BFA, CNRS UMR 8251, 75013 Paris, France
| | - Alain F Chaffotte
- Institut Pasteur, Unité de Résonance Magnétique Nucléaire des Biomolécules, 75015 Paris, France
| | - Jean Marie Dupret
- Université Paris Diderot, Sorbonne Paris Cité, Unité BFA, CNRS UMR 8251, 75013 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Plateforme de Cristallographie, CNRS UMR 3528, 75015 Paris, France
| | - Fernando Rodrigues-Lima
- Université Paris Diderot, Sorbonne Paris Cité, Unité BFA, CNRS UMR 8251, 75013 Paris, France
| |
Collapse
|
35
|
Manevski N, Swart P, Balavenkatraman KK, Bertschi B, Camenisch G, Kretz O, Schiller H, Walles M, Ling B, Wettstein R, Schaefer DJ, Itin P, Ashton-Chess J, Pognan F, Wolf A, Litherland K. Phase II metabolism in human skin: skin explants show full coverage for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation. Drug Metab Dispos 2015; 43:126-39. [PMID: 25339109 DOI: 10.1124/dmd.114.060350] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although skin is the largest organ of the human body, cutaneous drug metabolism is often overlooked, and existing experimental models are insufficiently validated. This proof-of-concept study investigated phase II biotransformation of 11 test substrates in fresh full-thickness human skin explants, a model containing all skin cell types. Results show that skin explants have significant capacity for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation. Novel skin metabolites were identified, including acyl glucuronides of indomethacin and diclofenac, glucuronides of 17β-estradiol, N-acetylprocainamide, and methoxy derivatives of 4-nitrocatechol and 2,3-dihydroxynaphthalene. Measured activities for 10 μM substrate incubations spanned a 1000-fold: from the highest 4.758 pmol·mg skin(-1)·h(-1) for p-toluidine N-acetylation to the lowest 0.006 pmol·mg skin(-1)·h(-1) for 17β-estradiol 17-glucuronidation. Interindividual variability was 1.4- to 13.0-fold, the highest being 4-methylumbelliferone and diclofenac glucuronidation. Reaction rates were generally linear up to 4 hours, although 24-hour incubations enabled detection of metabolites in trace amounts. All reactions were unaffected by the inclusion of cosubstrates, and freezing of the fresh skin led to loss of glucuronidation activity. The predicted whole-skin intrinsic metabolic clearances were significantly lower compared with corresponding whole-liver intrinsic clearances, suggesting a relatively limited contribution of the skin to the body's total systemic phase II enzyme-mediated metabolic clearance. Nevertheless, the fresh full-thickness skin explants represent a suitable model to study cutaneous phase II metabolism not only in drug elimination but also in toxicity, as formation of acyl glucuronides and sulfate conjugates could play a role in skin adverse reactions.
Collapse
Affiliation(s)
- Nenad Manevski
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Piet Swart
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Kamal Kumar Balavenkatraman
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Barbara Bertschi
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Gian Camenisch
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Olivier Kretz
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Hilmar Schiller
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Markus Walles
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Barbara Ling
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Reto Wettstein
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Peter Itin
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Joanna Ashton-Chess
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Francois Pognan
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Armin Wolf
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| | - Karine Litherland
- Drug Metabolism and Pharmacokinetics (N.M., P.S., G.C., O.K., H.S., M.W., K.L.), Pre-clinical Safety (K.K.B., B.B., F.P., A.W.), and Clinical Sciences and Innovation Translational Medicine (J.A.-C.), Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland; and Department of Plastic, Reconstructive, Aesthetic and Hand Surgery (B.L., R.W., D.J.S.), and Department of Dermatology (P.I.), University Hospital Basel, Basel, Switzerland
| |
Collapse
|
36
|
Sim E, Abuhammad A, Ryan A. Arylamine N-acetyltransferases: from drug metabolism and pharmacogenetics to drug discovery. Br J Pharmacol 2014; 171:2705-25. [PMID: 24467436 PMCID: PMC4158862 DOI: 10.1111/bph.12598] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 08/15/2013] [Accepted: 08/26/2013] [Indexed: 12/12/2022] Open
Abstract
Arylamine N-acetyltransferases (NATs) are polymorphic drug-metabolizing enzymes, acetylating arylamine carcinogens and drugs including hydralazine and sulphonamides. The slow NAT phenotype increases susceptibility to hydralazine and isoniazid toxicity and to occupational bladder cancer. The two polymorphic human NAT loci show linkage disequilibrium. All mammalian Nat genes have an intronless open reading frame and non-coding exons. The human gene products NAT1 and NAT2 have distinct substrate specificities: NAT2 acetylates hydralazine and human NAT1 acetylates p-aminosalicylate (p-AS) and the folate catabolite para-aminobenzoylglutamate (p-abaglu). Human NAT2 is mainly in liver and gut. Human NAT1 and its murine homologue are in many adult tissues and in early embryos. Human NAT1 is strongly expressed in oestrogen receptor-positive breast cancer and may contribute to folate and acetyl CoA homeostasis. NAT enzymes act through a catalytic triad of Cys, His and Asp with the architecture of the active site-modulating specificity. Polymorphisms may cause unfolded protein. The C-terminus helps bind acetyl CoA and differs among NATs including prokaryotic homologues. NAT in Salmonella typhimurium supports carcinogen activation and NAT in mycobacteria metabolizes isoniazid with polymorphism a minor factor in isoniazid resistance. Importantly, nat is in a gene cluster essential for Mycobacterium tuberculosis survival inside macrophages. NAT inhibitors are a starting point for novel anti-tuberculosis drugs. Human NAT1-specific inhibitors may act in biomarker detection in breast cancer and in cancer therapy. NAT inhibitors for co-administration with 5-aminosalicylate (5-AS) in inflammatory bowel disease has prompted ongoing investigations of azoreductases in gut bacteria which release 5-AS from prodrugs including balsalazide.
Collapse
Affiliation(s)
- E Sim
- Faculty of Science Engineering and Computing, Kingston University, Kingston, UK; Department of Pharmacology, Oxford University, Oxford, UK
| | | | | |
Collapse
|
37
|
Laurieri N, Kawamura A, Westwood IM, Varney A, Morris E, Russell AJ, Stanley LA, Sim E. Differences between murine arylamine N-acetyltransferase type 1 and human arylamine N-acetyltransferase type 2 defined by substrate specificity and inhibitor binding. BMC Pharmacol Toxicol 2014; 15:68. [PMID: 25432241 PMCID: PMC4258814 DOI: 10.1186/2050-6511-15-68] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/18/2014] [Indexed: 12/18/2022] Open
Abstract
Background The mouse has three arylamine N-acetyltransferase genes, (MOUSE)Nat1, (MOUSE)Nat2 and (MOUSE)Nat3. These are believed to correspond to (HUMAN)NAT1, (HUMAN)NAT2 and NATP in humans. (MOUSE)Nat3 encodes an enzyme with poor activity and human NATP is a pseudogene. (MOUSE)Nat2 is orthologous to (HUMAN)NAT1 and their corresponding proteins are functionally similar, but the relationship between (MOUSE)Nat1 and (HUMAN)NAT2 is less clear-cut. Methods To determine whether the (MOUSE)NAT1 and (HUMAN)NAT2 enzymes are functionally equivalent, we expressed and purified (MOUSE)NAT1*1 and analysed its substrate specificity using a panel of arylamines and hydrazines. To understand how specific residues contribute to substrate selectivity, three site-directed mutants of (MOUSE)NAT2*1 were prepared: these were (MOUSE)NAT2_F125S, (MOUSE)NAT2_R127G and (MOUSE)NAT2_R127L. All three exhibited diminished activity towards “(MOUSE)NAT2-specific” arylamines but were more active against hydrazines than (MOUSE)NAT1*1. The inhibitory and colorimetric properties of a selective naphthoquinone inhibitor of (HUMAN)NAT1 and (MOUSE)NAT2 were investigated. Results Comparing (MOUSE)NAT1*1 with other mammalian NAT enzymes demonstrated that the substrate profiles of (MOUSE)NAT1 and (HUMAN)NAT2 are less similar than previously believed. Three key residues (F125, R127 and Y129) in (HUMAN)NAT1*4 and (MOUSE)NAT2*1 were required for enzyme inhibition and the associated colour change on naphthoquinone binding. In silico modelling of selective ligands into the appropriate NAT active sites further implicated these residues in substrate and inhibitor specificity in mouse and human NAT isoenzymes. Conclusions Three non-catalytic residues within (HUMAN)NAT1*4 (F125, R127 and Y129) contribute both to substrate recognition and inhibitor binding by participating in distinctive intermolecular interactions and maintaining the steric conformation of the catalytic pocket. These active site residues contribute to the definition of substrate and inhibitor selectivity, an understanding of which is essential for facilitating the design of second generation (HUMAN)NAT1-selective inhibitors for diagnostic, prognostic and therapeutic purposes. In particular, since the expression of (HUMAN)NAT1 is related to the development and progression of oestrogen-receptor-positive breast cancer, these structure-based tools will facilitate the ongoing design of candidate compounds for use in (HUMAN)NAT1-positive breast tumours. Electronic supplementary material The online version of this article (doi:10.1186/2050-6511-15-68) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edith Sim
- Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Mathieu C, Duval R, Xu X, Rodrigues-Lima F, Dupret JM. Effects of pesticide chemicals on the activity of metabolic enzymes: focus on thiocarbamates. Expert Opin Drug Metab Toxicol 2014; 11:81-94. [PMID: 25391334 DOI: 10.1517/17425255.2015.975691] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Thiocarbamates are chemicals widely used as pesticides. Occupational exposure is associated with acute intoxication. Populations can be exposed through food and water. Moreover, certain thiocarbamates are used clinically. The widespread use of thiocarbamates raises many issues regarding their toxicological and pharmacological impact. AREAS COVERED Thiocarbamates and their metabolites can modify biological macromolecules functions, in particular enzymes, through modification of cysteine residues, chelation of metal ions or modulation of the oxidative stress. Loss of enzyme activity can lead to the disruption of metabolic pathways, and explain, at least in part, the effects of these pesticides. Additionally, their reactivity and ability to easily cross biological barrier confer them a great interest for development of clinical applications. EXPERT OPINION Many advances in the study of thiocarbamates metabolism and reactivity have led to a better knowledge of biological effects of these compounds. However, more data are needed on the determination of targets and specificity. Only few data concerning the exposure to a cocktail of pesticides/chemicals are available, raising the need to evaluate the toxic side effects of representative pesticides mixtures. Moreover, the dithiocarbamate Disulfiram has shown great potential in therapeutic applications and leads to the development of pharmacological thiocarbamates derivatives, highly specific to their target and easily distributed.
