bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Loss of multiple enzyme activities due to the human genetic variation P284T in NADPH cytochrome P450 oxidoreductase.

Shaheena Parween1,2, Maria Natalia Rojas Velazquez1,2,3, Sameer S. Udhane 1,2,#, Norio Kagawa4, and Amit V. Pandey1,2*

1Pediatric , Diabetology, and Metabolism, Department of Pediatrics, University Children’s Hospital Bern, 2Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland. 3Laboratorio de Genética Molecular, Departamento de Genética, Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asunción, Campus San Lorenzo, Central, Paraguay. 4School of Medicine, Nagoya University, 65 Tsurumai-Cho, Showa-Ku, Nagoya 466-8550, Japan

# Current address: Department of Pathology, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA.

* Address for correspondence: PD Dr. Amit V Pandey KIKL C837, Pediatric Endocrinology, Diabetology & Metabolism, University Children’s Hospital Bern, Freiburgstrasse 15, CH-3010 Bern Switzerland. Tel: 0041 31 632 9637. Email: [email protected]

Abstract: Cytochromes P450 located in the endoplasmic reticulum require NADPH cytochrome P450 oxidoreductase (POR) for their catalytic activities. Mutations in POR cause multiple disorders in humans related to the of hormones and also affect drug-metabolizing cytochrome P450 activities. Here we are reporting the effects of a POR genetic variant P284T which is located in the hinge region of POR that is necessary for the flexibility of domain movements. Human wild-type and P284T mutant of POR, as well as cytochrome P450 proteins, were expressed in bacteria, purified and then reconstituted in liposomes for enzyme kinetic assays. Quality of POR proteins was checked by cytochrome c, ferricyanide and tetrazolium dye reduction assay and measurements flavin content. We found that for the P284T variant of POR the cytochrome c reduction activity was reduced to 47% of the WT and MTT reduction was reduced to only 15% of the WT. No impact on ferricyanide reduction activity was observed, but a severe loss of CYP19A1 () activity was observed (9% of WT). In the assays of drug metabolizing cytochrome P450 enzymes, the P284T variant of POR showed 26% activity for CYP2C9, 44% activity for CYP2C19, 23% activity for CYP3A4 and 44% activity in CYP3A5 assays compared to the WT POR. These results indicate a severe effect on several cytochrome P450 activities due to the P284T variation in POR which suggests a negative impact on both the steroid as well as drug metabolism in the individuals carrying this variation.

Keywords: POR, P450 oxidoreductase, CYP2C9, CYP2C19, CYP3A4, CYP3A5, CYP19A1, polymorphisms, protein-protein interaction, cytochrome P450. 2007;Omura, 2010;Zanger and Schwab, 2013). 1 Introduction There are two different types of cytochrome P450 proteins in mammals (Omura, 2010;McLean et al., The NADPH cytochrome P450 reductase (POR, 2015). The type 1 P450s are found in the NCBI# NP_000932, UniProt# P16435) is the mitochondria use adrenodoxin/adrenodoxin obligate electron donor for all cytochromes P450 reductase system as their redox partner (Omura, proteins located in the endoplasmic reticulum 2006;Zalewski et al., 2016). The type 2 cytochrome (Pandey and Flück, 2013;Flück and Pandey, 2019). P450s are located in the endoplasmic reticulum and Cytochrome P450 proteins are responsible for the use a single protein, POR, as their redox partner (Lu metabolism of numerous endogenous and exogenous et al., 1969;Masters, 2005). POR contains both the chemicals, including most of the drugs, and flavin mononucleotide (FMN) and the flavin adenine other xenobiotics (Schuster and Bernhardt, dinucleotide (FAD) which are bound to distinct

1

bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. domains and supplies electrons to cytochromes P450 crystallography (Zhao et al., 1999), and more and other proteins via protein-protein interactions recently many structures of soluble POR proteins (Iyanagi and Mason, 1973;Iyanagi et al., containing the NADPH, FAD, and FMN binding 1974;Matsubara et al., 1976;Vermilion and Coon, domains but lacking the membrane-bound amino 1978;Oprian and Coon, 1980) (Figure 1). The three- terminus sequence has been determined by several dimensional structure of the FMN binding domain of groups (Wang et al., 1997;Aigrain et al., 2009;Xia et human POR has been solved by x-ray al., 2011a;Xia et al., 2011b).

Figure 1: POR as electron transfer partner to cytochromes P450 proteins. Transfer of electrons from NADPH to redox partners of POR. Co-factor NADPH binds to the POR located into the endoplasmic reticulum and donates electrons which are received by FAD. Electron transfer to FAD causes a conformational change in POR that brings together the FAD, and FMN domains and electrons are then transferred from FAD to FMN. The FMN domain of POR interacts with the P450s and other redox partners and completes the final step of electron transfer. From Pandey, A. V. and Sproll, P (2014). “Pharmacogenomics of human P450 oxidoreductase.” Frontiers in Pharmacology 5, 103. hyperplasia is an inborn error of adrenal steroid Human POR deficiency (PORD, OMIM: biosynthesis affecting the production of 613537 and 201750) is a form of congenital adrenal glucocorticoids (Miller and Fluck, 2014;Fluck, hyperplasia, initially described in patients with 2017). Sequencing of the POR gene in patients with altered steroidogenesis (Flück et al., 2004;Miller et symptoms of mixed oxidase deficiency revealed al., 2004;Flück et al., 2007;Flück and Pandey, mutations in POR linked to disorders of steroid 2011;Flück and Pandey, 2013;Pandey and Flück, biosynthesis (Flück et al., 2004;Miller et al., 2013;Fukami and Ogata, 2014;Pandey and Sproll, 2004;Pandey et al., 2004). Afterward, several 2014;Flück and Pandey, 2019) followed by several different laboratories reported mutations in POR in reports with a broad spectrum of disorders (Adachi patients with steroid biosynthesis disorders and bone et al., 2004;Arlt et al., 2004;Pandey and Flück, malformation syndromes (Adachi et al., 2004;Arlt et 2013;Flück and Pandey, 2014;Pandey and Sproll, al., 2004;Fukami et al., 2005;Pandey and Flück, 2014;Oldani et al., 2015;Bonamichi et al., 2013;Riddick et al., 2013). While the initial studies 2016;Nakanishi et al., 2016;Burkhard et al., on mutations in POR were on the metabolism of 2017;Flück and Pandey, 2019). Congenital adrenal steroid hormones, effects of POR on other redox

2 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. partners including heme oxygenase and drug NADPH binding site mutations in POR (Pandey et metabolizing cytochromes P450 have been studied in al., 2007;Flück et al., 2011;Flück and Pandey, 2017). later reports (Marohnic et al., 2006;Agrawal et al., 2008;Kranendonk et al., 2008;Agrawal et al., In the current study, we have investigated the effects 2010;Flück et al., 2010;Marohnic et al., 2010;Nicolo of POR variant P284T located near the hinge region et al., 2010;Pandey et al., 2010;Marohnic et al., of POR that is crucial for interactions with redox 2011;McCammon et al., 2016;Udhane et al., 2017). partner proteins, including P450s, to transfer A mouse knockout (KO) model of Por-/- had been electrons from NADPH. The POR variant P284T generated before the human mutations in POR were (rs72557937) has been identified in the Yoruba identified. The Por-/- was found to be embryonic population through the sequencing of DNA obtained lethal (Shen et al., 2002;Nicholas et al., 2017), and from a particular community in Ibadan, Nigeria resembled the KO mouse model of retinoic acid- (https://www.coriell.org/0/Sections/Collections/NH metabolizing Cyp26a1-/- (Gaston-Massuet et al., GRI/Yoruba.aspx?PgId=128). Based on information 2016). Liver specific deletion of POR has been available through the 1000 genome project, the reported to cause lipidosis and increased liver weight samples were from parent-adult child trios, and all (Gu et al., 2003;Wu et al., 2003). Deletion of POR parents in the trios had identified themselves as gene in cardiomyosite cells of mouse lead to reduced having four Yoruba grandparents. The P284T variant P450 activity in heart but there was no indication of of POR was found in the African American embryonic lethality or early morbidity and no population in the POR sequencing study carried out difference in heart function was observed (Fang et by Miller al., 2008). (www.pharmagenomics.unsf.edu/populations.html).

Large-scale sequencing projects have now identified 2 Materials and methods many variations in the POR gene, in several different human subpopulations, and multiple POR variants 2.1 Recombinant Expression of POR and have been linked to altered drug metabolism (Gomes Membrane Purification et al., 2008;Huang et al., 2008;Gomes et al., 2009;Agrawal et al., 2010;Nicolo et al., 2010;Pandey The human POR WT and variant forms of proteins et al., 2010). Since POR is necessary for several (NCBI# NP_000932, Uniprot# P16435) were biochemical reactions of steroid biosynthesis that are recombinantly expressed in bacteria using the catalyzed by cytochrome P450 proteins, a complex previously described methods and expression disorder is caused by mutations in POR. Several constructs (Huang et al., 2005;Pandey et al., variants in the POR gene have been found in patients 2007;Huang et al., 2008;Nicolo et al., 2010). The and non-clinical samples, have been tested for protocol for expression of an N-27 form of POR enzymatic activities (Adachi et al., 2004;Arlt et al., variants and subsequent membrane purification have 2004;Fukami et al., 2005;Fukami et al., been described and adopted from our previous 2006;Homma et al., 2006;Pandey, 2006;Huang et al., publications (Huang et al., 2005;Pandey et al., 2008;Sim et al., 2009). 2007;Pandey et al., 2010;Parween et al., 2016;Flück and Pandey, 2017). The cDNAs for WT or mutant While the mutations in POR may be located in all POR in a pET22b vector were transformed into regions of the protein, based on analysis of Escherichia coli BL21(DE3). Single colonies were previously identified mutations, some definite selected for growth on ampicillin and grown in patterns have emerged. Mutations that are located terrific broth pH 7.4 supplemented with 40 mM near the co-factor binding sites (FMN, FAD, and FeCl3, 4 mM ZnCl2, 2 mM CoCl2, 2 mM Na2MoO4, NADPH) generally give rise to a severe form of the 2 mM CaCl2, 2 mM CuCl2, 2 mM H3BO3, 0.5 mg/ml disease with the mutations causing severe loss of riboflavin, 100 µg/ml carbenicillin at 37 °C to an FMN or FAD showing most significant inhibitory optical density (OD) 600 nm of 0.6 and temperature effects on activities of all redox partners. We have was reduced to 25 °C for 16 h. The bacterial cells previously shown that aromatase (CYP19A1) were collected by centrifugation, washed with PBS activity responsible for the metabolism of and suspended in 100 mM Tris-acetate (pH 7.6), 0.5 to form is more susceptible to changes in M sucrose, and 1 mM EDTA and treated with

3 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. lysozyme (0.5 mg/ml) and EDTA (0.1mM [pH 8.0]) at 4 ◦C for 1 h with slow stirring to generate For Western blots, 1 μg of POR-WT and POR- spheroplasts. The spheroplasts were pelleted by P284T bacterial membrane proteins were separated centrifugation at 5000×g for 15 min; and suspended on an SDS-PAGE gel and blotted on to polyvinyl in 100 mM potassium phosphate (pH 7.6), 6 mM difluoride (PVDF) membranes. Blots were first Magnesium acetate, 0.1 mM DTT, 20% (v/v) incubated with a rabbit polyclonal antibody against glycerol, 0.2 mM PMSF, and 0.1 mM DNase I; and wild-type human POR from Genscript (Genscript, disrupted by sonication. A clear lysate devoid of NJ, USA) at a dilution of 1:1000. We then used a cellular debris was obtained by centrifugation at secondary goat anti-rabbit antibody labeled with a 12,000×g for 10 min, and then the membranes were phthalocyanine infrared dye (IRDye 700DX, LI- collected by centrifugation at 100,000×g for 60 min COR Bioscience Inc., NE, USA) at a 1:10000 at 4 °C. Membranes containing POR were mixed in dilution. Signals were analyzed with the green 50 mM Potassium phosphate buffer (pH 7.8) and fluorescent channel (700 nm) on an Odyssey Infrared 20% (v/v) glycerol and kept at −70 ºC. The Imaging System (LI-COR Bioscience Inc., NE, concentrations of proteins were measured by the RC- USA), and protein bands were measured using the DC protein assay method (Protein Assay Dye Odyssey software (LI-COR Bioscience Inc., NE, Reagent, Bio-Rad, Hercules, CA) and POR content USA). POR content of each membrane preparation in membrane proteins was measured by western blot was measured, and all samples were normalized analysis. against purified wild-type POR used as a standard. In all experiments described in this report, the 2.3 Western Blot Analysis of POR Content in normalized amount of POR content was used for the the Bacterial Membranes mutants as well as WT POR protein.

Table 1. Kinetic parameters for activities of cytochrome c, MTT, ferricyanide reduction and CYP19A1 activity supported by POR-WT and POR-P284T variant. For the cytochrome c, MTT and Ferricyanide reduction assay, the NADPH concentration was fixed at 100 M and varying concentrations of the substrate were used for the analysis. For the conversion of to estrone, NADPH was fixed at 1 mM, and variable concentrations (10–1000 nM) of androstenedione were used. Data are shown as mean ± SEM of independent replicates.

Km, Vmax Vmax/Km % WT (µM) nmol/min/mg Cytochrome c reduction assay WT 4.6±0.7 128.2±6.1 27.6 100 P284T 3.7±0.8 48.7±3 13 47 MTT reduction assay WT 19.9±1.8 179.5±4 9 100 P284T 48.3±7.5 64.8±3.1 1.3 15 Ferricyanide reduction assay WT 18.9±2.4 600±19 31.8 100 P284T 6.9±1.6 248.1±11 36 113

Km, Androstenedione Vmax Vmax/Km % WT (nM) pmol/min/nmol CYP19A1; aromatase (androstenedione to estrone) WT 80±14.5 0.72±0.03 0.009 100 P284T 165.7±147.9 0.12±0.037 0.0008 9

4 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

2.4 Small molecule Reduction Assay by WT followed by centrifugation at 14000 x g for 10 min and POR-P284T to remove coagulated protein. The FMN and FAD ratio was determined by measurement of Assay of cytochrome c reduction by bacterially fluorescence of the supernatant at pH 7.7 and pH 2.6 expressed WT or POR-284T was performed as (excitation at 450 nm, emission at 535 nm) (Faeder described previously by measuring the change in and Siegel, 1973). absorbance at 550 nm (ε=21.1 cm−1 mM−1) (Guengerich et al., 2009). In brief, the POR reduction reaction was performed in 96-well plates, in triplicate, with 5 µg membrane preparation containing POR in each well in 100 mM Tris-HCl (pH 7.5), using a microplate reader (Spectramax M2e, Molecular Devices, Sunnyvale, CA). The concentration of NADPH used was 100 M and concentrations of cytochrome c (1.3–40 μM) were varied for kinetic analysis. The POR reduction reaction was initiated by the addition of NADPH and the change in absorbance at 550 nm was monitored over 6 minutes. Data were fitted based on Michaelis- Menten kinetics (Michaelis and Menten, 1913) using GraphPad Prism (GraphPad Software, La Jolla, CA USA) to determine the Vmax and Km.

The NADPH-dependent MTT [3-(4,5- dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium] reduction rate was measured as the rate of increase in absorbance at 610 nm using an extinction coefficient -1 -1 of ε610=11 mM cm (Yim et al., 2005). The assay mixture contained 5 g of bacterial membranes containing POR in 100 mM phosphate buffer (pH 7.6), 100M NADPH and concentration of MTT varied from 0-500 M. Similarly, the ferricyanide reduction rate was measured as the rate of decrease -1 -1 in absorbance at 420 nm (ε420=1.02 mM cm ). The concentration of ferricyanide varied over a range from 0 to 500 M and the reactions were started by adding 100M NADPH (Marohnic et al., 2010). Activities represent the mean of at least triplicate determinations.

Figure 2: Cytochrome c, MTT and Ferricyanide reduction 2.5 Flavin Content Analysis of WT and assay with WT and POR-P284T. (A) Cytochrome c, (B) MTT Mutant POR and (C) ferricyanide reduction assays were performed with the WT and POR-P284T variant. Kinetic assays were performed by monitoring the changes in absorbance at 550 nm for Protein-bound flavin molecules were released by cytochrome c, 610 nm for MTT, and 420 nm for ferricyanide thermal denaturation of POR proteins (Faeder and reduction. Data were fitted to the Michaelis-Menten kinetics Siegel, 1973). Flavin content of WT and mutant POR model and analyzed using GraphPad Prism. The calculated Km proteins (100 µg/ml) was determined by boiling and Vmax values are presented in Table 1. protein samples at 95 °C for 10 min in the dark,

5

bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

2.6 Expression and Purification of human 2.7 Assay of cytochrome P450 CYP19A1 in CYP19A1 Reconstituted Liposome System

The vector for bacterial expression of human Purified recombinant CYP19A1 using the bacterial CYP19A1was transformed in E. coli BL21(DE3) expression system was used to test the effect of POR- cells, and the recombinant protein was expressed and 284T to support the aromatase activity of CYP19A1. purified following previously published protocols Standard tritiated water release assay for the (Kagawa et al., 2003;Kagawa, 2011), with slight CYP19A1 activity was performed in a reconstituted modifications. Briefly, a single transformed colony liposome system using androstenedione as a was selected for protein expression at 25 °C. After 4 substrate. Bacterial membranes containing POR and h of incubation, 1 mM δ-aminolevulinic acid (a heme purified CYP19A1 were reconstituted into DLPC- precursor) and 4 mg/ml arabinose (for induction of DLPG liposomes. The liposomes were prepared molecular chaperones GroEL/GroES) were added to previously (Udhane et al., 2017), and aromatase the culture and further incubated for 20 h. Cells were activity was measured by the tritiated water release harvested, and spheroplasts were prepared with 0.2 assay originally described by Lephart and Simpson mg/ml lysozyme in 50 mM Tris-Acetate (pH 7.6), (Lephart and Simpson, 1991) using androstenedione 250 mM sucrose and 0.5 mM EDTA at stored at -80 as the substrate. Reaction mixture consisted of 100 °C. For protein purification, spheroplasts were lysed pmol of CYP19A1, 400 pmol of POR, 100 mM NaCl using 10XCellLytic B (Sigma-Aldrich) in buffer and 3H labeled androstenedione ([1ᵝ-3H(N)]-andros- containing 100mM potassium phosphate (pH 7.4), tene-3,17-dione; ~20,000 cpm) in 100 mM 500 mM sodium acetate, 0.1 mM EDTA, 0.1 mM potassium phosphate buffer (pH 7.4). Different DTT, 20% glycerol, and 1 mM PMSF. The cell lysate concentrations (10–1000 nM) of androstenedione was centrifuged, and the supernatants were pooled were used for kinetic analysis. The catalytic reaction for purification by Ni2+ affinity chromatography. was initiated by the addition of 1 mM NADPH, and Purification was performed at 4 °C and protein the reaction tube was incubated for 1 h under concentration after the dialysis was determined by shaking. Data were fitted based on Michaelis- DC protein assay (Protein Assay Dye Reagent, Bio- Menten kinetics using GraphPad Prism (GraphPad Rad, Hercules, CA) using BSA as standard. Software, La Jolla, CA USA).

Figure 3: Flavin content of POR-WT and POR-P284T variant. Flavin content of the POR proteins was analyzed by boiling the proteins under selective pH conditions. Relative fluorescence unit (RFU) of the flavins released from the POR variants measured at (A) pH 7.7 (FMN or F7.7) and (B) pH 2.6 (FAD or F2.6) are shown. The RFU of WT POR was fixed as a hundred percent. Data are shown as mean ± SEM of three independent replicates.

