1
|
Cashman JR. Practical Aspects of Flavin-Containing Monooxygenase-Mediated Metabolism. Chem Res Toxicol 2024. [PMID: 39485380 DOI: 10.1021/acs.chemrestox.4c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Hepatic flavin-containing monooxygenase 3 (FMO3) is arguably the most important FMO in humans from the standpoint of drug metabolism. Recently, adult hepatic FMO3 has been linked to several conditions including cardiometabolic diseases, aging, obesity, and atherosclerosis in small animals. Despite the importance of FMO3 in drug and chemical metabolism, relative to cytochrome P-450 (CYP), fewer studies have been published describing drug and chemical metabolism. This may be due to the properties of human hepatic FMO3. For example, FMO3 is thermally labile, and often methods reported in the study of human hepatic FMO3 are not optimal. Herein, I describe some practical aspects for studying human hepatic FMO3 and other FMOs.
Collapse
Affiliation(s)
- John R Cashman
- Human BioMolecular Research Institute. 6351 Nancy Ridge Road, Suite B, San Diego, California 92121, United States
| |
Collapse
|
2
|
Maksymiuk KM, Szudzik M, Samborowska E, Chabowski D, Konop M, Ufnal M. Mice, rats, and guinea pigs differ in FMOs expression and tissue concentration of TMAO, a gut bacteria-derived biomarker of cardiovascular and metabolic diseases. PLoS One 2024; 19:e0297474. [PMID: 38266015 PMCID: PMC10807837 DOI: 10.1371/journal.pone.0297474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/06/2024] [Indexed: 01/26/2024] Open
Abstract
INTRODUCTION Increased plasma trimethylamine oxide (TMAO) is observed in cardiovascular and metabolic diseases, originating from the gut microbiota product, trimethylamine (TMA), via flavin-containing monooxygenases (FMOs)-dependent oxidation. Numerous studies have investigated the association between plasma TMAO and various pathologies, yet limited knowledge exists regarding tissue concentrations of TMAO, TMAO precursors, and interspecies variability. METHODS Chromatography coupled with mass spectrometry was employed to evaluate tissue concentrations of TMAO and its precursors in adult male mice, rats, and guinea pigs. FMO mRNA and protein levels were assessed through PCR and Western blot, respectively. RESULTS Plasma TMAO levels were similar among the studied species. However, significant differences in tissue concentrations of TMAO were observed between mice, rats, and guinea pigs. The rat renal medulla exhibited the highest TMAO concentration, while the lowest was found in the mouse liver. Mice demonstrated significantly higher plasma TMA concentrations compared to rats and guinea pigs, with the highest TMA concentration found in the mouse renal medulla and the lowest in the rat lungs. FMO5 exhibited the highest expression in mouse liver, while FMO3 was highly expressed in rats. Guinea pigs displayed low expression of FMOs in this tissue. CONCLUSION Despite similar plasma TMAO levels, mice, rats, and guinea pigs exhibited significant differences in tissue concentrations of TMA, TMAO, and FMO expression. These interspecies variations should be considered in the design and interpretation of experimental studies. Furthermore, these findings may suggest a diverse importance of the TMAO pathway in the physiology of the evaluated species.
Collapse
Affiliation(s)
- Klaudia M. Maksymiuk
- Laboratory of the Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Szudzik
- Laboratory of the Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| | - Emilia Samborowska
- Mass spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Dawid Chabowski
- Laboratory of the Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Konop
- Laboratory of the Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Ufnal
- Laboratory of the Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
Steinke I, Ghanei N, Govindarajulu M, Yoo S, Zhong J, Amin RH. Drug Discovery and Development of Novel Therapeutics for Inhibiting TMAO in Models of Atherosclerosis and Diabetes. Front Physiol 2020; 11:567899. [PMID: 33192565 PMCID: PMC7658318 DOI: 10.3389/fphys.2020.567899] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus exists as a comorbidity with congestive heart failure (CHF). However, the exact molecular signaling mechanism linking CHF as the major form of mortality from diabetes remains unknown. Type 2 diabetic patients display abnormally high levels of metabolic products associated with gut dysbiosis. One such metabolite, trimethylamine N-oxide (TMAO), has been observed to be directly related with increased incidence of cardiovascular diseases (CVD) in human patients. TMAO a gut-liver metabolite, comes from the metabolic degenerative product trimethylamine (TMA) that is produced from gut microbial metabolism. Elevated levels of TMAO in diabetics and obese patients are observed to have a direct correlation with increased risk for major adverse cardiovascular events. The pro-atherogenic effect of TMAO is attributed to enhancing inflammatory pathways with cholesterol and bile acid dysregulation, promoting foam cell formation. Recent studies have revealed several potential therapeutic strategies for reducing TMAO levels and will be the central focus for the current review. However, few have focused on developing rational drug therapeutics and may be due to the gaps in knowledge for understanding the mechanism by which microbial TMA producing enzymes and hepatic flavin-containing monoxygenase (FMO) can work together in preventing elevation of TMAO levels. Therefore, it is critical to understand the advantages of developing a novel rational drug design strategy that manipulates FMO production of TMAO and TMA production by microbial enzymes. This review will focus on the inspection of FMO manipulation, as well as gut microbiota dysbiosis and its influence on metabolic disorders including cardiovascular disease and describe novel potential pharmacological therapeutic development.
Collapse
Affiliation(s)
- Ian Steinke
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Nila Ghanei
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Manoj Govindarajulu
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Sieun Yoo
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Juming Zhong
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Rajesh H Amin
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| |
Collapse
|
4
|
Zhang S, Xu Z, Cao X, Xie Y, Lin L, Zhang X, Zou B, Liu D, Cai Y, Liao Q, Xie Z. Shenling Baizhu San improves functional dyspepsia in rats as revealed by 1H-NMR based metabolomics. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:2363-2375. [PMID: 32930262 DOI: 10.1039/d0ay00580k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Functional dyspepsia (FD), a common gastrointestinal disorder around the world, is driven by multiple factors, making prevention and treatment a major challenge. Shenling Baizhu San (SBS), a classical prescription of traditional Chinese medicine, has been proven to be effective in gastrointestinal disorders. However, studies on SBS improving FD are few. Thus, our study aimed to evaluate the effect of SBS on FD and further to explore the mechanism underlying the interactions between FD and SBS by the metabolomics approach. A FD rat model was induced by multiple forms of mild stimulation, and proton nuclear magnetic resonance (1H-NMR) spectroscopy and multivariate data analysis were used to profile the fecal and urinary metabolome in the FD rats during SBS intervention. Significant dyspeptic symptoms such as weight loss, poor appetite, reduced gastrointestinal motility and decreased absorptive capacity were observed in the FD rats, which were subsequently improved by SBS. Additionally, the levels of citrate, branched chain acids and pyruvate decreased, and the levels of choline, trimethylamine and taurine increased in the FD rats. Furthermore, the metabolic disorders were amended with SBS intervention mainly by modulating the metabolic pathways involved in energy metabolism, amino acid metabolism, and gut microbiota and host co-metabolism. Overall, our study highlighted the effect of SBS on the disturbed metabolic pathways in the FD rats, providing new insight into the mechanism of SBS treatment for FD from the perspective of metabolomics.
Collapse
Affiliation(s)
- Shaobao Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| | - Zengmei Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Xueqing Cao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yuzhen Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Lei Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| | - Xiao Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Baorong Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Deliang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Ying Cai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Qiongfeng Liao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China
| |
Collapse
|
5
|
Sakurai E. [Elucidation of New Function in Endothelial Cells for Efficient Delivery Strategy of Drug to Tissues]. YAKUGAKU ZASSHI 2020; 140:51-62. [PMID: 31902886 DOI: 10.1248/yakushi.19-00179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The author has described two new functions of endothelial cells for efficient delivery of drugs to tissues. First, it was indicated that tight junction (TJ)-associated protein, claudin-1, exerts potent paracellular barrier function in cultured mouse lung microvascular endothelial cells (LMECs). This barrier was instantly and reversibly opened by reduction of TJ proteins expression via histamine H1 and H2 receptors. Histamine was biosynthesized by l-histidine decarboxylase from uptaken l-histidine, and biotransformed by type B of monoamine oxidase, suggesting that histamine concentration is controlled in rat brain MECs (BMECs) and LMECs. Moreover, uptake of l-histidine into BMECs and LMECs markedly increased with addition of ZnSO4. Second, it was suggested that drug-metabolizing enzymes such as CYP and flavin-containing monooxygenase exist in vascular endothelial cells exposed to blood and to aerobic conditions. These cells have the same ability to metabolize drugs as hepatocytes, demonstrating that vascular endothelial cells are a metabolic barrier against tissue transfer of drugs. From these results, it was suggested that reversible opening of TJ and selective inhibition of drug metabolism in vascular endothelial cells may be efficient delivery strategies of drugs to tissues. Finally, I hope that this research will lead to development of new drugs and possible re-evaluation of discontinued drugs.
Collapse
Affiliation(s)
- Eiichi Sakurai
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
6
|
Comparison between the enzymatic activity, structure and substrate binding of mouse and human lecithin retinol acyltransferase. Biochem Biophys Res Commun 2019; 519:832-837. [PMID: 31561851 DOI: 10.1016/j.bbrc.2019.09.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 11/22/2022]
Abstract
Lecithin retinol acyltransferase (LRAT) is involved in the visual cycle where it catalyzes the formation of all-trans retinyl ester. The mouse animal model has been widely used to study LRAT. Primary sequence alignment shows 80% identity and 90% similarity between human and mouse LRAT. However, human LRAT has a proline at position 173 (hLRAT (P173)) while an arginine can be found at this position for the mouse protein (mLRAT (R173)). Moreover, residue 173 is important for the human protein since a substitution mutation of this residue to a leucine (P173L-hLRAT) caused night blindness in a patient. The present study was thus undertaken to determine whether mouse and human LRAT have a similar enzymatic activity, structure and substrate binding affinity using a truncated form of LRAT (tLRAT). The enzymatic activity and binding affinity to the substrate, all-trans retinol, of mtLRAT (R173) were found to be 2.7- and 3.9-fold lower, respectively, than that of htLRAT (P173). Moreover, the enzymatic activity of P173L-htLRAT is 6.3-fold lower compared to that of htLRAT (P173). Furthermore, a significant difference was observed between the intrinsic fluorescence emission as well as between the circular dichroism spectra of mtLRAT (R173) and htLRAT (P173). In addition, mtLRAT proteins are less thermostable than htLRAT proteins, which suggests that structural differences exist between the mouse and human proteins. Altogether, these data strongly suggest that the much lower catalytic activity of mtLRAT (R173) compared to that of htLRAT (P173) mostly results from differences between their structure, predominantly revealed by their dissimilar thermal stability, as well as their efficiency to bind all-trans retinol. Therefore, conclusions regarding the behavior of human LRAT based on measurements performed with mouse LRAT must be made with caution. Also, the much lower enzymatic activity of P173L-htLRAT compared to that of htLRAT (P173) might explain the night blindness of a patient carrying this mutation.
Collapse
|
7
|
Tian X, Zhao S, Guo Z, Hu B, Wei Q, Tang Y, Su J. Molecular characterization, expression pattern and metabolic activity of flavin-dependent monooxygenases in Spodoptera exigua. INSECT MOLECULAR BIOLOGY 2018; 27:533-544. [PMID: 29749684 DOI: 10.1111/imb.12392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Enhanced detoxification is one of the important mechanisms for insecticide resistance. Most research in this field to date has focused on the role of cytochrome P450s. Our previous work revealed that flavin-dependent monooxygenases (FMOs) were involved in metabolic resistance of Spodoptera exigua. In the present study we investigated the molecular characteristics, expression patterns and oxidative activities of SeFMO on insecticides. Three FMO genes, which encode proteins with the typical FMO motifs, were cloned from S. exigua. The oxidative activities of eukaryotically expressed SeFMO enzymes were verified with the model substrate of FMO. Importantly, the SeFMOs had significantly higher oxidative activities on metaflumizone and lambda-cyhalothrin than on model substrates and other insecticides tested. The three SeFMOs were mainly expressed in the midgut, fat body and Malpighian tubules. The tissues responsible for xenobiotic metabolism and their expression characteristics were similar to those of P450s acting as detoxification genes. The study also revealed that the expression of SeFMOs could be induced by insecticide exposure, and that SeFMOs were over-expressed in a metaflumizone-resistant strain of S. exigua. These results suggest that SeFMOs are important insecticide detoxifying enzymes, and that over-expression of FMO genes may be one of the mechanisms for metabolic resistance in S. exigua.
Collapse
Affiliation(s)
- X Tian
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - S Zhao
- Zoonbio Biotechnology Co., Ltd, Nanjing, China
| | - Z Guo
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - B Hu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Q Wei
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Y Tang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - J Su
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
8
|
Hoyles L, Jiménez-Pranteda ML, Chilloux J, Brial F, Myridakis A, Aranias T, Magnan C, Gibson GR, Sanderson JD, Nicholson JK, Gauguier D, McCartney AL, Dumas ME. Metabolic retroconversion of trimethylamine N-oxide and the gut microbiota. MICROBIOME 2018; 6:73. [PMID: 29678198 PMCID: PMC5909246 DOI: 10.1186/s40168-018-0461-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/13/2018] [Indexed: 05/24/2023]
Abstract
BACKGROUND The dietary methylamines choline, carnitine, and phosphatidylcholine are used by the gut microbiota to produce a range of metabolites, including trimethylamine (TMA). However, little is known about the use of trimethylamine N-oxide (TMAO) by this consortium of microbes. RESULTS A feeding study using deuterated TMAO in C57BL6/J mice demonstrated microbial conversion of TMAO to TMA, with uptake of TMA into the bloodstream and its conversion to TMAO. Microbial activity necessary to convert TMAO to TMA was suppressed in antibiotic-treated mice, with deuterated TMAO being taken up directly into the bloodstream. In batch-culture fermentation systems inoculated with human faeces, growth of Enterobacteriaceae was stimulated in the presence of TMAO. Human-derived faecal and caecal bacteria (n = 66 isolates) were screened on solid and liquid media for their ability to use TMAO, with metabolites in spent media analysed by 1H-NMR. As with the in vitro fermentation experiments, TMAO stimulated the growth of Enterobacteriaceae; these bacteria produced most TMA from TMAO. Caecal/small intestinal isolates of Escherichia coli produced more TMA from TMAO than their faecal counterparts. Lactic acid bacteria produced increased amounts of lactate when grown in the presence of TMAO but did not produce large amounts of TMA. Clostridia (sensu stricto), bifidobacteria, and coriobacteria were significantly correlated with TMA production in the mixed fermentation system but did not produce notable quantities of TMA from TMAO in pure culture. CONCLUSIONS Reduction of TMAO by the gut microbiota (predominantly Enterobacteriaceae) to TMA followed by host uptake of TMA into the bloodstream from the intestine and its conversion back to TMAO by host hepatic enzymes is an example of metabolic retroconversion. TMAO influences microbial metabolism depending on isolation source and taxon of gut bacterium. Correlation of metabolomic and abundance data from mixed microbiota fermentation systems did not give a true picture of which members of the gut microbiota were responsible for converting TMAO to TMA; only by supplementing the study with pure culture work and additional metabolomics was it possible to increase our understanding of TMAO bioconversions by the human gut microbiota.
Collapse
Affiliation(s)
- Lesley Hoyles
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Maria L. Jiménez-Pranteda
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Julien Chilloux
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Francois Brial
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Antonis Myridakis
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Thomas Aranias
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Christophe Magnan
- Sorbonne Paris Cité, Université Denis Diderot, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, 75205 Paris, France
| | - Glenn R. Gibson
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Jeremy D. Sanderson
- Department of Gastroenterology, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Jeremy K. Nicholson
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Dominique Gauguier
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Anne L. McCartney
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Marc-Emmanuel Dumas
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| |
Collapse
|
9
|
Zhang T, Yang P, Wei J, Li W, Zhong J, Chen H, Cao J. Overexpression of flavin-containing monooxygenase 5 predicts poor prognosis in patients with colorectal cancer. Oncol Lett 2018; 15:3923-3927. [PMID: 29456741 DOI: 10.3892/ol.2018.7724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the expression and clinical significance of flavin-containing monooxygenase 5 (FMO5) in colorectal cancer (CRC). The expression of FMO5 was detected by immunohistochemistry in 208 colon cancer tissues and 8 normal colon tissues. Then, the correlations of FMO5 expression with several clinicopathological features were evaluated. FMO5 mRNA expression from The Cancer Genome Atlas dataset was assessed for further validation. In addition, the association of the expression of FMO5 with prognosis was further evaluated by Kaplan-Meier survival curves and Cox proportional hazards model. The FMO5 protein level in colon cancer tissues was significantly higher than that in normal colon tissues (P<0.001). Overexpression of FMO5 was associated with an advanced clinical stage of cancer (P=0.018) and lymph node metastasis (P=0.03). The TCGA dataset also demonstrated that FMO5 was upregulated in CRC with advanced clinical stage (P=0.047), lymph node metastasis (P=0.045) and distant metastasis (P=0.030). The Kaplan-Meier survival curves showed that higher FMO5 mRNA indicated a shorter overall survival in patients with CRC compared with a low expression of FMO5 (P=0.029). Cox proportional hazards regression revealed that a high FMO5 mRNA level served as an independent prognostic factor for patients with CRC (hazard ratio, 2.865; 95% confidence interval, 1.116-7.355; P=0.029). A high expression of FMO5 may serve roles in colorectal carcinogenesis and distant metastasis. FMO5 may be an independent predictive factor for the prognosis of CRC.
Collapse
Affiliation(s)
- Tong Zhang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Ping Yang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Jianchang Wei
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Wanglin Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Junbin Zhong
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Huacui Chen
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
10
|
Veeravalli S, Karu K, Scott F, Fennema D, Phillips IR, Shephard EA. Effect of Flavin-Containing Monooxygenase Genotype, Mouse Strain, and Gender on Trimethylamine N-oxide Production, Plasma Cholesterol Concentration, and an Index of Atherosclerosis. Drug Metab Dispos 2017; 46:20-25. [PMID: 29070510 PMCID: PMC5733448 DOI: 10.1124/dmd.117.077636] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/19/2017] [Indexed: 11/22/2022] Open
Abstract
The objectives of the study were to determine the contribution, in mice, of members of the flavin-containing monooxygenase (FMO) family to the production of trimethylamine (TMA) N-oxide (TMAO), a potential proatherogenic molecule, and whether under normal dietary conditions differences in TMAO production were associated with changes in plasma cholesterol concentration or with an index of atherosclerosis (Als). Concentrations of urinary TMA and TMAO and plasma cholesterol were measured in 10-week-old male and female C57BL/6J and CD-1 mice and in mouse lines deficient in various Fmo genes (Fmo1−/−, 2−/−, 4−/−, and Fmo5−/−). In female mice most TMA N-oxygenation was catalyzed by FMO3, but in both genders 11%–12% of TMA was converted to TMAO by FMO1. Gender-, Fmo genotype-, and strain-related differences in TMAO production were accompanied by opposite effects on plasma cholesterol concentration. Plasma cholesterol was negatively, but weakly, correlated with TMAO production and urinary TMAO concentration. Fmo genotype had no effect on Als. There was no correlation between Als and either TMAO production or urinary TMAO concentration. Our results indicate that under normal dietary conditions TMAO does not increase plasma cholesterol or act as a proatherogenic molecule.
Collapse
Affiliation(s)
- Sunil Veeravalli
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Kersti Karu
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Flora Scott
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Diede Fennema
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Ian R Phillips
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology (S.V., F.S., D.F., I.R.P., E.A.S.) and Mass Spectrometry Facility, Department of Chemistry (K.K.), University College London, London, United Kingdom; and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| |
Collapse
|
11
|
Tanino T, Bando T, Komada A, Nojiri Y, Okada Y, Ueda Y, Sakurai E. Hepatic Flavin-Containing Monooxygenase 3 Enzyme Suppressed by Type 1 Allergy-Produced Nitric Oxide. Drug Metab Dispos 2017; 45:1189-1196. [DOI: 10.1124/dmd.117.076570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
|
12
|
Scott F, Gonzalez Malagon SG, O'Brien BA, Fennema D, Veeravalli S, Coveney CR, Phillips IR, Shephard EA. Identification of Flavin-Containing Monooxygenase 5 (FMO5) as a Regulator of Glucose Homeostasis and a Potential Sensor of Gut Bacteria. Drug Metab Dispos 2017. [PMID: 28646079 PMCID: PMC5539585 DOI: 10.1124/dmd.117.076612] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We have previously identified flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic aging. The aim of the present study was to investigate the role of FMO5 in glucose homeostasis and the impact of diet and gut flora on the phenotype of mice in which the Fmo5 gene has been disrupted (Fmo5−/− mice). In comparison with wild-type (WT) counterparts, Fmo5−/− mice are resistant to age-related changes in glucose homeostasis and maintain the higher glucose tolerance and insulin sensitivity characteristic of young animals. When fed a high-fat diet, they are protected against weight gain and reduction of insulin sensitivity. The phenotype of Fmo5−/− mice is independent of diet and the gut microbiome and is determined solely by the host genotype. Fmo5−/− mice have metabolic characteristics similar to those of germ-free mice, indicating that FMO5 plays a role in sensing or responding to gut bacteria. In WT mice, FMO5 is present in the mucosal epithelium of the gastrointestinal tract where it is induced in response to a high-fat diet. In comparison with WT mice, Fmo5−/− mice have fewer colonic goblet cells, and they differ in the production of the colonic hormone resistin-like molecule β. Fmo5−/− mice have lower concentrations of tumor necrosis factor α in plasma and of complement component 3 in epididymal white adipose tissue, indicative of improved inflammatory tone. Our results implicate FMO5 as a regulator of body weight and of glucose disposal and insulin sensitivity and, thus, identify FMO5 as a potential novel therapeutic target for obesity and insulin resistance.
Collapse
Affiliation(s)
- Flora Scott
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sandra G Gonzalez Malagon
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Brett A O'Brien
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Diede Fennema
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Sunil Veeravalli
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Clarissa R Coveney
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Ian R Phillips
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom (F.S., S.G.G.M., B.A.O., D.F., S.V., C.R.C., I.R.P., E.A.S.); and School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom (I.R.P.)
| |
Collapse
|
13
|
Phillips IR, Shephard EA. Drug metabolism by flavin-containing monooxygenases of human and mouse. Expert Opin Drug Metab Toxicol 2016; 13:167-181. [PMID: 27678284 DOI: 10.1080/17425255.2017.1239718] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Flavin-containing monooxygenases (FMOs) play an important role in drug metabolism. Areas covered: We focus on the role of FMOs in the metabolism of drugs in human and mouse. We describe FMO genes and proteins of human and mouse; the catalytic mechanism of FMOs and their significance for drug metabolism; differences between FMOs and CYPs; factors contributing to potential underestimation of the contribution of FMOs to drug metabolism; the developmental and tissue-specific expression of FMO genes and differences between human and mouse; and factors that induce or inhibit FMOs. We discuss the contribution of FMOs of human and mouse to the metabolism of drugs and how genetic variation of FMOs affects drug metabolism. Finally, we discuss the utility of animal models for FMO-mediated drug metabolism in humans. Expert opinion: The contribution of FMOs to drug metabolism may be underestimated. As FMOs are not readily induced or inhibited and their reactions are generally detoxifications, the design of drugs that are metabolized predominantly by FMOs offers clinical advantages. Fmo1(-/-),Fmo2(-/-),Fmo4(-/-) mice provide a good animal model for FMO-mediated drug metabolism in humans. Identification of roles for FMO1 and FMO5 in endogenous metabolism has implications for drug therapy and initiates an exciting area of research.
Collapse
Affiliation(s)
- Ian R Phillips
- a Institute of Structural and Molecular Biology , University College London , London , UK.,b School of Biological and Chemical Sciences , Queen Mary University of London , London , UK
| | - Elizabeth A Shephard
- a Institute of Structural and Molecular Biology , University College London , London , UK
| |
Collapse
|
14
|
Fiorentini F, Geier M, Binda C, Winkler M, Faber K, Hall M, Mattevi A. Biocatalytic Characterization of Human FMO5: Unearthing Baeyer-Villiger Reactions in Humans. ACS Chem Biol 2016; 11:1039-48. [PMID: 26771671 DOI: 10.1021/acschembio.5b01016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Flavin-containing mono-oxygenases are known as potent drug-metabolizing enzymes, providing complementary functions to the well-investigated cytochrome P450 mono-oxygenases. While human FMO isoforms are typically involved in the oxidation of soft nucleophiles, the biocatalytic activity of human FMO5 (along its physiological role) has long remained unexplored. In this study, we demonstrate the atypical in vitro activity of human FMO5 as a Baeyer-Villiger mono-oxygenase on a broad range of substrates, revealing the first example to date of a human protein catalyzing such reactions. The isolated and purified protein was active on diverse carbonyl compounds, whereas soft nucleophiles were mostly non- or poorly reactive. The absence of the typical characteristic sequence motifs sets human FMO5 apart from all characterized Baeyer-Villiger mono-oxygenases so far. These findings open new perspectives in human oxidative metabolism.
Collapse
Affiliation(s)
- Filippo Fiorentini
- Department
of Biology and Biotechnology, University of Pavia, via Ferrata
9, 27100 Pavia, Italy
- Austrian
Centre of Industrial Biotechnology, c/o Institute of Molecular Biotechnology, Graz University of Technology, Petersgasse 14, 8010 Graz, Austria
| | - Martina Geier
- Austrian
Centre of Industrial Biotechnology, c/o Institute of Molecular Biotechnology, Graz University of Technology, Petersgasse 14, 8010 Graz, Austria
| | - Claudia Binda
- Department
of Biology and Biotechnology, University of Pavia, via Ferrata
9, 27100 Pavia, Italy
| | - Margit Winkler
- Austrian
Centre of Industrial Biotechnology, c/o Institute of Molecular Biotechnology, Graz University of Technology, Petersgasse 14, 8010 Graz, Austria
| | - Kurt Faber
- Department
of Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz, Austria
| | - Mélanie Hall
- Department
of Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz, Austria
| | - Andrea Mattevi
- Department
of Biology and Biotechnology, University of Pavia, via Ferrata
9, 27100 Pavia, Italy
| |
Collapse
|
15
|
Gonzalez Malagon SG, Melidoni AN, Hernandez D, Omar BA, Houseman L, Veeravalli S, Scott F, Varshavi D, Everett J, Tsuchiya Y, Timms JF, Phillips IR, Shephard EA. The phenotype of a knockout mouse identifies flavin-containing monooxygenase 5 (FMO5) as a regulator of metabolic ageing. Biochem Pharmacol 2015; 96:267-77. [PMID: 26049045 PMCID: PMC4509511 DOI: 10.1016/j.bcp.2015.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
Abstract
We report the production and metabolic phenotype of a mouse line in which the Fmo5 gene is disrupted. In comparison with wild-type (WT) mice, Fmo5(-/-) mice exhibit a lean phenotype, which is age-related, becoming apparent after 20 weeks of age. Despite greater food intake, Fmo5(-/-) mice weigh less, store less fat in white adipose tissue (WAT), have lower plasma glucose and cholesterol concentrations and enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure, with no increase in physical activity. An increase in respiratory exchange ratio during the dark phase, the period in which the mice are active, indicates a switch from fat to carbohydrate oxidation. In comparison with WT mice, the rate of fatty acid oxidation in Fmo5(-/-) mice is higher in WAT, which would contribute to depletion of lipid stores in this tissue, and lower in skeletal muscle. Five proteins were down regulated in the liver of Fmo5(-/-) mice: aldolase B, ketohexokinase and cytosolic glycerol 3-phosphate dehydrogenase (GPD1) are involved in glucose or fructose metabolism and GPD1 also in production of glycerol 3-phosphate, a precursor of triglyceride biosynthesis; HMG-CoA synthase 1 is involved in cholesterol biosynthesis; and malic enzyme 1 catalyzes the oxidative decarboxylation of malate to pyruvate, in the process producing NADPH for use in lipid and cholesterol biosynthesis. Down regulation of these proteins provides a potential explanation for the reduced fat deposits and lower plasma cholesterol characteristic of Fmo5(-/-) mice. Our results indicate that disruption of the Fmo5 gene slows metabolic ageing via pleiotropic effects.
Collapse
Affiliation(s)
| | - Anna N Melidoni
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Diana Hernandez
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Bilal A Omar
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Lyndsey Houseman
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Sunil Veeravalli
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Flora Scott
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Dorsa Varshavi
- Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
| | - Jeremy Everett
- Medway Metabonomics Research Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
| | - Yugo Tsuchiya
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - John F Timms
- Women's Cancer, Institute for Women's Health, University College London, London WC1E 6BT, UK
| | - Ian R Phillips
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Elizabeth A Shephard
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
16
|
Rudraiah S, Moscovitz JE, Donepudi AC, Campion SN, Slitt AL, Aleksunes LM, Manautou JE. Differential Fmo3 gene expression in various liver injury models involving hepatic oxidative stress in mice. Toxicology 2014; 325:85-95. [PMID: 25193093 DOI: 10.1016/j.tox.2014.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/29/2014] [Accepted: 08/31/2014] [Indexed: 12/12/2022]
Abstract
Flavin-containing monooxygenase-3 (FMO3) catalyzes metabolic reactions similar to cytochrome P450 monooxygenase, however, most metabolites of FMO3 are considered non-toxic. Recent findings in our laboratory demonstrated Fmo3 gene induction following toxic acetaminophen (APAP) treatment in mice. The goal of this study was to evaluate Fmo3 gene expression in other diverse mouse models of hepatic oxidative stress and injury. Fmo3 gene regulation by Nrf2 was also investigated using Nrf2 knockout (Nrf2 KO) mice. In our studies, male C57BL/6J mice were treated with toxic doses of hepatotoxicants or underwent bile duct ligation (BDL, 10 days). Hepatotoxicants included APAP (400 mg/kg, 24-72 h), alpha-naphthyl isothiocyanate (ANIT; 50 mg/kg, 2-48 h), carbon tetrachloride (CCl4; 10 or 30 μL/kg, 24 and 48 h) and allyl alcohol (AlOH; 30 or 60 mg/kg, 6 and 24 h). Because oxidative stress activates nuclear factor (erythroid-derived 2)-like 2 (Nrf2), additional studies investigated Fmo3 gene regulation by Nrf2 using Nrf2 knockout (Nrf2 KO) mice. At appropriate time-points, blood and liver samples were collected for assessment of plasma alanine aminotransferase (ALT) activity, plasma and hepatic bile acid levels, as well as liver Fmo3 mRNA and protein expression. Fmo3 mRNA expression increased significantly by 43-fold at 12 h after ANIT treatment, and this increase translates to a 4-fold change in protein levels. BDL also increased Fmo3 mRNA expression by 1899-fold, but with no change in protein levels. Treatment of mice with CCl4 decreased liver Fmo3 gene expression, while no change in expression was detected with AlOH treatment. Nrf2 KO mice are more susceptible to APAP (400mg/kg, 72 h) treatment compared to their wild-type (WT) counterparts, which is evidenced by greater plasma ALT activity. The Fmo3 mRNA and protein expression increased in Nrf2 KO mice after APAP treatment. Collectively, not all hepatotoxicants that produce oxidative stress alter Fmo3 gene expression. Along with APAP, toxic ANIT treatment in mice markedly increased Fmo3 gene expression. While BDL increased the Fmo3 mRNA expression, the protein level did not change. The discrepancy with Fmo3 induction in cholestatic models, ANIT and BDL, is not entirely clear. Results from Nrf2 KO mice with APAP suggest that the transcriptional regulation of Fmo3 during liver injury may not involve Nrf2.
Collapse
Affiliation(s)
- Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA.
| | - Jamie E Moscovitz
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA.
| | - Ajay C Donepudi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.
| | - Sarah N Campion
- Drug Safety Research and Development, Pfizer Inc., Groton, USA.
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA.
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
17
|
Rudraiah S, Rohrer PR, Gurevich I, Goedken MJ, Rasmussen T, Hines RN, Manautou JE. Tolerance to acetaminophen hepatotoxicity in the mouse model of autoprotection is associated with induction of flavin-containing monooxygenase-3 (FMO3) in hepatocytes. Toxicol Sci 2014; 141:263-77. [PMID: 24973094 DOI: 10.1093/toxsci/kfu124] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acetaminophen (APAP) pretreatment with a hepatotoxic dose (400 mg/kg) in mice results in resistance to a second, higher dose (600 mg/kg) of APAP (APAP autoprotection). Recent microarray work by our group showed a drastic induction of liver flavin containing monooxygenase-3 (Fmo3) mRNA expression in our mouse model of APAP autoprotection. The role of liver Fmo3, which detoxifies xenobiotics, in APAP autoprotection is unknown. The purpose of this study was to characterize the gene regulation and protein expression of liver Fmo3 during APAP hepatotoxicity. The functional consequences of Fmo3 induction were also investigated. Plasma and livers were collected from male C57BL/6J mice over a period of 72 h following a single dose of APAP (400 mg/kg) to measure Fmo3 mRNA and protein expression. Although Fmo3 mRNA levels increased significantly following APAP treatment, protein expression changed marginally. In contrast, both Fmo3 mRNA and protein expression were significantly higher in APAP autoprotected livers. Unlike male C57BL/6J mice, female mice have ∼80-times higher constitutive Fmo3 mRNA levels and are highly resistant to APAP hepatotoxicity. Coadministration of APAP with the FMO inhibitor methimazole rendered female mice susceptible to APAP hepatotoxicity, with no changes in susceptibility detected in male mice. Furthermore, a human hepatocyte cell line (HC-04) clone over-expressing human FMO3 showed enhanced resistance to APAP cytotoxicity. Taken together, these findings establish for the first time induction of Fmo3 protein expression and function by xenobiotic treatment. Our results also indicate that Fmo3 expression and function plays a role in protecting the liver from APAP-induced toxicity. Although the mechanism(s) of this protection remains to be elucidated, this work describes a novel protective function for this enzyme.
Collapse
Affiliation(s)
- Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Philip R Rohrer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Igor Gurevich
- Cellular Dynamics International, Madison, Wisconsin 53711
| | - Michael J Goedken
- Rutgers University, Office of Translational Science, New Brunswick, New Jersey 08901
| | - Theodore Rasmussen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| | - Ronald N Hines
- US EPA, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina 27711
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
| |
Collapse
|
18
|
Effect of dietary α-lipoic acid on the mRNA expression of genes involved in drug metabolism and antioxidation system in rat liver. Br J Nutr 2014; 112:295-308. [DOI: 10.1017/s0007114514000841] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the present study, the mRNA levels of hepatic proteins involved in the drug metabolism of rats fed α-lipoic acid were evaluated by DNA microarray and real-time PCR analyses. Experimental diets containing 0, 0·1, 0·25 and 0·5 % (w/w) α-lipoic acid were fed to four groups of rats consisting of seven animals each for 21 d. DNA microarray analysis revealed that the diet containing 0·5 % α-lipoic acid significantly (P< 0·05) increased the mRNA levels of various phase I drug-metabolising enzymes up to 15-fold and phase II enzymes up to 52-fold in an isoenzyme-specific manner. α-Lipoic acid also up-regulated the mRNA levels of some members of the ATP-binding cassette transporter superfamily, presumed to be involved in the exportation of xenobiotics, up to 6·6-fold. In addition, we observed that α-lipoic acid increased the mRNA levels of many proteins involved in antioxidation, such as members of the thiol redox system (up to 5·5-fold), metallothioneins (up to 12-fold) and haeme oxygenase 1 (1·5-fold). These results were confirmed using real-time PCR analysis, and α-lipoic acid dose dependently increased the mRNA levels of various proteins involved in drug metabolism and antioxidation. Consistent with these observations, α-lipoic acid dose dependently increased the hepatic concentration of glutathione and the activities of glutathione reductase and glutathione transferase measured using 1-chloro-2,4-dinitrobenzene and 1,2-dichloro-4-nitrobenzene as substrates, but decreased the hepatic and serum concentrations of malondialdehyde. In conclusion, the present study unequivocally demonstrated that α-lipoic acid increases the mRNA expression of proteins involved in drug metabolism and antioxidation in the liver.
Collapse
|
19
|
Seyfried F, Li JV, Miras AD, Cluny NL, Lannoo M, Fenske WK, Sharkey KA, Nicholson JK, le Roux CW, Holmes E. Urinary Phenotyping Indicates Weight Loss-Independent Metabolic Effects of Roux-en-Y Gastric Bypass in Mice. J Proteome Res 2013; 12:1245-53. [DOI: 10.1021/pr300909v] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Florian Seyfried
- Department of General- and Visceral,
Vascular- and Pediatric Surgery, University Hospital of Wuerzburg, Germany
- Department of Investigative Medicine, Imperial College London, London, United Kingdom
| | - Jia V. Li
- Department of Surgery and Cancer,
Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Alexander D. Miras
- Department of Investigative Medicine, Imperial College London, London, United Kingdom
- Molecular
and Metabolic Imaging Group,
Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Nina L. Cluny
- Hotchkiss Brain Institute and
Snyder Institute for Chronic Diseases, Department of Physiology and
Pharmacology, University of Calgary, Calgary,
Alberta, Canada
| | - Matthias Lannoo
- Department of
Abdominal Surgery, University of Leuven, Belgium
| | - Wiebke K. Fenske
- Department of
Internal Medicine, University Hospital of Wuerzburg, Germany
| | - Keith A Sharkey
- Hotchkiss Brain Institute and
Snyder Institute for Chronic Diseases, Department of Physiology and
Pharmacology, University of Calgary, Calgary,
Alberta, Canada
| | - Jeremy K. Nicholson
- Department of Surgery and Cancer,
Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Carel W. le Roux
- Department of Investigative Medicine, Imperial College London, London, United Kingdom
- Experimental
Pathology, Conway
Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland
| | - Elaine Holmes
- Department of Surgery and Cancer,
Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
pH dependence on functional activity of human and mouse flavin-containing monooxygenase 5. Biochem Pharmacol 2012; 83:962-8. [DOI: 10.1016/j.bcp.2012.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 11/18/2022]
|
21
|
Lai WG, Farah N, Moniz GA, Wong YN. A Baeyer-Villiger Oxidation Specifically Catalyzed by Human Flavin-Containing Monooxygenase 5. Drug Metab Dispos 2010; 39:61-70. [DOI: 10.1124/dmd.110.035360] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
22
|
Reddy RR, Ralph EC, Motika MS, Zhang J, Cashman JR. Characterization of human flavin-containing monooxygenase (FMO) 3 and FMO5 expressed as maltose-binding protein fusions. Drug Metab Dispos 2010; 38:2239-45. [PMID: 20810540 DOI: 10.1124/dmd.110.033639] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The flavin-containing monooxygenase (FMO) family of enzymes oxygenates nucleophilic xenobiotics and endogenous substances. Human FMO3 and FMO5 are the predominant FMO forms in adult liver. These enzymes are naturally membrane-bound, and recombinant proteins are commercially available as microsomal preparations from insect cells (i.e., Supersome FMO). As an alternative, FMO3 has previously been expressed as a soluble protein, through use of an N-terminal maltose-binding protein (MBP) fusion. In the current study, MBP fusions of both human FMO3 and FMO5 were prepared to >90% purity in the presence of detergent and characterized for biochemical and kinetic parameters, and the parameters were compared with those of Supersome FMO samples. Although MBP-FMO enzymes afforded lower rates of turnover than the corresponding Supersome FMOs, both types of FMO showed identical substrate dependencies and similar responses to changes in assay conditions. Of interest, the FMO3 enzymes showed a 2-fold activation of k(cat)/K(m) in the presence of Triton X-100. Oligomeric analysis of MBP-FMO3 also showed disassociation from a high-order oligomeric form to a monomeric status in the presence of Triton X-100. This report serves as the first direct comparison between Supersome FMOs and the corresponding MBP fusions and the first report of a detergent-based activation of k(cat)/K(m) that corresponds to changes in oligomerization.
Collapse
Affiliation(s)
- Robert R Reddy
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | | | | | | | | |
Collapse
|
23
|
Motika MS, Zhang J, Zheng X, Riedler K, Cashman JR. Novel variants of the human flavin-containing monooxygenase 3 (FMO3) gene associated with trimethylaminuria. Mol Genet Metab 2009; 97:128-35. [PMID: 19321370 PMCID: PMC2739593 DOI: 10.1016/j.ymgme.2009.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 02/19/2009] [Accepted: 02/19/2009] [Indexed: 11/20/2022]
Abstract
The disorder trimethylaminuria (TMAu) often manifests itself in a body odor for individuals affected. TMAu is due to decreased metabolism of dietary-derived trimethylamine (TMA). In a healthy individual, 95% or more of TMA is converted by the flavin-containing monooxygenase 3 (FMO3, EC 1.14.13.8) to non-odorous trimethylamine N-oxide (TMA N-oxide). Several single nucleotide polymorphisms (SNPs) of the FMO3 gene have been described and result in an enzyme with decreased or abolished functional activity for TMA N-oxygenation thus leading to TMAu. Herein, we report two novel mutations observed from phenotyping and genotyping two self-reporting individuals. Sequence analysis of the exon regions of the FMO3 gene of a young woman with severe TMAu revealed heterozygous mutations at positions 187 (V187A), 158 (E158K), 308 (E308G), and 305 (E305X). Familial genetic analysis showed that the E158K/V187A/E308G derived from the same allele from the mother, and the E305X was derived from the father. FMO3 variants V187A and V187A/E158K were characterized for oxygenation of several common FMO3 substrates (i.e., 5- and 8-DPT, mercaptoimidazole (MMI), TMA, and sulindac sulfide) and for its thermal stability. Our findings show that with the combination of V187A/E158K mutations in FMO3, the enzyme activity is severely affected and possibly contributes to the TMAu observed. In another study, genotyping analysis of a 17 year old female revealed a mutation that caused a frame shift after K415 and resulted in a protein variant with only 486 amino acid residues that was associated with severe TMAu.
Collapse
Affiliation(s)
- Meike S Motika
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA
| | | | | | | | | |
Collapse
|
24
|
Cashman JR. Role of flavin-containing monooxygenase in drug development. Expert Opin Drug Metab Toxicol 2009; 4:1507-21. [PMID: 19040327 DOI: 10.1517/17425250802522188] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This review summarizes some recent observations and information related to the role of the flavin-containing monooxygenase (FMO) in preclinical drug development. Flavin-containing monooxygenase is a complimentary enzyme system to the cytochrome P450 (CYP) family of enzymes and oxygenates several soft, highly polarizable nucleophilic heteroatom-containing chemicals and drugs. The products of FMO-mediated metabolism are generally benign and highly polar, readily excreted materials. There may be some advantages in designing drugs that are metabolized in part by FMO and not exclusively by CYP. In this review, I describe the practical aspects for the participation of FMO in drug and chemical metabolism including: i) the study of FMO using in vitro preparations; ii) some observations about metabolism of drugs and chemicals by FMO in vivo; and iii) the consequences of studying FMO-related metabolism in various small animal models. Some of the preclinical research and development areas related to FMO are not fully mature areas and there are certain gaps in our knowledge. However, I include discussion of these areas to stimulate further work and invite further discussion.
Collapse
Affiliation(s)
- John R Cashman
- Human BioMolecular Research Institute, 5310 Eastgate Mall, San Diego, CA 92121, USA.
| |
Collapse
|
25
|
Systemic multicompartmental effects of the gut microbiome on mouse metabolic phenotypes. Mol Syst Biol 2008; 4:219. [PMID: 18854818 PMCID: PMC2583082 DOI: 10.1038/msb.2008.56] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 07/15/2008] [Indexed: 02/08/2023] Open
Abstract
To characterize the impact of gut microbiota on host metabolism, we investigated the multicompartmental metabolic profiles of a conventional mouse strain (C3H/HeJ) (n=5) and its germ-free (GF) equivalent (n=5). We confirm that the microbiome strongly impacts on the metabolism of bile acids through the enterohepatic cycle and gut metabolism (higher levels of phosphocholine and glycine in GF liver and marked higher levels of bile acids in three gut compartments). Furthermore we demonstrate that (1) well-defined metabolic differences exist in all examined compartments between the metabotypes of GF and conventional mice: bacterial co-metabolic products such as hippurate (urine) and 5-aminovalerate (colon epithelium) were found at reduced concentrations, whereas raffinose was only detected in GF colonic profiles. (2) The microbiome also influences kidney homeostasis with elevated levels of key cell volume regulators (betaine, choline, myo-inositol and so on) observed in GF kidneys. (3) Gut microbiota modulate metabotype expression at both local (gut) and global (biofluids, kidney, liver) system levels and hence influence the responses to a variety of dietary modulation and drug exposures relevant to personalized health-care investigations.
Collapse
|
26
|
Celius T, Roblin S, Harper PA, Matthews J, Boutros PC, Pohjanvirta R, Okey AB. Aryl hydrocarbon receptor-dependent induction of flavin-containing monooxygenase mRNAs in mouse liver. Drug Metab Dispos 2008; 36:2499-505. [PMID: 18765683 DOI: 10.1124/dmd.108.023457] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Flavin-containing monooxygenases (FMOs) are important in detoxication but generally are considered not to be inducible by xenobiotics. Our recent microarray studies revealed induction of FMO2 and FMO3 mRNAs by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in liver of mice with wild-type aryl hydrocarbon receptor (AHR) but not in Ahr-null mice. The aim of the present study was to delineate mechanisms of FMO regulation. In adult male mice, basal FMO3 mRNA is low but was induced 6-fold at 4 h and 6000-fold at 24 h. The ED50 was approximately 1 microg/kg for FMO2 and FMO3, similar to that for the classic AHR-regulated gene, Cyp1a1. In adult female mice basal FMO3 mRNA is high and was not induced at 4 h but was elevated 8-fold at 24 h. FMO5 mRNA was significantly down-regulated by TCDD in both male and female adult mice. Juvenile mice show no sex difference in response to TCDD; FMO3 was induced 4 to 6-fold by TCDD in both sexes. Chromatin immunoprecipitation demonstrated recruitment of AHR and aryl hydrocarbon nuclear translocator proteins to Fmo3 regulatory regions, suggesting that induction by TCDD is a primary AHR-mediated event. Although FMO2 and FMO3 mRNAs were highly induced by TCDD in adult males, overall FMO catalytic activity increased only modestly. In contrast to the striking up-regulation of FMO2 and FMO3 in mouse liver, TCDD has little effect on FMO mRNA in rat liver. However, FMO2 and FMO3 mRNAs were highly induced in transgenic mice that express wild-type rat AHR, indicating that lack of induction in rat is not due to an incompetent AHR in this species.
Collapse
Affiliation(s)
- Trine Celius
- Department of Pharmacology and Toxicology, Medical Sciences Building, 1 King's College Circle, University of Toronto, Toronto, ON, Canada M5S1A8
| | | | | | | | | | | | | |
Collapse
|