Collapse
Affiliation(s)
- Cécile Mathieu
- Paris Diderot University, Life Sciences , Paris , France
| | | | | | | | | |
Collapse
|
39
|
Sedigh-Ardekani M, Saadat M. Evaluation of chromosomal aberrations induced by hydralazine in Chinese hamster ovary cells. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2014. [DOI: 10.1016/j.ejmhg.2014.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
40
|
PharmGKB summary: very important pharmacogene information for N-acetyltransferase 2. Pharmacogenet Genomics 2014; 24:409-25. [PMID: 24892773 DOI: 10.1097/fpc.0000000000000062] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Salazar-González R, Gómez R, Romano-Moreno S, Medellín-Garibay S, Núñez-Ruíz A, Magaña-Aquino M, Milán-Segovia RC, Portales-Pérez DP. Expression of NAT2 in immune system cells and the relation of NAT2 gene polymorphisms in the anti-tuberculosis therapy in Mexican mestizo population. Mol Biol Rep 2014; 41:7833-43. [PMID: 25163630 DOI: 10.1007/s11033-014-3677-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/19/2014] [Indexed: 10/24/2022]
Abstract
Arylamine N-acetyltransferase 2 (NAT2) metabolizes isoniazid (INH) and Single Nucleotide Polymorphisms (SNP) responsible for its activity has been reported. The aim of this study in the Mexican mestizo population was to evaluate NAT2 expression at the protein level in immune cells, as well as the distribution and frequency of six NAT2 SNPs and their association with anti-TB therapy, by measuring the plasma levels of INH and Acetyl-INH (AcINH). We performed genotyping assays of NAT2 SNPs in 40 TB patients and 121 healthy volunteers by real-time PCR. A method for detecting NAT2 in immune cells using flow cytometry was developed. Plasma concentrations of INH and AcINH were obtained by HPLC in TB patients and the Metabolic Ratio (MR) was calculated. The phenotypes obtained in the healthy volunteers were as follows; 18.87 % of subjects had the rapid acetylator phenotype, 45.45 % had the intermediate phenotype and 39.66 % exhibited the slow acetylator phenotype. In the TB patient group, 35 % of patients had the rapid acetylator phenotype, 32.5 % were intermediate and 32.5 % showed the slow acetylator phenotype. A higher expression level of NAT2 in innate immune cells from TB patients compared to those from healthy volunteers was detected (P < 0.013). In TB patients the MR showed a bimodal distribution with an antimode of 0.7, which was used as a threshold value for acetylator classification. A high correspondence between the rapid and slow acetylator phenotype with MR was demonstrated. In conclusion, the 282C>T, 341T>C, 481C>T, 590G>A, 803A>G, 857G>A SNPs of NAT2 gene provides accurate for prediction of the acetylator phenotype in Mexican mestizo population. A statistical difference was found in frequency of rapid metabolizer phenotype, which was higher in TB patients. In addition, the expression of NAT2 protein in immune cells can lead to further studies related to its functional role in the innate immune response against M. tuberculosis and other xenobiotics metabolized by this enzyme.
Collapse
Affiliation(s)
- R Salazar-González
- Laboratorio de Biofarmacia y Farmacocinética, Facultad de Ciencias Químicas, UASLP, San Luis Potosí, SLP, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Vilčková M, Jurečeková J, Dobrota D, Habalová V, Klimčáková L, Waczulíková I, Slezák P, Kliment J, Sivoňová MK. Variation in N-acetyltransferase 2 (NAT2), smoking and risk of prostate cancer in the Slovak population. Med Oncol 2014; 31:987. [PMID: 24816842 DOI: 10.1007/s12032-014-0987-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/27/2014] [Indexed: 12/14/2022]
Abstract
N-acetyltransferase 2 (NAT2) is an enzyme involved in the biotransformation of xenobiotics, mainly aromatic and heterocyclic amines and hydrazines, all of which represent an important class of carcinogens found in tobacco smoke. Polymorphism in NAT2 gene is reported to be associated with susceptibility to various types of cancer. This study investigated the relationship between the NAT2 polymorphism and the risk of prostate cancer with reference to the link between cigarette smoking and the xenobiotic-metabolizing enzyme NAT2. Overall, 281 cases and 395 controls from Slovakia were studied using polymerase chain reaction-restriction fragment length polymorphism assay. We found no statistically significant association between NAT2 genotypes and prostate cancer risk (slow acetylation vs. rapid acetylation: OR 1.13; 95 % CI 0.83-1.55). We report here a statistically significant correlation between the NAT2*5C/NAT2*6A slow acetylator genotype and the risk for developing prostate cancer (OR 2.91; 95 % CI 1.43-5.94; p = 0.003) when compared with the rapid phenotype. Smokers with NAT2 rapid phenotype had a five percent (5 %) reduced risk of prostate cancer compared with non-smokers carrying the rapid acetylator genotype. The association was reversed among smokers and non-smokers with NAT2 slow phenotype. On the basis of the foregoing, we conclude that the NAT2 phenotypes whether alone or in association with smoking do not correlate with susceptibility to prostate cancer within the Slovak population.
Collapse
Affiliation(s)
- Marta Vilčková
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4, 03601, Martin, Slovakia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Howard JT, Baynes RE, Brooks JD, Yeatts JL, Bellis B, Ashwell MS, Routh P, O'Nan AT, Maltecca C. The effect of breed and sex on sulfamethazine, enrofloxacin, fenbendazole and flunixin meglumine pharmacokinetic parameters in swine. J Vet Pharmacol Ther 2014; 37:531-41. [PMID: 24731191 DOI: 10.1111/jvp.12128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/28/2014] [Indexed: 12/01/2022]
Abstract
Drug use in livestock has received increased attention due to welfare concerns and food safety. Characterizing heterogeneity in the way swine populations respond to drugs could allow for group-specific dose or drug recommendations. Our objective was to determine whether drug clearance differs across genetic backgrounds and sex for sulfamethazine, enrofloxacin, fenbendazole and flunixin meglumine. Two sires from each of four breeds were mated to a common sow population. The nursery pigs generated (n = 114) were utilized in a random crossover design. Drugs were administered intravenously and blood collected a minimum of 10 times over 48 h. A non-compartmental analysis of drug and metabolite plasma concentration vs. time profiles was performed. Within-drug and metabolite analysis of pharmacokinetic parameters included fixed effects of drug administration date, sex and breed of sire. Breed differences existed for flunixin meglumine (P-value<0.05; Cl, Vdss ) and oxfendazole (P-value<0.05, AUC0→∞ ). Sex differences existed for oxfendazole (P-value < 0.05; Tmax ) and sulfamethazine (P-value < 0.05, Cl). Differences in drug clearance were seen, and future work will determine the degree of additive genetic variation utilizing a larger population.
Collapse
Affiliation(s)
- J T Howard
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gorain B, Chakraborty S, Pal MM, Sarkar R, Samanta SK, Karmakar S, Sen T. Arylamine N-acetyl Transferase (NAT) in the blue secretion of Telescopium telescopium: xenobiotic metabolizing enzyme as a biomarker for detection of environmental pollution. SPRINGERPLUS 2014; 3:666. [PMID: 26034680 PMCID: PMC4447851 DOI: 10.1186/2193-1801-3-666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/24/2014] [Indexed: 11/25/2022]
Abstract
Telescopium telescopium, a marine mollusc collected from Sundarban mangrove, belongs to the largest mollusca phylum in the world and exudes a blue secretion when stimulated mechanically. The blue secretion was found to metabolize (preferentially) para-amino benzoic acid, a substrate for N-acetyl transferase (NAT), thereby indicating acetyl transferase like activity of the secretion. Attempts were also made to characterise bioactive fraction of the blue secretion and to further use this as a biomarker for monitoring of marine pollution. NAT like enzyme from marine mollusc is a potential candidate for detoxification of different harmful chemicals. A partially purified extract of blue secretion was obtained by fractional precipitation with (NH4)2SO4. From different fractions obtained by precipitation, the 0–30% fraction (30S) displayed NAT like activity (using para amino benzoic acid as a substrate with para nitrophenyl phosphate or acetyl coenzyme A as acetyl group donors). Maximum NAT like enzyme activity was attained at 25°C and at a pH of 6. The enzyme activity was found to be inhibited by 5 mM phenyl methyl sulfonyl fluoride. The divalent metal ions reduced NAT like activity of 30S. Moreover, Cu2+ and Zn2+ (at concentration of 1 mM) completely inhibited NAT activity. The thermal stability and bench-top stability studies were performed and it was found that the enzyme was stable at room temperature for more than 24 hours. Results from the present study further indicate that heavy metal content in blue secretion gradually decreased from pre-monsoon to post-monsoon season, which also corresponded to the change in NAT like activity. Therefore, this article stresses the importance of biomarker research for monitoring pollution.
Collapse
Affiliation(s)
- Bapi Gorain
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Sumon Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Murari Mohan Pal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Ratul Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India ; School of Natural Product Studies, Jadavpur University, Kolkata, 700032 India
| | - Samir Kumar Samanta
- Calcutta Institute of Pharmaceutical Technology and AHS, Banitabla, Ulluberia, Howrah 711316 India
| | - Sanmoy Karmakar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Tuhinadri Sen
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India ; School of Natural Product Studies, Jadavpur University, Kolkata, 700032 India
| |
Collapse
|
45
|
Molecular characterization of a novel N-acetyltransferase from Chryseobacterium sp. Appl Environ Microbiol 2013; 80:1770-6. [PMID: 24375143 DOI: 10.1128/aem.03449-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
N-Acetyltransferase from Chryseobacterium sp. strain 5-3B is an acetyl coenzyme A (acetyl-CoA)-dependent enzyme that catalyzes the enantioselective transfer of an acetyl group from acetyl-CoA to the amino group of l-2-phenylglycine to produce (2S)-2-acetylamino-2-phenylacetic acid. We purified the enzyme from strain 5-3B and deduced the N-terminal amino acid sequence. The gene, designated natA, was cloned with two other hypothetical protein genes; the three genes probably form a 2.5-kb operon. The deduced amino acid sequence of NatA showed high levels of identity to sequences of putative N-acetyltransferases of Chryseobacterium spp. but not to other known arylamine and arylalkylamine N-acetyltransferases. Phylogenetic analysis indicated that NatA forms a distinct lineage from known N-acetyltransferases. We heterologously expressed recombinant NatA (rNatA) in Escherichia coli and purified it. rNatA showed high activity for l-2-phenylglycine and its chloro- and hydroxyl-derivatives. The Km and Vmax values for l-2-phenylglycine were 0.145 ± 0.026 mM and 43.6 ± 2.39 μmol · min(-1) · mg protein(-1), respectively. The enzyme showed low activity for 5-aminosalicylic acid and 5-hydroxytryptamine, which are reported as good substrates of a known arylamine N-acetyltransferase and an arylalkylamine N-acetyltransferase. rNatA had a comparatively broad acyl donor specificity, transferring acyl groups to l-2-phenylglycine and producing the corresponding 2-acetylamino-2-phenylacetic acids (relative activity with acetyl donors acetyl-CoA, propanoyl-CoA, butanoyl-CoA, pentanoyl-CoA, and hexanoyl-CoA, 100:108:122:10:<1).
Collapse
|
46
|
Khlifi R, Messaoud O, Rebai A, Hamza-Chaffai A. Polymorphisms in the human cytochrome P450 and arylamine N-acetyltransferase: susceptibility to head and neck cancers. BIOMED RESEARCH INTERNATIONAL 2013; 2013:582768. [PMID: 24151610 PMCID: PMC3787584 DOI: 10.1155/2013/582768] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 06/25/2013] [Accepted: 07/24/2013] [Indexed: 12/12/2022]
Abstract
The occurrence of head and neck cancer (HNC) is associated with smoking and alcohol drinking. Tobacco smoking exposes smokers to a series of carcinogenic chemicals. Cytochrome P450 enzymes (CYP450s), such as CYP1A1, CYP1B1, and CYP2D6, usually metabolize carcinogens to their inactive derivatives, but they occasionally convert the chemicals to more potent carcinogens. In addition, via CYP450 (CYP2E1) oxidase, alcohol is metabolized to acetaldehyde, a highly toxic compound, which plays an important role in carcinogenesis. Furthermore, two N-acetyltransferase isozymes (NATs), NAT1 and NAT2, are polymorphic and catalyze both N-acetylation and O-acetylation of aromatic and heterocyclic amine carcinogens. Genetic polymorphisms are associated with a number of enzymes involved in the metabolism of carcinogens important in the induction of HNC. It has been suggested that such polymorphisms may be linked to cancer susceptibility. In this paper, we select four cytochrome P450 enzymes (CYP1A1, CYP1BA1, CYP2D6, and CYP2E1), and two N-acetyltransferase isozymes (NAT1 and NAT2) in order to summarize and analyze findings from the literature related to HNC risk by focusing on (i) the interaction between these genes and the environment, (ii) the impact of genetic defect on protein activity and/or expression, and (iii) the eventual involvement of race in such associations.
