Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Grover P. Miller is active.

Publication


Featured researches published by Grover P. Miller.


Chemistry & Biology | 1998

Stretching exercises — flexibility in dihydrofolate reductase catalysis

Grover P. Miller; Stephen J. Benkovic

As an enzyme, dihydrofolate reductase performs two tasks: transformation of its substrate dihydrofolate or folate to tetrahydrofolate, using NADPH as a cofactor, and regeneration of the enzyme for a subsequent round of catalysis. Studies discussed in this review highlight the role of conformational flexibility in both of these enzymatic functions.


Journal of Biological Chemistry | 2008

CYP2E1 Substrate Inhibition: MECHANISTIC INTERPRETATION THROUGH AN EFFECTOR SITE FOR MONOCYCLIC COMPOUNDS

Samuel L. Collom; Ryan M. Laddusaw; Amber M. Burch; Petr Kuzmic; Martin D. Perry; Grover P. Miller

In this study we offer a mechanistic interpretation of the previously known but unexplained substrate inhibition observed for CYP2E1. At low substrate concentrations, p-nitrophenol (pNP) was rapidly turned over (47 min-1) with relatively low Km (24 μm); nevertheless, at concentrations of >100 μm, the rate of pNP oxidation gradually decreased as a second molecule bound to CYP2E1 through an effector site (Kss = 260 μm), which inhibited activity at the catalytic site. 4-Methylpyrazole (4MP) was a potent inhibitor for both sites through a mixed inhibition mechanism. The Ki for the catalytic site was 2.0 μm. Although we were unable to discriminate whether an EIS or ESI complex formed, the respective inhibition constants were far lower than Kss. Bicyclic indazole (IND) inhibited catalysis through a single CYP2E1 site (Ki = 0.12 μm). Similarly, 4MP and IND yielded type II binding spectra that reflected the association of either two 4MP or one IND molecule(s) to CYP2E1, respectively. Based on computational docking studies with a homology model for CYP2E1, the two sites for monocyclic molecules, pNP and 4MP, exist within a narrow channel connecting the active site to the surface of the enzyme. Because of the presence of the heme iron, one site supports catalysis, whereas the other more distal effector site binds molecules that can influence the binding orientation and egress of molecules for the catalytic site. Although IND did not bind these sites simultaneously, the presence of IND at the catalytic site blocked binding at the effector site.


Drug Metabolism and Disposition | 2009

Characterization of Human Hepatic and Extrahepatic UDP-Glucuronosyltransferase Enzymes Involved in the Metabolism of Classic Cannabinoids

Anna Mazur; Cheryl F. Lichti; Paul L. Prather; Agnieszka K. Zielinska; Stacie M. Bratton; Anna Gallus-Zawada; Moshe Finel; Grover P. Miller; Anna Radominska-Pandya; Jeffery H. Moran

Tetrahydrocannabinol (Δ9-THC), the primary psychoactive ingredient in marijuana, is subject to cytochrome P450 oxidation and subsequent UDP-glucuronosyltransferase (UGT)-dependent glucuronidation. Many studies have shown that CYP2C9 and CYP3A4 are the primary enzymes responsible for these cytochrome P450-dependent oxidations, but little work has been done to characterize phase II metabolic pathways. In this study, we test the hypothesis that there are specific human UGTs responsible for classic cannabinoid metabolism. The activities of 12 human recombinant UGTs toward classic cannabinoids [cannabinol (CBN), cannabidiol (CBD), (–)-Δ8-THC, (–)-Δ9-THC, (±)-11-hydroxy-Δ9-THC (THC-OH), and (–)-11-nor-9-carboxy-Δ9-THC (THC-COOH)] were evaluated using high-performance liquid chromatography-tandem mass spectrometry and labeling assays. Despite activity by UGT1A1, 1A3, 1A8, 1A9, 1A10, and 2B7 toward CBN, CBD, THC-OH, and THC-COOH, only selected UGTs demonstrate sufficient activity for further characterization of steady-state kinetics. CBN was the most recognized substrate as evidenced by activities from hepatic UGT1A9 and extrahepatic UGT1A7, UGT1A8, and UGT1A10. These results may reflect the introduction of an aromatic ring to Δ9-THC, leading to favorable π stacking with phenylalanines in the UGT active site. Likewise, oxidation of Δ9-THC to THC-OH results in UGT1A9 and UGT1A10 activity toward the cannabinoid. Further oxidation to THC-COOH surprisingly leads to a loss in metabolism by UGT1A9 and UGT1A10, while creating a substrate recognized by UGT1A1 and UGT1A3. The resulting glucuronide of THC-COOH is the main metabolite found in urine, and thus these hepatic enzymes play a critical role in the metabolic clearance of cannabinoids. Taken together, glucuronidation of cannabinoids depends on upstream processing including enzymes such as CYP2C9 and CYP3A4.


Journal of Pharmacology and Experimental Therapeutics | 2007

Glucuronidation of Monohydroxylated Warfarin Metabolites by Human Liver Microsomes and Human Recombinant UDP-Glucuronosyltransferases

Agnieszka K. Zielinska; Cheryl F. Lichti; Stacie M. Bratton; Neil C. Mitchell; Anna Gallus-Zawada; Vi-Huyen Le; Moshe Finel; Grover P. Miller; Anna Radominska-Pandya; Jeffery H. Moran

Our understanding of human phase II metabolic pathways which facilitate detoxification and excretion of warfarin (Coumadin) is limited. The goal of this study was to test the hypothesis that there are specific human hepatic and extrahepatic UDP-glucuronosyltransferase (UGT) isozymes, which are responsible for conjugating warfarin and hydroxylated metabolites of warfarin. Glucuronidation activity of human liver microsomes (HLMs) and eight human recombinant UGTs toward (R)- and (S)-warfarin, racemic warfarin, and major cytochrome P450 metabolites of warfarin (4′-, 6-, 7-, 8-, and 10-hydroxywarfarin) has been assessed. HLMs, UGT1A1, 1A8, 1A9, and 1A10 showed glucuronidation activity toward 4′-, 6-, 7-, and/or 8-hydroxywarfarin with Km values ranging from 59 to 480 μM and Vmax values ranging from 0.03 to 0.78 μM/min/mg protein. Tandem mass spectrometry studies and structure comparisons suggested glucuronidation was occurring at the C4′-, C6-, C7-, and C8-positions. Of the hepatic UGT isozymes tested, UGT1A9 exclusively metabolized 8-hydroxywarfarin, whereas UGT1A1 metabolized 6-, 7-, and 8-hydroxywarfarin. Studies with extrahepatic UGT isoforms showed that UGT1A8 metabolized 7- and 8-hydroxywarfarin and that UGT1A10 glucuronidated 4′-, 6-, 7-, and 8-hydroxywarfarin. UGT1A4, 1A6, 1A7, and 2B7 did not have activity with any substrate, and none of the UGT isozymes evaluated catalyzed reactions with (R)- and (S)-warfarin, racemic warfarin, or 10-hydroxywarfarin. This is the first study identifying and characterizing specific human UGT isozymes, which glucuronidate major cytochrome P450 metabolites of warfarin with similar metabolic rates known to be associated with warfarin metabolism. Continued characterization of these pathways may enhance our ability to reduce life-threatening and costly complications associated with warfarin therapy.


ACS central science | 2015

Modeling Epoxidation of Drug-like Molecules with a Deep Machine Learning Network.

Tyler B. Hughes; Grover P. Miller; S. Joshua Swamidass

Drug toxicity is frequently caused by electrophilic reactive metabolites that covalently bind to proteins. Epoxides comprise a large class of three-membered cyclic ethers. These molecules are electrophilic and typically highly reactive due to ring tension and polarized carbon–oxygen bonds. Epoxides are metabolites often formed by cytochromes P450 acting on aromatic or double bonds. The specific location on a molecule that undergoes epoxidation is its site of epoxidation (SOE). Identifying a molecule’s SOE can aid in interpreting adverse events related to reactive metabolites and direct modification to prevent epoxidation for safer drugs. This study utilized a database of 702 epoxidation reactions to build a model that accurately predicted sites of epoxidation. The foundation for this model was an algorithm originally designed to model sites of cytochromes P450 metabolism (called XenoSite) that was recently applied to model the intrinsic reactivity of diverse molecules with glutathione. This modeling algorithm systematically and quantitatively summarizes the knowledge from hundreds of epoxidation reactions with a deep convolution network. This network makes predictions at both an atom and molecule level. The final epoxidation model constructed with this approach identified SOEs with 94.9% area under the curve (AUC) performance and separated epoxidized and non-epoxidized molecules with 79.3% AUC. Moreover, within epoxidized molecules, the model separated aromatic or double bond SOEs from all other aromatic or double bonds with AUCs of 92.5% and 95.1%, respectively. Finally, the model separated SOEs from sites of sp2 hydroxylation with 83.2% AUC. Our model is the first of its kind and may be useful for the development of safer drugs. The epoxidation model is available at http://swami.wustl.edu/xenosite.


Journal of Chromatography B | 2011

Novel multi-mode ultra performance liquid chromatography-tandem mass spectrometry assay for profiling enantiomeric hydroxywarfarins and warfarin in human plasma.

Drew R. Jones; Gunnar Boysen; Grover P. Miller

Coumadin (R/S-warfarin) is a commonly prescribed anticoagulant for over ∼20 million Americans. Although highly efficacious, positive clinical outcomes during warfarin therapy depend on maintaining a narrow therapeutic range for the drug. This goal is challenging due to large inter-individual variability in patient response, which has been attributed to diversity in drug metabolism. Warfarin is given as a racemic mixture and evidence suggest differences of R and S-warfarin in their therapeutic activities and metabolism. Previous investigation of warfarin metabolism has been hampered by the inability to quantify the individual enantiomers. To overcome this limitation a multi-mode LC-MS/MS method is reported. This strategy combines phenyl based reverse phase chromatography with chiral phase chromatography prior to quantitation by liquid chromatography tandem mass spectrometry. This approach was made possible through advances in UPLC technology producing narrow peaks suitable for transferring to a second column. The reported method separated individual R and S enantiomers of hydroxywarfarin and warfarin. All four possible isomers of 10-hydroxywarfarin were resolved to reveal unprecedented insights into the stereo-specific metabolism of warfarin. Characterization of the method demonstrated that it is robust and sensitive with inter-day coefficients of error between <7% and a detection limit of 2 nM in sample or 10 fmol on column for each analyte. Individual metabolites may be suitable surrogate biomarkers or predictive markers that predict warfarin dose, adverse interactions, or other important clinical outcomes during anticoagulant therapy. Consequently, the metabolite profiles obtained through this dual phase UPLC-MS/MS method are expected to increase our understanding of the role warfarin metabolism plays in patient response to therapy and yield new strategies to improve patient outcomes.


Expert Opinion on Drug Metabolism & Toxicology | 2008

Advances in the interpretation and prediction of CYP2E1 metabolism from a biochemical perspective.

Grover P. Miller

Background: Cytochrome P450 2E1 (CYP2E1) plays a central role in the metabolism and metabolic activation of a large number of small, mostly xenobiotic compounds. These qualities distinguish CYP2E1 from traditional enzymes and pose significant challenges to understanding the role and consequences of CYP2E1-mediated metabolism. Objective: This review discusses recent advances in kinetic profiling, quantitative structure–activity relationships and structural studies that have furthered the development of tools to interpret and predict CYP2E1 metabolism. Methods: Analysis of kinetic profiles by specific mechanisms produces important parameters describing specificity, stoichiometry and metabolism of molecules. Quantitative structure–activity relationships reveal a more specific basis for molecular recognition by CYP2E1. The corresponding protein structures imparting these interactions are the focus of chemical modifications, site-directed mutagenesis and homology modeling studies. Results: Compilation of kinetic profiling for CYP2E1 substrates established the selectivity for small substrates, whose characteristics could be generalized in parameters for hydrophobicity and steric properties as determined by quantitative structure–activity relationships. The possibility of an effector site for monocyclic compounds added an interesting variable to these modeling efforts. Various structural studies identified important residues contributing to binding and catalysis as well as the volume and location of the active site relative to the heme moiety. Pressure and carbon monoxide-binding experiments also demonstrated the inherent conformational flexibility of CYP2E1 that may contribute to rate-limiting steps during catalytic turnover. Conclusion: Although combinations of these approaches have reinforced important observations, more work is needed to verify findings and seek broader impacts for various interpretative and predictive tools.


Drug Metabolism Reviews | 2009

Warfarin and UDP-glucuronosyltransferases: writing a new chapter of metabolism

Drew R. Jones; Jeffery H. Moran; Grover P. Miller

The widely prescribed anticoagulant, Coumadin (racemic R/S-warfarin), Bristol-Myers Squibb Company, Clinton, NY has a narrow therapeutic range and wide interindividual response due, in part, to drug metabolism. Early identification of hydroxywarfarins (OHWARs), especially S-7-OHWAR, as major metabolites fostered studies characterizing cytochrome P450s responsible for those reactions. Nevertheless, phase II metabolism by sulfotransferases and, especially uridine diphosphate (UDP)-glucuronosyltransferases (UGTs), marks the next chapter in warfarin inactivation and clearance. Rodents converted OHWARs to glucuronides (O-GLUC), including high levels of 4’-, 7-, and 8-O-GLUC. Similarly, humans generated significant levels of glucuronides following treatment with warfarin. 7-O-GLUC was a major metabolite, while 6- and 8-O-GLUC were minor ones. Surprisingly, warfarin glucuronidation accounted for up to 13% of metabolites. This capacity in humans derives from several UGTs, as shown by studies with recombinant enzymes and racemic warfarin and OHWARs. 7-OHWAR was a high-affinity substrate for UGT1A1, compared to other UGTs. UGT1A1 and UGT1A10 also glucuronidated 6-OHWAR. Of five UGT1A enzymes, UGT1A10 was approximately 7-fold more efficient than the rest. Broad substrate specificity for UGT1A10 derives, in part, from an active site-binding motif, specifically F90-M91-V92-F93. Unlike glucuronidation, less is known about sulfonation of warfarin and its metabolites, except that low capacities are shown by rats and, possibly, humans. Collectively, phase I and II metabolic steps create pathways for inactivating and eliminating warfarin that require elucidation. These findings will ultimately enrich our understanding of warfarin metabolism and facilitate the interpreting of metabolic profiles of patients. This knowledge will possibly avoid complications during warfarin therapy related to metabolism by personalizing therapy for the patient.


Chemical Research in Toxicology | 2015

Site of reactivity models predict molecular reactivity of diverse chemicals with glutathione.

Tyler B. Hughes; Grover P. Miller; S. Joshua Swamidass

Drug toxicity is often caused by electrophilic reactive metabolites that covalently bind to proteins. Consequently, the quantitative strength of a molecules reactivity with glutathione (GSH) is a frequently used indicator of its toxicity. Through cysteine, GSH (and proteins) scavenges reactive molecules to form conjugates in the body. GSH conjugates to specific atoms in reactive molecules: their sites of reactivity. The value of knowing a molecules sites of reactivity is unexplored in the literature. This study tests the value of site of reactivity data that identifies the atoms within 1213 reactive molecules that conjugate to GSH and builds models to predict molecular reactivity with glutathione. An algorithm originally written to model sites of cytochrome P450 metabolism (called XenoSite) finds clear patterns in molecular structure that identify sites of reactivity within reactive molecules with 90.8% accuracy and separate reactive and unreactive molecules with 80.6% accuracy. Furthermore, the model output strongly correlates with quantitative GSH reactivity data in chemically diverse, external data sets. Site of reactivity data is nearly unstudied in the literature prior to our efforts, yet it contains a strong signal for reactivity that can be utilized to more accurately predict molecule reactivity and, eventually, toxicity.


Drug Metabolism and Disposition | 2012

CYP2E1 metabolism of styrene involves allostery.

Jessica H. Hartman; Gunnar Boysen; Grover P. Miller

We are the first to report allosterism during styrene oxidation by recombinant CYP2E1 and human liver microsomes. At low styrene concentrations, oxidation is inefficient because of weak binding to CYP2E1 (Ks = 830 μM). A second styrene molecule then binds CYP2E1 with higher affinity (Kss = 110 μM) and significantly improves oxidation to achieve a kcat of 6.3 nmol · min−1 · nmol CYP2E1−1. The transition between these metabolic cycles coincides with reported styrene concentrations in blood from exposed workers; thus, this CYP2E1 mechanism may be relevant in vivo. Scaled modeling of the in vitro-positive allosteric mechanism for styrene metabolism to its in vivo clearance led to significant deviations from the traditional model based on Michaelis-Menten kinetics. Low styrene levels were notably much less toxic than generally assumed. We interrogated the allosteric mechanism using the CYP2E1-specific inhibitor and drug 4-methylpyrazole, which we have shown binds two CYP2E1 sites. From the current studies, styrene was a positive allosteric effector on 4-methylpyrazole binding, based on a 10-fold increase in 4-methylpyrazole binding affinity from Ki 0.51 to Ksi 0.043 μM. The inhibitor was a negative allosteric effector on styrene oxidation, because kcat decreased 6-fold to 0.98 nmol · min−1 · nmol CYP2E1−1. Consequently, mixtures of styrene and other molecules can induce allosteric effects on binding and metabolism by CYP2E1 and thus mitigate the efficiency of their metabolism and corresponding effects on human health. Taken together, our elucidation of mechanisms for these allosteric reactions provides a powerful tool for further investigating the complexities of CYP2E1 metabolism of drugs and pollutants.

Collaboration


Dive into the Grover P. Miller's collaboration.

Top Co-Authors

Avatar

Jessica H. Hartman

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Gunnar Boysen

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Drew R. Jones

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Anna Radominska-Pandya

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Moshe Finel

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar

Anna Mazur

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

S. Joshua Swamidass

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Stacie M. Bratton

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Benkovic

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Tyler B. Hughes

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge