Network


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

Hotspot


Dive into the research topics where Stacie M. Bratton is active.

Publication


Featured researches published by Stacie M. Bratton.


Drug Metabolism and Disposition | 2012

Cytochrome P450-mediated Oxidative Metabolism of Abused Synthetic Cannabinoids Found in "K2/Spice": Identification of Novel Cannabinoid Receptor Ligands

Krishna C. Chimalakonda; Kathryn A. Seely; Stacie M. Bratton; Lisa K. Brents; Cindy L. Moran; Gregory W. Endres; Laura P. James; Paul F. Hollenberg; Paul L. Prather; Anna Radominska-Pandya; Jeffery H. Moran

Abuse of synthetic cannabinoids (SCs), such as [1-naphthalenyl-(1-pentyl-1H-indol-3-yl]-methanone (JWH-018) and [1-(5-fluoropentyl)-1H-indol-3-yl]-1-naphthalenyl-methanone (AM2201), is increasing at an alarming rate. Although very little is known about the metabolism and toxicology of these popular designer drugs, mass spectrometric analysis of human urine specimens after JWH-018 and AM2201 exposure identified monohydroxylated and carboxylated derivatives as major metabolites. The present study extends these initial findings by testing the hypothesis that JWH-018 and its fluorinated counterpart AM2201 are subject to cytochrome P450 (P450)-mediated oxidation, forming potent hydroxylated metabolites that retain significant affinity and activity at the cannabinoid 1 (CB1) receptor. Kinetic analysis using human liver microsomes and recombinant human protein identified CYP2C9 and CYP1A2 as major P450s involved in the oxidation of the JWH-018 and AM2201. In vitro metabolite formation mirrored human urinary metabolic profiles, and each of the primary enzymes exhibited high affinity (Km = 0.81–7.3 μM) and low to high reaction velocities (Vmax = 0.0053–2.7 nmol of product · min−1 · nmol protein−1). The contribution of CYP2C19, 2D6, 2E1, and 3A4 in the hepatic metabolic clearance of these synthetic cannabinoids was minimal (fm = <0.2). In vitro studies demonstrated that the primary metabolites produced in humans display high affinity and intrinsic activity at the CB1 receptor, which was attenuated by the CB1 receptor antagonist (6aR,10aR)-3-(1-methanesulfonylamino-4-hexyn-6-yl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran (O-2050). Results from the present study provide critical, missing data related to potential toxicological properties of “K2” parent compounds and their human metabolites, including mechanism(s) of action at cannabinoid receptors.


Drug Metabolism and Disposition | 2011

Conjugation of Synthetic Cannabinoids JWH-018 and JWH-073, Metabolites by Human UDP-Glucuronosyltransferases

Krishna C. Chimalakonda; Stacie M. Bratton; Vi-Huyen Le; Kan Hui Nicole Yiew; Anna Dineva; Cindy L. Moran; Laura P. James; Jeffery H. Moran; Anna Radominska-Pandya

K2, a synthetic cannabinoid (SC), is an emerging drug of abuse touted as “legal marijuana” and marketed to young teens and first-time drug users. Symptoms associated with K2 use include extreme agitation, syncope, tachycardia, and visual and auditory hallucinations. One major challenge to clinicians is the lack of clinical, pharmacological, and metabolic information for the detection and characterization of K2 and its metabolites in human samples. Information on the metabolic pathway of SCs is very limited. However, previous reports have shown the metabolites of these compounds are excreted primarily as glucuronic acid conjugates. Based on this information, this study evaluates nine human recombinant uridine diphosphate-glucuronosyltransferase (UGT) isoforms and human liver and intestinal microsomes for their ability to glucuronidate hydroxylated metabolites of 1-naphthalenyl-1(1-pentyl-1H-indol-3-yl)-methanone (JWH-018) and (1-butyl-1H-indol-3-yl)-1-naphthalenyl-methanone (JWH-073), the two most common SCs found in K2 products. Conjugates were identified and characterized using liquid chromatography/tandem mass spectrometry, whereas kinetic parameters were quantified using high-performance liquid chromatography-UV-visible methods. UGT1A1, UGT1A3, UGT1A9, UGT1A10, and UGT2B7 were shown to be the major enzymes involved, showing relatively high affinity with Km ranging from 12 to 18 μM for some hydroxylated K2s. These UGTs also exhibited a high metabolic capacity for these compounds, which indicates that K2 metabolites may be rapidly glucuronidated and eliminated from the body. Studies of K2 metabolites will help future development and validation of a specific assay for K2 and its metabolites and will allow researchers to fully explore their pharmacological actions.


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.


Drug Metabolism Reviews | 2010

The crystal structure of human UDP-glucuronosyltransferase 2B7 C-terminal end is the first mammalian UGT target to be revealed: the significance for human UGTs from both the 1A and 2B families.

Anna Radominska-Pandya; Stacie M. Bratton; Matthew R. Redinbo; Michael J. Miley

Human UDP-glucuronosyltransferases (EC 2.4.1.17) (UGTs) are major phase II metabolism enzymes that detoxify a multitude of endo- and xenobiotics through the covalent addition of a glucuronic acid moiety. UGTs are promiscuous enzymes that regulate the levels of numerous important endobiotics in a range of tissues, and inactivate most therapeutic compounds in concert with phase I enzymes. In spite of the importance of these enzymes, we have only a limited understanding of the molecular mechanisms governing their substrate specificity and catalytic activity. Until recently, no three-dimensional structural information was available for any mammalian UGT. The 1.8-å resolution apo crystal structure of the UDP-glucuronic acid binding domain of human UGT2B7 (2B7CT) is the only structure of a mammalian UGT target determined to date. In this review, we summarize what has been learned about human UGT function from the analysis of this and other related glycosyltransferase (GT) crystal structures.


Current Drug Metabolism | 2005

A Historical Overview of the Heterologous Expression of Mammalian UDP-Glucuronosyltransferase Isoforms Over the Past Twenty Years

Anna Radominska-Pandya; Stacie M. Bratton; Joanna M. Little

UDP-Glucuronosyltransferases (UGTs) are actively involved in detoxification of xenobiotics and endogenous compounds and are a major source of drug inactivation and drug-drug interactions. UGTs are membrane-bound enzymes mostly localized in the endoplasmic reticulum (ER) and inner and outer nuclear membranes. UGT activities are totally dependent on the phospholipid content of the membrane and, as a result, are usually inactive when isolated from the ER in the presence of detergent. Several UGT expression systems have been described by different laboratories. They include expression in mammalian cells such as COS, V79 and HEK293. Also, baculovirus-infected insect cells systems have been developed and allow the expression of UGT isoforms with or without histidine molecule tags (His-tags). Moreover, as for CYP450, UGT isoforms have been expressed in E.coli. This review concentrates on a detailed description of all these expression systems in terms of their use for substrate specificity studies and the preparation of pure UGT proteins for active site identification and other structural studies. The effect of detergents and alamethicin on UGT catalytic activity in different expression systems will be discussed. Moreover, extensive comparative studies on the characterization of recombinant UGTs in terms of substrate specificity, evaluation of kinetic parameters, and the effect of inhibitors will be presented in this review. An overall picture of the use of different UGT expression systems will help in selecting the best one for identification of the individual UGT isoforms involved in the glucuronidation of drugs, environmental pollutants and physiologically important endogenous compounds. Especially important is an expression system where UGTs are biosynthesized with His-tags. UGTs expressed in this system can be easily purified to homogeneity, which will result in significant development of structure-function relationship studies, including the identification of substrate active sites and eventual crystallization. These are underdeveloped areas of UGT research and the availability of these recombinant UGTs will allow these gaps to be filled.


International Journal of Nanomedicine | 2011

Ethylenediamine functionalized-single-walled nanotube (f-SWNT)-assisted in vitro delivery of the oncogene suppressor p53 gene to breast cancer MCF-7 cells

Alokita Karmakar; Stacie M. Bratton; Enkeleda Dervishi; Anindya Ghosh; Meena Mahmood; Yang Xu; Lamya Saeed; Thikra Mustafa; Dan Casciano; Anna Radominska-Pandya; Alexandru S. Biris

A gene delivery concept based on ethylenediamine-functionalized single-walled carbon nanotubes (f-SWCNTs) using the oncogene suppressor p53 gene as a model gene was successfully tested in vitro in MCF-7 breast cancer cells. The f-SWCNTs-p53 complexes were introduced into the cell medium at a concentration of 20 μg mL−1 and cells were exposed for 24, 48, and 72 hours. Standard ethidium bromide and acridine orange assays were used to detect apoptotic cells and indicated that a significantly larger percentage of the cells (approx 40%) were dead after 72 hours of exposure to f-SWCNTs-p53 as compared to the control cells, which were exposed to only p53 or f-SWCNTs, respectively. To further support the uptake and expression of the genes within the cells, green fluorescent protein-tagged p53, attached to the f-SWCNTs was added to the medium and the complex was observed to be strongly expressed in the cells. Moreover, caspase 3 activity was found to be highly enhanced in cells incubated with the f-SWCNTs-p53 complex, indicating strongly induced apoptosis. This system could be the foundation for novel gene delivery platforms based on the unique structural and morphological properties of multi-functional nanomaterials.


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.


Steroids | 2007

Phenylalanine90 and phenylalanine93 are crucial amino acids within the estrogen binding site of the human UDP-glucuronosyltransferase 1A10

Athena Starlard-Davenport; Yan Xiong; Stacie M. Bratton; Anna Gallus-Zawada; Moshe Finel; Anna Radominska-Pandya

Human UDP-glucuronosyltransferase 1A10 has been identified as the major isoform involved in the biotransformation of a wide range of phenolic substrates, including native estrogens and their oxidized metabolites. Our recent studies point to the F(90)-M(91)-V(92)-F(93) amino acid motif of UGT1A10, which was identified using photoaffinity labeling followed by LC-MS/MS analysis, as a key determinant of the binding of phenolic substrates. In this report, we have evaluated the role of F(90), V(92), and F(93) in the recognition of estrogens by UGT1A10 using site-directed mutagenesis. Kinetic studies using five mutants revealed that F(90) and F(93) are critical residues for the recognition of all estrogen substrates. The substitution of F(90) with alanine totally abolished the activity of this enzyme toward all the estrogens investigated. Overall, sequential removal for the aromatic ring (F to L) and of the hydrophobic chain (F to A and V to A) from amino acids 90, 92, and 93 effectively alters estrogen recognition. This demonstrates that individual features of the native and hydroxylated estrogens determine the specific binding properties of the compound within the binding site of the human UGT1A10 and the mutants. The resulting activities are completely abolished, unchanged, increased, or decreased depending on the structures of both the mutant and the substrate. The novel identification of UGT1A10 as the major isoform involved in the glucuronidation of all estrogens and the discovery of the importance of the FMVF motif in the binding of steroids will help to elucidate the molecular mechanism of glucuronidation, resulting in the design of more effective estrogen-based therapies.


Journal of Applied Toxicology | 2014

Single-walled carbon nanotube and graphene nanodelivery of gambogic acid increases its cytotoxicity in breast and pancreatic cancer cells.

Lamya Saeed; Meena Mahmood; Sebastian J. Pyrek; Tariq Fahmi; Yang Xu; Thikra Mustafa; Zeid A. Nima; Stacie M. Bratton; Dan Casciano; Enkeleda Dervishi; Anna Radominska-Pandya; Alexandru S. Biris

Graphene and single‐walled carbon nanotubes were used to deliver the natural low‐toxicity drug gambogic acid (GA) to breast and pancreatic cancer cells in vitro, and the effectiveness of this complex in suppressing cellular integrity was assessed. Cytotoxicity was assessed by measuring lactate dehydrogenase release, mitochondria dehydrogenase activity, mitochondrial membrane depolarization, DNA fragmentation, intracellular lipid content, and membrane permeability/caspase activity. The nanomaterials showed no toxicity at the concentrations used, and the antiproliferative effects of GA were significantly enhanced by nanodelivery. The results suggest that these complexes inhibit human breast and pancreatic cancer cells grown in vitro. This analysis represents a first step toward assessing their effectiveness in more complex, targeted, nanodelivery systems. Copyright


Xenobiotica | 2012

Natural prenylated resveratrol analogs arachidin-1 and -3 demonstrate improved glucuronidation profiles and have affinity for cannabinoid receptors

Lisa K. Brents; Fabricio Medina-Bolivar; Kathryn A. Seely; Vipin Nair; Stacie M. Bratton; Luis Nopo-Olazabal; Ronak Y. Patel; Haining Liu; Robert J. Doerksen; Paul L. Prather; Anna Radominska-Pandya

Rationale. The therapeutic promise of trans-resveratrol (tRes) is limited by poor bioavailability following rapid metabolism. We hypothesise that trans-arachidin-1 (tA1) and trans-arachidin-3 (tA3), peanut hairy root-derived isoprenylated analogs of tRes, will exhibit slower metabolism/enhanced bioavailability and retain biological activity via cannabinoid receptor (CBR) binding relative to their non-prenylated parent compounds trans-piceatannol (tPice) and tRes, respectively. Results. The activities of eight human UDP-glucuronosyltransferases (UGTs) toward these compounds were evaluated. The greatest activity was observed for extrahepatic UGTs 1A10 and 1A7, followed by hepatic UGTs 1A1 and 1A9. Importantly, an additional isoprenyl and/or hydroxyl group in tA1 and tA3 slowed overall glucuronidation. CBR binding studies demonstrated that all analogs bound to CB1Rs with similar affinities (5–18 µM); however, only tA1 and tA3 bound appreciably to CB2Rs. Molecular modelling studies confirmed that the isoprenyl moiety of tA1 and tA3 improved binding affinity to CB2Rs. Finally, although tA3 acted as a competitive CB1R antagonist, tA1 antagonised CB1R agonists by both competitive and non-competitive mechanisms. Conclusions. Prenylated stilbenoids may be preferable alternatives to tRes due to increased bioavailability via slowed metabolism. Similar structural analogs might be developed as novel CB therapeutics for obesity and/or drug dependency.

Collaboration


Dive into the Stacie M. Bratton's collaboration.

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
Top Co-Authors

Avatar

Anna Gallus-Zawada

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Agnieszka K. Zielinska

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Grover P. Miller

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Alexandru S. Biris

University of Arkansas at Little Rock

View shared research outputs
Top Co-Authors

Avatar

Anna Mazur

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Cheryl F. Lichti

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Aleksandra K. Greer

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge