Network


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

Hotspot


Dive into the research topics where Jenny L. Wiley is active.

Publication


Featured researches published by Jenny L. Wiley.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo

Jonathan Z. Long; Daniel K. Nomura; Robert E. Vann; D. Matthew Walentiny; Lamont Booker; Xin Jin; James J. Burston; Laura J. Sim-Selley; Aron H. Lichtman; Jenny L. Wiley; Benjamin F. Cravatt

Δ9-Tetrahydrocannabinol (THC), the psychoactive component of marijuana, and other direct cannabinoid receptor (CB1) agonists produce a number of neurobehavioral effects in mammals that range from the beneficial (analgesia) to the untoward (abuse potential). Why, however, this full spectrum of activities is not observed upon pharmacological inhibition or genetic deletion of either fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), enzymes that regulate the two major endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG), respectively, has remained unclear. Here, we describe a selective and efficacious dual FAAH/MAGL inhibitor, JZL195, and show that this agent exhibits broad activity in the tetrad test for CB1 agonism, causing analgesia, hypomotilty, and catalepsy. Comparison of JZL195 to specific FAAH and MAGL inhibitors identified behavioral processes that were regulated by a single endocannabinoid pathway (e.g., hypomotility by the 2-AG/MAGL pathway) and, interestingly, those where disruption of both FAAH and MAGL produced additive effects that were reversed by a CB1 antagonist. Falling into this latter category was drug discrimination behavior, where dual FAAH/MAGL blockade, but not disruption of either FAAH or MAGL alone, produced THC-like responses that were reversed by a CB1 antagonist. These data indicate that AEA and 2-AG signaling pathways interact to regulate specific behavioral processes in vivo, including those relevant to drug abuse, thus providing a potential mechanistic basis for the distinct pharmacological profiles of direct CB1 agonists and inhibitors of individual endocannabinoid degradative enzymes.


European Journal of Pharmacology | 2001

Assessment of the role of CB1 receptors in cannabinoid anticonvulsant effects

Melisa J. Wallace; Jenny L. Wiley; Billy R. Martin; Robert J. DeLorenzo

The cannabinoid CB1 receptor has been shown to be the primary site of action for cannabinoid-induced effects on the central nervous system. Activation of this receptor has proven to dampen neurotransmission and produce an overall reduction in neuronal excitability. Cannabinoid compounds like delta9-tetrahydrocannabinol and cannabidiol have been shown to be anticonvulsant in maximal electroshock, a model of partial seizure with secondary generalization. However, until now, it was unknown if these anticonvulsant effects are mediated by the cannabinoid CB1 receptor. Likewise, (R)-(+)-[2,3-Dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone (WIN 55,212-2), a cannabimimetic compound that has been shown to decrease hyperexcitability in cell culture models via the cannabinoid CB1 receptor, has never been evaluated for anticonvulsant activity in an animal seizure model. We first show that the cannabinoid compounds delta9-tetrahydrocannabinol (ED50 = 42 mg/kg), cannabidiol (ED50 = 80 mg/kg), and WIN 55,212-2 (ED50 = 47 mg/kg) are anticonvulsant in maximal electroshock. We further establish, using the cannabinoid CB1 receptor specific antagonist N-(piperidin-1-yl-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride (SR141716A) (AD50 = 2.5 mg/kg), that the anticonvulsant effects of delta9-tetrahydrocannabinol and WIN 55,212-2 are cannabinoid CB1 receptor-mediated while the anticonvulsant activity of cannabidiol is not. This study establishes a role for the cannabinoid CB1 receptor in modulating seizure activity in a whole animal model.


British Journal of Pharmacology | 2005

CB1 cannabinoid receptor-mediated modulation of food intake in mice

Jenny L. Wiley; James J. Burston; Darnica C Leggett; Olga O. Alekseeva; Raj K. Razdan; Anu Mahadevan; Billy R. Martin

1 Marijuanas appetite‐increasing effects have long been known. Recent research suggests that the CB1 cannabinoid receptor antagonist SR141716A may suppress appetite. This study represents a further, systematic investigation of the role of CB1 cannabinoid receptors in the pharmacological effects of cannabinoids on food intake. 2 Mice were food‐restricted for 24 h and then allowed access to their regular rodent chow for 1 h. Whereas the CB1 antagonist SR141716A dose‐dependently decreased food consumption at doses that did not affect motor activity, Δ9‐tetrahydrocannabinol (Δ9‐THC) increased food consumption at doses that had no effect on motor activity. O‐3259 and O‐3257, structural analogs of SR141716A, produced effects similar to those of the parent compound. 3 Amphetamine (a known anorectic) and diazepam (a benzodiazepine and CNS depressant) decreased food consumption, but only at doses that also increased or decreased motor activity, respectively. The CB2 cannabinoid receptor antagonist SR144528 and the nonpsychoactive cannabinoid cannabidiol did not affect food intake nor activity. 4 SR141716A decreased feeding in wild‐type mice, but lacked pharmacological activity in CB1 knockout mice; however, basal food intake was lower in CB1 knockout mice. Amphetamine decreased feeding in both mouse genotypes. 5 These results suggest that SR141716A may affect the actions of endogenous cannabinoids in regulating appetite or that it may have effects of its own aside from antagonism of cannabinoid effects (e.g., decreased feeding behavior and locomotor stimulation). In either case, these results strongly suggest that CB1 receptors may play a role in regulation of feeding behavior.


Bioorganic & Medicinal Chemistry | 2003

3-Indolyl-1-naphthylmethanes: new cannabimimetic indoles provide evidence for aromatic stacking interactions with the CB1 cannabinoid receptor

John W. Huffman; Ross Mabon; Ming-Jung Wu; Jianzhong Lu; Richard E. Hart; Dow P. Hurst; Patricia H. Reggio; Jenny L. Wiley; Billy R. Martin

A series of 1-pentyl-1H-indol-3-yl-(1-naphthyl)methanes (9-11) and 2-methyl-1-pentyl-1H-indol-3-yl-(1-naphthyl)methanes (12-14) have been synthesized to investigate the hypothesis that cannabimimetic 3-(1-naphthoyl)indoles interact with the CB(1) receptor by hydrogen bonding to the carbonyl group. Indoles 9-11 have significant (K(i)=17-23nM) receptor affinity, somewhat less than that of the corresponding naphthoylindoles (5, 15, 16). 2-Methyl-1-indoles 12-14 have little affinity for the CB(1) receptor, in contrast to 2-methyl-3-(1-naphthoyl)indoles 17-19, which have affinities comparable to those of 5, 15, 16. A cannabimimetic indene hydrocarbon (26) was synthesized and found to have K(i)=26+/-4nM. Molecular modeling and receptor docking studies of naphthoylindole 16, its 2-methyl congener (19) and indolyl-1-naphthylmethanes 11 and 14, combined with the receptor affinities of these cannabimimetic indoles, strongly suggest that these cannabinoid receptor ligands bind primarily by aromatic stacking interactions in the transmembrane helix 3-4-5-6 region of the CB(1) receptor.


European Journal of Pharmacology | 1995

Effects of site-selective NMDA receptor antagonists in an elevated plus-maze model of anxiety in mice

Jenny L. Wiley; Anthony F. Cristello; Robert L. Balster

NMDA receptor antagonists have been shown to be anxiolytic in animal models of anxiety, although they have not been tested extensively. These compounds bind to several specific sites within the NMDA-receptor complex, including the NMDA site itself, the phencyclidine site, and the strychnine-insensitive glycine site. The purpose of the present study was to examine potential anxiolytic effects of site-selective NMDA receptor antagonists in the elevated plus-maze. Drug-naive albino mice were placed in the center of an elevated maze shaped like a plus sign. Two opposing arms were enclosed by high walls; the crossing arms were open. Following injection with drug or vehicle, the number of entries and time spent in each type of arm were measured during 5-min tests. Analysis of results showed that the benzodiazepine, diazepam, and the competitive NMDA receptor antagonist, NPC 17742 (2R,4R,5S 2-amino-4,5-(1,2-cyclohexyl)-7-phosphono-heptanoic acid), increased number of open arm entries and open arm time. N-Nitro-L-arginine methyl ester, a nitric oxide synthase inhibitor which may interfere with the transduction of NMDA receptor activation, also increased open arm entries and time; however, the magnitude of these increases was small. The phencyclidine-site NMDA receptor antagonist, phencyclidine, increased open arm entries, but failed to significantly increase open arm time. ACEA 1021 (5-nitro-6,7-dichloro-1,4-dihydro-2,3-quinoxalinedione), a putative glycine-site antagonist, had significant effects only on open arm entries at the highest dose tested. These results suggest that NMDA receptor antagonists show promise as potential anxiolytic agents, but that differences among antagonists acting at different cellular sites may be expected.


European Journal of Pharmacology | 2003

Cannabinoid pharmacological properties common to other centrally acting drugs.

Jenny L. Wiley; Billy R. Martin

Cannabinoids produce a characteristic profile of in vivo effects in mice, including suppression of spontaneous activity, antinociception, hypothermia, and catalepsy. Measurement of these four properties, commonly referred to as the tetrad test, has played a key role in establishing the structure-activity relationship of cannabinoids acting at cannabinoid CB(1) receptors. The purpose of this study was to determine whether drugs acting at noncannabinoid CB(1) receptors produced a similar pharmacological profile. Mice were tested in this paradigm after being injected with Delta(9)-tetrahydrocannabinol and selected drugs from other drug classes. Delta(9)-Tetrahydrocannabinol dose-dependently produced all four effects with reversal by the cannabinoid CB(1) receptor antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride (SR 141716A). Amphetamine, scopolamine, morphine, desipramine, pimozide, pentobarbital, ethanol, and diazepam were not fully active in at least one of the tests. Antipsychotics showed the greatest similarity to those of cannabinoids in the tetrad tests, although there were also distinct differences. Clozapine, haloperidol, thioridazine, and chlorpromazine (but not pimozide) were fully active in all four tests; however, unlike with Delta(9)-tetrahydrocannabinol, their effects were not blocked by SR 141716A. Further, whereas antipsychotics produced nearly 100% catalepsy, maximal catalepsy produced by Delta(9)-tetrahydrocannabinol was 60%. The mechanism through which antipsychotics produce these effects in mice is uncertain, but it differs from cannabinoid CB(1) receptor activation that mediates the effects of cannabinoids. While results of previous research suggest that the tetrad tests are a useful tool in examination of structure-activity relationships of cannabinoid CB(1) receptor agonists, the present results suggest that they must be used cautiously in the search for novel cannabinoid receptors.


Neuropharmacology | 1995

Discriminative stimulus effects of CP 55,940 and structurally dissimilar cannabinoids in rats

Jenny L. Wiley; Rl Barrett; J Lowe; Robert L. Balster; Billy R. Martin

CP 55,940 is a potent synthetic bicyclic cannabinoid analog that has been used in a number of studies as a radioligand for the cannabinoid receptor. This compound shares behavioral and biochemical properties with naturally occurring cannabinoids such as delta 9-THC. The purpose of the present study was 3-fold: to establish the ability of CP 55,940 to serve as a discriminative stimulus, to determine whether this discriminative stimulus is identical to that of delta 9-THC, and to examine whether a newly developed cannabinoid antagonist, SR141716A, would antagonize the discriminative stimulus effects of CP 55,940. Rats were trained to discriminate 0.1 mg/kg CP 55,940 from vehicle in standard 2-lever operant conditioning chambers. CP 55,940 produced dose-dependent generalization from the training dose in dose-effect determinations conducted before and after testing with other drugs. The effects of the training dose of CP 55,940 were dose-dependently antagonized by co-administration of SR141716A. Results of substitution tests showed that delta 9-THC, WIN 55,212-2, and cannabinol substituted completely for CP 55,940 in a dose-dependent manner; however, CP 55,940 was approx 10-fold more potent than any of the other drugs in producing CP 55,940-like discriminative stimulus effects. Several drugs with CNS depressant properties (phencyclidine, haloperidol and diazepam) failed to produce reliable substitution for CP 55,940. These results demonstrate that CP 55,940 has discriminative stimulus effects and that it shares these effects with structurally dissimilar compounds that, like CP 55,940, bind to the cannabinoid receptor. Further, these effects are blocked by SR141716A, a cannabinoid receptor antagonist.(ABSTRACT TRUNCATED AT 250 WORDS)


Chemistry and Physics of Lipids | 2002

Cannabinoid pharmacology: implications for additional cannabinoid receptor subtypes.

Jenny L. Wiley; Billy R. Martin

Delta(9)-Tetrahydrocannabinol (delta(9)-THC), the primary psychoactive constituent of marijuana (Cannabis sativa), is known to bind to two cannabinoid receptors: CB(1) receptors, located primarily in the brain, and CB(2) receptors, located primarily in the periphery. Recent research has suggested that other cannabinoids, including anandamide and WIN 55212-2, may also act at novel non-CB(1), non-CB(2) cannabinoid receptor(s). Anandamide produces a number of in vivo pharmacological effects in CB(1) knockout mice that are not produced by delta(9)-THC and cannot be explained by anandamides rapid metabolism. In addition, in vitro anandamide and WIN 55212-2 stimulate [35S]GTPgammaS binding in both CB(1) knockout and wildtype mice while delta(9)-THC stimulates this binding only in wildtype mice. Although anandamide and vanilloid agonists share pharmacological effects, anandamides actions in CB(1) knockout mice do not appear to be mediated by vanilloid VR(1) receptors. While not yet conclusive, these results suggest the possibility of additional cannabinoid receptors in the brain and periphery.


European Journal of Pharmacology | 1995

Discriminative stimulus effects of anandamide in rats

Jenny L. Wiley; Robert L. Balster; Billy R. Martin

Anandamide (arachidonylethanolamide), a putative endogenous ligand for the cannabinoid receptor, produces a tetrad of behavioral effects in mice characteristic of psychoactive cannabinoids including catalepsy, antinociception, hypothermia, and hypomobility. The present study examined the discriminative stimulus effects of anandamide in rats trained to discriminate delta 9-tetrahydrocannabinol or the potent cannabinoid receptor ligand CP 55,940 [(-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)-phenyl]-trans-4-(3- hydroxypropyl)cyclohexanol)] from vehicle. Intraperitoneal injections of anandamide substituted for delta 9-tetrahydrocannabinol and for CP 55,940; however, unlike substitution dose-effect curves with the training drugs, anandamide substitution occurred at a single dose (30 or 45 mg/kg) and was accompanied by severe decreases in response rates. The results of the present study suggest that, although systemic anandamide administration may have cannabimimetic effects similar to those of delta 9-tetrahydrocannabinol and CP 55,940, some differences in the behavioral effects of anandamide and other psychoactive cannabinoids also are apparent.


The Journal of Neuroscience | 2014

Baths Salts, Spice, and Related Designer Drugs: The Science Behind the Headlines

Michael H. Baumann; Ernesto Solis; Lucas R. Watterson; Julie A. Marusich; William E. Fantegrossi; Jenny L. Wiley

The abuse of synthetic psychoactive substances known as “designer drugs,” or “new psychoactive substances” (NPS), is increasing at an alarming rate. NPS are purchased as alternatives to traditional illicit drugs of abuse and are manufactured to circumvent laws regulating the sale and use of controlled substances. Synthetic cathinones (i.e., “bath salts”) and synthetic cannabinoids (i.e., “spice”) are two types of NPS that have received substantial media attention. Although low recreational doses of bath salts or spice compounds can produce desirable effects, high doses or chronic exposure often leads to dangerous medical consequences, including psychosis, violent behaviors, tachycardia, hyperthermia, and even death. Despite the popularity of NPS, there is a paucity of scientific data about these drugs. Here we provide a brief up-to-date review describing the mechanisms of action and neurobiological effects of synthetic cathinones and cannabinoids.

Collaboration


Dive into the Jenny L. Wiley's collaboration.

Top Co-Authors

Avatar

Billy R. Martin

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert L. Balster

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aron H. Lichtman

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Robert E. Vann

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge