Network


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

Hotspot


Dive into the research topics where Kay Ahn is active.

Publication


Featured researches published by Kay Ahn.


Journal of Biological Chemistry | 2010

SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1

Michelle Pacholec; John E. Bleasdale; Boris A. Chrunyk; David Cunningham; Declan Flynn; Robert S. Garofalo; David A. Griffith; Matt Griffor; Pat Loulakis; Brandon Pabst; Xiayang Qiu; Brian J. Stockman; Venkataraman Thanabal; Alison H. Varghese; Jessica Ward; Jane M. Withka; Kay Ahn

Sirtuins catalyze NAD+-dependent protein deacetylation and are critical regulators of transcription, apoptosis, metabolism, and aging. There are seven human sirtuins (SIRT1–7), and SIRT1 has been implicated as a key mediator of the pathways downstream of calorie restriction that have been shown to delay the onset and reduce the incidence of age-related diseases such as type 2 diabetes. Increasing SIRT1 activity, either by transgenic overexpression of the Sirt1 gene in mice or by pharmacological activation by small molecule activators resveratrol and SRT1720, has shown beneficial effects in rodent models of type 2 diabetes, indicating that SIRT1 may represent an attractive therapeutic target. Herein, we have assessed purported SIRT1 activators by employing biochemical assays utilizing native substrates, including a p53-derived peptide substrate lacking a fluorophore as well as the purified native full-length protein substrates p53 and acetyl-CoA synthetase1. SRT1720, its structurally related compounds SRT2183 and SRT1460, and resveratrol do not lead to apparent activation of SIRT1 with native peptide or full-length protein substrates, whereas they do activate SIRT1 with peptide substrate containing a covalently attached fluorophore. Employing NMR, surface plasmon resonance, and isothermal calorimetry techniques, we provide evidence that these compounds directly interact with fluorophore-containing peptide substrates. Furthermore, we demonstrate that SRT1720 neither lowers plasma glucose nor improves mitochondrial capacity in mice fed a high fat diet. SRT1720, SRT2183, SRT1460, and resveratrol exhibit multiple off-target activities against receptors, enzymes, transporters, and ion channels. Taken together, we conclude that SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1.


Chemical Reviews | 2008

Enzymatic Pathways That Regulate Endocannabinoid Signaling in the Nervous System

Kay Ahn; Michele K. McKinney; Benjamin F. Cravatt

Chemical signals, or neurotransmitters, represent the fundamental mode for intercellular communication in the nervous system. (1) The classical model for neurotransmitter action involves the uptake and storage of these small molecules into synaptic vesicles, release of vesicular contents into the synaptic cleft in response to depolarization of the presynaptic terminal by an action potential, binding of released neurotransmitters to cognate protein receptors on the postsynaptic (and presynaptic) terminal, and, finally, termination of signaling by protein-mediated uptake and degradation of neurotransmitters from the synaptic cleft. This model applies to a large number of well-studied neurotransmitters, including glutamate, γ-amino butyric acid (GABA), acetylcholine, and the monoamines, all of which represent aqueous solution-soluble molecules. More recently, lipids have emerged as an important class of chemical messengers in the nervous system that operate by a distinct mechanism. The hydrophobic nature of lipids precludes their stable uptake and storage into synaptic vesicles. Instead, lipid messengers appear to be biosynthesized and released by neurons at the moment of their intended action, which is often referred to as “on-demand” production. Similarly, the capacity of lipids to freely cross cell membranes places the burden of signal termination largely on the action of degradative enzymes. Lipid signaling systems are thus embedded within an elaborate collection of metabolic pathways, the composition and regulation of which ultimately establish the magnitude and duration of transmitter action. Here, we will review these general concepts as they relate to a specific class of lipid transmitters, the endogenous cannabinoids (endocannabinoids), and highlight how delineation of their cognate metabolic enzymes has been translated into the development of chemical and genetic tools to test the role that the endocannabinoid system plays in nervous system signaling and behavior. Endocannabinoids are defined as endogenous small molecules that activate the cannabinoid receptors CB1 and CB2, which are G-protein-coupled receptors that also recognize Δ9-tetrahydrocannabinol (THC), the psychoactive component of marijuana. (2, 3) The CB1 receptor is the major cannabinoid receptor in the nervous system and is responsible for mediating most of the neurobehavioral effects of THC. (4, 5) The CB2 receptor is predominantly expressed in immune cells, (6) where it appears to play a role in mediating the immunosuppressive effects of cannabinoids. Two principal endocannabinoids have been identified in mammals, N-arachidonoyl ethanolamine (anandamide) (7) and 2-arachidonoylglycerol (2-AG) (8, 9) (Figure 1). Each endocannabinoid also belongs to a much larger class of lipids, termed N-acyl ethanolamines (NAEs) and monoacylglycerols (MAGs), respectively, where individual members differ in the length and degree of unsaturation of their acyl chains (Figure 1). Several NAEs and MAGs have been ascribed potential biological activities in vivo; (10) however, most of these lipids do not serve as ligands for cannabinoid receptors, a property that appears to be restricted to polyunsaturated derivatives such as anandamide and 2-AG. Figure 1 Two principle endocannabinoids, N-arachidonoyl ethanolamine (anandamide) and 2-arachidonoylglycerol (2-AG), which are members of theN-acyl ethanolamine (NAE) and monoacylglycerol (MAG) classes of lipids, respectively. In the nervous system, endocannabinoids are hypothesized to act as retrograde messengers, being released by postsynaptic neurons and traversing the synaptic cleft to stimulate CB1 receptors on presynaptic termini (11, 12) (Figure 2). This model is supported by a large number of in vitro electrophysiological studies, (12) as well as by the restricted localization of the CB1 receptor to presynaptic structures in many regions of the nervous system. (13, 14) Once activated by endocannabinoids, CB1 receptors couple principally through the G i/G o class of G proteins to regulate calcium and potassium channels and reduce the probability of neurotransmitter release. (3) This suppression of neurotransmitter release can result in the inhibition or, paradoxically, disinhibition of neuronal circuits, depending on whether the CB1 receptor is expressed on glutamatergic or GABergic neurons. Figure 2 General model for endocannabinoid-based retrograde signaling. Upon release of neurotransmitter (e.g., glutamate), postsynaptic receptors (e.g., α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl- d-aspartic acid (NMDA)) and ... Despite sharing a common receptor and considerable structural similarity, anandamide and 2-AG can be distinguished by multiple factors. First, these endocannabinoids activate cannabinoid receptors to a differing degree in vitro, with anandamide displaying lower intrinsic efficacy than 2-AG, which results in the former lipid acting as a partial agonist (15) (it should be specified that the relevance of this distinction for signaling in vivo is unclear, especially when one considers that THC also acts as only a partial agonist for cannabinoid receptors in vitro (16)). Second, the endogenous quantities of anandamide and 2-AG differ dramatically, with the latter lipid being found at more than 100-fold higher concentrations in the nervous system. (16) Of course, these values are based on bulk tissue measurements of endocannabinoids, which almost certainly reflect a combination of metabolic and signaling pools of these lipids. Indeed, recent microdialysis studies have revealed that the extracellular concentrations of anandamide and 2-AG are nearly equivalent (within 2 5-fold), (17, 18) suggesting that, at least for the latter endocannabinoid, a large fraction of bulk tissue concentration may correspond to intracellular metabolic pools. Finally, and of greatest relevance for the subject of this review, anandamide and 2-AG are regulated by distinct biosynthetic and degradative pathways. Over the past decade, several excellent reviews have appeared that discuss endocannabinoid metabolism and signaling. (10, 19-23) Here, we will focus on the most recent advances in our understanding of the composition and regulation of endocannabinoid metabolic pathways, especially as pertains to the nervous system. A pervasive theme throughout this review will be the importance of developing selective genetic and pharmacological tools to specifically perturb individual enzymatic pathways to test their contribution to endocannabinoid metabolism, nervous system function, and, ultimately, mammalian physiology and behavior.


Chemistry & Biology | 2009

Discovery and Characterization of a Highly Selective FAAH Inhibitor that Reduces Inflammatory Pain

Kay Ahn; Douglas S. Johnson; Mauro Mileni; David Beidler; Jonathan Z. Long; Michele K. McKinney; Eranthie Weerapana; Nalini Sadagopan; Marya Liimatta; Sarah E. Smith; Scott E. Lazerwith; Cory Michael Stiff; Satwik Kamtekar; Keshab Bhattacharya; Yanhua Zhang; Stephen Swaney; Keri Van Becelaere; Raymond C. Stevens; Benjamin F. Cravatt

Endocannabinoids are lipid signaling molecules that regulate a wide range of mammalian behaviors, including pain, inflammation, and cognitive/emotional state. The endocannabinoid anandamide is principally degraded by the integral membrane enzyme fatty acid amide hydrolase (FAAH), and there is currently much interest in developing FAAH inhibitors to augment endocannabinoid signaling in vivo. Here, we report the discovery and detailed characterization of a highly efficacious and selective FAAH inhibitor, PF-3845. Mechanistic and structural studies confirm that PF-3845 is a covalent inhibitor that carbamylates FAAHs serine nucleophile. PF-3845 selectively inhibits FAAH in vivo, as determined by activity-based protein profiling; raises brain anandamide levels for up to 24 hr; and produces significant cannabinoid receptor-dependent reductions in inflammatory pain. These data thus designate PF-3845 as a valuable pharmacological tool for in vivo characterization of the endocannabinoid system.


Journal of Pharmacology and Experimental Therapeutics | 2011

Mechanistic and Pharmacological Characterization of PF-04457845: A Highly Potent and Selective Fatty Acid Amide Hydrolase Inhibitor That Reduces Inflammatory and Noninflammatory Pain

Kay Ahn; Sarah E. Smith; Marya Liimatta; David Beidler; Nalini Sadagopan; David T. Dudley; Tim Young; Paul Wren; Yanhua Zhang; Steven Swaney; Keri Van Becelaere; Jacqueline L. Blankman; Daniel K. Nomura; Shoba N. Bhattachar; Cory Michael Stiff; Tyzoon K. Nomanbhoy; Eranthie Weerapana; Douglas S. Johnson; Benjamin F. Cravatt

The endogenous cannabinoid (endocannabinoid) anandamide is principally degraded by the integral membrane enzyme fatty acid amide hydrolase (FAAH). Pharmacological blockade of FAAH has emerged as a potentially attractive strategy for augmenting endocannabinoid signaling and retaining the beneficial effects of cannabinoid receptor activation, while avoiding the undesirable side effects, such as weight gain and impairments in cognition and motor control, observed with direct cannabinoid receptor 1 agonists. Here, we report the detailed mechanistic and pharmacological characterization of N-pyridazin-3-yl-4-(3-{[5-(trifluoromethyl)pyridin-2-yl]oxy}benzylidene)piperidine-1-carboxamide (PF-04457845), a highly efficacious and selective FAAH inhibitor. Mechanistic studies confirm that PF-04457845 is a time-dependent, covalent FAAH inhibitor that carbamylates FAAHs catalytic serine nucleophile. PF-04457845 inhibits human FAAH with high potency (kinact/Ki = 40,300 M−1s−1; IC50 = 7.2 nM) and is exquisitely selective in vivo as determined by activity-based protein profiling. Oral administration of PF-04457845 produced potent antinociceptive effects in both inflammatory [complete Freunds adjuvant (CFA)] and noninflammatory (monosodium iodoacetate) pain models in rats, with a minimum effective dose of 0.1 mg/kg (CFA model). PF-04457845 displayed a long duration of action as a single oral administration at 1 mg/kg showed in vivo efficacy for 24 h with a concomitant near-complete inhibition of FAAH activity and maximal sustained elevation of anandamide in brain. Significantly, PF-04457845-treated mice at 10 mg/kg elicited no effect in motility, catalepsy, and body temperature. Based on its exceptional selectivity and in vivo efficacy, combined with long duration of action and optimal pharmacokinetic properties, PF-04457845 is a clinical candidate for the treatment of pain and other nervous system disorders.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Structure-guided inhibitor design for human FAAH by interspecies active site conversion

Mauro Mileni; Douglas S. Johnson; Zhigang Wang; Daniel S. Everdeen; Marya Liimatta; Brandon Pabst; Keshab Bhattacharya; Richard A. Nugent; Satwik Kamtekar; Benjamin F. Cravatt; Kay Ahn; Raymond C. Stevens

The integral membrane enzyme fatty acid amide hydrolase (FAAH) hydrolyzes the endocannabinoid anandamide and related amidated signaling lipids. Genetic or pharmacological inactivation of FAAH produces analgesic, anxiolytic, and antiinflammatory phenotypes but not the undesirable side effects of direct cannabinoid receptor agonists, indicating that FAAH may be a promising therapeutic target. Structure-based inhibitor design has, however, been hampered by difficulties in expressing the human FAAH enzyme. Here, we address this problem by interconverting the active sites of rat and human FAAH using site-directed mutagenesis. The resulting humanized rat (h/r) FAAH protein exhibits the inhibitor sensitivity profiles of human FAAH but maintains the high-expression yield of the rat enzyme. We report a 2.75-Å crystal structure of h/rFAAH complexed with an inhibitor, N-phenyl-4-(quinolin-3-ylmethyl)piperidine-1-carboxamide (PF-750), that shows strong preference for human FAAH. This structure offers compelling insights to explain the species selectivity of FAAH inhibitors, which should guide future drug design programs.


Expert Opinion on Drug Discovery | 2009

Fatty acid amide hydrolase as a potential therapeutic target for the treatment of pain and CNS disorders.

Kay Ahn; Douglas S. Johnson; Benjamin F. Cravatt

Background: Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme that hydrolyzes the endocannabinoid anandamide and related amidated signaling lipids. Genetic or pharmacological inactivation of FAAH produces analgesic, anti-inflammatory, anxiolytic and antidepressant phenotypes without showing the undesirable side effects of direct cannabinoid receptor agonists, indicating that FAAH may be a promising therapeutic target. Objectives: This review highlights advances in the development of FAAH inhibitors of different mechanistic classes and their in vivo efficacy. Also highlighted are advances in technology for the in vitro and in vivo selectivity assessment of FAAH inhibitors using activity-based protein profiling and click chemistry-activity-based protein profiling, respectively. Recent reports on structure-based drug design for human FAAH generated by protein engineering using interspecies active site conversion are also discussed. Methods: The literature searches of Medline and SciFinder databases were used. Conclusions: There has been tremendous progress in our understanding of FAAH and development of FAAH inhibitors with in vivo efficacy, selectivity and drug-like pharmacokinetic properties.


Journal of Lipid Research | 2012

Evidence for regulated monoacylglycerol acyltransferase expression and activity in human liver

Angela M. Hall; Kou Kou; Zhouji Chen; Terri Pietka; Mrudula Kumar; Kevin M. Korenblat; Kyuha Lee; Kay Ahn; Elisa Fabbrini; Samuel Klein; Bryan Goodwin; Brian N. Finck

Intrahepatic lipid accumulation is extremely common in obese subjects and is associated with the development of insulin resistance and diabetes. Hepatic diacylglycerol and triacylglycerol synthesis predominantly occurs through acylation of glycerol-3-phosphate. However, an alternative pathway for synthesizing diacylglycerol from monoacylglycerol acyltransferases (MGAT) could also contribute to hepatic glyceride pools. MGAT activity and the expression of the three genes encoding MGAT enzymes (MOGAT1, MOGAT2, and MOGAT3) were determined in liver biopsies from obese human subjects before and after gastric bypass surgery. MOGAT expression was also assessed in liver of subjects with nonalcoholic fatty liver disease (NAFLD) or control livers. All MOGAT genes were expressed in liver, and hepatic MGAT activity was readily detectable in liver lysates. The hepatic expression of MOGAT3 was highly correlated with MGAT activity, whereas MOGAT1 and MOGAT2 expression was not, and knockdown of MOGAT3 expression attenuated MGAT activity in a liver-derived cell line. Marked weight loss following gastric bypass surgery was associated with a significant reduction in MOGAT2 and MOGAT3 expression, which were also overexpressed in NAFLD subjects. These data suggest that the MGAT pathway is active and dynamically regulated in human liver and could be an important target for pharmacologic intervention for the treatment of obesity-related insulin resistance and NAFLD.


Bioorganic & Medicinal Chemistry Letters | 2009

Benzothiophene piperazine and piperidine urea inhibitors of fatty acid amide hydrolase (FAAH)

Douglas S. Johnson; Kay Ahn; Suzanne Ross Kesten; Scott E. Lazerwith; Yuntao Song; Mark Morris; Lorraine Kathleen Fay; Tracy Fay Gregory; Cory Michael Stiff; James B. Dunbar; Marya Liimatta; David Beidler; Sarah E. Smith; Tyzoon K. Nomanbhoy; Benjamin F. Cravatt

The synthesis and structure-activity relationships (SAR) of a series of benzothiophene piperazine and piperidine urea FAAH inhibitors is described. These compounds inhibit FAAH by covalently modifying the enzymes active site serine nucleophile. Activity-based protein profiling (ABPP) revealed that these urea inhibitors were completely selective for FAAH relative to other mammalian serine hydrolases. Several compounds showed in vivo activity in a rat complete Freunds adjuvant (CFA) model of inflammatory pain.


Journal of Pharmacology and Experimental Therapeutics | 2015

PF-1355, a Mechanism-Based Myeloperoxidase Inhibitor, Prevents Immune Complex Vasculitis and Anti–Glomerular Basement Membrane Glomerulonephritis

Wei Zheng; Roscoe L. Warner; Roger Benjamin Ruggeri; Chunyan Su; Christian Cortes; Athanasia Skoura; Jessica Ward; Kay Ahn; Amit S. Kalgutkar; Dexue Sun; Tristan S. Maurer; Paul D. Bonin; Carlin Okerberg; Walter F. Bobrowski; Thomas T. Kawabe; Yanwei Zhang; Timothy M. Coskran; Sammy Bell; Bhupesh Kapoor; Kent J. Johnson; Leonard Buckbinder

Small vessel vasculitis is a life-threatening condition and patients typically present with renal and pulmonary injury. Disease pathogenesis is associated with neutrophil accumulation, activation, and oxidative damage, the latter being driven in large part by myeloperoxidase (MPO), which generates hypochlorous acid among other oxidants. MPO has been associated with vasculitis, disseminated vascular inflammation typically involving pulmonary and renal microvasculature and often resulting in critical consequences. MPO contributes to vascular injury by 1) catabolizing nitric oxide, impairing vasomotor function; 2) causing oxidative damage to lipoproteins and endothelial cells, leading to atherosclerosis; and 3) stimulating formation of neutrophil extracellular traps, resulting in vessel occlusion and thrombosis. Here we report a selective 2-thiouracil mechanism-based MPO inhibitor (PF-1355 [2-(6-(2,5-dimethoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide) and demonstrate that MPO is a critical mediator of vasculitis in mouse disease models. A pharmacokinetic/pharmacodynamic response model of PF-1355 exposure in relation with MPO activity was derived from mouse peritonitis. The contribution of MPO activity to vasculitis was then examined in an immune complex model of pulmonary disease. Oral administration of PF-1355 reduced plasma MPO activity, vascular edema, neutrophil recruitment, and elevated circulating cytokines. In a model of anti–glomerular basement membrane disease, formerly known as Goodpasture disease, albuminuria and chronic renal dysfunction were completely suppressed by PF-1355 treatment. This study shows that MPO activity is critical in driving immune complex vasculitis and provides confidence in testing the hypothesis that MPO inhibition will provide benefit in treating human vasculitic diseases.


Nuclear Medicine and Biology | 2012

The synthesis and in vivo evaluation of [18F]PF-9811: a novel PET ligand for imaging brain fatty acid amide hydrolase (FAAH).

Marc B. Skaddan; Lei Zhang; Douglas S. Johnson; Aijun Zhu; Kenneth Zasadny; Richard V. Coelho; Kyle Kuszpit; Gwen Currier; Kuo-Hsien Fan; Elizabeth Mary Beck; Laigao Chen; Susan E. Drozda; Gayatri Balan; Micah J. Niphakis; Benjamin F. Cravatt; Kay Ahn; Thomas Bocan; Anabella Villalobos

INTRODUCTION Fatty acid amide hydrolase (FAAH) is responsible for the enzymatic degradation of the fatty acid amide family of signaling lipids, including the endogenous cannabinoid (endocannabinoid) anandamide. The involvement of the endocannabinoid system in pain and other nervous system disorders has made FAAH an attractive target for drug development. Companion molecular imaging probes are needed, however, to assess FAAH inhibition in the nervous system in vivo. We report here the synthesis and in vivo evaluation of [(18)F]PF-9811, a novel PET ligand for non-invasive imaging of FAAH in the brain. METHODS The potency and selectivity of unlabeled PF-9811 were determined by activity-based protein profiling (ABPP) both in vitro and in vivo. [(18)F]PF-9811 was synthesized in a 3-step, one-pot reaction sequence, followed by HPLC purification. Biological evaluation was performed by biodistribution and dynamic PET imaging studies in male rats. The specificity of [(18)F]PF-9811 uptake was evaluated by pre-administration of PF-04457845, a potent and selective FAAH inhibitor, 1h prior to radiotracer injection. RESULTS Biodistribution studies show good uptake (SUV~0.8 at 90 min) of [(18)F]PF-9811 in rat brain, with significant reduction of the radiotracer in all brain regions (37%-73% at 90 min) in blocking experiments. Dynamic PET imaging experiments in rat confirmed the heterogeneous uptake of [(18)F]PF-9811 in brain regions with high FAAH enzymatic activity, as well as statistically significant reductions in signal following pre-administration of the blocking compound PF-04457845. CONCLUSIONS [(18)F]PF-9811 is a promising PET imaging agent for FAAH. Biodistribution and PET imaging experiments show that the tracer has good uptake in brain, regional heterogeneity, and specific binding as determined by blocking experiments with the highly potent and selective FAAH inhibitor, PF-04457845.

Collaboration


Dive into the Kay Ahn's collaboration.

Researchain Logo
Decentralizing Knowledge