Collapse
Affiliation(s)
- Rim Khlifi
- Research Unit on Toxicology and Environment, Sfax University, 3018 Sfax, Tunisia
- Bioinformatics Unit, Centre of Biotechnology of Sfax, Sfax University, 3018 Sfax, Tunisia
| | - Olfa Messaoud
- Biomedical Genomics and Oncogenetics Laboratory LR11IPT05, University of Tunis El Manar, 1002 Tunis, Tunisia
| | - Ahmed Rebai
- Bioinformatics Unit, Centre of Biotechnology of Sfax, Sfax University, 3018 Sfax, Tunisia
| | - Amel Hamza-Chaffai
- Research Unit on Toxicology and Environment, Sfax University, 3018 Sfax, Tunisia
| |
Collapse
|
47
|
Kurogi K, Liu TA, Sakakibara Y, Suiko M, Liu MC. The use of zebrafish as a model system for investigating the role of the SULTs in the metabolism of endogenous compounds and xenobiotics. Drug Metab Rev 2013; 45:431-40. [DOI: 10.3109/03602532.2013.835629] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
48
|
Bui LC, Manaa A, Xu X, Duval R, Busi F, Dupret JM, Rodrigues-Lima F, Dairou J. Acrolein, an α,β-unsaturated aldehyde, irreversibly inhibits the acetylation of aromatic amine xenobiotics by human arylamine N-acetyltransferase 1. Drug Metab Dispos 2013; 41:1300-5. [PMID: 23633528 DOI: 10.1124/dmd.113.052258] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Acrolein is an electrophilic α,β-unsaturated aldehyde of industrial, pharmaceutic, and toxicologic importance to which we are exposed in environmental, occupational, and therapeutic situations. Acrolein is known to exert different biologic effects through reactions with cellular macromolecules such as DNA, certain proteins, or glutathione. In many situations (such as in tobacco smoke or other fumes), exposure to acrolein occurs concomitantly with other compounds such as aromatic amine chemicals. Interestingly, it has been shown that acrolein could impact the cellular metabolism of aromatic xenobiotics through an indirect mechanism based on the transcriptional induction of phase II xenobiotic-metabolizing enzymes. Here we report a novel mechanism by which acrolein acts on the metabolism of aromatic foreign chemicals. We provide molecular, kinetic, and cellular evidence that acrolein can react directly and irreversibly with arylamine N-acetyltransferases, a major family of xenobiotic-metabolizing enzymes involved in the metabolization of aromatic amine chemicals. Formation of an acrolein adduct with a catalytic cysteine residue in the active site is responsible for the impairment of aromatic amine acetylation by the enzyme. This biochemical process may represent an additional mechanism by which acrolein impacts the metabolism and fate of aromatic amine drugs and pollutants.
Collapse
Affiliation(s)
- Linh C Bui
- Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC 4413, 75013, Paris. France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kubiak X, Li de la Sierra-Gallay I, Chaffotte AF, Pluvinage B, Weber P, Haouz A, Dupret JM, Rodrigues-Lima F. Structural and biochemical characterization of an active arylamine N-acetyltransferase possessing a non-canonical Cys-His-Glu catalytic triad. J Biol Chem 2013; 288:22493-505. [PMID: 23770703 DOI: 10.1074/jbc.m113.468595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Arylamine N-acetyltransferases (NATs), a class of xenobiotic-metabolizing enzymes, catalyze the acetylation of aromatic amine compounds through a strictly conserved Cys-His-Asp catalytic triad. Each residue is essential for catalysis in both prokaryotic and eukaryotic NATs. Indeed, in (HUMAN)NAT2 variants, mutation of the Asp residue to Asn, Gln, or Glu dramatically impairs enzyme activity. However, a putative atypical NAT harboring a catalytic triad Glu residue was recently identified in Bacillus cereus ((BACCR)NAT3) but has not yet been characterized. We report here the crystal structure and functional characterization of this atypical NAT. The overall fold of (BACCR)NAT3 and the geometry of its Cys-His-Glu catalytic triad are similar to those present in functional NATs. Importantly, the enzyme was found to be active and to acetylate prototypic arylamine NAT substrates. In contrast to (HUMAN) NAT2, the presence of a Glu or Asp in the triad of (BACCR)NAT3 did not significantly affect enzyme structure or function. Computational analysis identified differences in residue packing and steric constraints in the active site of (BACCR)NAT3 that allow it to accommodate a Cys-His-Glu triad. These findings overturn the conventional view, demonstrating that the catalytic triad of this family of acetyltransferases is plastic. Moreover, they highlight the need for further study of the evolutionary history of NATs and the functional significance of the predominant Cys-His-Asp triad in both prokaryotic and eukaryotic forms.
Collapse
Affiliation(s)
- Xavier Kubiak
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC4413, 75013 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Rapid birth-and-death evolution of the xenobiotic metabolizing NAT gene family in vertebrates with evidence of adaptive selection. BMC Evol Biol 2013; 13:62. [PMID: 23497148 PMCID: PMC3601968 DOI: 10.1186/1471-2148-13-62] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/27/2013] [Indexed: 11/10/2022] Open
Abstract
Background The arylamine N-acetyltransferases (NATs) are a unique family of enzymes widely distributed in nature that play a crucial role in the detoxification of aromatic amine xenobiotics. Considering the temporal changes in the levels and toxicity of environmentally available chemicals, the metabolic function of NATs is likely to be under adaptive evolution to broaden or change substrate specificity over time, making NATs a promising subject for evolutionary analyses. In this study, we trace the molecular evolutionary history of the NAT gene family during the last ~450 million years of vertebrate evolution and define the likely role of gene duplication, gene conversion and positive selection in the evolutionary dynamics of this family. Results A phylogenetic analysis of 77 NAT sequences from 38 vertebrate species retrieved from public genomic databases shows that NATs are phylogenetically unstable genes, characterized by frequent gene duplications and losses even among closely related species, and that concerted evolution only played a minor role in the patterns of sequence divergence. Local signals of positive selection are detected in several lineages, probably reflecting response to changes in xenobiotic exposure. We then put a special emphasis on the study of the last ~85 million years of primate NAT evolution by determining the NAT homologous sequences in 13 additional primate species. Our phylogenetic analysis supports the view that the three human NAT genes emerged from a first duplication event in the common ancestor of Simiiformes, yielding NAT1 and an ancestral NAT gene which in turn, duplicated in the common ancestor of Catarrhini, giving rise to NAT2 and the NATP pseudogene. Our analysis suggests a main role of purifying selection in NAT1 protein evolution, whereas NAT2 was predicted to mostly evolve under positive selection to change its amino acid sequence over time. These findings are consistent with a differential role of the two human isoenzymes and support the involvement of NAT1 in endogenous metabolic pathways. Conclusions This study provides unequivocal evidence that the NAT gene family has evolved under a dynamic process of birth-and-death evolution in vertebrates, consistent with previous observations made in fungi.
Collapse
|