Hepatic Microsomal Bile Acid Biotransformation Studies Carried out So Far Seem to Be Restricted by Concentrating Only on CYP3A-Mediated Bile Acid Biotransformation

Total Page:16

File Type:pdf, Size:1020Kb

Hepatic Microsomal Bile Acid Biotransformation Studies Carried out So Far Seem to Be Restricted by Concentrating Only on CYP3A-Mediated Bile Acid Biotransformation HEPATIC MICROSOMAL BILE ACID BIOTRANSFORMATION: IDENTIFICATION OF METABOLITES AND CYTOCHROME P450 ENZYMES INVOLVED by Anand K. Deo B. Pharm., University of Pune, India, 2000 M. Pharm. Sci., University of Mumbai, 2002 M.Sc., The University of British Columbia, 2005 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in THE FACULTY OF GRADUATE STUDIES (PHARMACEUTICAL SCIENCES) THE UNIVERSITY OF BRITISH COLUMBIA Vancouver February 2009 © ANAND K. DEO, 2009 ABSTRACT Bile acids are end-products of cholesterol metabolism and essential for absorption of dietary lipids in the body. Impaired bile flow leads to hepatic bile acid accumulation and liver damage. Hepatic microsomal oxidation offers a potential mechanism for efficient elimination of bile acids. The present study investigated the cytochrome P450 (P450)-mediated hepatic microsomal biotransformation profiles of lithocholic acid, cholic acid and chenodeoxycholic acid using a liquid chromatography-mass spectrometry (LCIMS) based assay. Incubation of lithocholic acid with rat hepatic microsomes resulted in the formation of a major 6J3-hydroxylated metabolite, murideoxycholic acid, followed by isolithocholic acid and 3- ketocholanoic acid. Ursodeoxycholic acid, hyodeoxycholic acid and 6-ketolithocholic acid were identified as minor metabolites. Studies using P450-specific antibodies, chemical inducers, and rat recombinant enzymes showed that formation of murideoxycholic acid and 3-ketocholanoic acid were mediated by CYP3A2 and CYP2C 11. Similar metabolite profiles were obtained by incubation of lithocholic acid with mouse hepatic microsomes generating murideoxycholic acid as the major metabolite. Studies using P450 inducers and chemical inhibitors suggested the involvement of murine CYP3A in murideoxycholic acid and 3-ketocholanoic acid formation, and CYP1A, CYP2B and CYP3A enzymes in ursodeoxycholic acid, hyodeoxycholic acid and 6-ketolithocholic acid formation by mouse liver microsomes. Biotransformation of lithocholic acid by human hepatic microsomes generated 3-ketocholanoic acid as the major metabolite, and hyodeoxycholic acid, ursodeoxycholic acid, 6-ketolithocholic acid and murideoxycholic acid, as minor metabolites. Studies with chemical inhibitors and human recombinant enzymes demonstrated that CYP3A4 catalyzed the formation of all five metabolites. The biotransformation of cholic acid and chenodeoxycholic acid by human hepatic microsomes revealed the formation of a single cholic acid metabolite, 3-dehydrocholic acid. 11 Chenodeoxycholic acid biotransformation generated 7ct-hydroxy-3 -oxo-5 3-cholan-24-oic acid as the major metabolite followed by ‘y-muricholic acid, 7-ketolithocholic acid and cholic acid, respectively. CYP3A4 was found to be the major enzyme involved in the biotransformation of cholie acid and chenodeoxycholic acid in human liver microsomes. A comparison of metabolite profiles demonstrated the dominant role of human CYP3A4 in the oxidation of bile acids at the C-3 position. In contrast, 63-hydroxy1ation catalyzed by multiple P450 (CYP1A, CYP2B, CYP2C and CYP3A) enzymes was the preferred biotransformation pathway in rodent liver microsomes. 111 TABLE OF CONTENTS ABSTRACT ii TABLE OF CONTENTS iv LIST OF TABLES ix LIST OF FIGURES x LIST OF ABBREVIATIONS xiii ACKNOWLEDGEMENTS xiv DEDICATION xv CO-AUTHORSHIP STATEMENT xvi 1. 1 1.1 Bile Acids: Structure And Physicochemical Properties 2 1.2 Bile Acid Biosynthesis: Steps Involved 3 1.2.1 Biosynthesis of primary bile acids 4 1.2.2 Biosynthesis of secondary bile acids 8 1.3 Biliary Bile Acids: Composition And Physiological Relevance 9 1.4 Bile Acids: Enterohepatic Circulation 10 1.5 Bile Acid Toxicity 12 1.6 Cholestasis And Bile Acid Toxicity 13 1.7 Nuclear Receptors In Bile Acid Regulation 18 1.8 Bile Acid Biotransformation By Conjugation 22 1.9 Bile Acid Biotransformation By P450 Enzymes 24 1.10 Rationale 28 1.11 Hypotheses 31 1.1 1.1 Specific research objectives 32 1.12 References 50 iv 2. BIOTRANSFORMATION OF LITHOCHOLIC ACID BY RAT HEPATIC MICROSOMES: METABOLITE ANALYSIS BY LIQUID CHROMATOGRAPHY/MASS SPECTROMETRY 67 2.1 Summary 68 2.2 Introduction 69 2.3 Materials And Methods 71 2.3.1 Chemicals and reagents 71 2.3.2 Animal treatment and preparation of hepatic microsomes 72 2.3.3 Lithocholic acid biotransformation assay 72 2.3.4 Antibody inhibition 74 2.3.5 Analytical methods 74 2.3.6 Data analysis and calculation of enzyme kinetic parameters 75 2.3.7 Statistical analysis 76 2.4 Results 76 2.4.1 Biotransformation of lithocholic acid and metabolite identification 76 2.4.2 Kinetic analysis of hepatic microsomal metabolite formation 78 2.4.3 Effect of P450 inducers on lithocholic acid biotransformation 80 2.4.4 Antibody inhibition studies 80 2.4.5 Biotransformation studies with recombinant P450 enzymes 81 2.5 Discussion 82 2.6 References 99 3. IDENTIFICATION OF HUMAN HEPATIC CYTOCHROME P450 ENZYMES INVOLVED IN THE BIOTRANSFORMATION OF CHOLIC AND CHENODEOXYCHOLIC ACID 104 3.1 Summary 105 3.2 Introduction 106 3.3 Materials And Methods 108 3.3.1 Chemicals and reagents 108 V 3.3.2 Cholic acid and chenodeoxycholic acid biotransformation assays 109 3.3.3 Analytical methods 110 3.3.4 Data analysis and calculation of enzyme kinetic parameters 111 3.4 Results 112 3.4.1 Biotransformation and kinetic analysis of cholic acid metabolites 112 3.4.2 Biotransformation and kinetic analysis of chenodeoxycholic acid metabolites 113 3.4.3 Biotransformation studies with human recombinant P450 enzymes 114 3.5 Discussion 116 3.6 References 130 4. 3-KETOCHOLANOIC ACID IS TifE MAJOR CYP3A4 MEDIATED LITHOCHOLIC ACID METABOLITE IN HUMAN HEPATIC 133 4.1 Summary 134 4.2 Introduction 135 4.3 Materials And Methods 137 4.3.1 Chemicals and Reagents 137 4.3.2 Lithocholic acid biotransformation assay 138 4.3.3 Analytical methods 139 4.3.4 Data analysis and calculation of enzyme kinetic parameters 139 4.3.5 Chemical inhibition studies 140 4.3.6 Statistical analysis 141 4.4 Results 141 4.4.1 Lithocholic acid biotransformation by human liver microsomes 141 4.4.2 Lithocholic acid biotransformation by human recombinant P450 enzymes 142 4.4.3 Chemical inhibition studies 143 4.4.4 Lithocholic acid biotransformation in human hepatic microsomes with varying CYP3A4 levels 143 4.5 Discussion 144 4.6 References 158 vi 5. HEPATIC MICROSOMAL P450 MEDIATED BIOTRANSFORMATION OF LITHOCHOLIC ACID BY MOUSE HIPi4.TIC 1’II1SO1VIES 162 5.1 Summary 163 5.2 Introduction 164 5.3 Materials And Methods 165 5.3.1 Chemicals and reagents 165 5.3.2 Animal treatment and preparation of microsomes 166 5.3.3 Lithocholic acid biotransformation assay 166 5.3.4 Chemical inhibition studies 166 5.3.5 Statistical analysis 167 5.4 Results 167 5.4.1 Hepatic biotransformation of lithocholic acid by mouse hepatic microsomes 167 5.4.2 Effect of P450 inducers in lithocholic acid biotransformation 167 5.4.3 Chemical inhibition studies 168 5.5 Discussion 170 5.8 References 178 6. ENE1J4.IJ DISCIJSSIO1i 181 6.1 Overview 182 6.2 Cholic Acid Biotransformation By Human Hepatic Microsomes 183 6.3 Chenodeoxycholic Acid Biotransformation By Human Hepatic Microsomes 183 6.4 Comparison of Lithocholic Acid Metabolite Formation Patterns By Rat, Human And Mouse Liver Microsomes 184 6.5 Limitations 188 6.6 Overall Significance of Thesis Research 190 6.7 Future Studies 191 6.8 References 200 vii ITIA £OZJ )CIGtsJaJcI’r LIST OF TABLES Table 1.1 Oxidative metabolites of bile acids 33 Table 1.2 Different P450 enzymes and their levels in human and rat hepatic microsomes 34 Table 2.1 Kinetic parameters of lithocholic acid metabolite formation by rat hepatic microsomes 88 Table 2.2 Effect of sex and treatment with P450 inducers on lithocholie acid metabolite formation by rat hepatic microsomes 89 Table 4.1 Kinetic parameters of lithocholic acid metabolite formation by human hepatic microsomes 149 Table 4.2 Kinetic parameters of lithocholic acid metabolite formation by recombinant CYP3A4 150 Table 4.3 Rate of formation of lithocholic acid metabolites in human liver microsomes with varying CYP3A4 activities 151 Effect of treatment with P450 inducers on lithocholic acid metabolite Table 5 1 formation by mouse hepatic microsomes 174 Table 6.1 Regio-selective oxidation of lithocholic acid by human, rat and mouse hepatic microsomes 195 Table 6.2 Comparison of P450 enzymes involved in lithocholic acid metabolite formation in human and rodent hepatic microsomes 196 ix LIST OF FIGURES Figure 1.1 Cholesterol and bile acid structure 35 Figure 1.2 Chemical structures of unconjugated and conjugated bile acids 36 Figure 1.3 Bile acid levels in human, rat and mouse liver 37 Figure 1.4 Neutral and acidic pathways in bile acid formation 38 Figure 1.5 Bile acid levels in human, rat and mouse bile 39 Figure 1.6 Enterohepatic circulation of bile acids 40 Figure 1.7 Amphipathic nature of bile acids 41 Figure 1.8 Obstruction to the flow of bile acids from the liver to the intestine is termed as cholestasis 42 Figure 1.9 Comparison of bile acid levels in livers of patients with end-stage cholestasis and normal humans 43 Figure 1.10 Bile acid regulation: A complex process involving various receptors, enzymes, and transporters 44 Figure 1.11 Regulation of bile acids by nuclear receptors 45 Figure 1.12 PXR and CAR cross-talk mediated metabolism of lithocholic acid 46 Figure 1.13 Major hepatic P450 enzymes involved in drug metabolism 47 Figure 1.14 Biotransformation of bile acids 48 Figure 2.1 General bile acid structure showing positions available for hydroxylation 90 Figure 2.2 Representative
Recommended publications
  • Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid
    Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 231 _____________________________ _____________________________ Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid Cloning, Expression and Catalytic Properties of CYP4F8 and CYP4F21 BY JOHAN BYLUND ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2000 Dissertation for the Degree of Doctor of Philosophy (Faculty of Pharmacy) in Pharmaceutical Pharmacology presented at Uppsala University in 2000 ABSTRACT Bylund, J. 2000. Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid: Cloning, Expression and Catalytic Properties of CYP4F8 and CYP4F21. Acta Universitatis Upsaliensis. Comprehensive Summaries of Uppsala Dissertations from Faculty of Pharmacy 231 50 pp. Uppsala. ISBN 91-554-4784-8. Cytochrome P450 (P450 or CYP) is an enzyme system involved in the oxygenation of a wide range of endogenous compounds as well as foreign chemicals and drugs. This thesis describes investigations of P450-catalyzed oxygenation of prostaglandins, linoleic and arachidonic acids. The formation of bisallylic hydroxy metabolites of linoleic and arachidonic acids was studied with human recombinant P450s and with human liver microsomes. Several P450 enzymes catalyzed the formation of bisallylic hydroxy metabolites. Inhibition studies and stereochemical analysis of metabolites suggest that the enzyme CYP1A2 may contribute to the biosynthesis of bisallylic hydroxy fatty acid metabolites in adult human liver microsomes. 19R-Hydroxy-PGE and 20-hydroxy-PGE are major components of human and ovine semen, respectively. They are formed in the seminal vesicles, but the mechanism of their biosynthesis is unknown. Reverse transcription-polymerase chain reaction using degenerate primers for mammalian CYP4 family genes, revealed expression of two novel P450 genes in human and ovine seminal vesicles.
    [Show full text]
  • Inhibition of Cytochrome P450 2C8-Mediated Drug Metabolism by the Flavonoid Diosmetin
    Drug Metab. Pharmacokinet. 26 (6): 559­568 (2011). Copyright © 2011 by the Japanese Society for the Study of Xenobiotics (JSSX) Regular Article Inhibition of Cytochrome P450 2C8-mediated Drug Metabolism by the Flavonoid Diosmetin Luigi QUINTIERI1,PietroPALATINI1,StefanoMORO2 and Maura FLOREANI1,* 1Department of Pharmacology and Anaesthesiology, University of Padova, Italy 2Molecular Modeling Section (MMS), Department of Pharmaceutical Sciences, University of Padova, Italy Full text of this paper is available at http://www.jstage.jst.go.jp/browse/dmpk Summary: The aim of this study was to assess the effects of diosmetin and hesperetin, two flavonoids present in various medicinal products, on CYP2C8 activity of human liver microsomes using paclitaxel oxidation to 6¡-hydroxy-paclitaxel as a probe reaction. Diosmetin and hesperetin inhibited 6¡-hydroxy- paclitaxel production in a concentration-dependent manner, diosmetin being about 16-fold more potent than hesperetin (mean IC50 values 4.25 « 0.02 and 68.5 « 3.3 µM for diosmetin and hesperetin, respectively). Due to the low inhibitory potency of hesperetin, we characterized the mechanism of diosmetin-induced inhibition only. This flavonoid proved to be a reversible, dead-end, full inhibitor of CYP2C8, its mean inhibition constant (Ki)being3.13« 0.11 µM. Kinetic analysis showed that diosmetin caused mixed-type inhibition, since it significantly decreased the Vmax (maximum velocity) and increased the Km value (substrate concentration yielding 50% of Vmax) of the reaction. The results of kinetic analyses were consistent with those of molecular docking simulation, which showed that the putative binding site of diosmetin coincided with the CYP2C8 substrate binding site. The demonstration that diosmetin inhibits CYP2C8 at concentrations similar to those observed after in vivo administration (in the low micromolar range) is of potential clinical relevance, since it may cause pharmacokinetic interactions with co- administered drugs metabolized by this CYP.
    [Show full text]
  • Synonymous Single Nucleotide Polymorphisms in Human Cytochrome
    DMD Fast Forward. Published on February 9, 2009 as doi:10.1124/dmd.108.026047 DMD #26047 TITLE PAGE: A BIOINFORMATICS APPROACH FOR THE PHENOTYPE PREDICTION OF NON- SYNONYMOUS SINGLE NUCLEOTIDE POLYMORPHISMS IN HUMAN CYTOCHROME P450S LIN-LIN WANG, YONG LI, SHU-FENG ZHOU Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P. R. China (LL Wang & Y Li) Discipline of Chinese Medicine, School of Health Sciences, RMIT University, Bundoora, Victoria 3083, Australia (LL Wang & SF Zhou). 1 Copyright 2009 by the American Society for Pharmacology and Experimental Therapeutics. DMD #26047 RUNNING TITLE PAGE: a) Running title: Prediction of phenotype of human CYPs. b) Author for correspondence: A/Prof. Shu-Feng Zhou, MD, PhD Discipline of Chinese Medicine, School of Health Sciences, RMIT University, WHO Collaborating Center for Traditional Medicine, Bundoora, Victoria 3083, Australia. Tel: + 61 3 9925 7794; fax: +61 3 9925 7178. Email: [email protected] c) Number of text pages: 21 Number of tables: 10 Number of figures: 2 Number of references: 40 Number of words in Abstract: 249 Number of words in Introduction: 749 Number of words in Discussion: 1459 d) Non-standard abbreviations: CYP, cytochrome P450; nsSNP, non-synonymous single nucleotide polymorphism. 2 DMD #26047 ABSTRACT Non-synonymous single nucleotide polymorphisms (nsSNPs) in coding regions that can lead to amino acid changes may cause alteration of protein function and account for susceptivity to disease. Identification of deleterious nsSNPs from tolerant nsSNPs is important for characterizing the genetic basis of human disease, assessing individual susceptibility to disease, understanding the pathogenesis of disease, identifying molecular targets for drug treatment and conducting individualized pharmacotherapy.
    [Show full text]
  • Human Cytochrome P450 CYP2A13
    [CANCER RESEARCH 60, 5074–5079, September 15, 2000] Human Cytochrome P450 CYP2A13: Predominant Expression in the Respiratory Tract and Its High Efficiency Metabolic Activation of a Tobacco-specific Carcinogen, 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone1 Ting Su, Ziping Bao, Qing-Yu Zhang, Theresa J. Smith, Jun-Yan Hong,2 and Xinxin Ding2 Wadsworth Center, New York State Department of Health, Albany, New York 12201 [T. S., Q-Y. Z., X. D.]; School of Public Health, State University of New York at Albany, Albany, New York [T. S., X. D.]; and Environmental and Occupational Health Sciences Institute, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854 [Z. B., T. J. S., J-Y. H.] ABSTRACT However, heterologously expressed CYP2A7 showed no catalytic activity (17, 18). CYP2A13 cDNA has not been isolated previously; The human CYP2A subfamily comprises three genes, CYP2A6, the reported protein sequence was deduced from the predicted coding CYP2A7, and CYP2A13. CYP2A6 is active toward many carcinogens and region of a CYP2A13 genomic clone (1). On the basis of its sequence is the major coumarin 7-hydroxylase and nicotine C-oxidase in the liver, whereas CYP2A7 is not functional. The function of CYP2A13 has not been features that resemble the nonfunctional CYP2A7 and CYP2A6v1 (a characterized. In this study, a CYP2A13 cDNA was prepared by RNA- genetic variant of CYP2A6) proteins, the CYP2A13 protein was PCR from human nasal mucosa and was translated using a baculovirus predicted to be nonfunctional in coumarin 7-hydroxylation (1). Be- expression system. In a reconstituted system, the expressed CYP2A13 was cause the deduced amino acid sequence of CYP2A13 shares a 95.4% more active than CYP2A6 in the metabolic activation of hexamethylphos- identity with that of CYP2A6 (1), antibodies and chemical probes for phoramide, N,N-dimethylaniline, 2؅-methoxyacetophenone, and N-nitro- CYP2A6 may interact with CYP2A13.
    [Show full text]
  • Colorectal Cancer and Omega Hydroxylases
    1 The differential expression of omega-3 and omega-6 fatty acid metabolising enzymes in colorectal cancer and its prognostic significance Abdo Alnabulsi1,2, Rebecca Swan1, Beatriz Cash2, Ayham Alnabulsi2, Graeme I Murray1 1Pathology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25, 2ZD, UK. 2Vertebrate Antibodies, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, UK. Address correspondence to: Professor Graeme I Murray Email [email protected] Phone: +44(0)1224 553794 Fax: +44(0)1224 663002 Running title: omega hydroxylases and colorectal cancer 2 Abstract Background: Colorectal cancer is a common malignancy and one of the leading causes of cancer related deaths. The metabolism of omega fatty acids has been implicated in tumour growth and metastasis. Methods: This study has characterised the expression of omega fatty acid metabolising enzymes CYP4A11, CYP4F11, CYP4V2 and CYP4Z1 using monoclonal antibodies we have developed. Immunohistochemistry was performed on a tissue microarray containing 650 primary colorectal cancers, 285 lymph node metastasis and 50 normal colonic mucosa. Results: The differential expression of CYP4A11 and CYP4F11 showed a strong association with survival in both the whole patient cohort (HR=1.203, 95% CI=1.092-1.324, χ2=14.968, p=0.001) and in mismatch repair proficient tumours (HR=1.276, 95% CI=1.095-1.488, χ2=9.988, p=0.007). Multivariate analysis revealed that the differential expression of CYP4A11 and CYP4F11 was independently prognostic in both the whole patient cohort (p = 0.019) and in mismatch repair proficient tumours (p=0.046). Conclusions: A significant and independent association has been identified between overall survival and the differential expression of CYP4A11 and CYP4F11 in the whole patient cohort and in mismatch repair proficient tumours.
    [Show full text]
  • Systemic Gut Microbial Modulation of Bile Acid Metabolism in Host Tissue Compartments
    Systemic gut microbial modulation of bile acid metabolism in host tissue compartments Jonathan R. Swanna,b, Elizabeth J. Wanta, Florian M. Geiera, Konstantina Spagoua, Ian D. Wilsonc, James E. Sidawayd, Jeremy K. Nicholsona,1, and Elaine Holmesa,1 aBiomolecular Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London SW7 2AZ, United Kingdom; bDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, The University of Reading, Reading RG6 6AP, United Kingdom; cAstraZeneca, Department of Clinical Pharmacology, Drug Metabolism and Pharmacokinetics; and dAstraZeneca, Global Safety Assessment, Cheshire SK10 4TG, United Kingdom Edited by Todd R. Klaenhammer, North Carolina State University, Raleigh, NC, and approved July 30, 2010 (received for review May 26, 2010) We elucidate the detailed effects of gut microbial depletion on the deconjugation, dehydrogenation, dehydroxylation, and sulfation bile acid sub-metabolome of multiple body compartments (liver, reactions (8) to produce secondary bile acids, which are reabsorbed kidney, heart, and blood plasma) in rats. We use a targeted ultra- and returned to the liver for further processing. performance liquid chromatography with time of flight mass-spec- Historically, bile acids have been primarily viewed as detergent trometry assay to characterize the differential primary and secondary molecules important for the absorption of dietary fats and lipid- bile acid profiles in each tissue and show a major increase in the soluble vitamins in the small intestine and the maintenance of proportion of taurine-conjugated bile acids in germ-free (GF) and cholesterol homeostasis in the liver. However, their role in the antibiotic (streptomycin/penicillin)-treated rats. Although conjugated mammalian system is much broader than this, and they are now bile acids dominate the hepatic profile (97.0 ± 1.5%) of conventional recognized as important signaling molecules with systemic endo- animals, unconjugated bile acids comprise the largest proportion of crine functions.
    [Show full text]
  • Supplementary Material
    Supplementary Material Table S1: Significant downregulated KEGGs pathways identified by DAVID following exposure to five cinnamon- based phenylpropanoids (p < 0.05). p-value Term: Genes (Benjamini) Cytokine-cytokine receptor interaction: FASLG, TNFSF14, CXCL11, IL11, FLT3LG, CCL3L1, CCL3L3, CXCR6, XCR1, 2.43 × 105 RTEL1, CSF2RA, TNFRSF17, TNFRSF14, CCNL2, VEGFB, AMH, TNFRSF10B, INHBE, IFNB1, CCR3, VEGFA, CCR2, IL12A, CCL1, CCL3, CXCL5, TNFRSF25, CCR1, CSF1, CX3CL1, CCL7, CCL24, TNFRSF1B, IL12RB1, CCL21, FIGF, EPO, IL4, IL18R1, FLT1, TGFBR1, EDA2R, HGF, TNFSF8, KDR, LEP, GH2, CCL13, EPOR, XCL1, IFNA16, XCL2 Neuroactive ligand-receptor interaction: OPRM1, THRA, GRIK1, DRD2, GRIK2, TACR2, TACR1, GABRB1, LPAR4, 9.68 × 105 GRIK5, FPR1, PRSS1, GNRHR, FPR2, EDNRA, AGTR2, LTB4R, PRSS2, CNR1, S1PR4, CALCRL, TAAR5, GABRE, PTGER1, GABRG3, C5AR1, PTGER3, PTGER4, GABRA6, GABRA5, GRM1, PLG, LEP, CRHR1, GH2, GRM3, SSTR2, Chlorogenic acid Chlorogenic CHRM3, GRIA1, MC2R, P2RX2, TBXA2R, GHSR, HTR2C, TSHR, LHB, GLP1R, OPRD1 Hematopoietic cell lineage: IL4, CR1, CD8B, CSF1, FCER2, GYPA, ITGA2, IL11, GP9, FLT3LG, CD38, CD19, DNTT, 9.29 × 104 GP1BB, CD22, EPOR, CSF2RA, CD14, THPO, EPO, HLA-DRA, ITGA2B Cytokine-cytokine receptor interaction: IL6ST, IL21R, IL19, TNFSF15, CXCR3, IL15, CXCL11, TGFB1, IL11, FLT3LG, CXCL10, CCR10, XCR1, RTEL1, CSF2RA, IL21, CCNL2, VEGFB, CCR8, AMH, TNFRSF10C, IFNB1, PDGFRA, EDA, CXCL5, TNFRSF25, CSF1, IFNW1, CNTFR, CX3CL1, CCL5, TNFRSF4, CCL4, CCL27, CCL24, CCL25, CCL23, IFNA6, IFNA5, FIGF, EPO, AMHR2, IL2RA, FLT4, TGFBR2, EDA2R,
    [Show full text]
  • Understanding Drug-Drug Interactions Due to Mechanism-Based Inhibition in Clinical Practice
    pharmaceutics Review Mechanisms of CYP450 Inhibition: Understanding Drug-Drug Interactions Due to Mechanism-Based Inhibition in Clinical Practice Malavika Deodhar 1, Sweilem B Al Rihani 1 , Meghan J. Arwood 1, Lucy Darakjian 1, Pamela Dow 1 , Jacques Turgeon 1,2 and Veronique Michaud 1,2,* 1 Tabula Rasa HealthCare Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; [email protected] (M.D.); [email protected] (S.B.A.R.); [email protected] (M.J.A.); [email protected] (L.D.); [email protected] (P.D.); [email protected] (J.T.) 2 Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada * Correspondence: [email protected]; Tel.: +1-856-938-8697 Received: 5 August 2020; Accepted: 31 August 2020; Published: 4 September 2020 Abstract: In an ageing society, polypharmacy has become a major public health and economic issue. Overuse of medications, especially in patients with chronic diseases, carries major health risks. One common consequence of polypharmacy is the increased emergence of adverse drug events, mainly from drug–drug interactions. The majority of currently available drugs are metabolized by CYP450 enzymes. Interactions due to shared CYP450-mediated metabolic pathways for two or more drugs are frequent, especially through reversible or irreversible CYP450 inhibition. The magnitude of these interactions depends on several factors, including varying affinity and concentration of substrates, time delay between the administration of the drugs, and mechanisms of CYP450 inhibition. Various types of CYP450 inhibition (competitive, non-competitive, mechanism-based) have been observed clinically, and interactions of these types require a distinct clinical management strategy. This review focuses on mechanism-based inhibition, which occurs when a substrate forms a reactive intermediate, creating a stable enzyme–intermediate complex that irreversibly reduces enzyme activity.
    [Show full text]
  • Simulation of Physicochemical and Pharmacokinetic Properties of Vitamin D3 and Its Natural Derivatives
    pharmaceuticals Article Simulation of Physicochemical and Pharmacokinetic Properties of Vitamin D3 and Its Natural Derivatives Subrata Deb * , Anthony Allen Reeves and Suki Lafortune Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA; [email protected] (A.A.R.); [email protected] (S.L.) * Correspondence: [email protected] or [email protected]; Tel.: +1-224-310-7870 or +1-305-760-7479 Received: 9 June 2020; Accepted: 20 July 2020; Published: 23 July 2020 Abstract: Vitamin D3 is an endogenous fat-soluble secosteroid, either biosynthesized in human skin or absorbed from diet and health supplements. Multiple hydroxylation reactions in several tissues including liver and small intestine produce different forms of vitamin D3. Low serum vitamin D levels is a global problem which may origin from differential absorption following supplementation. The objective of the present study was to estimate the physicochemical properties, metabolism, transport and pharmacokinetic behavior of vitamin D3 derivatives following oral ingestion. GastroPlus software, which is an in silico mechanistically-constructed simulation tool, was used to simulate the physicochemical and pharmacokinetic behavior for twelve vitamin D3 derivatives. The Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) Predictor and PKPlus modules were employed to derive the relevant parameters from the structural features of the compounds. The majority of the vitamin D3 derivatives are lipophilic (log P values > 5) with poor water solubility which are reflected in the poor predicted bioavailability. The fraction absorbed values for the vitamin D3 derivatives were low except for calcitroic acid, 1,23S,25-trihydroxy-24-oxo-vitamin D3, and (23S,25R)-1,25-dihydroxyvitamin D3-26,23-lactone each being greater than 90% fraction absorbed.
    [Show full text]
  • Diazepam Therapy and CYP2C19 Genotype
    NLM Citation: Dean L. Diazepam Therapy and CYP2C19 Genotype. 2016 Aug 25. In: Pratt VM, McLeod HL, Rubinstein WS, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012-. Bookshelf URL: https://www.ncbi.nlm.nih.gov/books/ Diazepam Therapy and CYP2C19 Genotype Laura Dean, MD1 Created: August 25, 2016. Introduction Diazepam is a benzodiazepine with several clinical uses, including the management of anxiety, insomnia, muscle spasms, seizures, and alcohol withdrawal. The clinical response to benzodiazepines, such as diazepam, varies widely between individuals (1, 2). Diazepam is primarily metabolized by CY2C19 and CYP3A4 to the major active metabolite, desmethyldiazepam. Approximately 3% of Caucasians and 15 to 20% of Asians have reduced or absent CYP2C19 enzyme activity (“poor metabolizers”). In these individuals, standard doses of diazepam may lead to a higher exposure to diazepam. The FDA-approved drug label for diazepam states that “The marked inter-individual variability in the clearance of diazepam reported in the literature is probably attributable to variability of CYP2C19 (which is known to exhibit genetic polymorphism; about 3-5% of Caucasians have little or no activity and are “poor metabolizers”) and CYP3A4” (1). Drug: Diazepam Diazepam is used in the management of anxiety disorders or for the short-term relief of the symptoms of anxiety. In acute alcohol withdrawal, diazepam may provide symptomatic relief from agitation, tremor, delirium tremens, and hallucinations. Diazepam is also useful as an adjunct treatment for the relief of acute skeletal muscle spasms, as well as spasticity caused by upper motor neuron disorders (3). There are currently 16 benzodiazepines licensed by the FDA.
    [Show full text]
  • Variation in CYP2A6 Activity and Personalized Medicine
    Journal of Personalized Medicine Review Variation in CYP2A6 Activity and Personalized Medicine Julie-Anne Tanner 1,2 and Rachel F. Tyndale 1,2,3,* 1 Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada; [email protected] 2 Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada 3 Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada * Correspondence: [email protected]; Tel.: +1-416-978-6374; Fax: +1-416-978-6395 Academic Editor: Stephen B. Liggett Received: 7 October 2017; Accepted: 27 November 2017; Published: 1 December 2017 Abstract: The cytochrome P450 2A6 (CYP2A6) enzyme metabolizes several clinically relevant substrates, including nicotine—the primary psychoactive component in cigarette smoke. The gene that encodes the CYP2A6 enzyme is highly polymorphic, resulting in extensive interindividual variation in CYP2A6 enzyme activity and the rate of metabolism of nicotine and other CYP2A6 substrates including cotinine, tegafur, letrozole, efavirenz, valproic acid, pilocarpine, artemisinin, artesunate, SM-12502, caffeine, and tyrosol. CYP2A6 expression and activity are also impacted by non-genetic factors, including induction or inhibition by pharmacological, endogenous, and dietary substances, as well as age-related changes, or interactions with other hepatic enzymes, co-enzymes, and co-factors. As variation in CYP2A6 activity is associated with smoking behavior, smoking cessation, tobacco-related lung cancer risk, and with altered metabolism and resulting clinical responses for several therapeutics, CYP2A6 expression and enzyme activity is an important clinical consideration. This review will discuss sources of variation in CYP2A6 enzyme activity, with a focus on the impact of CYP2A6 genetic variation on metabolism of the CYP2A6 substrates.
    [Show full text]
  • Time-Dependent Inhibition of CYP2C8 and CYP2C19 by Hedera Helix Extracts, a Traditional Respiratory Herbal Medicine
    molecules Article Time-dependent Inhibition of CYP2C8 and CYP2C19 by Hedera helix Extracts, A Traditional Respiratory Herbal Medicine Shaheed Ur Rehman 1, In Sook Kim 2, Min Sun Choi 2, Seung Hyun Kim 3, Yonghui Zhang 4 and Hye Hyun Yoo 2,* 1 Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan; [email protected] 2 Institute of Pharmaceutical Science and Technology and College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Korea; [email protected] (I.S.K.); [email protected] (M.S.C.) 3 College of Pharmacy, Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Korea; [email protected] 4 School of Pharmacy, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; [email protected] * Correspondence: [email protected]; Tel.: +82-31-400-5804; Fax: +82-31-400-5958 Received: 1 June 2017; Accepted: 20 July 2017; Published: 24 July 2017 Abstract: The extract of Hedera helix L. (Araliaceae), a well-known folk medicine, has been popularly used to treat respiratory problems, worldwide. It is very likely that this herbal extract is taken in combination with conventional drugs. The present study aimed to evaluate the effects of H. helix extract on cytochrome P450 (CYP) enzyme-mediated metabolism to predict the potential for herb–drug interactions. A cocktail probe assay was used to measure the inhibitory effect of CYP. H. helix extracts were incubated with pooled human liver microsomes or CYP isozymes with CYP-specific substrates, and the formation of specific metabolites was investigated to measure the inhibitory effects.
    [Show full text]