6 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

at an excitation wavelength of 415 nm and an 2.8 Assay of cytochrome P450 CYP2C9 Activity emission wavelength of 460 nm for BOMCC. in Reconstituted Liposome System 2.10 Assay of cytochrome P450 CYP3A4 The activity of CYP2C9 promoted by WT or mutant Activity in Reconstituted Liposome System POR was tested using the fluorogenic substrate BOMCC (Invitrogen Corp, Carlsbad, CA, United To compare the activities of CYP19A1 with another States). The purified CYP2C9 (CYPEX, Dundee, steroid binding cytochrome P450, we tested the Scotland, United Kingdom) was used to test the effect of POR mutations to support the enzyme activities of the POR variants using 20 µM BOMCC activity of CYP3A4. The activity of the major drug as substrate. In vitro CYP2C9 assays were performed metabolizing enzyme CYP3A4 supported by WT or using a reconstituted liposome system consisting of POR-284T was tested using the fluorogenic WT/mutant POR, CYP2C9 and cytochrome b5 at a substrates [BOMCC (7-Benzyloxy-4- ratio of 5:1:1. The final assay mixture consisted of 5 trifluoromethylcoumarin) (Invitrogen Corp, µg DLPC (1,2-Dilauroyl-sn-glycero-3- Carlsbad, CA) as described earlier (Flück et al., phosphocholine) and proteins (1 µM POR: 200 nM 2010). The purified CYP3A4 (CYPEX, Dundee, CYP2C9: 200 nM b5), 3 mM MgCl2, 20 µM Scotland, UK) was used to test the activities of the BOMCC in 100 mM Tris-HCl buffer PH 7.4 and the POR variants using 20 µM BOMCC. In-vitro reaction volume was 100 µL. The P450 reaction was CYP3A4 assays were performed using a started by addition of NADPH to a final reconstituted liposome system consisting of concentration of 1 mM, and fluorescence was WT/mutant POR, CYP3A4 and cytochrome b5 at a measured on a Spectramax M2e plate reader ratio of 4:1:1 (POR:CYP3A4:b5). The final assay (Molecular Devices, Sunnyvale, CA, United States) mixture consisted of liposomes and proteins (80 at an excitation wavelength of 415 nm and an pmol POR: 20 pmol CYP3A4: 20 pmol b5), 2.5 mM emission wavelength of 460 nm for BOMCC. MgCl2, 2.5 M GSH and 20 M BOMCC in 50 mM HEPES buffer and the reaction volume was 200 l. 2.9 Assay of cytochrome P450 CYP2C19 Activity The catalytic reaction was initiated by addition of in Reconstituted Liposome System NADPH to 1 mM final concentration, and fluorescence was monitored on a Spectramax M2e The activity of CYP2C19 promoted by WT or mutant plate reader (Molecular Devices, Sunnyvale, CA) at POR was tested using the fluorogenic substrate an excitation wavelength of 415 nm and an emission EOMCC (Invitrogen Corp, Carlsbad, CA, United wavelength of 460 nm. States). The purified CYP2C19 (CYPEX, Dundee, Scotland, United Kingdom) was used to test the 2.11 Assay of cytochrome P450 CYP3A5 activities of the POR variants using 20 µM EOMCC Activity in Reconstituted Liposome System as substrate. In vitro CYP2C19 assays were performed using a reconstituted liposome system The activity of CYP3A5 promoted by WT or mutant consisting of WT/mutant POR, CYP2C9 and POR was tested using the fluorogenic substrate cytochrome b5 at a ratio of 5:1:1. The final assay BOMCC (Invitrogen Corp, Carlsbad, CA, United mixture consisted of 2,5 µg DLPC (1,2-Dilauroyl-sn- States). The purified CYP3A5 (CYPEX, Dundee, glycero-3-phosphocholine) and proteins (0,5 µM Scotland, United Kingdom) was used to test the POR: 100 nM CYP2C9: 100 nM b5), 3 mM MgCl2, activities of the POR variants using 20 µM BOMCC 20 µM EOMCC in 100 mM Tris-HCl buffer PH 7.4 as substrate. In vitro CYP3A5 assays were and the reaction volume was 100 µL. The P450 performed using a reconstituted liposome system reaction was started by addition of NADPH to a final consisting of WT/mutant POR, CYP3A5 and concentration of 0.5 mM, and fluorescence was cytochrome b5 at a ratio of 5:1:1. The final assay measured on a Spectramax M2e plate reader mixture consisted of 5 µg DLPC (1,2-Dilauroyl-sn- (Molecular Devices, Sunnyvale, CA, United States) glycero-3-phosphocholine) and proteins (1 µM POR: 200 nM CYP2C9: 200 nM b5), 3 mM MgCl2, 20 µM 7

bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

BOMCC in 100 mM Tris-HCl buffer PH 7.4 and the POR-P284T affects both FMN as well as FAD reaction volume was 100 µL. The CYP3A5 reaction binding to POR (Figure 3). was started by addition of NADPH to 1 mM final concentration, and fluorescence was measured on a Spectramax M2e plate reader (Molecular Devices, Sunnyvale, CA, United States) at an excitation wavelength of 415 nm and an emission wavelength of 460 nm for BOMCC.

2.12 Statistical Analysis of results

Data are shown as mean, standard errors of the mean (SEM) in each group or replicates. Differences within the subsets of experiments were calculated using Student’s t-test. P values less than 0.05 were considered statistically significant.

3 Results Figure 4: Enzymatic activity of CYP19A1 supported by 3.1 Effect of POR-P284T on Small molecule POR-WT and POR-P284T variant. Bacterially expressed, Reduction Activity purified, recombinant CYP19A1 and the enriched bacterial membranes containing POR proteins were inserted into liposomes and their activity to convert [3H] labeled To study the effect of the P284T mutation on POR androstenedione to estrone was tested by the tritiated water activity, we expressed WT and POR-P284T in E.coli release assay. Data were analyzed using the Michaelis-Menten and measured their ability to transfer electrons from kinetics with GraphPad Prism. The calculated Km and Vmax NADPH to cytochrome c, MTT or ferricyanide. The values are shown in Table 1. POR-P284T mutation had a much lower capacity to reduce cytochrome c and MTT (Table 1, Figure 2A 3.3 CYP19A1- Aromatase Activity and 2B). Compared to WT POR, the P284T variant lost ~50% of its activity to reduce cytochrome c and The POR-P284T showed almost complete loss of had only 15 % of the WT POR activity to reduce CYP19A1 activity (Figure 4, Table 1). For the POR- MTT. However, the ferricyanide reduction activity P284T variant, the apparent Km for androstenedione (Table 1, Figure 2C) was not affected by the P284T was increased by 2- fold as compared to WT POR mutation. The loss of activities with cytochrome c suggesting that P284T mutation affects either and MTT indicates disruption of electron transport substrate interaction with CYP19A1 or the from NADPH to FAD, which could be due to CYP19A1-POR interaction. The apparent Vmax of conformational instability due to a P284T mutation POR-P284T was reduced by ~85%. The POR variant affecting domain movements and transfer of P284T showed only 9% residual activity in electrons from NADPH to FAD. supporting CYP19A1 as compared to WT POR.

3.2 Flavin Content 3.4 CYP2C9 Activity

To differentiate the conformational changes and We tested the activity of CYP2C9 supported by the effects of POR mutation P284T on flavin binding, we WT and P284T variant of POR in reconstituted evaluated the relative flavin content since the activity liposomes. Compared to WT POR activity of of POR may be affected by the changes in the CYP2C19 supported by P284T variant of POR was binding of cofactors FMN and FAD. As compared to reduced by 78% (Figure 5). This loss of more than WT POR, both the FMN and the FAD-binding was three-quarters of activity compared to WT POR affected due to P284T mutation. The FMN content indicated a severe effect on drug metabolism of POR-P284T was 64% as compared to WT while supported by CYP2C9. the FAD content was reduced by 35% suggesting that

8 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

3.5 CYP2C19 Activity 4 Discussion

The activity of CYP2C19 was tested with both the In previous reports, the effects of POR variants on WT and P284T variant of POR. We found that in drug metabolizing cytochrome P450 enzymes has CYP2C19 assays the P284T variant of POR showed not been studied in detail, and effects on most major only 44% of the WT POR activity (Figure 6). The drug metabolizing enzyme activities remain effect of P284T variation in POR on CYP2C19 was unknown. The cytochrome P450 subfamily 3A is the not as strong as in case of CYP2C9 activity, but the most versatile among all P450 enzymes and accounts loss of activity was still more than 50%, indicating a for the metabolism of a large number of xenobiotics reduced capacity of drug metabolism reactions and pharmaceutical compounds. Cytochrome P450 mediated by CYP2C19. 3A4 (CYP3A4) is the principal hepatic enzyme that metabolizes a large percentage of drugs and endogenous substrates (Klein and Zanger, 2013;Meyer et al., 2013;Zanger and Schwab, 2013). While CYP3A5 also has a significant role in xenobiotic metabolism in extrahepatic tissues like intestine and gut. The cytochrome P450 subfamily 2C is the second most relevant class of P450 proteins, and CYP2C9 and CYP2C19 are major drug- metabolizing enzymes.

Figure 5: Activity of cytochrome P450 CYP2C9 supported by POR-WT and POR-P284T variant. Assay of CYP3A4 activity was performed to compare POR-WT and POR-P284T by using 20 µM BOMCC as a substrate. Activity with the WT POR was fixed as a hundred percent, and results are given as a percentage of WT activity. Data are shown as mean ± SEM of three independent replicates.

3.6 CYP3A4 Activity

The P284T variant of POR showed only 22.8 % Figure 6: Activity of cytochrome P450 CYP2C19 supported activity as compared to the WT enzyme (Figure 7). by POR-WT and POR-P284T variant. Assay of CYP3A4 The loss of activities for drug metabolizing activity was performed to compare POR-WT and POR-P284T by using 20 µM EOMCC as a substrate. Activity with the WT cytochrome P450 enzyme-CYP3A4 by the POR POR was fixed as a hundred percent, and results are given as a variant P284T indicates problems with POR-P450 percentage of WT activity. Data are shown as mean ± SEM of interactions which seem to be different for different three independent replicates. cytochrome P450 partners. Our result shows that mutations in POR can affect 3.7 CYP3A5 Activity different activities of POR to varying extents and some cytochrome P450 proteins are affected more The 284T variant of POR had only 43.8% of the WT than others. Also, there may be differences due to activity in CYP3A5 assay (Figure 8). This was different allelic variations of cytochrome P450 different from the results obtained for the CYP3A4 proteins. The effects of different cytochrome P450 activity assays, indicating there are differences in the isoforms on the impact of POR activities has not interaction of POR with these two closely related been studied in detail. One study (Subramanian et al., cytochrome P450 proteins. 2012) tested different alleles of CYP2C9 (CYP2C9.1, CYP2C9.2, and CYP2C9.3) with a small

9 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. number of POR variants but patterns were similar for P284T mutation (9% of WT activity) indicated a all isoforms of CYP2C9 (Subramanian et al., 2012). significant effect on interaction with POR. In case of drug metabolizing cytochrome P450s, CYP2C9 and POR mutations leading to the significant loss of CYP3A4 lost more than 75% of activities while FAD/FMN co-factors hurt all redox partner CYP2C19 and CYP3A5 lost more than 50% of the activities. However, in most other cases, the effects WT POR activity due to P284T mutation in POR. of POR mutations on activities of the individual Taken together these results indicate variability in redox partner could be variable. For the mutation interaction with different cytochrome P450s due to P284T, we observed a small loss of cofactor binding, P284T mutation in POR. These results further which could be due to overall protein stability underscore the fact that effects of POR mutations can changes due to the location of the P284 residue near be quite variable for different redox partners and the hinge region of POR that joins FMN binding mutations in POR require detailed characterization domain with the FAD-binding domain. However, the with different partner proteins and small molecule activity of ferricyanide reduction which represents substrates to understand the biochemical direct electron transport through FAD without the consequences of individual POR mutations. involvement of FMN indicates that this loss of FAD did not severely reduce the overall quality of the protein. Cytochrome c reduction that involves protein-protein interaction and MTT reduction which a dual electron transfer system through FMN showed loss of activities.

Figure 8: Activity of cytochrome P450 CYP3A5 supported by POR-WT and POR-P284T variant. Assay of CYP3A5 activity was performed to compare POR-WT and POR-P284T by using BOMCC as a substrate. Activity with the WT POR was fixed as a hundred percent, and results are given as a percentage of WT activity. Data are shown as mean ± SEM of Figure 7: Activity of cytochrome P450 CYP3A4 supported three independent replicates. by POR-WT and POR-P284T variant. Assay of CYP3A4 activity was performed to compare POR-WT and POR-P284T Therefore, biochemical analysis using recombinant by using 20 µM BOMCC as a substrate. Activity with the WT proteins is necessary to confirm the damaging effects POR was fixed as a hundred percent, and results are given as a percentage of WT activity. Data are shown as mean ± SEM of of mutations with each redox partner separately three independent replicates. (Parween et al., 2016;Udhane et al., 2017;Flück and Pandey, 2019). Several cytochrome P450 proteins The location of the P284T mutation in the hinge for which POR is the redox partner, still have region indicated a potential impact on domain uncharacterized activities and their physiological movements which may change protein-protein roles are not defined. These, other partners of POR interaction which may differ based on which redox could also influence the metabolic profiles of partner is being tested. To test this hypothesis we patients with PORD. Preliminary computational tested POR-P284T with several different cytochrome docking and functional analysis suggest that the loss P450 proteins. The CYP19A1 activity requires the of activity caused by some POR variants may be transfer of three pairs of electrons, and therefore, is reversed by the introduction of external FMN more susceptible to change in protein interaction (Nicolo et al., 2010) or FAD (Marohnic et al., 2006). with POR (Pandey et al., 2007;Flück and Pandey, However, the effect of flavin treatment in PORD 2017). A severe loss of CYP19A1 activity due to needs to be tested in a clinical setting.

