51
JPET #172957 N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4- carboxamide (THIIC), a novel mGlu2 potentiator with potential anxiolytic/antidepressant properties: In vivo profiling suggests a link between behavioral and CNS neurochemical changes Matthew J. Fell, Jeffrey M. Witkin, Julie F. Falcone, Jason S. Katner, Kenneth W. Perry, John Hart, Linda Rorick-Kehn, Carl D. Overshiner, Kurt Rasmussen, Stephen F. Chaney, Mark J. Benvenga, Xia Li, Deanna L. Marlow, Linda K. Thompson, Susan K. Luecke, Keith A. Wafford, Wesley F. Seidel, Dale M. Edgar 1 , Anne T. Quets, Christian C. Felder, XuShan Wang, Beverly A. Heinz, Alexander Nikolayev, Ming- Shang Kuo, Daniel Mayhugh, Albert Khilevich, Deyi Zhang, Philip J Ebert, James A. Eckstein, Bradley L. Ackermann, Steven P. Swanson, John T. Catlow, Robert A. Dean, Kimberley Jackson, Sitra Tauscher- Wisniewski, Gerard J. Marek, Jeffrey M. Schkeryantz, Kjell A. Svensson Neuroscience Discovery Research, Eli Lilly and Company, Indianapolis, IN 46285, USA (MJF, JMW, JFF, JSK, KWP, JH, LRK, CDO, KR, SFC, MJB, XL, DLM, LKT, SKL, ATQ, CCF, STW, GJM, KAS) Neuroscience Discovery Research, Eli Lilly and Company, Erl Wood, Surrey, UK (KAW, WFS, DME). Quantitative and Structural Biology, Eli Lilly and Company, Indianapolis, IN 46285, USA (XSW, BAH) Translational Science, Eli Lilly and Company, Indianapolis, IN 46285, USA (AN, MSK, PJE) LRL Discovery Chemistry Research, Eli Lilly and Company, Indianapolis, IN 46285, USA (DM, AK, DZ, JMS) Laboratory for Experimental Medicine, Eli Lilly and Company, Indianapolis, IN 46285, USA (JAE, BLM) Drug Disposition, Eli Lilly and Company, Indianapolis, IN 46285, USA (SPS, JTC) Diagnostic & Experimental Medicine, Eli Lilly and Company, Indianapolis, IN 46285, USA (RAD) Global PK/PD & TS, Eli Lilly and Company, Erl Wood, Surrey, UK (KJ) JPET Fast Forward. Published on October 14, 2010 as DOI:10.1124/jpet.110.172957 Copyright 2010 by the American Society for Pharmacology and Experimental Therapeutics. This article has not been copyedited and formatted. The final version may differ from this version. JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957 at ASPET Journals on March 19, 2021 jpet.aspetjournals.org Downloaded from

N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

N-(4-((2-(trifluoromethyl)-3-hydroxy-4-

(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-

carboxamide (THIIC), a novel mGlu2 potentiator with potential

anxiolytic/antidepressant properties: In vivo profiling suggests a link

between behavioral and CNS neurochemical changes

Matthew J. Fell, Jeffrey M. Witkin, Julie F. Falcone, Jason S. Katner, Kenneth W. Perry, John Hart, Linda

Rorick-Kehn, Carl D. Overshiner, Kurt Rasmussen, Stephen F. Chaney, Mark J. Benvenga, Xia Li,

Deanna L. Marlow, Linda K. Thompson, Susan K. Luecke, Keith A. Wafford, Wesley F. Seidel, Dale M.

Edgar1, Anne T. Quets, Christian C. Felder, XuShan Wang, Beverly A. Heinz, Alexander Nikolayev, Ming-

Shang Kuo, Daniel Mayhugh, Albert Khilevich, Deyi Zhang, Philip J Ebert, James A. Eckstein, Bradley L.

Ackermann, Steven P. Swanson, John T. Catlow, Robert A. Dean, Kimberley Jackson, Sitra Tauscher-

Wisniewski, Gerard J. Marek, Jeffrey M. Schkeryantz, Kjell A. Svensson

Neuroscience Discovery Research, Eli Lilly and Company, Indianapolis, IN 46285, USA (MJF, JMW, JFF,

JSK, KWP, JH, LRK, CDO, KR, SFC, MJB, XL, DLM, LKT, SKL, ATQ, CCF, STW, GJM, KAS)

Neuroscience Discovery Research, Eli Lilly and Company, Erl Wood, Surrey, UK (KAW, WFS, DME).

Quantitative and Structural Biology, Eli Lilly and Company, Indianapolis, IN 46285, USA (XSW, BAH)

Translational Science, Eli Lilly and Company, Indianapolis, IN 46285, USA (AN, MSK, PJE)

LRL Discovery Chemistry Research, Eli Lilly and Company, Indianapolis, IN 46285, USA (DM, AK, DZ,

JMS)

Laboratory for Experimental Medicine, Eli Lilly and Company, Indianapolis, IN 46285, USA (JAE, BLM)

Drug Disposition, Eli Lilly and Company, Indianapolis, IN 46285, USA (SPS, JTC)

Diagnostic & Experimental Medicine, Eli Lilly and Company, Indianapolis, IN 46285, USA (RAD)

Global PK/PD & TS, Eli Lilly and Company, Erl Wood, Surrey, UK (KJ)

JPET Fast Forward. Published on October 14, 2010 as DOI:10.1124/jpet.110.172957

Copyright 2010 by the American Society for Pharmacology and Experimental Therapeutics.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 2: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Running Title Page

Running Title: mGlu2 Potentiator THIIC

Corresponding Author:

Dr. Matthew J. Fell, Neuroscience Discovery Research, Lilly Research Laboratories, Lilly Corporate

Center, DC0510, Indianapolis, IN 46285, USA; Tel: 317-433-1096; Fax: 317-276-7600;

Email: [email protected]

Number of Text Pages: 38

Number of Tables: 1

Number of Figures: 12

Number of References: 40

Number of Words in Abstract: 243

Number of Words in Introduction: 696

Number of Words in Discussion: 1476

JPET Section: Neuropharmacology

Abbreviations: mGlu, metabotropic glutamate; iGlu, ionotropic glutamate; THIIC, N-(4-((2-

(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide;

LY404039, (-)-(1R,4S,5S,6S)-4-amino-2 sulfonylbicyclo [3.1.0] hexane-4,6-dicarboxylic acid; LY379268,

(-)-2-oxa-4-aminobicyclo[3.1.0] hexane-4,6-dicarboxylic acid; LY341495, 2S-2-amino-2-(1S,2S-2-

carboxycycloprop-1-yl)-3-(xant-9-yl) propanoic acid; LY354740, 1S,2S,5R,6S-2

aminobicyclo[3.1.0]hexane-2, 6-bicaroxylate monohydrate; CBiPES, N-[4'-cyano-biphenyl-3-yl)-N-(3-

pyridinylmethyl)-ethanesulfonamide hydrochloride; BINA, Biphenyl-indanone A; LY487379, N-(4-

(2-methoxyphenoxy)-phenyl-N-(2,2,2-trifluoroethylsulfonyl)-pyrid-3-ylmethylamine; mPFC, medial

prefrontal cortex; t-MeHA, tele-methylhistamine;

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 3: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Abstract

The normalization of excessive glutamatergic neurotransmission through the activation of mGlu2

receptors may have therapeutic potential in a variety of psychiatric disorders including anxiety/depression

and schizophrenia. Here, we characterize the pharmacological properties of THIIC (N-(4-((2-

(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide)

a structurally novel, potent and selective allosteric potentiator of human and rat mGlu2 receptors (EC50’s

= 23 and 13 nM, respectively). THIIC produced anxiolytic-like efficacy in the rat stress induced

hyperthermia assay and in the mouse stress-induced elevation of cerebellar cGMP and marble-burying

assays. THIIC also produced robust activity in three assays that detect antidepressant-like activity

including the mouse forced-swim test, the rat DRL-72 sec assay, and the rat dominant-submissive test,

with a maximal response similar to that of imipramine. Effects of THIIC in the forced-swim test and

marble-burying were deleted in mGlu2 receptor null mice. Analysis of sleep EEG showed that THIIC had

a sleep-promoting profile with increased NREM and decreased REM sleep. THIIC also decreased the

dark phase increase in extracellular histamine in the mPFC and decreased levels of the histamine

metabolite tele-methylhistamine (t-MeHA) in rat CSF. Collectively, these results indicate that the novel

mGlu2 positive allosteric modulator, THIIC, has robust activity in models used to predict

anxiolytic/antidepressant efficacy substantiating, at least with this molecule, differentiation in the biological

impact of mGlu2 potentiation vs. mGlu2/3 orthosteric agonism. In addition, we provide evidence that

sleep EEG and CSF t-MeHA might function as viable biomarker approaches to facilitate the translational

development of THIIC and other mGlu2 potentiators.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 4: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Introduction

Glutamate is the principle excitatory neurotransmitter in the mammalian central nervous system and

mediates a diversity of physiological and behavioral actions through “ionotropic” and “metabotropic”

(mGlu) receptors. Of the eight mGlu receptor subtypes the group II mGlu receptors, which includes

mGlu2 and mGlu3, are particularly attractive as novel drug targets. mGlu2 and mGlu3 receptors function

to inhibit neurotransmitter release as autoreceptors on glutamatergic terminals, presynaptic

heteroreceptors (Schoepp, 2001), or via modulatory actions on glia (Conn and Pin, 1997). The mGlu2

and mGlu3 receptors are present throughout key forebrain and limbic areas such as the prefrontal cortex,

thalamus, striatum, hippocampus and the amygdala (Wright et al., 2001). Consequently, drugs which

activate group II mGlu receptors may represent a novel approach to treat a variety of psychiatric and

neurological disorders.

To date, only one structural class of selective agonists for group II mGlu receptors have been discovered

and are undergoing development. In preclinical studies, mGlu2/3 receptor agonists (LY379268,

LY354740 and LY404039) have behavioral and neurochemical effects in models predictive of anti-

stress/anxiolytic and antipsychotic efficacy (Schoepp and Marek, 2002; Swanson et al., 2005). Moreover,

the anxiolytic and antipsychotic potential of selective mGlu2/3 receptor agonists has been confirmed in

early clinical trials (Grillon et al., 2003; Schoepp et al., 2003; Patil et al., 2007; Dunayevich et al., 2008). In

addition, recent evidence also suggests that the activation of mGlu2/3 receptors may be beneficial in

depression. For example, mGlu2/3 receptors exert tonic control over the hypothalamic-pituitary-adrenal-

axis which is known to be disrupted in depressed patients. mGlu2/3 receptors agonists also induce

neurochemical and behavioral changes suggestive of potential antidepressant efficacy including

increased monoamine efflux in limbic regions (Cartmell et al., 2001; Rorick-Kehn et al., 2007b),

stimulation of neurotrophic factor production (Di Giorgi-Gerevini et al., 2005), neuroprotection (Bruno et

al., 2001) and an antidepressant-like suppression of REM sleep (Ahnaou et al., 2009). Finally, in-vitro and

in-vivo studies suggest the existence of a synergism between clinically effective antidepressants and

mGlu2/3 receptor agonists (Matrisciano et al., 2008).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 5: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Another approach toward enhancing group II mGlu receptor function is through the development of

allosteric modulators of mGlu2. Unlike orthosteric acting agonists, allosteric modulators do not activate

the receptor directly but act at an allosteric site to selectively potentiate the response of mGlu2 receptors

to glutamate. Using electrophysiological techniques, it has been shown that allosteric modulators have

little or no effects on glutamate release under “normal conditions” but act to potentiate negative feedback

control under conditions of excessive glutamate release (Johnson et al., 2005). This ability to recognize

“pathological states” or networks could result in therapeutic advantages in terms of safety and efficacy.

Given the selectively of mGlu2 potentiators for mGlu2 over mGlu3 receptors, such potentiators are also

valuable tools for helping to differentiate the biological substrates driven exclusively by physiologically-

relevant augmentation of mGlu2 receptors.

Multiple highly selective positive allosteric modulators of mGlu2 have been identified, of which MPPTS

(LY487379), CBiPES and BINA are the most widely characterized compounds (Galici et al., 2005;

Johnson et al., 2005). Like mGlu2/3 receptor agonists, these mGlu2 positive allosteric modulators have

shown efficacy in preclinical models used to predict anxiolytic and antipsychotic activity. However,

progress in the development of these positive allosteric modulators of mGlu2 has been slow primarily due

to their lack of good drug-like features including modest potency, poor metabolic stability and brain

penetration.

In the present study we report the discovery of N-(4-((2-(trifluoromethyl)-3-hydroxy-4-

(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC) (Khilevich et al., 2010),

a structurally novel and highly selective positive allosteric modulator of mGlu2 receptors (figure 1). In

addition to evaluating the in vitro pharmacology of THIIC, we have also characterized the effects of THIIC

in models used to predict anxiolytic and/or antidepressant activity including stress-induced hyperthermia,

reversal of stress-induced increases in cerebellar cGMP, marble-burying, dominant submissive test,

differential reinforcement of low rate 72-s (DRL-72) behavior and mouse forced swim test. A role for

mGlu2 receptor actions driving antidepressant- or anxiolytic-like effects were assessed by the use of

mGlu2 -/- mice. Finally, in an effort to develop a potential translational biomarker for this target, we

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 6: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

studied the effects of THIIC for changes in rat sleep EEG patterns and CNS neurochemistry with a focus

on histamine release in the brain and histamine metabolites in the CSF.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 7: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Methods

In vitro Assays

Human and Rat mGlu2 Potentiator FLIPR Assays

AV-12 cell lines stably expressing the human or rat mGlu2 receptor and co-transfected with the rat

glutamate transporter EAAT 1 (Excitatory Amino Acid Transporter 1) and the Gα15 subunit were used for

these studies. The expression of Gα15 allows Gi-coupled receptors to signal through the phospholipase

C pathway, resulting in the ability to measure receptor activation by a fluorometric calcium response

assay. The cell lines were maintained by culturing in Dulbecco’s Modified Eagle’s Medium (DMEM) with

high glucose and pyridoxine hydrochloride supplemented with 5% heat inactivated, dialyzed fetal bovine

serum, 1 mM sodium pyruvate , 10 mM HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid), 1

mM of L-glutamine, and 5 μg/mL blasticidin (all media purchased from Invitrogen). Confluent cultures

were passaged biweekly using an enzyme-free dissociation solution (Chemicon S-004-B). Cells were

harvested 24 hours prior to assay and dispensed using a Matrix Well-Mate cell seeder at 85,000 (human)

or 65,000 (rat) cells per well into 96-well, black-walled, poly-D-lysine-coated plates (BD BioCoat #354663)

in medium containing only 250 μM L-glutamine (freshly added).

Intracellular calcium levels were monitored before and after the addition of compounds using a

Fluorometric Imaging Plate Reader (FLIPR, Molecular Devices). The assay buffer was comprised of

Hank’s Buffered Salt Solution (HBSS; Sigma) supplemented with 20 mM HEPES. The medium was

removed and the cells were incubated with 8 μM Fluo-3AM (Molecular Probes, F-1241; 50 μL per well) in

assay buffer for 90 minutes at 25ºC. The dye solution was removed and replaced with fresh assay buffer

(50 μL per well). A single-addition FLIPR assay generating an 11-point concentration response curve for

the agonist glutamate (Fisher A125-100) was conducted prior to each experiment. The results were

analyzed using Prism v4.03 (GraphPad Software) to calculate the concentrations of glutamate needed to

induce the EC10 responses and to monitor the general sensitivity of the cells for the agonist.

The compound was tested in a two-addition FLIPR assay using a 10-point concentration response profile

starting at a final concentration of 25 μM (agonist mode) or 12.5 μM (potentiator mode). A 3-fold dilution

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 8: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

series in dimethyl sulfoxide (DMSO) was followed by a single dilution into assay buffer; the final

concentration of DMSO was 0.625%. After taking an initial 5-sec fluorescent read on the FLIPR

instrument, compound was added to the cell plate (50 ul per well). Data were collected every second for

the first 30 seconds and then every 3 seconds for a total of 90 seconds in order to detect agonist activity.

Immediately thereafter, the second addition consisting of 100 μl of glutamate in assay buffer (typically

about 1 μM) was added to the cell plate, generating an EC10 response. Following the second addition,

data were collected every second for 29 images and then every 3 seconds for 15 images. The maximal

response was defined as that induced by ECmax (100 μM glutamate). The compound effect was

measured as maximal minus minimal peak heights in relative fluorescent units (RFUs) corrected for basal

fluorescence measured in the absence of glutamate. Determinations were carried out using single plates.

Agonist effects were quantified as percent stimulation induced by compound alone relative to the maximal

glutamate response. Potentiation effects were quantified as percent increase in the presence of an EC10

response in glutamate relative to the ECmax response. Using calcium mobilization assays analogous to

those described above, THIIC was tested for activity in agonist, potentiator, or antagonist modes at the

human mGluR1, 3, 4, 5, and 8 receptor subtypes, each stably expressed in AV12 cells. Maximum

concentrations were 12.5 μM (antagonist and potentiator modes) and 25 μM (agonist mode). All data

were calculated as relative EC50 values using a four-parameter logistic curve fitting program

(ActivityBase v5.3.1.22).

The observed ability to potentiate the glutamate activity was confirmed using glutamate concentration-

response curves generated in the presence of increasing concentrations of THIIC. Glutamate was diluted

in assay buffer through 12 concentrations ranging from 100 μM to 49 nM, and the compound was 3X

serially diluted in DMSO for an 8-point concentration response profile starting at a final concentration of

12.5 μM. Data collection and analysis were as described previously.

GTPγ35S Binding Assay

GTPγ35S binding was determined using an antibody capture scintillation proximity technique in a 96 well

plate format. Briefly, 100 μl (20-40 fmol/well) of membrane preparations from AV12 cells that ectopically

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 9: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

express either human or rat metabotropic glutamate receptor 2 (mGluR2) were incubated for 30 minutes

with 50 μl of test compound. GDP (1 μM final concentration) was added to receptor membranes prior to

incubation. Dose response curves were achieved with increasing amounts of THIIC in the presence of

increasing amounts of the orthosteric agonist, glutamate (Tocris, Ellisville, MO). Following the incubation

period, 50 μl diluted GTP-γ-[35S] (500 pM final concentration; PerkinElmer Life and Analytical Sciences,

Boston, MA) was added to each well and incubated for 30 min. The labeled membranes were then

solubilized with 0.27% Nonidet P40 followed by a 3 hour incubation with 20 μl (final dilution of 1:200) of

anti-Gαi-3 rabbit polyclonal antibody (Covance, Princeton, NJ) and 50 μl (1.25 mg/well) anti-rabbit

scintillation proximity assay (SPA) beads (GE Healthcare, Piscataway, NJ). The plates were centrifuged

for 10 min. at 700 x g and counted for 1 min. per well using a Wallac MicroBeta TriLux scintillation counter

(PerkinElmer, Boston, MA). All incubations took place at room temperature in GTP-binding assay buffer

(20 mM Hepes, 100 mM NaCl, 5 mM MgC12, pH 7.4). Data were analyzed using nonlinear regression for

a sigmoidal concentration response curve (GraphPad Prism v. 4.03). Background was subtracted from all

wells and data were normalized to a full agonist response (100 μM glutamate) and efficacy was

expressed as a percentage of Maximal Response. Mean EC50 were calculated as a mean of three

independent determinations ± standard error of the mean (SEM). Kb and cooperativity factor were

calculated from EC50-generated dose ratios imported into an allosteric Schild regression algorithm;

Y=(Kb+α*B)/(Kb+B), where α =10^Logα, Kb=10^LogKb and B=10^X.

Selectivity Panel

THIIC was tested for binding to dopamine and serotonin receptors using receptor binding assays

performed according to methods reported previously by Rorick-Kehn et al., (2007a). An additional panel

of over 30 different biogenic amine receptors, neuropeptide receptors, ion channel binding sites, and

neurotransmitter transporter binding assays were run by CEREP Inc. (Celle L’Evescault, France). The

targets run were: human adenosine A3 receptors; acetylcholinesterase; angiotensin II; adrenergic

receptors α1 (nonselective), α2 (nonselective), β1 and β2 ; norepinephrine (NE) transporter; rat brain

benzodiazepine receptor (BZD); dopaminergic receptors D1 and D2s; histaminergic receptors H1 and H2;

muscarinic receptors M2 and M3; neurokinin receptor 1; opioid receptor μ; serotonin receptors 5HT2B

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 10: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

agonist site, rat Ca2+ channel verapamil site; rat brain voltage-gated potassium channel (K+V channel);

rat brain small-conductance Ca2+-activated K+ channel (SK+Ca channel); rat Na+ channel (site 2); and

rat Cl- channel. All assays were run using recombinant human receptors, except where noted. The full

methods and references can be found on the Cerep website (www.cerep.fr). The assays were run at 1

μM and 10 μM THIIC, and the percentage of inhibition is given as the average of three determinations.

When significant displacement of radioligand was observed (>50% inhib at 1 µM), complete

concentration-dependent displacement curves (in triplicate) were constructed to generate IC50 values.

Animals

All experiments were performed according to the policies of the Animal Care and Use Committee of Eli

Lilly and Company, in conjunction with the American Association for the Accreditation of Laboratory

Animal Care-approved guidelines and the Guide for Care and Use of Laboratory Animals (Institute of

Laboratory Animal Resources, 1996). Animals were individually or group housed depending on the assay

in an environmentally controlled facility where food and water were available ad libitum unless specified

otherwise. mGlu2 receptor -/- mice and their wild-type littermates were derived and bred as described in

Fell et al., (2008). For EEG studies, each rat was housed individually within a specially modified

microisolator cage having a custom polycarbonate riser to raise the filter top which held an ultra-low-

torque slip-ring commutator (Hypnion, Inc.). A custom flexible tether (Hypnion, Inc.) was connected at one

end to the commutator and at the other end to the animal’s cranial implant. Each cage was located within

a separate compartment of a stainless steel recording chamber, and had an infrared light source and

digital video camera to allow a minimum of twice daily remote visual monitoring. The ambient temperature

was 23±1°C, and relative humidity averaged 50%. A 24-hr light-dark cycle(LD 12:12) was maintained

throughout the study using fluorescent light. Light intensity averaged 35-40 lux at mid-level inside the

cage.

Materials

THIIC (Khilevic et al., 2010) and MTEP were synthesized at Lilly Research Laboratories. For in vivo

experiments, THIIC was suspended in 1% carboxymethylcellulose, 0.25% Tween 80, and 0.05% Dow

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 11: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

antifoam. MTEP was dissolved in sterile H2O. Imipramine and alprazolam were purchased from Sigma

Aldrich (St. Louis, MO). Chlordiazepoxide HCl (Sigma-Aldrich) was dissolved in 0.9% NaCl. Alprazolam

was suspended in 5% acacia while imipramine was dissolved in sterile 0.9% NaCl. For all experiments,

drugs were mixed fresh before use. Doses of THIIC were administered to rats in a volume of 4 ml/kg;

doses were administered to mice in a volume of 10 ml/kg. Imipramine and alprazolam were dosed in a

volume of 1 mg/kg to rats and 10 mg/kg to mice. Routes of administration are indicated separately for

each experiment.

Assays for Antidepressant-like Activity

Mouse Forced-Swim Test

Male, NIH Swiss mice (Harlan, Labs, USA) weighing 20 to 25 g were housed in plastic cages (40.6 × 20.3

× 15.2 cm) with 10 to 12 mice/cage in a vivarium at least 7 days before the experiments. Mice were

maintained on a 12-h light/dark cycle (6:00 AM/6:00 PM), and all procedures were performed between

12:00 PM and 4:00 PM. Animals were removed from the vivarium to the testing area in their home cages

and allowed to adapt to the new environment for at least 1 h before testing. The forced-swim test was

performed using the original method described by Porsolt et al. (1977). In brief, mice were placed

individually in clear plastic cylinders (10 cm in diameter by 25 cm in height) filled to 6 cm with 22 to 25°C

water for 6 min. The duration of immobility was recorded during the last 4 min of a 6-min trial. A mouse

was regarded as immobile when floating motionless or making only those movements necessary to keep

its head above the water. Dose-effect functions for THIIC were performed after both i.p.(30 min prior) and

oral dosing (60 min prior). Imipramine was used as a comparator standard (15 mg/kg,i.p., 30 min prior).

Statistical analyses were performed by ANOVA followed by Dunnett's test.

Differential Reinforcement of Low Rate 72-s (DRL 72-s) behavior in rats

Male Sprague-Dawley rats weighing between 300 and 350 g at the beginning of the behavioral

experiments (Holtzman, Madison, WI) were housed in pairs. The colony room was maintained at 20°C

and 60% relative humidity. The room was illuminated 12 h/day (7:00 AM–7:00 PM). All rats had free

access to laboratory chow (Teklad 4% Rat Diet; Harlan Teklad, Madison, WI) except during experimental

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 12: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

sessions. Water was available for a 20-min period following the daily behavioral session. Sixteen operant-

conditioning chambers (MED Associates, St. Albans, VT) were used. The lever in these chambers was

mounted on one wall with the water access next to the lever in the middle of the wall. A reinforced

response caused the dipper (0.02-ml cup) to be lifted from a water trough to an opening in the floor of the

access port for 4 s. The house light, which was mounted on the opposite wall, was turned on when the

session began, remained on throughout the entire session, and was turned off at the end of the session.

Each experimental chamber was enclosed in a separate sound-attenuating cubicle equipped with a white

noise generator to provide masking noise. Rats were water deprived for 22.5 h before each session. Each

rat was initially trained under an alternative fixed ratio 1, fixed time 1-min schedule for water

reinforcement where each response was reinforced, and water was also provided every minute if a

response did not occur. The few rats that did not acquire lever-pressing behavior after three daily 1-h

sessions under this schedule were trained by the experimenter using the method of successive

approximation. After the rats had acquired lever-pressing behavior, they were trained during daily DRL

18-s sessions for 2 weeks before moving directly to DRL 72-s sessions. Under the DRL schedules, a

response only produced water if it occurred at least 18 or 72 s after the last water delivery; responses

prior to the designated time period reset the timer. Responding on these sessions became stable after 8

weeks. Experimental sessions lasted for 1 h and were conducted 5 days/week during light hours. The

data analyses were conducted on the raw response and reinforcement data. All behavioral data are

expressed as the mean ± S.E.M. normalized to the vehicle or vehicle/vehicle condition. The effects of

THIIC and imipramine were analyzed with a one-way repeated measures analysis of variance (ANOVA).

The level of significance was set for p < 0.05 for all analyses.

Dominant Submissive Test in Rats

Experimentally-naïve male Sprague-Dawley rats, weighing 160-180 grams at the start of the study,

(Harlan Sprague Dawley) were assigned randomly to pairs and the paired animals remained separated

through the course of the experiment, except during the 5-minute testing periods. Rats were maintained

on a 12hr light/dark cycle (0600/1800 hours), and all procedures were performed between 12:00 PM and

4:00 PM in ambient room light. The testing apparatus was constructed from transparent plastic material

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 13: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

and consisted of two identical chambers (24 x 17 x 14 cm) connected by a round tunnel (4.5 cm diameter

x 52 cm long). A container (10 ml beaker) of sweetened milk (9 % sucrose) was placed in an opening in

the floor at the mid-point of the tunnel. Animals were food deprived over-night prior to the first test

session. During the testing period, each member of a pair was placed in the different chambers of the

testing apparatus, the gates were opened, and the time spent drinking was recorded for each animal for a

5 minute period. At the end of the 5-minute testing period, animals were returned to their home cage and

given free access to food for 1 hour. The animals were also given free access to food from Friday

afternoon (following the test session) to Sunday morning, when they were once again food-deprived.

During the first week of the testing (acclimation week), drinking time was not scored. During the second

week of testing (selection week), the time spent drinking was recorded. Pairs of animals that passed the

following criteria continued into the drug administration phase of the experiment: (1) the difference

between the average daily drinking scores of the two animals was significant (two-tailed t-test, p < 0.05);

and, (2) the dominant animal’s score was at least 25% greater than the submissive animal’s score. Any

pairs not passing the selection criteria were dropped from the study. The animal of a pair with the higher

drinking score was labeled as “dominant” and the animal of a pair with lower drinking score was labeled

as “submissive”. The submissive rat of each pair was treated with the THIIC (3 or 30 mg/kg) or

imipramine (10 mg/kg) while the dominant rat of each pair was treated with vehicle for the next 21 days.

The 5-minute test was repeated once daily, except weekends, for 21 days following the beginning of the

drug treatment. Dominance levels were calculated as the difference in daily drinking scores between

paired rats. The daily dominance level values were averaged over each week and statistical comparisons

were made by analysis of repeated measurements coupled Tukey’s post-hoc test.

Assays for Anxiolytic-like Activity

Marble-burying and rotorod performances. Male, CD1 mice were used as described (Li et al.,2006 ).

Briefly, mice were dosed with either THIIC (i.p.) or chlordiazepoxide (i.p.) and tested on a rotating rod

without prior training. Mice were scored as failing if they fell off the rotorod on two occasions during a 2

min test. Marble-burying behavior was assessed in the same mice immediately post rotorod testing. The

number of marbles buried out of 20 by sawdust bedding by at least 2/3 was measured. The number of

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 14: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

marbles buried was evaluated by ANOVA followed by Dunnett’s test; rotorod performances were

evaluated by Fisher’s exact probability test.

Rat Stress-Induced Hyperthermia

Male, Fischer F-344 rats (Harlan, Indianapolis, IN, USA) weighing between 275 – 350 g were individually-

housed with food and water available ad libitum, and maintained on a 12 h light/dark cycle (lights on at

06:00). The rats were fasted for approximately 12-18 hours before the experiment. Rats were

transported from the colony room in groups of n = 10, to a procedure room for dosing. Rats were dosed

p.o. in a dose volume of 2 mL/kg with THIIC (1, 3, 10, and 30 mg/kg). The mGlu5 receptor antagonist

MTEP, which has demonstrated robust anxiolytic-like activity in preclinical models, was used as a positive

control (10 mg/kg, PO). Immediately following dosing, rats were returned to their home cage, the lights

and left the room and dosing room (Room A) was darkened for the remainder of the 4 hr pretreatment

period. After the pretreatment period, rats were taken individually to a brightly lit adjacent room (Room B)

where baseline body temperatures were determined by insertion of a rectal probe lubricated with mineral

oil. Core body temperature was assessed using a Physitemp® BAT-12® Microprobe Thermometer with

a Physitemp® RET-2® rat rectal probe (Physitemp Instruments Inc., NJ) inserted approximately 2 cm into

the rectum and is designated as the baseline body temperature, T1, in degrees C. The rat was then

placed back in the homecage, and remained in Room B. Ten minutes later a second body temperature

measurement was recorded (T2). The difference between the first and second body temperature

measurements (T2 – T1) was used as an index of stress-induced hyperthermia. Data represent means

and SEM and were subjected to one-way ANOVA followed by Dunnett's test.

Reversal of Stress induced increase in cerebellar cGMP

Male CF-1 mice (18-20g) (Harlan Industries, Indianapolis, IN) were housed in an enriched environment in

groups of 8 mice per cage with food and automated water available ad libitum. Animals were kept under a

12-hr light/dark cycle with lights on at 6:00 am in a temperature and humidity-controlled environment for

5-7 days prior to their use. The mice were kept in their home cages to avoid stress and received an IP

injection of either vehicle, THIIC (3, 10 and 30 mg/kg) or and alprazolam (1 mg/kg) 30 minutes prior to

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 15: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

stressor (foot-shock). To evaluate the effects of the test compounds on basal cGMP levels, vehicle

control ‘no stress’ mice were sacrificed without being subjected to a foot-shock. Vehicle stress, THIIC and

alprazolam treated mice were subjected to a stressor of inescapable electric footshock (1 mA intensity for

10 sec) in a metal cage with stainless steel grid floor (Coulbourn Instruments, L.L.C. 7462 Penn Drive,

Allentown, PA 18106), and immediately sacrificed for estimation of basal cGMP levels. All mice were

euthanized using a beam of microwave radiation focused on the skull for 0.5 sec (Thermax Thermatron,

Louisville, KY). The cerebellum was quickly removed from the skull and the weight recorded. The tissue

was then placed in a polystyrene tube containing 2.0 ml of 1% perchloric acid (PCA) on ice. Tissues

were then homogenized and incubated on ice for 30 minutes after which the samples were boiled for 5

minutes. The tubes were then centrifuged at 11,700xg for 20 minutes at room temperature. The

supernatant was acetylated using a 2:1 ratio of triethylamine (EM Science, TX1200-5) and acetic

anhydride. The samples were immediately vortexed and then centrifuged at 13,000xg for 20 minutes at

4oC. The cGMP assay buffer included the following: 0.05 M sodium acetate, trihydrate (6.8 g/L) and 0.1%

sodium azide adjusted to pH 6.2 with acetic acid. For the radioligand tracer, a separate tracer buffer was

prepared using the cGMP assay buffer and adding 0.002 M EDTA disodium salt dihydrate (0.7 g/L), with

1% normal rabbit serum (Perkin Elmer, FP526), added just prior to use.

125I-cGMP (guanosine 3’,5’-cyclic phosphoric acid 2’-0-succinyl[125I]iodotyrosine methyl ester, obtained

from Perkin Elmer (SA 2200 Ci/mmol, 32.3 uCi/ml, NEX-131, Boston, MA), was prepared in the tracer

diluent, pH 6.2, at a concentration of 0.75 μCi/10 ml tracer buffer per FlashPlateTM (Perkin Elmer Life

Sciences, Boston, MA, SMP002). A cGMP standard curve was prepared using guanosine-3’:5’-cyclic

monophosphate lyophilized standard (Perkin Elmer, FP523) reconstituted with 2.0 ml of distilled water to

make a 2000 pm/ml solution (stored @ 4oC). This stock was diluted with assay buffer to generate a 9-

point standard curve. A 1.0 ml aliquot of each standard, including a blank, were acetylated and then

vortexed immediately. 100 µl of each standard was added, in duplicate, to flashplate wells, including two

100 µl aliquots directly from the stock cGMP standard bottle for nonspecific binding (NSB) determination.

40 µl of each sample, in duplicate, was added to flashplate wells containing 60 µl of cGMP assay buffer

for a total sample volume of 100 µl (yields a 1:50 final dilution). 100 µl of [125 I]-Cyclic GMP was added to

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 16: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

all flashplate wells and plates were covered before being shaken overnight (16-24 hours) at 2-8oC using a

titer plate shaker. Contents of the wells were aspirated and the plates resealed. Plates were then counted

for 1 minute/well in a Packard TopCountTM radioactive counter.

cGMP data calculations were analyzed using Excel and GraphPad Prism. Mean counts/min (cpms) from

duplicate samples were averaged and the average NSB cpms subtracted to get specific cpms. Specific

cpms were used to determine the Normalized % Bound. GraphPad Prism determined the nonlinear

regression curve fit from the Normalized % Bound for the standard curve and interpolated the values of

the unknown samples as log pmol cGMP. Results were normalized to tissue weights for each sample to

calculate pmol cGMP/gram tissue. Group averages were calculated as % Vehicle Control and % Vehicle

Stress Control and the SEM was determined. Statistical analyses were carried out by GraphPad Prism

using 1-factor (dose group) ANOVA followed by Dunnett’s Multiple Comparison Test versus vehicle and

vehicle stress groups indicating a p<0.05 value for significance.

Sleep EEG in the rat

Male, Wistar rats were surgically prepared with a cranial implant that permitted chronic electro-

encephalogram (EEG) and electromyogram (EMG) recording, in addition to an abdominally implanted

telemeter to measure temperature and locomotor activity. Following recovery animals were individually

housed and connected to low torque commutator via a flexible tether allowing access to all areas of the

cage. Animals were maintained on a 24 hr light/dark schedule (12:12) with adlib food and water. Animals

were undisturbed for 48 hr before and after each treatment. Sleep and wakefulness were determined

using SCORE2004™ — a microcomputer-based sleep-wake and physiological monitoring system. In the

present study, the system monitored amplified EEG (Х10,000, bandpass 1-30 Hz; initial digitization rate

400 Hz [Grass Corp., Quincy, MA]), integrated EMG (bandpass 10-100 Hz, RMS integration), telemetered

body temperature, non-specific locomotor activity (LMA), and drinking activity simultaneously. Arousal

states were classified on-line as NREM sleep, REM sleep, wake, or theta-dominated wake every 10

seconds using EEG period and amplitude feature extraction and ranked membership algorithms.

Individually taught EEG-arousal-state templates and EMG criteria differentiated states of arousal. Drug

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 17: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

was administered 6 hrs after lights off (circadian time denoted CT-18) to look for sleep promoting effects

of the drug at 10 or 30 mg/kg, or vehicle. Experiments were run as simultaneous parallel groups, all

doses including vehicle were tested in parallel on the same day in approximately equal sized dose groups

of 10-14 rats. Total NREM sleep and total REM sleep were computed for each animal. The primary

outcomes were the total NREM sleep and REM sleep during the first 12 hr and during the first 18 hr post-

treatment. To assess sleep continuity, the single longest bout of the 6 hourly longest sleep bouts during

the first 6 hr after treatment was calculated, as was the average sleep bout over this period. Each

outcome was analyzed by analysis of covariance using treatment group as the factor and the pre-

treatment period shown in Table 2 as the covariate. Sleep bouts were analyzed on the log scale to

stabilize the variation. Adjusted means and the change from vehicle means and their corresponding

standard errors were summarized for each treatment group. For longest sleep bouts results were

transformed back to the linear scale and represent fold changes over vehicle. Unadjusted and Dunnett’s

multiple-comparison adjusted P values are shown for each outcome in each period.

In vivo Neurochemistry

In-vivo Microdialysis in the rat

Implantation of a BAS guide cannula into the mPFC (A 3.2 mm; L 0.8 mm; and V, -2 mm) was carried out

by Taconic (Germantown, NY) as 5-7 days before the experiment previously described in Fell et al.,

(2010). A concentric type probe (BR-4) from BAS (West Lafayette, IN) to match the implanted cannula

with a 4 mm membrane tip extending below the cannula was flushed with water and carefully inserted

through the cannula 16 hours before the experiment began. Immediately after insertion of the probe the

rat was returned to the home cage. On the morning of the experiment the rat was moved to cage where

the experiment was to be conducted and allowed to acclimate for a period of 4 hours. After the

acclimation period the rat was connected to a fraction collection system for freely moving animals

(BioAnalytical Systems (BASi), West Lafayette, IN). The input tube of the dialysis probe was connected to

a syringe pump (BeeHive and BabyBee, BASi) which delivered an artificial cerebrospinal fluid containing

150 mM NaCl, 3 mM KCl, 1.7 mM CaCl2 and 0.9 mM MgCl2 (pH 6.0) to the probe at a rate of 1 µl/min.

The output tubes from the rats were attached to a refrigerated fraction collector (BASi). After a period of 2

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 18: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

hours for equilibration of the probe and establishment of stable histamine baseline levels, collection of 60

minute fractions was started. The flow from the output lines was collected in 400 µl plastic tubes which

contained 15 µl of an antioxidant solution (3 mM L-cysteine, 10 nM EDTA and 50 nM isoproterenol as an

internal standard, in 0.5 M acetic acid). The antioxidant serves to acidify the dialysate and prevents

degradation of the amines and metabolites during the 24 hour period that it takes to collect the samples

and complete the HPLC assays. Typically the total sample volume was 60 µl. Three baseline samples

were collected before injection of any drugs. At 6pm the lights were turned out in the experimental room

and not turned back on until 6am the next morning. All microdialysis data were calculated as percent

change from dialysate basal concentrations with 100% defined as the average of the three drug pre-

injection values and each group had 5-8 rats.

Histamine was separated on an HPLC column and quantified by fluorimetric detection after a postcolumn

derivatization with an o-phthaldialdehyde-containing reagent as follows. The mobile phase consisted of

0.16 M KH2PO4, 0.1 mM sodium octanesulfonic acid and 0.1 mM EDTA; the pH was adjusted to 4.6. The

flow rate of the mobile phase was 0.7 ml/min. The eluent line was connected by a T-piece for mixing with

a reagent line through which a 0.02% solution of o-phthaldialdehyde (OPA) in 0.15 M NaOH was

delivered at 0.6 ml/min. The OPA reagent was mixed with the eluent in a mixing coil of Teflon tubing (OD

1.1 mm, ID 0.55 mm; length 1 m) which was insulated to allow the derivatization reaction to proceed at

ambient temperature. The OPA reagent solution was prepared fresh daily, protected from light, and kept

cooled in ice. Histamine was separated on a reverse-phase column, a BDS Hypersil, 3 μm, C18 analytical

column (4.6×100 mm from Thermo-Fisher). The fluorescence of the reaction product was measured by a

fluorimeter (Jasco FP-920; excitation: 350 nm; emission: 450 nm) set at the highest sensitivity. The

sensitivity for histamine was about 20 fmol per sample (20 μl). All dialysis probe placements were

checked by perfusing a tetra red solution through the dialysis probe for subsequent histological

examination. Only results derived from rats with correctly positioned probes were included in the data

analysis. Time course data represent the percent change from baseline and were analyzed by two-way

analysis of variance (ANOVA) followed by Post-hoc Bonferroni corrected t-test. The level of significance

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 19: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

was set at p < 0.05. All analyses were performed using the GraphPad PRISM statistical program

(GraphPad, San Diego, CA).

Analysis of tele-methylhistamine (t-MeHA) in rat CSF

Male Harlan-Sprague Dawley rats (200 g) were dosed PO either acutely with THIIC at doses of 1-10

mg/kg. Six hours after dosing the rats were sacrificed with CO2 and CSF samples were collected from the

cisterna magna. Concentrations of histamine and tele-methylhistamine (t-MeHA) were determined using

HPLC separation and detected by tandem mass spectrometry. Cerebrospinal fluid (50-100 microliters)

samples were spiked with stable label internal standards for both analytes. The CSF is then passed

though an anion exchange solid phase extraction cartridge to remove protein and interferences. The

extracts for CSF were injected onto a Varian, Monochrome 100 X 2 mm HPLC column for separation. The

separation is achieved utilizing a mobile phase of 0.5% HFBA and 10% acetic acid/0.5% HFBA in

acetonitrile. A linear gradient is utilized for separation of histamine and its metabolite. Histamine, its tele-

methyl metabolite, and the stable label internal standards were detected by electrospray ionization with

the specific MS/MS transitions. Standard curves were analyzed from 0.150 to 50 ng/ml and unknown

concentrations were calculated using an internal standard method, detection limit for t-MeHA was 0.3

ng/ml. Data are expressed as mean±SEM and were subjected to a one-way analysis of variance

(ANOVA) followed by Post-hoc analyses using Dunnett’s corrected t-test.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 20: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Results

THIIC induces a Selective Allosteric Potentiation in vitro of mGluR2 in Recombinant systems.

Activation of mGluR2 was assessed by measuring glutamate-induced increases in intracellular calcium in

AV12 cells expressing human or rat mGluR2 co-expressed with the G protein (Gqi5). Consistent with the

effects of other allosteric potentiators of mGluR, THIIC positively modulated the mGluR2 receptor as

shown by the parallel leftward shift of a glutamate dose-response curve in the presence of increasing

concentrations of THIIC with no increase in the maximal response to glutamate (figure 2). This leftward

shift of the glutamate dose response represents a positive cooperativity between the allosteric binding

site for THIIC and the orthosteric glutamate binding site. In the presence of an EC10 concentration of

glutamate, the EC50 value for potentiation by THIIC was 22.5 ± 3.2 nM (mean ± SEM, n=7) for the human

mGluR2 receptor and EC50 = 12.8 ± 3.8 nM (mean ± SEM, n=4) for the rat mGluR2 receptor homolog

(data not shown).

The ability of THIIC to shift glutamate-induced [35S]GTPγS binding in membranes expressing human

mGluR2 is shown in Figure 3. Increasing concentrations of THIIC produced an approximately 25-fold

leftward shift in the glutamate-induced [35S]GTPγS in the human mGluR2 (positive cooperativity factor =

23.6, Kb = 824 nM). Interestingly, in the absence of glutamate, THIIC also increased [35S]GTPγS

binding in membranes expressing mGluR2 (EC50 1.5 µM). Agonist activity is also evident in the

increasing baselines and the progressively flattening Hill slopes of the glutamate dose response curves

as the concentration of THIIC is increased. Together, these data indicate allosteric agonist and

potentiation effects of THIIC in this particular assay.

THIIC showed no significant activity across a panel of mGlu receptors (mGluR's 1, 3, 4, 5, 6, 7, 8) and

GABAB Receptors in antagonist, agonist and potentiator modalities (see table 1.). In addition THIIC was

profiled further against a diverse panel of CNS receptors showing no significant radioligand displacement

at concentrations of 1uM. However, THIIC did show signs of radioligand displacement at the following

receptors and concentrations: hNET, (71% at 10 uM and 7% at 1uM, r Cl- channel, 80% at 10 uM and

12% at 1 uM, r Na+ channel, 78% at 10uM and 25% at 1 uM, h5HT2B agonist site 97% at 10 uM and 41%

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 21: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

at 1 uM with a Ki = 3.7 uM,). A follow-up study for agonist activity at the h5-HT2B using GTPγS binding

revealed no significant stimulation of binding at concentrations up to 10 uM.

THIIC engenders antidepressant-like effect in the mouse forced swim test that are dependent

upon mGlu2 receptors. The forced swim (behavioral despair) test in rodents in often used as a screen

assay for drugs with potential antidepressant activity. Orally administered THIIC decreased immobility in

the mouse forced-swim test in a dose-dependent manner after both acute and five day dosing (fig. 4).

However, somewhat greater potency and efficacy was achieved after repeat dosing than after acute

dosing despite the lack of significant changes in the plasma kinetics of THIIC after repeat dosing (data not

shown).

To determine whether mGlu2 receptors are responsible for the antidepressant-like effects of THIIC,

mGlu2 receptor knockout mice were studied. In this study, mGlu2 receptor KO mice showed a small

antidepressant-like phenotype on their own. When dosed acutely with 30 mg/kg THIIC (i.p., 30 min prior),

the antidepressant-like efficacy observed in the wild-type mice was eliminated in the mGlu2 receptor

knockout mice (fig. 4). In contrast, the antidepressant-like effects of imipramine occurred in both mGlu2 -

/- and mGlu2 +/+ mice (data not shown).

THIIC produces antidepressant-like activity in DRL-72s assay. The DRL-72 assay in rats is an

operant responding paradigm that is sensitive to clinically active anti-depressant drugs including SSRI’s

NRI’s and SNRI’s (Marek et al., 1989; Marek et al., 2005). Under this assay, THIIC increased the number

of water reinforcements received, and reduced responses emitted, in a dose-dependent manner (figure

5a). Although doses of 1 & 3 mg/kg were not significantly different from vehicle-treated animals, a dose of

10 mg/kg produced a significant decrease in responses emitted (p < 0.05), while producing an increase in

reinforcers received (though not statistically significant). The results on responses and reinforcers

matched well with the data produced by the positive control, imipramine which also produced a significant

decrease in responses emitted (p < 0.05), while producing a non-significant increase in reinforcers

received. In these animals, the magnitude of effect on responses and reinforcers was comparable. At 10

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 22: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

mg/kg, THIIC also produced a statistically significant increase in efficiency of responding by the animals

(p < 0.05) as with 10 mg/kg of imipramine. Following treatment of 10 mg/kg of THIIC for 5 days, there was

a significant increase in reinforcers received and a significant decrease in responses emitted (fig 5b)

compared to vehicle. These results showed a trend for a larger magnitude of effect in the DRL-72 assay

with THIIC following repeated treatment, compared to acute treatment.

THIIC produces antidepressant-like activity in the dominant-submissive behavior assay. The

dominant-submissive test (DST) is a chronic dosing assay which is sensitive to clinically active anti-

depressant drugs including SSRI’s and SNRI’s. In the DST, the dominance level was significantly lower

than baseline after oral treatment with THIIC at 3 mg/kg and 30 mg/kg (p < 0.05) with statistical significant

differences at week two and three (p < 0.05 in all cases). In contrast, 10 mg/kg of the tricyclic

antidepressant imipramine required 3 weeks of dosing for statistical significant alleviation of submissive

behavior (p < 0.05; figure 6).

Effects of THIIC on Sleep EEG in the rat. As shown in figure 7a, THIIC (10 and 30 mg/kg, PO) dosed

at CT18 (Circadian Time 18 = 18 hours after lights on), i.e during the dark, active phase produced a

significant increase in NREM sleep at 30mg/kg over the first 12hrs post dose, and an increase at both 10

and 30mg/kg over the first 18hrs, eliciting an extra 36.0 ± 10.9 min and 57.0 ± 10.9 min, respectively,

relative to vehicle control. A significant increase in NREM sleep of 30.8 ± 7.7 min was also seen after 10

mg/kg, PO after dosing at CT-5 (5 hours after lights on, i.e. during the light inactive phase (data not

shown). A significant decrease in REM sleep was also observed over the same time course with

30mg/kg THIIC resulting in a reduction of 11.2± 2.7 min over the first 12hrs and 15.1±3.6 min over the

first 18hrs compared to vehicle (figure 7b). Analysis of sleep bouts over the first 6 hrs after dosing

resulted in a dose dependent increase, more than doubling at 30mg/kg, for both average sleep bout

length (2.2 ± 0.3 fold) and longest sleep bout (2.1 ± 0.3 fold, (figure 7c), indicating the ability of the

compound to consolidate sleep at 10 and 30 mg/kg, PO. A lower dose of 3.0 mg/kg, PO did not produce

statistically significant changes in sleep parameters (data not shown).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 23: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

THIIC suppresses marble-burying in a genotype-dependent manner. Marble-burying behavior in the

mouse can detect both GABA and serotonin-based anxiolytic agents (Li et al., 2006). THIIC produced

dose-dependent decreases in marble-burying as did the anxiolytic agent chlordiazepoxide (fig. 8). THIIC

did not affect rotorod performance of mice whereas chlordiazepoxide did (fig. 8). The effect of THIIC was

eliminated in mGlu2 receptor null mice (fig. 8).

THIIC inhibits Stress-induced hyperthermia in rats. Hyperthermia is a general phenomenon that has

been reliably demonstrated in many species in response to stress, and is a component of the well-

characterized fight-or-flight response. Stress-induced hyperthermia is attenuated by clinical anxiolytics

and is widely used preclinically to predict anxiolytic efficacy of novel compounds (Spooren et al., 2002).

As is shown in figure 8, THIIC (1 – 30 mg/kg) produced a dose-dependent reduction in stress-induced

hyperthermia [F(5,53) = 7.06, p < 0.001], with significant reductions compared to vehicle observed at 3,

10 and 30 mg/kg (P < 0.05 in all cases). The effect of THIIC was comparable to the response of the

mGlu5 antagonist MTEP (10 mg/kg) . THIIC did not affect baseline core body temperature (data not

shown).

THIIC prevents stress-induced increases in cerebellar cGMP. Another animal model that is often used

to assess the potential anxiolytic/anti-stress effects of novel compounds is reversal of stress-induced

levels of cGMP in the cerebellum. As shown in figure 9, exposure to a brief foot-shock resulted in a

significant increase in cGMP (~80%) compared to the vehicle treated group which received no foot-shock

[F(5,41) = 6.227; p < 0.001]. This stress-induced increase in cGMP was completely reversed by THIIC at

the highest dose tested of 30 mg/kg (p < 0.05) although lower doses of 3 and 10 mg/kg were not

significantly different from the vehicle + foot-shock treated group. In the same experiment, alprazolam (1

mg/kg) produced a similar antagonism of the stress effect (p < 0.05).

THIIC decreases the dark phase increase in HA efflux in the mPFC and reduces t-MeHA levels in

rat CSF after acute and 5 day dosing. As is shown in figure 10, THIIC 10 and 30 mg/kg significantly

attenuated the dark phase increase in mPFC histamine efflux as measured by in vivo microdialysis. A

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 24: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

two-way ANOVA revealed a significant effect of drug [F(2, 208) = 34.52; P < 0.001], time [F(15, 208) =

10.73; p < 0.001] and a significant drug x time interaction [F(30, 208) = 16.11; P < 0.05]. THIIC showed a

dose-dependent inhibition of t-MeHA in the CSF with approximately 50% inhibition after 3 (p < 0.05) and

10 mg/kg (p < 0.05; figure 11a). After 5 days repeated dosing, levels of t-MeHA were completely

suppressed and were below the level of detection after both the 3 and 10 mg/kg, PO doses (figure 11b).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 25: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Discussion

The current report details the pharmacological properties of a structurally novel positive allosteric

modulator of mGlu2 receptors, THIIC. In rat and human recombinant systems, THIIC is a potent

potentiator of the response of mGlu2 receptors to glutamate and is highly selective for mGlu2 receptors

having no direct effect on the glutamate response of other mGlu receptors, ionotropic glutamate

receptors, glutamate transporters or receptors implicated in the actions of clinically effective psychiatric

medications including dopaminergic, serotonergic, muscarinic, adrenergic, GABAergic and histaminergic.

The selectivity of this molecule for mGlu2 receptors thus enables an evaluation of the biological

consequences of augmenting mGlu2 receptor function in vivo. THIIC showed robust activity in behavioral

models used to predict anxiolytic- and antidepressant-like activity. Importantly, in several assays, the

effects of THIIC were negated in mice without mGlu2 receptors providing in vivo support for the molecular

target of these biological impacts from THIIC. Encouragingly, these data also provide evidence that doses

of THIIC with anxiolytic/antidepressant-like activity also produced significant electrophysiological and

neurochemical changes including changes on sleep EEG parameters (NREM and REM sleep), and

decreased histamine release in the prefrontal cortex and t-MeHA in the CSF which may represent

potential translational biomarkers for this drug target.

THIIC demonstrated efficacy in three animal models used to predict antidepressant-like activity. Such

antidepressant activity of orthosteric mGlu2/3 receptor agonists have not been reported although

modification of antidepressant effects have been shown by one group (Matrisciano et al., 2008; see

Witkin et al., 2007 for an overview). Systemically administered THIIC dose dependently decreased

immobility time in the mouse forced swim test (an effect lost in mice deficient of mGlu2 receptors) and

was also efficacious in the rat DRL-72 assay, an operant responding paradigm that is sensitive to

clinically active anti-depressant drugs including SSRI’s, NRI’s and SNRI’s. In both assays the degree of

efficacy with THIIC was similar to that of the clinically efficacious tricyclic antidepressant imipramine.

Moreover, the antidepressant-like effects of THIIC were retained after 5 days of dosing suggesting that

there is a low potential for tolerance with this mechanism. These findings are further supported by our

finding that THIIC was efficacious in the rat dominant-submissive test, a chronic assay in which THIIC

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 26: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

was administered for a three-week treatment period. Although neurogenesis has been suggested to occur

with repeat dosing of antidepressants (c.f., Witkin et al., 2007), we did not study this phenomenon here.

Together, these data suggests that THIIC might produce antidepressant-like efficacy in humans.

To the best of our knowledge these are the first data showing that the potentiation of mGlu2 receptor

activity and a subsequent reduction in glutamatergic tone has efficacy in preclinical models that detect

acute or subchronic dosing with antidepressants in vivo. However, increasing glutamate tone also

produces antidepressant-like effects in a host of rodent models (Witkin et al., 2007). The seeming

disparity of mechanisms producing a common behavioral outcome is potentially puzzling. Resolution to

such potential mechanistic convergence could come from several lines of argument. First, mGlu2

receptor potentiation engages mGlu2 in a physiological-relevant manner whereas the orthosteric

antagonists do not. Secondly, mGlu2 potentiators influence mGlu2 in exclusion of mGlu3. Thirdly, only

one mGlu2 potentiator was exemplified here and might not be representative of a class effect. Fourthly, in

contrast to THIIC, mGlu2/3 receptor antagonists do not generally produce anxiolytic-like effects (Witkin

and Eiler, 2006), mGlu2/3 antagonists do not display antipsychotic activity as reported for mGlu2

potentiators (Galici et al., 2005), and mGlu2/3 antagonists are not active in some antidepressant-

detecting assays that showed positive results with THIIC and vis versa (Witkin et al., 2007). Importantly,

major depressive disorder is a composed of a heterogeneous set of symptoms and symptom clusters

which likely have distinct implications for therapeutics (Witkin et al., 2007).

Consistent with previous reports with mGlu2/3 receptor agonists and other mGlu2 receptor potentiators,

THIIC produced anxiolytic-like effects in pharmacological models predicative of anxiolytic efficacy. THIIC

produced a dose-related reduction in the stress-induced increase in cerebellar cGMP and reduced the

number of marbles buried in the mouse marble burying assay, an effect which was lost in mGlu2 receptor

deficient mice. THIIC also prevented the development of stress-induced hypothermia in rats and again

the magnitude of the observed decrease in body temperature was similar to effects observed previously

with structurally unrelated allosteric potentiators of mGlu2 receptor (CBiPES, BINA) and mGlu2/3 receptor

agonists (LY354740) (Galici et al., 2005; Johnson et al., 2005; Rorick-Kehn et al., 2006).

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 27: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

THIIC was also characterized in a series of in vivo and ex vivo neurochemical experiments in order to

shed further light on the mechanism of action of mGlu2 receptor potentiation and to identify potential

neurochemical markers of behavioral efficacy. Although not reported here, we did not observe any effects

of THIIC on extracellular (or brain tissue) monoamine levels (dopamine, serotonin and norepinephrine) or

their metabolites. Based on the finding that THIIC has significant sleep promoting effects and that we

recently observed significant interactions for CBiPES with the histamine system (Fell et al., 2010), we

assessed the effects of THIIC on extracellular histamine levels. THIIC clearly attenuated the increase in

extracellular histamine in the mPFC which occurs during the dark phase, suggesting that potentiation of

mGlu2 receptor activity can modulate central histaminergic neurotransmission in the brain. Although this

mechanism is not fully understood, previous studies have shown a stimulatory action of glutamate on

histaminergic neuronal activity (Okakura et al., 1992). Thus the ability of mGlu2 receptors to dampen

presynaptic glutamate release may account for the effect of THIIC on HA efflux in the mPFC. Further

studies are currently ongoing in our laboratory in order to elucidate this mechanism in more detail.

Nonetheless, that THIIC reduced HA efflux is intriguing and may contribute to the behavioral effects of

THIIC. Neuronal histamine is known to play a key modulatory role in a number of CNS functions including

arousal and has also been implicated in stress-related disorders such as anxiety (Frisch et al., 1998;

Zarrindast et al., 2005). Further, the ability to modulate histamergic neurotransmission is a feature shared

by clinically effective anxiolytic drugs, diazepam and buspirone (Oishi et al., 1986; Oishi et al., 1992).

Additionally, the relationship between extracellular histamine levels and sleep-wake activity is well

established, i.e histamine H3 antagonists which increase histamine release are wake-promoting in several

species including humans (Witkin and Nelson, 2004).

In recent years, an increasing emphasis has been placed on the alignment of preclinical and clinical

research in drug discovery in order to facilitate the development of novel therapeutic agents. Indeed, a

central aim of the present study was to identify potential biomarkers which could facilitate the translational

development of THIIC. This drive for a biomarker combined with the finding that THIIC modulates

histaminergic neurotransmission led us to evaluate the effects of THIIC on histamine and its metabolite t-

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 28: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

MeHA in the CSF of rats. The analysis of CSF neurotransmitter levels is a relatively underexploited but

potentially translatable assay which can be performed in both animals and human subjects. Moreover,

since CSF is in direct contact with the CNS, brain processes are more likely to be reflected in the CSF

than other in other body fluids. Indeed, CSF histamine levels in rats have been shown to reflect central

histamine neurotransmission (Soya et al., 2008). Consistent with the in-vivo microdialysis data, we

observed a significant reduction in CSF t-MeHA levels after acute and repeated dosing with THIIC which

likely reflects the effect of THIIC on central histaminergic activity. Although further studies are needed in

order to evaluate the role of mGlu2 receptors in the regulation of histaminergic activity, these findings

suggest that the analysis of the histamine metabolite t-MeHA in the CSF could represent a viable

translational biomarker approach for the clinical development of mGlu2 potentiators.

Another preclinical assay which has the potential to translate to the clinical setting is sleep EEG.

Abnormalities of REM sleep (shortening of REM latency, lengthening of the duration of the first REM

period) are frequently observed in patients with major depressive disorder and have been considered

specific of depressive disease (Feige et al., 2002; Chalon et al., 2005). In the present study, THIIC

decreased REM sleep and increased NREM sleep. These effects on REM sleep are consistent with a

previous report showing that the mGlu2 receptor potentiator BINA similarly inhibited REM sleep and

increased its onset latency (Ahnaou et al., 2009). Moreover, these changes are also similar to the effects

of many clinically effective antidepressants which are known to produce changes in sleep pattern

including reduced REM sleep and increased REM sleep onset latency (Mayers and Baldwin, 2005;

Thase, 2006).

In summary, convergent data from the present series of experiments demonstrates that allosteric

potentiation of the mGlu2 receptor with THIIC suggests a novel and potentially promising target for the

treatment of anxiety and/or depression. Our findings also provide evidence that sleep EEG and CSF t-

MeHA may represent viable translational biomarker approaches to the clinical development of mGlu2

potentiators.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 29: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Authorship Contributions

Participated in research design: Fell, Witkin, Perry, Rorick-Kehn, Rasmussen, Benvenga, Leuke, Edgar,

Wafford, Seidel, Quets, Felder, Heinz, Kuo, Ebert, Eckstein, Ackermann, Swanson, Catlow, Dean,

Jackson, Tauscher-Wisniewski, Marek, Schkeryantz, Svensson.

Conducted experiments: Fell, Falcone, Katner, Hart, Overshiner, Chaney, Li, Marlow, Thompson,

Wafford, Quets, Wang, Nikolayev, Eckstein, Ebert.

Contributed new reagents or analytic tools: Mayhugh, Khilevich, Zhang, Schkeryantz

Performed data analysis: Fell, Witkin, Falcone, Rorick-Kehn, Rasmussen, Benvenga, Li, Marlow,

Thompson, Seidel, Wafford, Wang, Heinz, Quets.

Wrote or contributed to the writing of the manuscript: Fell, Witkin, Rorick-Kehn, Rasmussen, Benvenga,

Wafford, Quets, Heinz, Schkeryantz, Svensson.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 30: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

References

Ahnaou A, Dautzenberg FM, Geys H, Imogai H, Gibelin A, Moechars D, Steckler T and Drinkenburg WH

(2009) Modulation of group II metabotropic glutamate receptor (mGlu2) elicits common changes

in rat and mice sleep-wake architecture. Eur J Pharmacol 603:62-72.

Bruno V, Battaglia G, Copani A, D'Onofrio M, Di Iorio P, De Blasi A, Melchiorri D, Flor PJ and Nicoletti F

(2001) Metabotropic glutamate receptor subtypes as targets for neuroprotective drugs. J Cereb

Blood Flow Metab 21:1013-1033.

Cartmell J, Perry KW, Salhoff CR, Monn JA and Schoepp DD (2001) Acute increases in monoamine

release in the rat prefrontal cortex by the mGlu2/3 agonist LY379268 are similar in profile to

risperidone, not locally mediated, and can be elicited in the presence of uptake blockade.

Neuropharmacology 40:847-855.

Chalon S, Pereira A, Lainey E, Vandenhende F, Watkin JG, Staner L and Granier LA (2005) Comparative

effects of duloxetine and desipramine on sleep EEG in healthy subjects. Psychopharmacology

(Berl) 177:357-365.

Conn PJ and Pin JP (1997) Pharmacology and functions of metabotropic glutamate receptors. Annu Rev

Pharmacol Toxicol 37:205-237.

Di Giorgi-Gerevini V, Melchiorri D, Battaglia G, Ricci-Vitiani L, Ciceroni C, Busceti CL, Biagioni F, Iacovelli

L, Canudas AM, Parati E, De Maria R and Nicoletti F (2005) Endogenous activation of

metabotropic glutamate receptors supports the proliferation and survival of neural progenitor

cells. Cell Death Differ 12:1124-1133.

Dunayevich E, Erickson J, Levine L, Landbloom R, Schoepp DD and Tollefson GD (2008) Efficacy and

tolerability of an mGlu2/3 agonist in the treatment of generalized anxiety disorder.

Neuropsychopharmacology 33:1603-1610.

Feige B, Voderholzer U, Riemann D, Dittmann R, Hohagen F and Berger M (2002) Fluoxetine and sleep

EEG: effects of a single dose, subchronic treatment, and discontinuation in healthy subjects.

Neuropsychopharmacology 26:246-258.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 31: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Fell MJ, Johnson BG, Svensson KA and Schoepp DD (2008) Evidence for the role of mGlu2 not mGlu3

receptors in the pre-clinical antipsychotic pharmacology of the mGlu2/3 receptor agonist

LY404039. J Pharmacol Exp Ther 326:209-231

Fell MJ, Katner JS, Johnson BG, Khilevich A, Schkeryantz JM, Perry KW and Svensson KA (2010)

Activation of metabotropic glutamate (mGlu)2 receptors suppresses histamine release in limbic

brain regions following acute ketamine challenge. Neuropharmacology 58:632-639.

Frisch C, Hasenohrl RU, Krauth J and Huston JP (1998) Anxiolytic-like behavior after lesion of the

tuberomammillary nucleus E2-region. Exp Brain Res 119:260-264.

Galici R, Echemendia NG, Rodriguez AL and Conn PJ (2005) A selective allosteric potentiator of

metabotropic glutamate (mGlu) 2 receptors has effects similar to an orthosteric mGlu2/3 receptor

agonist in mouse models predictive of antipsychotic activity. J Pharmacol Exp Ther 315:1181-

1187.

Grillon C, Cordova J, Levine LR and Morgan CA, 3rd (2003) Anxiolytic effects of a novel group II

metabotropic glutamate receptor agonist (LY354740) in the fear-potentiated startle paradigm in

humans. Psychopharmacology (Berl) 168:446-454.

Johnson MP, Barda D, Britton TC, Emkey R, Hornback WJ, Jagdmann GE, McKinzie DL, Nisenbaum ES,

Tizzano JP and Schoepp DD (2005) Metabotropic glutamate 2 receptor potentiators: receptor

modulation, frequency-dependent synaptic activity, and efficacy in preclinical anxiety and

psychosis model(s). Psychopharmacology (Berl) 179:271-283.

Khilevich A, Liu B, Mayhugh DR, Schkeryantz JM and Zhang D (2010) Imidazole carboxamides.

International Patent Application 2010/WO2010009062 A1, in.

Li X, Morrow D and Witkin JM (2006) Decreases in nestlet shredding of mice by serotonin uptake

inhibitors: comparison with marble burying. Life Sci 78:1933-1939.

Marek GJ, Li AA and Seiden LS (1989) Evidence for involvement of 5-hydroxytryptamine1 receptors in

antidepressant-like drug effects on differential-reinforcement-of-low-rate 72-second behavior. J

Pharmacol Exp Ther 250:60-71.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 32: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Marek GJ, Martin-Ruiz R, Abo A and Artigas F (2005) The selective 5-HT2A receptor antagonist

M100907 enhances antidepressant-like behavioral effects of the SSRI fluoxetine.

Neuropsychopharmacology 30:2205-2215.

Matrisciano F, Zusso M, Panaccione I, Turriziani B, Caruso A, Iacovelli L, Noviello L, Togna G, Melchiorri

D, Debetto P, Tatarelli R, Battaglia G, Nicoletti F, Giusti P and Girardi P (2008) Synergism

between fluoxetine and the mGlu2/3 receptor agonist, LY379268, in an in vitro model for

antidepressant drug-induced neurogenesis. Neuropharmacology 54:428-437.

Mayers AG and Baldwin DS (2005) Antidepressants and their effect on sleep. Hum Psychopharmacol

20:533-559.

Oishi R, Itoh Y and Saeki K (1992) Inhibition of histamine turnover by 8-OH-DPAT, buspirone and 5-

hydroxytryptophan in the mouse and rat brain. Naunyn Schmiedebergs Arch Pharmacol 345:495-

499.

Oishi R, Nishibori M, Itoh Y and Saeki K (1986) Diazepam-induced decrease in histamine turnover in

mouse brain. Eur J Pharmacol 124:337-342.

Okakura K, Yamatodani A, Mochizuki T, Horii A and Wada H (1992) Glutamatergic regulation of

histamine release from rat hypothalamus. Eur J Pharmacol 213:189-192.

Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM,

Gurovich IY, Morozova MA, Mosolov SN, Neznanov NG, Reznik AM, Smulevich AB, Tochilov VA,

Johnson BG, Monn JA and Schoepp DD (2007) Activation of mGlu2/3 receptors as a new

approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nat Med 13:1102-1107.

Porsolt RD, Bertin A and Jalfre M (1977) Behavioral despair in mice: a primary screening test for

antidepressants. Arch Int Pharmacodyn Ther 229:327-336.

Rorick-Kehn LM, Johnson BG, Burkey JL, Wright RA, Calligaro DO, Marek GJ, Nisenbaum ES, Catlow

JT, Kingston AE, Giera DD, Herin MF, Monn JA, McKinzie DL and Schoepp DD (2007a)

Pharmacological and Pharmacokinetic Properties of a Structurally Novel, Potent, and Selective

Metabotropic Glutamate 2/3 Receptor Agonist: In Vitro Characterization of Agonist (-)-

(1R,4S,5S,6S)-4-Amino-2-sulfonylbicyclo[3.1.0]-hexane-4,6-dicarboxylic Acid (LY404039). J

Pharmacol Exp Ther 321:308-317.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 33: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Rorick-Kehn LM, Johnson BG, Knitowski KM, Salhoff CR, Witkin JM, Perry KW, Griffey KI, Tizzano JP,

Monn JA, McKinzie DL and Schoepp DD (2007b) In vivo pharmacological characterization of the

structurally novel, potent, selective mGlu2/3 receptor agonist LY404039 in animal models of

psychiatric disorders. Psychopharmacology (Berl) 193:121-136.

Rorick-Kehn LM, Perkins EJ, Knitowski KM, Hart JC, Johnson BG, Schoepp DD and McKinzie DL (2006)

Improved bioavailability of the mGlu2/3 receptor agonist LY354740 using a prodrug strategy: in

vivo pharmacology of LY544344. J Pharmacol Exp Ther 316:905-913.

Schoepp DD (2001) Unveiling the functions of presynaptic metabotropic glutamate receptors in the

central nervous system. J Pharmacol Exp Ther 299:12-20.

Schoepp DD and Marek GJ (2002) Preclinical pharmacology of mGlu2/3 receptor agonists: novel agents

for schizophrenia? Curr Drug Targets CNS Neurol Disord 1:215-225.

Schoepp DD, Wright RA, Levine LR, Gaydos B and Potter WZ (2003) LY354740, an mGlu2/3 receptor

agonist as a novel approach to treat anxiety/stress. Stress 6:189-197.

Soya A, Song YH, Kodama T, Honda Y, Fujiki N and Nishino S (2008) CSF histamine levels in rats reflect

the central histamine neurotransmission. Neurosci Lett 430:224-229.

Spooren WP, Schoeffter P, Gasparini F, Kuhn R and Gentsch C (2002) Pharmacological and

endocrinological characterisation of stress-induced hyperthermia in singly housed mice using

classical and candidate anxiolytics (LY314582, MPEP and NKP608). Eur J Pharmacol 435:161-

170.

Swanson CJ, Bures M, Johnson MP, Linden AM, Monn JA and Schoepp DD (2005) Metabotropic

glutamate receptors as novel targets for anxiety and stress disorders. Nat Rev Drug Discov

4:131-144.

Thase ME (2006) Depression and sleep: pathophysiology and treatment. Dialogues Clin Neurosci 8:217-

226.

Witkin JM and Eiler WJA, II. (2006) Antagonism of metabotropic glutamate group II receptors in the

potential treatment of neurological and neuropsychiatric disorders. Drug Development Research

67:757-769.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 34: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Witkin JM, Marek GJ, Johnson BG and Schoepp DD (2007) Metabotropic glutamate receptors in the

control of mood disorders. CNS Neurol Disord Drug Targets 6:87-100.

Witkin JM and Nelson DL (2004) Selective histamine H3 receptor antagonists for treatment of cognitive

deficiencies and other disorders of the central nervous system. Pharmacol Ther 103:1-20.

Wright RA, Arnold MB, Wheeler WJ, Ornstein PL and Schoepp DD (2001) [3H]LY341495 binding to

group II metabotropic glutamate receptors in rat brain. J Pharmacol Exp Ther 298:453-460.

Zarrindast MR, Moghadam AH, Rostami P and Roohbakhsh A (2005) The effects of histaminergic agents

in the central amygdala of rats in the elevated plus-maze test of anxiety. Behav Pharmacol

16:643-649.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 35: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Footnotes

These studies were funded by Eli Lilly and Company, USA. All authors are or were employees of Eli Lilly

and Company and own stock in Eli Lilly and Company. GJM current address: Abbott Laboratories, GPRD,

R48B, AP04-01, 100 Abbott Park Road, Abbott Park, IL 60064-6075 USA.

Reprint requests:

Dr. Matthew J. Fell, Neuroscience Discovery Research, Lilly Research Laboratories, Lilly Corporate

Center, DC0510, Indianapolis, IN 46285, USA; Tel: 317-433-1096; Fax: 317-276-7600;

Email: [email protected]

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 36: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Figure Legends

Figure 1. Chemical structure of THIIC

Figure 2. Concentration-dependent potentiation of glutamate-stimulated mobilization of intracellular Ca2++

by THIIC using the FLIPR assay in clonal AV12 cells stably expressing the human mGluR2 receptor (a)

and typical FLIPR traces showing minimal impact from agonist activity of compound alone (b). The effects

of different concentrations of THIIC were measured in the presence of increasing concentrations of the

agonist glutamate. EC50 values were calculated using a 4-parameter logistic curve-fitting program.

Figure 3. Concentration-dependent stimulation of GTPγ35S binding by THIIC in clonal AV12 cells stably

expressing the human mGlu2 receptor.

Figure 4. Top Panel: Comparison of acute (open bars) and 5-day repeated dosing (filled bars) of THIIC

administered orally 60 minutes prior to testing on immobility in the mouse forced-swim test. Bars

represent mean + SEM of 8 mice/group: Imipramine (15 mg/kg) was dosed i.p. 30 min prior (n=4/group).

Data were analyzed by a one-way ANOVA followed by Dunnett’s test. *p < 0.05 compared to vehicle

treated group. Bottom Panel: Comparison of effects of THIIC (30 mg/kg, i.p., 30 min) in the forced-swim

test in wild-type (WT) and mGlu2 receptor knockout (KO) mice. * p < 0.05 compared to the WT vehicle

control mice.

Figure 5. The effect of (a) acute and (b) 5-days repeated dosing of THIIC on responses (●) and

reinforcers (○) on a DRL-72 schedule. Each point represents the mean (± SEM) of 10 rats at each dose

tested. Imipramine (10 mg/kg, i.p.) results are also shown. Data were analyzed by a one-way ANOVA

followed by Dunnett’s test. *p < 0.05 compared to vehicle treated group.

Figure 6. Dose-dependent decrease in difference in dominance level (i.e. decrease in difference in

feeding time) between the dominant and submissive rat after treatment with THIIC 3 and 30 mg/kg, PO

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 37: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

daily dosing and imipramine (10 mg/kg/day). Bars represent mean + SEM in 8-13 rat pairs. * p<0.05

compared to baseline with Tukey’s post-hoc test.

Figure 7. Effect of THIIC on (a) NREM sleep and (b) REM sleep and (c) sleep bout length in rats. (a)

Dose-dependent increase in NREM sleep over the first 12 hr and first 18 hr period after dosing PO at 10

and 30mg/kg. (b) Dose dependent decrease in REM sleep over the first 12 hr and first 18 hr period after

dosing PO at 10 and 30mg/kg. (c) Dose dependent increase in longest sleep bout length over the first 6

hrs after dosing at 10 and 30mg/kg. Bars represent mean ± SEM of 10-13 rats. NREM and REM sleep

values are cumulative minutes over the stated period. The longest sleep bout was analyzed on the log

scale and back-transformed to the linear scale to stabilize the variation. Each outcome was analyzed by

analysis of covariance using treatment group as the factor and the pre-treatment period as the covariate.

*p<0.01 compared to vehicle group, **p<0.0001 compared to vehicle group.

Figure 8. THIIC suppresses marble-burying of mice that is dependent upon mGlu2 receptors.

Chlordiazepoxide HCl was studied as an anxiolytic comparator. Each point represents effects in 8

mice/dose group with 12-16 mice/group in the vehicle control animals. Significant effects by ANOVA

were followed by Dunnett’s post-hoc test (** p < 0.01 vs. control). The inset shows the effects of 30

mg/kg THIIC and 30 mg/kg chlordiazepoxide on rotorod performance. * p < 0.05 by Fishers exact

probability test.

Figure 9. Dose-dependent inhibition of stress-induced hyperthermia in Fischer F-344 rats after oral

administration of THIIC. Data represent means and SEM for 9-10 rats/group. Data were analyzed by a

one-way ANOVA [F(5,53) = 7.06, p < 0.001] followed by Dunnett’s test *p < 0.05 compared to the vehicle

treated group. % values in each bar represent the percent inhibition relative to the vehicle control group.

Figure 10. Inhibition of stress-induced increase in cerebellar cGMP in male CF-1 mice after IP

administration of THIIC. Data represent means and SEM for 7-10 mice/group and were analyzed by a

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 38: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

one-way ANOVA followed by Dunnett’s test. # P< 0.05 compared to the vehicle-no stress group. * p <

0.05; **p < 0.01 compared to the vehicle + stress group.

Figure 11. Effect of THIIC on the dark phase increase in histamine efflux in the mPFC. THIIC (10 and 30

mg/kg) was dosed 60 minutes before the onset of the dark phase (6 pm and time 0 on graph). Data are

the mean + SEM of the dialysate levels, expressed as a percentage of the baseline and were analyzed by

a two-way ANOVA followed by a Post-hoc Bonferroni corrected t-test. N = 5-8 per group *p < 0.05

compared to the vehicle treated group.

Figure 12. Effects of THIIC on CSF levels of tele-methylhistamine. Animals were administered THIIC (1-

10 mg/kg PO) and CSF samples were collected 6 hours after dosing. Data were analyzed by a one-way

ANOVA followed by Dunnett’s test. N = 4-8 per group. *p < 0.05; **p < 0.01 compared to the vehicle

treated group.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 39: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

JPET #172957

Table 1. Summary of Potency Values (EC50 or IC50 in nM) for THIIC tested in a Panel of Human

Cloned mGlu and GABAB Receptors using the Calcium Mobilization Assay.

Mode mGlu1 mGlu2 mGlu3 mGlu4 mGlu5 mGlu7 mGlu8 GABAB

Agonist >25,000 >25,000 >25,000 >25,000 >25,000 >25,000 >25,000 >25,000

Potentiator >12,500 22.5 ± 3.2 (N=7)

>12,500 >12,500 >12,500 NT >12,500 >12,500

Antagonist >12,500 >12,500 >12,500 >12,500 >12,500 NT* >12,500 >12,500

Overall mean IC50 or EC50 + SEM as shown. NT = not tested. *Single point inhibition =58%.

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 40: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 41: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 42: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 43: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 44: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 45: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 46: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 47: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 48: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 49: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 50: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from

Page 51: N-(4-((2-(trifluoromethyl)-3-hydroxy-4- (isobutyryl ...jpet.aspetjournals.org/content/jpet/early/2010/10/... · 10/14/2010  · Dr. Matthew J. Fell, Neuroscience Discovery Research,

This article has not been copyedited and formatted. The final version may differ from this version.JPET Fast Forward. Published on October 14, 2010 as DOI: 10.1124/jpet.110.172957

at ASPE

T Journals on M

arch 19, 2021jpet.aspetjournals.org

Dow

nloaded from