6 research outputs found

    Deorphanizing Human Cytochrome P450 Enzymes CYP4A22 and CYP4Z1 through Mechanistic in silico Modeling

    Get PDF
    Cytochrome P450 (CYP) enzymes are monooxygenases that catalyze the oxidation of structurally diverse substrates and are present in various lifeforms, including humans. Human CYPs catalyze the metabolism of xenobiotics including drugs and are involved in the essential biosynthesis of steroids, vitamins, and lipids. CYP-catalyzed metabolism and biosynthesis has been extensively studied recently, but several CYPs remain understudied despite their potential role in key biotransformation pathways. For these so-called ‘orphaned CYPs’, physiological function and structure are yet unknown, such as for CYP4A22 and 4Z1. CYP4A22 catalyzes the ω-hydroxylation of arachidonic acid to the angiogenic 20-hydroxyeicosatetraenoic acid. CYP4Z1 is overexpressed in breast cancer and other malignancies, which is correlated with tumor progression. Hence, CYP4Z1 is considered a promising breast cancer target that was not previously addressed by small molecule inhibitors. Here, we report our efforts to deorphanize CYP4A22 and 4Z1 together with our experimental partner Prof. Bureik. We were the first to predict the structure of CYP4A22 and 4Z1 by homology modeling and overcame the challenge of low-sequence similarity templates by incorporating substrate activities. We applied substrate docking and 3D pharmacophore modeling to rationalize how the binding site structure determines structure-activity relationships (SAR) trends. The well-known structural flexibility of CYPs was partially accounted for by molecular dynamics simulations. For the first time, enzyme-substrate interactions dynamics were analyzed with our novel dynamic pharmacophore approach, which led to the prediction of key residues. For CYP4A22, a residue influencing ω-hydroxylation (Phe320) and two binding residues (Arg96 and Arg233) were predicted. For CYP4Z1, the key role of Arg487 and assisting role of Asn381 for substrate binding were predicted, which was validated by in vitro mutational studies. The thereby validated CYP4Z1 model and substrate SAR were used in a virtual screening campaign resulting in a new potent and selective CYP4Z1 inhibitor (IC50: 63 ± 19 nM). Taken together, we established an in vitro/in silico deorphanization protocol that shed light on the structure-function relationships of CYP4A22 and 4Z1. This enabled us to discover a potent inhibitor of CYP4Z1 that will allow further studies on the physiological and pathophysiological role of the enzyme and might be further improved to target CYP4Z1 in a new therapeutical approach. Similar workflows could easily be applied to study other neglected enzymes in metabolism and other biotransformation pathways.Cytochrom P450 (CYP)-Enzyme sind Monooxygenasen, die die Oxidation strukturell diverser Substrate katalysieren und in verschiedenen Lebensformen, einschließlich des Menschen, vorkommen. Menschliche CYPs katalysieren den Metabolismus von Xenobiotika einschließlich Arzneistoffen und sind an der essenziellen Biosynthese von Steroiden, Vitaminen und Lipiden beteiligt. CYP-katalysierter Metabolismus und Biosynthese wurden in der Vergangenheit intensiv untersucht, aber einige CYPs sind trotz ihrer potenziellen Rolle in wichtigen Biotransformationswegen noch wenig erforscht. Für diese so genannten „orphaned“ oder „verwaisten“ CYPs, sind physiologische Funktion und Struktur noch unbekannt, wie z.B. CYP4A22 und 4Z1. CYP4A22 katalysiert die ω-Hydroxylierung von Arachidonsäure zu der angiogenen 20-Hydroxyeicosatetraensäure. CYP4Z1 wird bei Brustkrebs und anderen malignen Erkrankungen überexprimiert, was mit der Tumorprogression korreliert ist. Daher wird CYP4Z1 als ein vielversprechendes Brustkrebs-Target angesehen, das bisher nicht durch niedermolekulare Inhibitoren adressiert wurde. Hier berichten wir über unsere Bemühungen, CYP4A22 und 4Z1 zusammen mit unserem experimentellen Partner Prof. Bureik zu deorphanisieren. Wir waren die Ersten, die die Struktur von CYP4A22 und 4Z1 durch Homologiemodellierung vorhersagten und überwanden die Herausforderung der Templates mit geringer Sequenzähnlichkeit, indem wir Substrataktivitäten mit einbezogen. Wir wendeten Substrat-Docking und 3D-Pharmakophor-Modellierung an, um zu rationalisieren, wie die Struktur der Bindungstasche die Trends der Struktur-Aktivitäts-Beziehungen (SAR) bestimmt. Die bekannte strukturelle Flexibilität von CYPs wurde partiell durch Molekulardynamik-Simulationen berücksichtigt. Zum ersten Mal wurde die Dynamik der Enzym-Substrat-Interaktionen mit unserem neuartigen dynamischen Pharmakophor-Ansatz analysiert, was zur Vorhersage von wichtigen Aminosäuren führte. Für CYP4A22 wurde eine Aminosäure, die die ω-Hydroxylierung beeinflusst (Phe320) und zwei Bindungsaminosäuren (Arg96 und Arg233) vorhergesagt. Für CYP4Z1 wurde die Schlüsselrolle von Arg487 und die unterstützende Rolle von Asn381 für die Substratbindung vorhergesagt, welche durch in vitro Mutationsstudien validiert wurde. Das dadurch validierte CYP4Z1-Modell und die Substrat-SAR wurden in einer virtuellen Screening-Kampagne verwendet, die zu einen neuen potenten und selektiven CYP4Z1-Inhibitor führte (IC50: 63 ± 19 nM). Zusammengenommen haben wir ein in vitro/in silico Deorphanisierungsprotokoll etabliert, welches die Struktur-Funktionsbeziehungen von CYP4A22 und 4Z1 beleuchtet. Dies versetzte uns in die Lage einen potenten Inhibitor von CYP4Z1 zu entdecken, der weitere Studien über die physiologische und pathophysiologische Rolle des Enzyms ermöglichen wird und möglicherweise weiter verbessert werden kann, um CYP4Z1 in einem neuen therapeutischen Ansatz zu adressieren. Ähnliche Arbeitsabläufe könnte leicht angewendet werden, um andere vernachlässigte Enzyme im Metabolismus und anderen Biotransformationswegen zu untersuchen

    Functional Expression of All Human Sulfotransferases in Fission Yeast, Assay Development, and Structural Models for Isoforms SULT4A1 and SULT6B1

    Get PDF
    Cytosolic sulfotransferases (SULTs) catalyze phase II (conjugation) reactions of drugs and endogenous compounds. A complete set of recombinant fission yeast strains each expressing one of the 14 human SULTs was generated, including SULT4A1 and SULT6B1. Sulfation of test substrates by whole-cell biotransformation was successfully demonstrated for all enzymes for which substrates were previously known. The results proved that the intracellular production of the cofactor 3′-phosphoadenosine 5′-phosphosulfate (PAPS) necessary for SULT activity in fission yeast is sufficiently high to support metabolite production. A modified variant of sulfotransferase assay was also developed that employs permeabilized fission yeast cells (enzyme bags). Using this approach, SULT4A1-dependent sulfation of 1-naphthol was observed. Additionally, a new and convenient SULT activity assay is presented. It is based on the sulfation of a proluciferin compound, which was catalyzed by SULT1E1, SULT2A1, SULT4A1, and SULT6B1. For the latter two enzymes this study represents the first demonstration of their enzymatic functionality. Furthermore, the first catalytically competent homology models for SULT4A1 and SULT6B1 in complex with PAPS are reported. Through mechanistic molecular modeling driven by substrate docking, we pinned down the increased activity levels of these two isoforms to optimized substrate binding

    No Direct Hydroxylation of Pregnenolone by Steroid 21-Hydroxylase

    Get PDF
    Cytochrome P450s (CYPs) are an essential family of enzymes in the human body. They play a crucial role in metabolism, especially in human steroid biosynthesis. Reactions catalyzed by these enzymes are highly stereo- and regio-specific. Lack or severe malfunctions of CYPs can cause severe diseases and even shorten life. Hence, investigations on metabolic reactions and structural requirements of substrates are crucial to gain further knowledge on the relevance of different enzymes in the human body functions and the origin of diseases. One key enzyme in the biosynthesis of gluco- and mineralocorticoids is CYP21A2, also known as steroid 21-hydroxylase. To investigate the steric and regional requirements of substrates for this enzyme, we performed whole-cell biotransformation assays using a strain of fission yeast Schizosaccharomyces pombe recombinantly expressing CYP21A2. The progestogens progesterone, pregnenolone, and their 17α-hydroxy-derivatives were used as substrates. After incubation, samples were analyzed using gas chromatography coupled to mass spectrometry. For progesterone and 17α-hydroxyprogesterone, their corresponding 21-hydroxylated metabolites 11-deoxycorticosterone and 11-deoxycortisol were detected, while after incubation of pregnenolone and 17α-hydroxypregnenolone, no hydroxylated product was observed. Findings were confirmed with authentic reference material. Molecular docking experiments agree with these results and suggest that interaction between the 3-oxo group and arginine-234 of the enzyme is a strict requirement. The presented results demonstrate once more that the presence of an oxo-group in position 3 of the steroid is indispensable, while a 3-hydroxy group prevents hydroxylation in position C-21 by CYP21A2. This knowledge may be transferred to other CYP21A2 substrates and hence help to gain essential insights into steroid metabolism

    Next generation 3D pharmacophore modeling

    Get PDF
    3D pharmacophore models are three‐dimensional ensembles of chemically defined interactions of a ligand in its bioactive conformation. They represent an elegant way to decipher chemically encoded ligand information and have therefore become a valuable tool in drug design. In this review, we provide an overview on the basic concept of this method and summarize key studies for applying 3D pharmacophore models in virtual screening and mechanistic studies for protein functionality. Moreover, we discuss recent developments in the field. The combination of 3D pharmacophore models with molecular dynamics simulations could be a quantum leap forward since these approaches consider macromolecule–ligand interactions as dynamic and therefore show a physiologically relevant interaction pattern. Other trends include the efficient usage of 3D pharmacophore information in machine learning and artificial intelligence applications or freely accessible web servers for 3D pharmacophore modeling. The recent developments show that 3D pharmacophore modeling is a vibrant field with various applications in drug discovery and beyond

    Functional Expression of All Human Sulfotransferases in Fission Yeast, Assay Development, and Structural Models for Isoforms SULT4A1 and SULT6B1

    No full text
    Cytosolic sulfotransferases (SULTs) catalyze phase II (conjugation) reactions of drugs and endogenous compounds. A complete set of recombinant fission yeast strains each expressing one of the 14 human SULTs was generated, including SULT4A1 and SULT6B1. Sulfation of test substrates by whole-cell biotransformation was successfully demonstrated for all enzymes for which substrates were previously known. The results proved that the intracellular production of the cofactor 3′-phosphoadenosine 5′-phosphosulfate (PAPS) necessary for SULT activity in fission yeast is sufficiently high to support metabolite production. A modified variant of sulfotransferase assay was also developed that employs permeabilized fission yeast cells (enzyme bags). Using this approach, SULT4A1-dependent sulfation of 1-naphthol was observed. Additionally, a new and convenient SULT activity assay is presented. It is based on the sulfation of a proluciferin compound, which was catalyzed by SULT1E1, SULT2A1, SULT4A1, and SULT6B1. For the latter two enzymes this study represents the first demonstration of their enzymatic functionality. Furthermore, the first catalytically competent homology models for SULT4A1 and SULT6B1 in complex with PAPS are reported. Through mechanistic molecular modeling driven by substrate docking, we pinned down the increased activity levels of these two isoforms to optimized substrate binding

    Androgen-and estrogen-receptor mediated activities of 4-hydroxytestosterone, 4-hydroxyandrostenedione and their human metabolites in yeast based assays

    No full text
    4-Hydroxyandrost-4-ene-3,17-dione, also named formestane, is an irreversible aromatase inhibitor and therapeutically used as anti-breast cancer medication in post-menopausal women. Currently, no therapeutical indication led to approval of its 17-hydroxylated analog 4-hydroxytestosterone, an anabolic steroid. However, it is currently investigated in a clinical trial for breast cancer. In context with sports doping, aromatase inhibitors are administered to reduce estrogenic side effects of misused anabolic substances or their metabolites. Therefore, both substances are prohibited in sports by the World Anti-Doping Agency (WADA). Analysis of urinary phase I and phase II metabolites showed similar results for both compounds. In the current investigation, 4-hydroxyandrost-4-ene-3,17-dione, 4-hydroxytestosterone and seven of their described urinary metabolites as well as 2 alpha-hydroxyandrostenedione were tested in the yeast androgen screen and the yeast estrogen screen. Androgenic effects were observed for all tested substances, except for one, which showed anti-androgenic properties. With regard to the yeast estrogen screen, estrogenic effects were observed for only two metabolites at rather high concentrations, while six out of the ten substances tested showed anti-estrogenic properties. In terms of the strong androgenic effect observed for 4-hydroxytestosterone (10(-8) M), 4-hydroxyandrost-4-ene-3,17-dione (10(-8) M) and two more urinary metabolites, the yeast androgen assay may also be used to trace abuse in urine samples
    corecore