10 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

PORD is a complex disorder affecting both the steroid and the drug metabolism in the affected individual with varying effects. Different POR 9 References variations can cause highly variable effects on different metabolic reactions depending on the Adachi, M., Tachibana, K., Asakura, Y., Yamamoto, unique nature of the interaction of POR with T., Hanaki, K., and Oka, A. (2004). different redox partners. A careful examination of Compound heterozygous mutations of affected target proteins by use of urine/serum steroid cytochrome P450 oxidoreductase gene analysis is required for initial verification of PORD. (POR) in two patients with Antley-Bixler In cases of severe loss of drug metabolizing syndrome. Am J Med Genet A 128A, 333- cytochrome P450 enzyme activities, further 339. considerations about the clearance of drugs in Agrawal, V., Choi, J.H., Giacomini, K.M., and patients with PORD would require evaluation. Effect Miller, W.L. (2010). Substrate-specific of altered clearance of kidney transplant drug modulation of CYP3A4 activity by genetic tacrolimus has been indicated in several studies variants of cytochrome P450 oxidoreductase. among the people with POR*28 allele (Zhang et al., Pharmacogenet Genomics 20, 611-618. 2015). Effects of different POR variations found in Agrawal, V., Huang, N., and Miller, W.L. (2008). non-clinical populations should also be investigated Pharmacogenetics of P450 oxidoreductase: in detail, and our recent work has shown that some effect of sequence variants on activities of of these variations in POR could lead to severe CYP1A2 and CYP2C19. Pharmacogenet changes in activities of enzymes dependent on POR Genomics 18, 569-576. (Udhane et al., 2017). Aigrain, L., Pompon, D., Morera, S., and Truan, G. (2009). Structure of the open conformation of a functional chimeric NADPH cytochrome 5 Conflict of Interest : The authors declare P450 reductase. EMBO Rep 10, 742-747. that the research was conducted in the absence of Arlt, W., Walker, E.A., Draper, N., Ivison, H.E., any commercial or financial relationships that Ride, J.P., Hammer, F., Chalder, S.M., could be construed as a potential conflict of Borucka-Mankiewicz, M., Hauffa, B.P., interest. Malunowicz, E.M., Stewart, P.M., and Shackleton, C.H. (2004). Congenital adrenal 6 Author Contributions hyperplasia caused by mutant P450 oxidoreductase and human Participated in research design: AP synthesis: analytical study. Lancet 363, 2128- Conducted experiments: SP, MV, SU 2135. Contributed new reagents or analytical tools: NK Bonamichi, B.D.S.F., Santiago, S.L.M., Bertola, Performed data analysis: SP, MV, AP D.R., Kim, C.A., Alonso, N., Mendonca, Overall supervision of the project: AP B.B., Bachega, T.a.S.S., and Gomes, L.G. Wrote or contributed the writing of the manuscript: (2016). Long-term follow-up of a female SP, MV, SU, NK, AP with congenital adrenal hyperplasia due to P450-oxidoreductase deficiency. Archives of 7 Funding: This work was supported by the Endocrinology Metabolism 60, 500-504. Swiss National Science Foundation (31003A- Burkhard, F.Z., Parween, S., Udhane, S.S., Flück, 135926), the Novartis Foundation for Medical- C.E., and Pandey, A.V. (2017). P450 Biological Research (18A053) and a grant from Oxidoreductase deficiency: Analysis of Burgergemiende Bern to AVP. MNRV was mutations and polymorphisms. J Steroid supported by Consejo Nacional de Ciencia y Biochem Mol Biol 165, 38-50. Tecnología, Paraguay. Faeder, E.J., and Siegel, L.M. (1973). A rapid micromethod for determination of FMN and 8 Acknowledgments FAD in mixtures. Anal Biochem 53, 332-336. Fang, C., Gu, J., Xie, F., Behr, M., Yang, W., Abel, Prof. Walter L. Miller (UCSF, San Francisco, CA, E.D., and Ding, X.X. (2008). Deletion of the USA) provided several POR cDNA clones.

11 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

NADPH-cytochrome P450 reductase gene in Encyclopedia of Endocrine Diseases (Second cardiomyocytes does not protect mice against Edition), eds. I. Huhtaniemi & L. Martini. doxorubicin-mediated acute cardiac toxicity. (Oxford: Academic Press), 431-443. Drug Metabolism and Disposition 36, 1722- Flück, C.E., Tajima, T., Pandey, A.V., Arlt, W., 1728. Okuhara, K., Verge, C.F., Jabs, E.W., Fluck, C.E. (2017). MECHANISMS IN Mendonca, B.B., Fujieda, K., and Miller, ENDOCRINOLOGY: Update on W.L. (2004). Mutant P450 oxidoreductase pathogenesis of primary adrenal causes disordered steroidogenesis with and insufficiency: beyond steroid enzyme without Antley-Bixler syndrome. Nat Genet deficiency and autoimmune adrenal 36, 228-230. destruction. European journal of Fukami, M., Hasegawa, T., Horikawa, R., Ohashi, endocrinology 177, R99-R111. T., Nishimura, G., Homma, K., and Ogata, T. Flück, C.E., Mallet, D., Hofer, G., Samara-Boustani, (2006). Cytochrome P450 oxidoreductase D., Leger, J., Polak, M., Morel, Y., and deficiency in three patients initially regarded Pandey, A.V. (2011). Deletion of P399_E401 as having 21-hydroxylase deficiency and/or in NADPH cytochrome P450 oxidoreductase aromatase deficiency: diagnostic value of results in partial mixed oxidase deficiency. urine steroid hormone analysis. Pediatr Res Biochem Biophys Res Commun 412, 572- 59, 276-280. 577. Fukami, M., Horikawa, R., Nagai, T., Tanaka, T., Flück, C.E., Mullis, P.E., and Pandey, A.V. (2010). Naiki, Y., Sato, N., Okuyama, T., Nakai, H., Reduction in hepatic drug metabolizing Soneda, S., Tachibana, K., Matsuo, N., Sato, CYP3A4 activities caused by P450 S., Homma, K., Nishimura, G., Hasegawa, oxidoreductase mutations identified in T., and Ogata, T. (2005). Cytochrome P450 patients with disordered steroid metabolism. oxidoreductase gene mutations and Antley- Biochem Biophys Res Commun 401, 149- Bixler syndrome with abnormal genitalia 153. and/or impaired steroidogenesis: molecular Flück, C.E., Nicolo, C., and Pandey, A.V. (2007). and clinical studies in 10 patients. J Clin Clinical, structural and functional Endocrinol Metab 90, 414-426. implications of mutations and Fukami, M., and Ogata, T. (2014). Cytochrome P450 polymorphisms in human NADPH P450 oxidoreductase deficiency: Rare congenital oxidoreductase. Fundamental and Clinical disorder leading to skeletal malformations Pharmacology 21, 399-410. and steroidogenic defects. Pediatrics Flück, C.E., and Pandey, A.V. (2011). Clinical and International 56, 805-808. biochemical consequences of p450 Gaston-Massuet, C., Mccabe, M.J., Scagliotti, V., oxidoreductase deficiency. Endocr Dev 20, Young, R.M., Carreno, G., Gregory, L.C., 63-79. Jayakody, S.A., Pozzi, S., Gualtieri, A., Basu, Flück, C.E., and Pandey, A.V. (2013). "P450 B., Koniordou, M., Wu, C.I., Bancalari, R.E., Oxidoreductase Deficiency (PORD) " in Rahikkala, E., Veijola, R., Lopponen, T., Genetic Steroid Disorders, eds. M.I. New, O. Graziola, F., Turton, J., Signore, M., Lekarev, A. Parsa, T.T. Yuen, B.W. O'malley Mousavy Gharavy, S.N., Charolidi, N., & G.D. Hammer. (San Diego: Academic Sokol, S.Y., Andoniadou, C.L., Wilson, Press), 125-143. S.W., Merrill, B.J., Dattani, M.T., and Flück, C.E., and Pandey, A.V. (2014). Martinez-Barbera, J.P. (2016). Transcription Steroidogenesis of the testis -- new genes and factor 7-like 1 is involved in hypothalamo- pathways. Ann Endocrinol (Paris) 75, 40-47. pituitary axis development in mice and Flück, C.E., and Pandey, A.V. (2017). Impact on humans. Proc Natl Acad Sci U S A 113, CYP19A1 activity by mutations in NADPH E548-557. cytochrome P450 oxidoreductase. J Steroid Gomes, A.M., Winter, S., Klein, K., Turpeinen, M., Biochem Mol Biol 165, 64-70. Schaeffeler, E., Schwab, M., and Zanger, Flück, C.E., and Pandey, A.V. (2019). "Human P450 U.M. (2009). Pharmacogenomics of human Oxidoreductase Deficiency," in liver cytochrome P450 oxidoreductase:

12 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

multifactorial analysis and impact on Iyanagi, T., and Mason, H.S. (1973). Some microsomal drug oxidation. Properties of Hepatic Reduced Nicotinamide Pharmacogenomics 10, 579-599. Adenine-Dinucleotide Phosphate - Gomes, L.G., Huang, N., Agrawal, V., Mendonca, Cytochrome-C Reductase. Biochemistry 12, B.B., Bachega, T.A., and Miller, W.L. 2297-2308. (2008). The common P450 oxidoreductase Kagawa, N. (2011). Efficient expression of human variant A503V is not a modifier gene for 21- aromatase (CYP19) in E. coli. Methods Mol hydroxylase deficiency. J Clin Endocrinol Biol 705, 109-122. Metab 93, 2913-2916. Kagawa, N., Cao, Q., and Kusano, K. (2003). Gu, J., Weng, Y., Zhang, Q.Y., Cui, H.D., Behr, M., Expression of human aromatase (CYP19) in Wu, L., Yang, W.Z., Zhang, L., and Ding, Escherichia coli by N-terminal replacement X.X. (2003). Liver-specific deletion of the and induction of cold stress response. NADPH-cytochrome P450 reductase gene - Steroids 68, 205-209. Impact on plasma homeostasis Klein, K., and Zanger, U.M. (2013). and the function and regulation of Pharmacogenomics of Cytochrome P450 microsomal cytochrome P450 and heme 3A4: Recent Progress Toward the "Missing oxygenase. Journal of Biological Chemistry Heritability" Problem. Front Genet 4, 12. 278, 25895-25901. Kranendonk, M., Marohnic, C.C., Panda, S.P., Guengerich, F.P., Martin, M.V., Sohl, C.D., and Duarte, M.P., Oliveira, J.S., Masters, B.S.S., Cheng, Q. (2009). Measurement of and Rueff, J. (2008). Impairment of human cytochrome P450 and NADPH-cytochrome CYP1A2-mediated xenobiotic metabolism P450 reductase. Nat Protoc 4, 1245-1251. by Antley-Bixler syndrome variants of Homma, K., Hasegawa, T., Nagai, T., Adachi, M., cytochrome P450 oxidoreductase. Archives Horikawa, R., Fujiwara, I., Tajima, T., of Biochemistry and Biophysics 475, 93-99. Takeda, R., Fukami, M., and Ogata, T. Lephart, E.D., and Simpson, E.R. (1991). Assay of (2006). Urine steroid hormone profile aromatase activity. Methods Enzymol 206, analysis in cytochrome P450 oxidoreductase 477-483. deficiency: implication for the backdoor Lu, A.Y., Junk, K.W., and Coon, M.J. (1969). pathway to dihydrotestosterone. J Clin Resolution of the cytochrome P-450- Endocrinol Metab 91, 2643-2649. containing -hydroxylation system of liver Huang, N., Agrawal, V., Giacomini, K.M., and microsomes into three components. J Biol Miller, W.L. (2008). Genetics of P450 Chem 244, 3714-3721. oxidoreductase: sequence variation in 842 Marohnic, C.C., Huber Iii, W.J., Patrick Connick, J., individuals of four ethnicities and activities Reed, J.R., Mccammon, K., Panda, S.P., of 15 missense mutations. Proc Natl Acad Sci Martasek, P., Backes, W.L., and Masters, U S A 105, 1733-1738. B.S. (2011). Mutations of human cytochrome Huang, N., Pandey, A.V., Agrawal, V., Reardon, W., P450 reductase differentially modulate heme Lapunzina, P.D., Mowat, D., Jabs, E.W., Van oxygenase-1 activity and oligomerization. Vliet, G., Sack, J., Flück, C.E., and Miller, Arch Biochem Biophys 513, 42-50. W.L. (2005). Diversity and function of Marohnic, C.C., Panda, S.P., Martasek, P., and mutations in p450 oxidoreductase in patients Masters, B.S. (2006). Diminished FAD with Antley-Bixler syndrome and disordered binding in the Y459H and V492E Antley- steroidogenesis. Am J Hum Genet 76, 729- Bixler syndrome mutants of human 749. cytochrome P450 reductase. J Biol Chem Iyanagi, T., Makino, N., and Mason, H.S. (1974). 281, 35975-35982. Redox Properties of Reduced Nicotinamide Marohnic, C.C., Panda, S.P., Mccammon, K., Rueff, Adenine-Dinucleotide Phosphate- J., Masters, B.S.S., and Kranendonk, M. Cytochrome P-450 and Reduced (2010). Human Cytochrome P450 Nicotinamide Adenine Dinucleotide- Oxidoreductase Deficiency Caused by the Cytochrome B5 Reductases. Biochemistry Y181D Mutation: Molecular Consequences 13, 1701-1710.

13 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

and Rescue of Defect. Drug Metabolism and Nicolo, C., Flück, C.E., Mullis, P.E., and Pandey, Disposition 38, 332-340. A.V. (2010). Restoration of mutant Masters, B.S.S. (2005). The journey from NADPH- cytochrome P450 reductase activity by cytochrome P450 oxidoreductase to nitric external flavin. Mol Cell Endocrinol 321, oxide synthases. Biochemical and 245-252. Biophysical Research Communications 338, Oldani, E., Garel, C., Bucourt, M., and Carbillon, L. 507-519. (2015). Prenatal Diagnosis of Antley-Bixler Matsubara, T., Baron, J., Peterson, L.L., and Syndrome and POR Deficiency. Am J Case Peterson, J.A. (1976). NADPH-cytochrome Rep 16, 882-885. P450 reductase. Arch Biochem Biophys 172, Omura, T. (2006). Mitochondrial P450s. Chem Biol 463-469. Interact 163, 86-93. Mccammon, K.M., Panda, S.P., Xia, C., Kim, J.J., Omura, T. (2010). Structural diversity of cytochrome Moutinho, D., Kranendonk, M., Auchus, P450 enzyme system. Journal of R.J., Lafer, E.M., Ghosh, D., Martasek, P., Biochemistry 147, 297-306. Kar, R., Masters, B.S., and Roman, L.J. Oprian, D.D., and Coon, M.J. (1980). Redox States (2016). Instability of the Human Cytochrome of Fmn and Fad in Nadph-Cytochrome P-450 P450 Reductase A287P Variant Is the Major Reductase during Reduction by Nadph. Contributor to Its Antley-Bixler Syndrome- Federation Proceedings 39, 2054-2054. like Phenotype. J Biol Chem 291, 20487- Pandey, A.V. (2006). Biochemical analysis of 20502. mutations in P450 oxidoreductase. Biochem Mclean, K.J., Luciakova, D., Belcher, J., Tee, K.L., Soc Trans 34, 1186-1191. and Munro, A.W. (2015). Biological Pandey, A.V., and Flück, C.E. (2013). NADPH P450 diversity of cytochrome P450 redox partner oxidoreductase: structure, function, and systems. Adv Exp Med Biol 851, 299-317. pathology of diseases. Pharmacol Ther 138, Meyer, U.A., Zanger, U.M., and Schwab, M. (2013). 229-254. Omics and drug response. Annu Rev Pandey, A.V., Fluck, C.E., Huang, N., Tajima, T., Pharmacol Toxicol 53, 475-502. Fujieda, K., and Miller, W.L. (2004). P450 Michaelis, L., and Menten, M.L. (1913). Die Kinetik oxidoreductase deficiency: a new disorder of der Invertinwirkung. Biochemische steroidogenesis affecting all microsomal Zeitschrift 49, 333-369. P450 enzymes. Endocr Res 30, 881-888. Miller, W.L., and Fluck, C.E. (2014). "Adrenal Pandey, A.V., Flück, C.E., and Mullis, P.E. (2010). cortex and its disorders," in Pediatric Altered heme catabolism by heme Endocrinology, ed. M.A. Sperling. 4th ed: oxygenase-1 caused by mutations in human Saunders). NADPH cytochrome P450 reductase. Miller, W.L., Huang, N., Flück, C.E., and Pandey, Biochem Biophys Res Commun 400, 374- A.V. (2004). P450 oxidoreductase 378. deficiency. Lancet 364, 1663. Pandey, A.V., Kempna, P., Hofer, G., Mullis, P.E., Nakanishi, K., Yamashita, A., Miyamoto, T., and Flück, C.E. (2007). Modulation of Takeguchi, R., Furuya, A., Matsuo, K., human CYP19A1 activity by mutant Tanahashi, Y., Kawamura, M., and Sengoku, NADPH P450 oxidoreductase. Mol K. (2016). P450 oxidoreductase deficiency Endocrinol 21, 2579-2595. with maternal during pregnancy. Pandey, A.V., and Sproll, P. (2014). Clinical and Experimental Obstetrics & Pharmacogenomics of human P450 Gynecology 43, 902-904. oxidoreductase. Front Pharmacol 5, 103. Nicholas, A.K., Jaleel, S., Lyons, G., Schoenmakers, Parween, S., Roucher-Boulez, F., Flück, C.E., E., Dattani, M.T., Crowne, E., Bernhard, B., Lienhardt-Roussie, A., Mallet, D., Morel, Y., Kirk, J., Roche, E.F., Chatterjee, V.K., and and Pandey, A.V. (2016). P450 Schoenmakers, N. (2017). Molecular Oxidoreductase Deficiency: Loss of Activity spectrum of TSHbeta subunit gene defects in Caused by Protein Instability From a Novel central hypothyroidism in the UK and L374H Mutation. J Clin Endocrinol Metab Ireland. Clin Endocrinol (Oxf) 86, 410-418. 101, 4789-4798.

14 bioRxiv preprint doi: https://doi.org/10.1101/643825; this version posted May 20, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Riddick, D.S., Ding, X., Wolf, C.R., Porter, T.D., Q., and Ding, X.X. (2003). Conditional Pandey, A.V., Zhang, Q.Y., Gu, J., Finn, knockout of the mouse NADPH-cytochrome R.D., Ronseaux, S., Mclaughlin, L.A., p450 reductase gene. Genesis 36, 177-181. Henderson, C.J., Zou, L., and Flück, C.E. Xia, C., Hamdane, D., Shen, A.L., Choi, V., Kasper, (2013). NADPH-cytochrome P450 C.B., Pearl, N.M., Zhang, H., Im, S.C., oxidoreductase: roles in physiology, Waskell, L., and Kim, J.J. (2011a). pharmacology, and toxicology. Drug Metab Conformational changes of NADPH- Dispos 41, 12-23. cytochrome P450 oxidoreductase are Schuster, I., and Bernhardt, R. (2007). Inhibition of essential for catalysis and cofactor binding. J cytochromes p450: existing and new Biol Chem 286, 16246-16260. promising therapeutic targets. Drug Metab Xia, C., Panda, S.P., Marohnic, C.C., Martasek, P., Rev 39, 481-499. Masters, B.S., and Kim, J.J. (2011b). Shen, A.L., O'leary, K.A., and Kasper, C.B. (2002). Structural basis for human NADPH- Association of multiple developmental cytochrome P450 oxidoreductase deficiency. defects and embryonic lethality with loss of Proc Natl Acad Sci U S A 108, 13486-13491. microsomal NADPH-cytochrome P450 Yim, S.K., Yun, C.H., Ahn, T., Jung, H.C., and Pan, oxidoreductase. J Biol Chem 277, 6536- J.G. (2005). A continuous 6541. spectrophotometric assay for NADPH- Sim, S.C., Miller, W.L., Zhong, X.B., Arlt, W., cytochrome P450 reductase activity using 3- Ogata, T., Ding, X., Wolf, C.R., Flück, C.E., (4,5-dimethylthiazol-2-yl)-2,5- Pandey, A.V., Henderson, C.J., Porter, T.D., diphenyltetrazolium bromide. J Biochem Mol Daly, A.K., Nebert, D.W., and Ingelman- Biol 38, 366-369. Sundberg, M. (2009). Nomenclature for Zalewski, A., Ma, N.S., Legeza, B., Renthal, N., alleles of the cytochrome P450 Flück, C.E., and Pandey, A.V. (2016). oxidoreductase gene. Pharmacogenet Vitamin D-Dependent Rickets Type 1 Genomics 19, 565-566. Caused by Mutations in CYP27B1 Affecting Subramanian, M., Agrawal, V., Sandee, D., Tam, Protein Interactions With Adrenodoxin. J H.K., Miller, W.L., and Tracy, T.S. (2012). Clin Endocrinol Metab 101, 3409-3418. Effect of P450 oxidoreductase variants on the Zanger, U.M., and Schwab, M. (2013). Cytochrome metabolism of model substrates mediated by P450 enzymes in drug metabolism: CYP2C9.1, CYP2C9.2, and CYP2C9.3. regulation of gene expression, enzyme Pharmacogenet Genomics 22, 590-597. activities, and impact of genetic variation. Udhane, S.S., Parween, S., Kagawa, N., and Pandey, Pharmacol Ther 138, 103-141. A.V. (2017). Altered CYP19A1 and Zhang, J.J., Liu, S.B., Xue, L., Ding, X.L., Zhang, CYP3A4 Activities Due to Mutations H., and Miao, L.Y. (2015). The genetic A115V, T142A, Q153R and P284L in the polymorphisms of POR*28 and CYP3A5*3 Human P450 Oxidoreductase. Frontiers in significantly influence the pharmacokinetics Pharmacology 8. of tacrolimus in Chinese renal transplant Vermilion, J.L., and Coon, M.J. (1978). recipients. Int J Clin Pharmacol Ther 53, Identification of the High and Low Potential 728-736. Flavins of Liver Microsomal Nadph- Zhao, Q., Modi, S., Smith, G., Paine, M., Mcdonagh, Cytochrome P-450 Reductase. Journal of P.D., Wolf, C.R., Tew, D., Lian, L.Y., Biological Chemistry 253, 8812-8819. Roberts, G.C., and Driessen, H.P. (1999). Wang, M., Roberts, D.L., Paschke, R., Shea, T.M., Crystal structure of the FMN-binding domain Masters, B.S., and Kim, J.J. (1997). Three- of human cytochrome P450 reductase at 1.93 dimensional structure of NADPH- A resolution. Protein Sci 8, 298-306. cytochrome P450 reductase: prototype for FMN- and FAD-containing enzymes. Proc Natl Acad Sci U S A 94, 8411-8416. Wu, L., Gu, J., Weng, Y., Kluetzman, K., Swiatek, P., Behr, M., Zhang, Q.Y., Zhuo, X.L., Xie,

15