16
GENETIC DELETION OF MT 1 /MT 2 MELATONIN RECEPTORS ENHANCES MURINE COGNITIVE AND MOTOR PERFORMANCE G. O’NEAL-MOFFITT, * J. PILLI, S. S. KUMAR AND J. OLCESE Florida State University Program in Neuroscience, College of Medicine, Department of Biomedical Sciences, 1115 West Call Street, Tallahassee, FL 32306, USA Abstract—Melatonin, an indoleamine hormone secreted into circulation at night primarily by the brain’s pineal gland, has been shown to have a wide variety of actions on the devel- opment and physiology of neurons in the CNS. Acting via two G-protein-coupled membrane receptors (MT 1 and MT 2 ), melatonin modulates neurogenesis, synaptic functions, neuronal cytoskeleton and gene expression. In the present studies, we sought to characterize the behavior and neuro- nal biology of transgenic mice lacking both of these melato- nin receptors as a way to understand the hormone’s receptor versus non-receptor-mediated actions in CNS- dependent activities, such as learning and memory, anxiety, general motor performance and circadian rhythmicity. Assessment of these behaviors was complemented by molecular analyses of gene expression in the brain. Our results demonstrate mild behavioral hyperactivity and a lengthened circadian period of free-running motor activity in melatonin receptor-deficient mice as compared to recep- tor-intact control mice beginning at an early age. Significant improvement in cognitive performance was found using the Barnes Maze and the Y-Maze. No behavioral changes in anxiety levels were found. Electrophysiological measures in hippocampal slices revealed a clear enhancement of long-term potentiation in mice lacking melatonin receptors with no significant differences in paired-pulse facilitation. Quantitative analysis of brain protein expression levels of phosphoCREB and phosphoERK1/2 and key markers of synaptic activity (synapsin, glutamate receptor 1, spinophi- lin, and glutamic acid decarboxylase 1) revealed significant differences between the double-knockout and wild-type ani- mals, consistent with the behavioral findings. Thus, genetic deletion of melatonin receptors produces mice with enhanced cognitive and motor performance, supporting the view that these receptors play an important role in neurobehavioral development. Ó 2014 Published by Elsevier Ltd. on behalf of IBRO. Key words: melatonin, learning, memory, activity, LTP, mice. INTRODUCTION Knockout mice lacking both the MT 1 and the MT 2 (encoded by the MTNR1a and MTNR1b genes respectively) melatonin receptors have been used in a variety of studies to examine the mechanisms of melatonin action in specific disorders and conditions, including focal cerebral ischemia (Kilic et al., 2012), methamphetamine-induced locomotor sensitization (Hutchinson et al., 2012), sleep–wake characterization (Comai et al., 2013), nicotine sensitivity (Mexal et al., 2012), and blood glucose regulation (Mu¨hlbauer et al., 2009); however, no comprehensive characterization of baseline differences between these transgenic animals and their wild-type counterparts has been published. Sim- ilarly, aspects of neurodevelopment have not been addressed before, despite the fact that these animals’ brains develop not only without the receptor-mediated influence of melatonin, but they also lack the impact of the normally constituently active MT 1 receptor (Dubocovich et al., 2010). The genetic absence of specific hormone receptors can result in a variety of neurobiological disorders, e.g., oxytocin receptors and social behavior (cf. Donaldson and Young, 2008), vasoactive intestinal peptide receptors and circadian behavior (Harmar et al., 2002), androgen receptors and reproductive systems (Chang et al., 2013). Functionally significant polymorphisms in the cod- ing sequences of both the human MT 1 and the human MT 2 melatonin receptors have been identified and found to be associated with some diseases and disorders (Li et al., 2013; Comai and Gobbi, 2014). A few of the http://dx.doi.org/10.1016/j.neuroscience.2014.07.018 0306-4522/Ó 2014 Published by Elsevier Ltd. on behalf of IBRO. * Corresponding author. Address: 1115 West Call Street, Florida State University, College of Medicine, Tallahassee, FL 32306, USA. Tel: +1-850-645-1479. E-mail addresses: gmoffi[email protected] (G. O’Neal-Moffitt), jyotsna. [email protected] (J. Pilli), [email protected] (S. S. Kumar), [email protected] (J. Olcese). Abbreviations: aCSF, artificial cerebrospinal fluid; ADHD, attention deficit/hyperactivity disorder; ANOVA, analysis of variance; C3B6, mice with a combination C3H/He and C57BL/6 background; cDNA, complementary deoxyribonucleic acid; CAT, catalase; CREB, cAMP response element-binding protein; DBL-KO, double-knockout; DD, dark–dark or constant dark; DNA, deoxyribonucleic acid; EDTA, ethylenediaminetetraacetic acid; EPM, elevated plus maze; EPSP, excitatory post-synaptic potential; ERK, extracellular signal-regulated kinases; GAD1, glutamic acid decarboxylase 1; GluR1, glutamate receptor subunit 1; GPx-1, glutathione peroxidase 1; HEPES, 2-[4-(2- hydroxyethyl)piperazin-1-yl]ethanesulfonic acid; HPC, hippocampus; LD, light–dark; LTP, long-term potentiation; MAPKs, mitogen-activated protein kinases; mRNA, messenger ribonucleic acid; MTNR, melatonin receptor; MT 1 , melatonin receptor type 1; MT 2 , melatonin receptor type 2; MTNR1a, gene encoding the MT 1 receptor; MTNR1b, gene encoding the MT 2 receptor; NIR, near-infrared; Nrf2, nuclear factor erythroid 2-related factor 2; NT, non-transgenic; PGK, phosphoglycerate kinase promoter; PCR, polymerase chain reaction; qPCR, quantitative polymerase chain reaction; SCN, suprachiasmatic nuclei; SOD1, superoxide dismutase 1; SDS–PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis. Neuroscience 277 (2014) 506–521 506

Genetic deletion of MT₁/MT₂ melatonin receptors enhances murine cognitive and motor performance

  • Upload
    fsu

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Neuroscience 277 (2014) 506–521

GENETIC DELETION OF MT1/MT2 MELATONIN RECEPTORSENHANCES MURINE COGNITIVE AND MOTOR PERFORMANCE

G. O’NEAL-MOFFITT, * J. PILLI, S. S. KUMAR ANDJ. OLCESE

Florida State University Program in Neuroscience, College of

Medicine, Department of Biomedical Sciences, 1115 West

Call Street, Tallahassee, FL 32306, USA

Abstract—Melatonin, an indoleamine hormone secreted into

circulation at night primarily by the brain’s pineal gland, has

been shown to have a wide variety of actions on the devel-

opment and physiology of neurons in the CNS. Acting via

two G-protein-coupled membrane receptors (MT1 and MT2),

melatonin modulates neurogenesis, synaptic functions,

neuronal cytoskeleton and gene expression. In the present

studies, we sought to characterize the behavior and neuro-

nal biology of transgenic mice lacking both of these melato-

nin receptors as a way to understand the hormone’s

receptor versus non-receptor-mediated actions in CNS-

dependent activities, such as learning and memory, anxiety,

general motor performance and circadian rhythmicity.

Assessment of these behaviors was complemented by

molecular analyses of gene expression in the brain. Our

results demonstrate mild behavioral hyperactivity and a

lengthened circadian period of free-running motor activity

in melatonin receptor-deficient mice as compared to recep-

tor-intact control mice beginning at an early age. Significant

improvement in cognitive performance was found using the

Barnes Maze and the Y-Maze. No behavioral changes in

anxiety levels were found. Electrophysiological measures

in hippocampal slices revealed a clear enhancement of

http://dx.doi.org/10.1016/j.neuroscience.2014.07.0180306-4522/� 2014 Published by Elsevier Ltd. on behalf of IBRO.

*Corresponding author. Address: 1115 West Call Street, Florida StateUniversity, College of Medicine, Tallahassee, FL 32306, USA. Tel:+1-850-645-1479.

E-mail addresses: [email protected] (G. O’Neal-Moffitt), [email protected] (J. Pilli), [email protected] (S. S. Kumar),[email protected] (J. Olcese).Abbreviations: aCSF, artificial cerebrospinal fluid; ADHD, attentiondeficit/hyperactivity disorder; ANOVA, analysis of variance; C3B6, micewith a combination C3H/He and C57BL/6 background; cDNA,complementary deoxyribonucleic acid; CAT, catalase; CREB, cAMPresponse element-binding protein; DBL-KO, double-knockout; DD,dark–dark or constant dark; DNA, deoxyribonucleic acid; EDTA,ethylenediaminetetraacetic acid; EPM, elevated plus maze; EPSP,excitatory post-synaptic potential; ERK, extracellular signal-regulatedkinases; GAD1, glutamic acid decarboxylase 1; GluR1, glutamatereceptor subunit 1; GPx-1, glutathione peroxidase 1; HEPES, 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid; HPC, hippocampus;LD, light–dark; LTP, long-term potentiation; MAPKs, mitogen-activatedprotein kinases; mRNA, messenger ribonucleic acid; MTNR, melatoninreceptor; MT1, melatonin receptor type 1; MT2, melatonin receptor type2; MTNR1a, gene encoding the MT1 receptor; MTNR1b, geneencoding the MT2 receptor; NIR, near-infrared; Nrf2, nuclear factorerythroid 2-related factor 2; NT, non-transgenic; PGK,phosphoglycerate kinase promoter; PCR, polymerase chain reaction;qPCR, quantitative polymerase chain reaction; SCN, suprachiasmaticnuclei; SOD1, superoxide dismutase 1; SDS–PAGE, sodium dodecylsulfate–polyacrylamide gel electrophoresis.

506

long-term potentiation in mice lacking melatonin receptors

with no significant differences in paired-pulse facilitation.

Quantitative analysis of brain protein expression levels of

phosphoCREB and phosphoERK1/2 and key markers of

synaptic activity (synapsin, glutamate receptor 1, spinophi-

lin, and glutamic acid decarboxylase 1) revealed significant

differences between the double-knockout and wild-type ani-

mals, consistent with the behavioral findings. Thus, genetic

deletion of melatonin receptors produces mice with

enhanced cognitive and motor performance, supporting

the view that these receptors play an important role in

neurobehavioral development. � 2014 Published by Elsevier

Ltd. on behalf of IBRO.

Key words: melatonin, learning, memory, activity, LTP, mice.

INTRODUCTION

Knockout mice lacking both the MT1 and the MT2

(encoded by the MTNR1a and MTNR1b genes

respectively) melatonin receptors have been used in a

variety of studies to examine the mechanisms of

melatonin action in specific disorders and conditions,

including focal cerebral ischemia (Kilic et al., 2012),

methamphetamine-induced locomotor sensitization

(Hutchinson et al., 2012), sleep–wake characterization

(Comai et al., 2013), nicotine sensitivity (Mexal et al.,

2012), and blood glucose regulation (Muhlbauer et al.,

2009); however, no comprehensive characterization of

baseline differences between these transgenic animals

and their wild-type counterparts has been published. Sim-

ilarly, aspects of neurodevelopment have not been

addressed before, despite the fact that these animals’

brains develop not only without the receptor-mediated

influence of melatonin, but they also lack the impact of

the normally constituently active MT1 receptor

(Dubocovich et al., 2010).

The genetic absence of specific hormone receptors

can result in a variety of neurobiological disorders, e.g.,

oxytocin receptors and social behavior (cf. Donaldson

and Young, 2008), vasoactive intestinal peptide receptors

and circadian behavior (Harmar et al., 2002), androgen

receptors and reproductive systems (Chang et al.,

2013). Functionally significant polymorphisms in the cod-

ing sequences of both the human MT1 and the human

MT2 melatonin receptors have been identified and found

to be associated with some diseases and disorders (Li

et al., 2013; Comai and Gobbi, 2014). A few of the

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 507

reported receptor polymorphism associated diseases

include, but are not limited to, breast cancer (MT1 and

MT2; Deming et al., 2012), type 2 diabetes mellitus

(MT2; Bonnefond et al., 2012) and multiple sclerosis

(MT2; Natarajan et al., 2012). As little is known about

the consequences of complete melatonin membrane

receptor deletion on brain function and behavior, our

primary objective was to identify any behavioral, neuro-

biological or molecular differences in melatonin receptor-

deficient mice as compared to mice expressing both

receptors. Our findings point to a significant role for mel-

atonin receptor signaling in brain development and in

the regulation of behavioral activity through actions on

major intracellular signal transduction pathways and on

both excitatory and inhibitory synaptic communication.

EXPERIMENTAL PROCEDURES

Ethics statement

All mice were housed and handled in accordance with

Federal animal welfare guidelines and in compliance

with the Public Health Service Policy on Humane Care

and Use of Laboratory Animals (2002) and the Guide for

the Care and Use of Laboratory Animals (8th Edition).

Experiments were reviewed and approved prior to being

carried out by the Institutional Animal Use and Care

Committee (IACUC) of the Florida State University

(Protocols #1016, 1135; Association for Assessment

and Accreditation of Laboratory Animal Care

International accreditation unit #001031; Office of

Laboratory Animal Welfare Assurance #A3854-01).

Animals

Mice in this study were progeny of a University of

Massachusetts Medical School mouse colony, where

the melatonin receptor knockout strains were generated

by a genetic mutation introduced by homologous

recombination (Liu et al., 1997; Jin et al., 2003). Sites

within exon 1 of both the melatonin type 1 receptor gene

(MT1) and the melatonin type 2 receptor gene (MT2) were

replaced with phosphoglycerate kinase promoter (PGK-

neomycin) cassettes. In each case, the clones carrying

the targeted allele were injected into C57BL/6 mouse

blastocytes. The resulting chimeric animals were bred to

C57BL/6 animals to obtain offspring that were heterozy-

gous for the knockout allele. These animals were then

backcrossed for 10 generations to C3H/He mice because

others have used the C3H strain extensively in examining

circadian behavioral responses to melatonin. The C3H

strain also has rhythmic melatonin production (Ebihara

et al., 1986) unlike most other inbred strains of mice,

including the C57BL/6 (Roseboom et al., 1998;

Kasahara et al., 2010). The resultant C3B6 (C3H/

He + C57BL/6) melatonin type 1 receptor-deficient

homozygous mice (MT1�/�) were crossed with C3B6

melatonin type 2 receptor-deficient mice (MT2�/�) to obtain

MT1�/�/MT2

�/� double-mutant mice, hereafter referred to

as double-knockout (DBL-KO) mice, and their non-trans-

genic (NT) wild-type counterparts, hereafter referred to

as NT mice.

Multiple cohorts of 6–10 male mice were randomly

selected from breeders of the DBL-KO and NT lines for

multiple aspects of this study: circadian, behavioral, a

melatonin challenge, or serum collection. Some animals

were sacrificed at specific ages for tissue analysis (e.g.,

long-term potentiation (LTP) at 6 months of age), while

other cohorts were maintained for behavioral testing at

3, 6, 12, and 15 months of age. Prior to experimentation

at each age, animal weights in grams were 24.2 ± 2.4,

29.8 ± 3.8, 33.6 ± 4.9, 32.9 ± 3.5 respectively, and

there were no significant differences between genotype

groups at any age. The animals were housed

individually in a polycarbonate cage (Ancare, Bellmore,

NY; 19 cm � 29 cm � 13 cm) with hardwood laboratory

bedding chips (Nepco Beta Chip�, Warrensburg, NY),

nesting material (Ancare Nestlet), and a polycarbonate

mouse igloo (Bio-Serv, Flemington, NJ) for enrichment.

They were maintained under a 12-h light–dark cycle

(7 am to 7 pm) at 21.1 �C and given ad libitum access to

LabDiet� 5001 Rodent Chow and water. Daily water

intake was averaged from bottle weight measurements

every 5–7 days for 4 weeks.

Genotyping by polymerase chain reaction (PCR)

At approximately 21 days of age, an IACUC and

veterinarian recommended inhalation anesthetic,

isoflurane (Butler Schein, Dublin, OH; 029405) was used

to sedate the animal. Once anesthetized, 2-mm terminal

segment of tail was removed with a sterile scissors.

Hemostasis was achieved using a silver nitrate applicator

stick (Butler Schein; 005383) and potential pain and

discomfort were alleviated by applying bupivacaine

hydrochloride in sterile isotonic solution (Sigma, St.

Louis, MO; B5274; 2.5 lg/mL) locally to the excision site

for long-acting pain management. Post-surgical excision

site monitoring occurred for 10 days. The tail biopsy was

placed in 250 lL of DirectPCR Tail lysis buffer (Viagen

101-T) with 10 lL Proteinase K solution (Viagen 501-K)

and lysed for 6 h at 55 �C. The sample was then

incubated at 85 �C for 45 min and briefly centrifuged. The

supernatant containing the genetic material was

collected and stored at �20 �C for subsequent PCR

analysis. Primers used to amplify the MT1 receptors

included mMT1R-FW-WT (50-GAAGTTTTCTCAGTGTC

CCGCAATGG-30), mMT1R-REV-WT (50-GAGTCCAAGT

TGCTGGGCAGTGGA-30), and mMT1R-NEO-FW-KO

(50-CCAGCTCATTCCTCCACTCATGATCTA-30). The

genomic DNA (deoxyribonucleic acid) was subjected to a

3-min hot start at 94 �C and then 35 cycles of 45 s at

94 �C, 45 s at 60 �C, and then 3 min at 72 �C, with a final

stage of 7 min at 72 �C. Primers used to detect the MT2

receptor alleles included a common forward primer

(mMT2R-FW-WT 50-CCAGGCCCCCTGTGACTGCCCG

GG-30), a gene-specific reverse primer from intron 1

(mMT2R-REV-WT 50-CCTGCCACTGAGGACAGAACAG

GG-30), and a reverse primer based on the PGK-neo

cassette (mMT2R-NEO-REV-KO 50-TGCCCCAAAGGC

CTACCCGCTTCC-30). These genomic DNA samples

were subjected to a 3-min hot start at 94 �C, 35 cycles of

30 s at 94 �C, 30 s at 68 �C, and 1 min at 72 �C, with a

final 7-min stage at 72 �C. The resulting samples were

508 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

loaded on to a 1.5% agarose gel (EMDMillipore OmniPur�

2120-OP) for 25 min of electrophoresis at 110 V and

subsequently imaged on a Bio-Rad Gel Doc� XR (Fig. 1).

Melatonin determination

To validate that both the DBL-KO and NT mice used in

this study were endogenously synthesizing melatonin in

a rhythmic fashion, blood serum was collected for

analysis by radioimmunoassay. Collection occurred

during the night under red light conditions at 7 pm,

10 pm, 1 am, and 4 am and during the day in white light

at 7 am and 1 pm. Three littermates of each genotype

were assigned to a single time point. Mice were

anesthetized with isoflurane and blood was collected via

a puncture to the vascular bed at the back of the jaw

where the orbital veins, the submandibular veins, and

the other veins draining the facial area coalesce to form

the jugular vein. The exuding droplets of blood (�0.3–0.4 mL) were collected into an EDTA tube, after which,

a sterile gauze compress was applied to the puncture

site for 20 s. This rapid blood collection method is more

humane than retro-orbital or cardiac puncture, leaving

the animals unaffected with no signs of distress (Golde

et al. 2005). Whole blood was centrifuged at 2000g for

3 min to separate the serum, which was sent frozen in

light-protective vials to a commercial diagnostics com-

pany (Pharmasan, Inc. Osceola, WI, USA) for melatonin

radioimmunoassay.

Behavioral testing

DBL-KO and NT mice were tested in either six-week

circadian running wheel study or in a behavioral battery,

which included Open Field activity, elevated plus maze

(EPM), Rota-rod, Y-Maze, Platform Recognition, and

Barnes Maze. For all behavioral testing, testers were

blinded to the animal identification number and their

respective genotypes and were only allowed to know

the artificial 5-digit alphanumeric scheme created to

identify the animals. Behavioral tests were performed

either during the 12-h light phase in white light or during

the 12-h dark phase, corresponding to the active phase

for mice, under red light or white light conditions.

Fig. 1. PCR analysis of mouse genomic DNA from wild-type, heterozygous,

PCR using a cocktail of three primers: mMT1R-REV-WT (primer A), mMT1R

distinct bands representing the wild-type and disrupted alleles. The wild-type

allele (primers A–C) produces a 240-bp product. (MT2) Genomic DNA PCR

(primer B), and mMT2R-NEO-REV-KO (primer C) to produce distinct bands

(primers A–B) produces a 272-bp product, while the disrupted allele (primer

Light-phase testing occurred during the hours of 8 am to

noon. Dark-phase testing occurred during the hours of

7 pm to midnight, and the rodents were shielded from all

non-red light sources during transport to and from the

testing chamber during the dark phase. For any

behavioral tests conducted both during the active and

inactive phases at a single age, the same mice were

used for testing in both the phases. Video recording

and/or computer monitoring was utilized to provide

accurate analysis of results at a later date. Testing was

done in male mice at multiple ages in independent

cohorts (3mo n= 3–5; 6mo n= 5–6; 12mo n= 10–11;

15mo n= 4–6).

Circadian behavior. For assessment of circadianrhythmicity, 6-month-old mice of both genotypes were

singly housed in cages equipped with running wheels.

All mice were entrained in a cycle of 12-h light and 12-h

dark (LD) for at least 1 week before they were released

into constant darkness (DD). After 2 weeks in DD mice

were returned to LD for 1 week before another exposure

to DD. Circadian period duration, or tau, was

determined by using a best-fit regression line followed

by a chi-squared (v2) periodogram analysis function in

Clocklab (Actimetrics, Evanston, IL, USA). Tau was

calculated from activity onset during the final DD

episode. Time to re-entrain was determined by number

of days required to obtain a 24-h rhythm in LD

conditions after the first DD exposure.

Anxiety behavior. To assess anxiety, animals were

evaluated using a near-infrared (NIR) backlit Elevatedplus maze (EPM; Med Associates, Inc., St. Albans, VT,

USA) consisting of an elevated plus-shaped maze with

two opposite open arms (50 cm � 10 cm) and two

opposite closed arms (50 cm � 10 cm with 40-cm walls).

The tests were conducted at night from 7:30 pm to

10 pm under red lighting, with NIR backlight for animal

tracking. The task was initiated by placing the test

subject into the center of the maze facing an open arm.

Activity was monitored via a mounted overhead camera

and video tracking system for a 5-min period. Entries

and homozygous mutant mice. (MT1) Genomic DNA was subjected to

-FW-WT (primer B), and mMT1R-NEO-FW-KO (primer C) to produce

allele (primers A–B) produces a 480-bp product, while the disrupted

with three primers: mMT2R-FW-WT (primer A), mMT2R-REV-WT

representing the wild-type and disrupted alleles. The wild-type allele

s A–C) produces a 550-bp product.

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 509

into each arm, and the time spent in open arms, closed

arms, and center of maze were evaluated.

Sensorimotor/locomotor behavior. An accelerating

Rota-rod treadmill (Med Associates, Inc., St. Albans,

VT, USA) was used to assess motor and coordination

deficits. In this test, the mouse was placed on a rotating

horizontal rod, which was 3.2 cm in diameter. The test

was conducted at night from 8 pm to 10:30 pm under

red light conditions. In each trial, the animal was placed

on the rod and allowed to acclimate while the rod

rotated at a constant 4 rotations per minute (rpm).

Subsequently, the speed was accelerated from 4 to

40 rpm. Latency in seconds was recorded when either

the mouse dropped to the platform (16.5 cm) or until

5 min elapsed.

Platform Recognition is a swimming-based

sensorimotor task often used to measure the animal’s

ability to visually identify/recognize a variably placed

elevated platform, and its ability to approach and

ascend on to the surface of the platform. Specifically,

this test was utilized as a means of determining if any

animal had visual deficits that could impair their ability to

perform in later cognitive tests, as mice on a combined

C3H background can have retinal degeneration. The

test was conducted at night from 8 pm to 11 pm under

red lighting in a pool that measured 88 cm in diameter

and 21 cm deep and was separated into four equal

quadrants by lines suspended above the pool. The

temperature of the water was held at 26 �C. A 9 cm

circular platform with a prominent black ensign attached

was raised 0.8 cm above the surface of the water. The

platform had a textured surface to aid the mouse in

climbing onto the surface and to encourage the mouse

to stay on the platform. A 61-cm high circular barrier

was placed around the pool to lessen the escape of the

mice. The mice were given four, 60-s maximum trials

per day for 4 days. On each day, the first two trials were

consecutive and were separated by a 30-min gap before

the next two consecutive trials were performed. The

mice began each trial facing the wall of the pool in

the same quadrant for each trial for that particular day.

The platform was placed in a different quadrant at the

start of each day and the mice were started from a

different quadrant than the previous day. For each trial,

the escape latency was recorded in seconds, concluding

when the mouse obtained the platform. A 30-s stay on

the platform between consecutive trials was

encouraged. A full 60 s was recorded for any animal

that did not obtain the platform and the mouse was

gently guided toward the platform and encouraged to

remain on the platform for 30 s. During the 30-min gap

between consecutive trials and after daily testing, the

mice were washed and placed under warming lamps to

dry. For statistical analysis, escape latencies were

averaged for the first two trials and the second two trials

per day.

An Open Field activity test was used to measure

exploratory behavior and general activity. The mice

were individually placed into an open field box (45 cm

long � 43.9 cm wide � 30 cm high). The area was

divided into a 4 � 4 grid (16 squares) with each square

measuring 11.8 cm � 11.2 cm. Testing was conducted

during both the active dark phase (7 pm–10 pm) and the

inactive light phase (10 am–12 pm). Mice were given

one 4-min trial in which they were free to move around

the box. Activity was scored as the number of line

crossings by the mouse during the trial. A line crossing

was defined as all four limbs entering into a new square.

The Y-Maze was used also to measure general

activity. The maze consisted of three equal arms (47 cm

long � 16 cm wide � 30.1 cm high) with an equilateral

19-cm triangular center compartment. The center could

be blocked off from all three arms by the insertion of

sliding panels. Visual cues were placed around the

room as well as on the maze arms. Each mouse was

given one minute in the blocked-off center compartment

to acclimate to the maze. At the end of one minute, the

panels blocking entry into the three arms were removed

to allow the mice full access to the maze for 8 min. The

number of entries into each arm and the sequence of

entries were recorded. An entry was defined as all four

limbs entering an arm. Testing was conducted during

both the active dark phase (7:30 pm–11:30 pm) and the

inactive light phase (8 am–11 am). Total number of

entries was used to determine activity.

Cognitive behavior. The Barnes Maze (Med

Associates, Inc., St. Albans, VT, USA) is a type of

delayed match-to-place experiment used to assess

spatial/reference learning and memory. Visual objects

arranged around the maze served as spatial cues and

testing occurred from 7:30 pm to 10 pm. The circular

platform was 121.8 cm in diameter and 140.2 cm high

with 40 equally spaced holes on the periphery of the

platform. A dark box filled with bedding was positioned

under one of the holes on the platform to allow the

mouse to escape from aversive stimuli. Each mouse

was assigned a specific unique escape hole for the

duration of the experiment. For each trial, the mouse

was placed in the center of the platform facing away

from its target escape hole. The aversive stimuli

included one high-intensity fan blowing at the level of

the platform and two 120-watt flood lamps hung from

the ceiling near to and aimed at the platform. Each

mouse was given one 600-s trial per day for 4 days and

the escape latency in seconds was recorded for each

trial. A full 600 s was recorded for any animal that did

not find its target escape hole during a trial.

The number of alternating entries in the Y-Maze was

also used to assess basic memory function (refer to

protocol above). For this, the number of entries that

were different from the two preceding entries was

divided by the total number of entries. This normalizes

the results to account for differences in speed through

the maze during the 8-min trials.

LTP

Hippocampal slices (350-lm thick) were cut from brains

of C3B6 mice at 6–7 months of age in a chilled (4 �C)low-Ca2+, low-Na+ ‘‘cutting solution’’ containing the

following (in mM): 230 sucrose, 10 D-glucose, 26

510 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

NaHCO3, 2.5 KCl, 1.25 NaH2PO4, 10 MgSO4, and 0.5

CaCl2 (equilibrated with 95% O2/5% CO2). Slices were

allowed to equilibrate in oxygenated artificial

cerebrospinal fluid (aCSF; in mM: 126 NaCl, 26

NaHCO3, 3 KCl, 1.25 NaH2PO4, 2 MgSO4, 2 CaCl2, and

10 D-glucose, pH 7.4), first at 32 �C for 1 h and

subsequently at room temperature before being

transferred to the recording chamber. Recordings were

obtained at 32 ± 1 �C from neurons of the CA1 region,

visualized through a 63� objective under Infra-red

Differential Interference Contrast (IR-DIC) optics.

Recording electrodes (1.2–2-lm-tip diameters; 3–6 MX)

contained the following (in mM): for current-clamp

experiments, 105 potassium gluconate, 30 KCl, 10

HEPES, 10 phosphocreatine, 4 MgATP, 0.3 GTP, and

20 biocytin (adjusted to pH 7.3 with KOH). Slices were

maintained in oxygenated (95% O2/5% CO2) aCSF.

Stimulating electrodes placed on the Schaffer collaterals

(S: bipolar CE-2C75, FHC) delivered constant current

pulses 50 ls in duration and 100–500 lA in amplitude

at low frequencies (0.1–0.3 Hz) to activate the Schaffer

collaterals. Tetanus was applied in bursts consisting of

5–8 stimuli at 100 Hz, burst interval ranged from 250 ms

to 5 s. Excitatory postsynaptic responses (EPSPs) were

recorded with an Axon� MultiClamp� 700B amplifier and

pClamp� software (Molecular Devices), filtered at

1–2 kHz (10 kHz for current-clamp), digitized at

10–20 kHz, and stored digitally.

Tissue collection for protein and transcript analysis

All mice were euthanized between the hours of 8am and

11am by sterile intraperitoneal overdose injection of

ketamine (Butler Schein; 100 mg/kg) and xylazine

(Vedco, St. Joseph, MO; 10 mg/kg), followed by

transcardial 0.9% sterile saline perfusion. Brains were

quickly removed and placed on an ice-cold plate for

dissection. The brain was initially bisected along the

sagittal plane and the left hemisphere was dissected for

the hippocampus (HPC) and frontal cortex. The

dissected samples were then flash frozen for

subsequent protein analysis. The right hemisphere was

similarly dissected and preserved in QIAGEN RNAlater�

RNA stabilization reagent for later mRNA (messenger

ribonucleic acid) analysis by quantitative polymerase

chain reaction (qPCR).

Isolation of protein from brain tissue andimmunoblotting

Hippocampi or frontal cortices (�10 mg per sample) were

disrupted and homogenized in 300 lL standard HEPES

protein extraction buffer containing Thermo Scientific

Halt� phosphatase inhibitor cocktail and Roche

Complete protease inhibitor cocktail. Samples were

centrifuged at 10,000g for 10 min at 4 �C. The

supernatant containing the solubilized proteins was

transferred to a new tube for a second spin. The final

supernatant was analyzed on a Thermo Scientific

Nanodrop photometer to determine protein

concentration. 2� SDS (sodium dodecyl sulfate) dye

was added in an equal volume and samples were

stored at �80 �C for later immunoblotting. SDS–PAGE

(sodium dodecyl sulfate–polyacrylamide gel

electrophoresis) immunoblotting was performed and

proteins were detected via the LI-COR Odyssey�

infrared imaging system with secondary antibodies

sensitive to 685- and 785-nm excitation wavelengths. All

primary antibodies were rabbit polyclonals from Cell

Signaling Technologies, with the exception of goat anti-

b-actin (Abcam, Cambridge, MA), rabbit anti-GluR1

(Abcam) and rabbit anti-spinophilin, courtesy of

Dr. Charles Ouimet (Allen et al., 1997). Levels of b-actinremained consistent for both DBL-KO and NT samples.

Isolation of mouse brain mRNA and real-time qPCR

Total cellular RNA was extracted from homogenized

frontal cortices with QIAShredder� columns in

QIAGEN’s buffer RLT and b-mercaptoethanol in a 100:1

ratio followed by processing with the QIAGEN� RNeasy

Kit� and DNase Set treatment kits according to the

manufacturer’s protocols. The RNA concentration was

measured with a Thermo Scientific Nanodrop

photometer. For analysis of transcript levels, one lg of

total RNA was reverse-transcribed to cDNA

(complementary deoxyribonucleic acid) by means of the

Bio-rad iScript� reverse-transcription system.

Amplification of the cDNA was performed on a Bio-Rad

CFX96 Touch� Real-Time PCR Detection System using

the iQ SYBR� Green Supermix protocol according to

the manufacturer’s specifications. The following thermal

cycling parameters were used: initial heat activation of

the DNA-polymerase was performed at 95 �C for 5 min.

Thereafter, 40 cycles of 95 �C (10 s), 57–60.5 �C (10 s)

and 72 �C (30 s) were run. The primer sequences and

annealing temperatures for the gene transcripts that

were analyzed are listed in Table 1.

Statistics

All calculations, comparisons and statistical analysis were

performed using GraphPad Prism version 6.0a software

for Mac OS X (GraphPad Software, La Jolla, California,

USA, www.graphpad.com). The values shown on the

graphs represent the means ± S.E.M. from independent

experiments. Molecular or behavioral differences in

genotype for a single test conducted only at night or

only in the day for a specific age were computed with

an independent (between subjects) Student’s t-test.Behavioral tests comparing differences at night versus

differences during the day were conducted with the

same animals and computed with a 2-way analysis of

variance (ANOVA) followed by Bonferroni’s multiple

comparison of means. Tests conducted with multiple

trials or time points were analyzed with a 2-way ANOVA

for repeated measures (mixed-design) and followed by

the Bonferroni test for post hoc comparisons. For

behavioral tests conducted longitudinally with different

cohorts (3mo, 6mo, 12mo, 15mo), Student’s t-test wasused to evaluate each age independently. Statistical

differences for the LTP data were measured with the

t-test (paired where indicated). Statistical values

reaching p 6 0.05 were considered significant.

Table 1. Gene-specific primers used for qPCR experiments

Gene Gene-specific primers 50–30 GenBank# Amplicon size Annealing

SOD1 S: ATG GCG ATG AAA GCG GTG TG NM_011434 460 59 �CAS: GCG CAA TCC CAA TCA CTC CA

GPx1 S: ATG TGT GCT GCT CGG CTC TC NM_008160 590 60 �CAS: TGC TGG GAC AGC AGG GTT TC

CAT S: AGG TTT GGC CTC ACA AGG AC NM_009804 239 58 �CAS: GCG GTA GGG ACA GTT CAC AG

Nrf2 S: AAC GAC AGA AAC CTC CAT CTA C NM_010902 94 57 �CAS: AGT AAG GCT TTC CAT CCT CAT C

Rp27 S: CCA GGA TAA GGA AGG AAT TCC TCC TG NM_024277 297 59 �CAS: CCA GCA CCA CAT TCA TCA GAA GG

MT1R S: GGA TAT GGG TCC TGG TCC TT NM_008639.2 215 58.5 �CAS: ACT AGC CAC GAA CAG CCA CT

MT2R S: ACA TCA CAG CCA TTG CCA TCA ACC NM_145712.2 186 60.5 �CAS: AGG TGC AGG AAT AGA TGC GTG GAT

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 511

RESULTS

Genotyping

Confirmation of genotypes for the MT1�/�/MT2

�/� DBL-KO

mice and their NT wild-type counterparts was performed

on tail snips of each mouse at approximately 21 days of

age (Fig. 1). Subsequently, RNA-Sequencing was

performed on the mouse frontal cortex and hippocampal

samples per previously published protocols (Mortazavi

et al., 2008). Sequencing was performed on an Illumina

HiSeq 2500 and confirmed that no melatonin receptor

expression was present in the DBL-KO mice, while both

MT1 and MT2 expression were clearly evident in NT sam-

ples. The RNA-Sequencing data were confirmed by

qPCR for both MT1 and MT2.

To further confirm a phenotype difference between the

two groups, we utilized a melatonin or vehicle challenge.

As part of a parallel study, we administered either

melatonin (100 lg/mL; Sigma M5250) or vehicle (0.1%

ethyl alcohol; Electron Microscopy Sciences 15058) for

10 months in the drinking water of both NT and DBL-KO

mice (n= 4–6). No differences were seen in health,

weight, or appearance post-treatment. For tissues

collected at 12 months, expression levels of superoxide

dismutase 1 (SOD1), glutathione peroxidase 1 (GPx-1),

and catalase (CAT) were determined in the frontal cortex

by quantitative PCR. Additionally, we measured the

transcript levels of nuclear factor erythroid 2-related

factor 2 (Nrf2), a transcription activator that binds to

antioxidant response elements in many genes involved

in cellular responses to oxidative stress. Fig. 2 shows

that, in all cases, the strong melatonin-receptor mediated

effects of melatonin treatment versus vehicle (0.1%

ethanol) on antioxidant signaling which are present in

wild-type mice are absent in melatonin receptor-deficient

mice. All evaluations: a 2-way ANOVA using between-

group experimental design; genotype � treatment;

n= 4–6 for all groups. Fig. 2A SOD1: genotype:

F(1,15) = 16.07, p= 0.0011; treatment: F(1,15) = 8.91,

p= 0.0093; interaction: F(1,15) = 19.50, p= 0.0005;

Bonferroni post hoc analysis revealed a difference

between Mel and EtOH treatment only in animals with

their receptors (p= 0.0001). Fig. 2B GPx-1: genotype:

F(1,17) = 1.644, p= 0.2170; treatment: F(1,17) = 7.507,

p= 0.0140; interaction: F(1,17) = 10.77, p= 0.0044;

Bonferroni post hoc analysis revealed a difference

between Mel and EtOH treatment only in animals with

their receptors (p= 0.0013). Fig. 2C CAT: genotype:

F(1,16) = 0.08874, p= 0.7696; treatment: F(1,16) =

5.412, p= 0.0335; interaction: F(1,16) = 3.190, p=

0.0931; Bonferroni post hoc analysis revealed a

difference between Mel and EtOH treatment only in

animals with their receptors (p= 0.0273). Fig. 2D Nrf2:

genotype: F(1,18) = 18.75, p= 0.0004; treatment:

F(1,18) = 14.00, p= 0.0015; interaction: F(1,18) = 3.948,

p= 0.0624; Bonferroni post hoc analysis revealed a

difference between Mel and EtOH treatment only in

animals with their receptors (p= 0.0022).

Serum melatonin determination by commercial RIA

Commercial radioimmunoassay of blood serum from both

DBL-KO and NT mice (n= 3 per genotype per time point)

confirmed that endogenous melatonin was rhythmic,

ranging from 10 pg/mL in the light to 100 pg/mL in the

dark (a 2-way mixed-design ANOVA for repeated

measures; time: F(5,20) = 4.940, p= 0.0042) and

present in similar levels in both genotypes at each time

point (genotype F(1,4) = 0.00046, p= 0.9839; Fig. 3).

The peak value for melatonin was at Zeitgeber time 15,

corresponding to 3 h after lights off. Lowest melatonin

levels were recorded between Zeitgeber time 6 and 12,

or 6–12 h after lights came on as reported by others

(Vivien-Roels et al. 1998; Kennaway et al. 2002).

Behavior

Circadian behavior. Six-month male NT and DBL-KO

mice were assessed for free-running periods under

constant dark conditions (DD) and for their ability to

entrain to LD cycles (Fig. 4). The free-running period

(tau) for NT mice was determined to be 23.25 h as

compared to 23.73 h for the DBL-KO mice (NT n= 4

and DBL-KO n= 5; Student’s t-test; p= 0.028).

Re-entrainment to LD for DBL-KO mice was

Fig. 2. qPCR analysis after melatonin or vehicle challenge in the frontal cortex of 12-month NT and DBL-KO mice. Four key elements of the

antioxidant pathway respond to melatonin administration (MEL; shaded) vs. vehicle (EtOH; unshaded) in mice expressing the melatonin receptors

(NT; blue) but not in mice lacking the receptors (DBL-KO; red). The results show RP27 normalized mRNA expression levels of (A) superoxide

dismutase (SOD1), (B) glutathione peroxidase (GPx-1), (C) catalase and (D) nuclear factor erythroid 2-related factor 2 (Nrf2) in NT vs DBL-KO mice

(a 2-way ANOVA, p 6 0.05 for all comparisons; error bars represent SEM; n= 4–6 for all groups). (For interpretation of the references to color in

this figure legend, the reader is referred to the web version of this article.)

512 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

1.80 ± 0.84 days compared to 3.75 ± 2.22 days for NT

mice (Student’s t-test; p= 0.109). Light onset for re-

entrainment was initiated during the subjective day for

both the DBL-KO mice and NT mice to compensate for

differences in phase after 2 weeks in DD.

Anxiety behavior. Assessment of anxiety/emotionality

in rodents is commonly made using the EPM, which

incorporates both open and closed arms, the mice

generally preferring the closed arms. Thus, the greater

the time spent in the open arms is considered a

measure of decreased anxiety. As seen in Fig. 5, mice

tested at 6, 12 and 15 months of age showed no

significant differences (p> 0.05), despite a clear trend

to less anxiety (more time spent in the open arms) in

the DBL-KO mice at 6 months of age. Striped bars

indicate nighttime testing. (Student’s t-test; 6mo: NT

n= 6 and DBL-KO n= 5, p= 0.1818; 12mo

NT n= 11 and DBL-KO n= 10, p= 0.7311; 15mo NT

n= 6 and DBL-KO n= 4, p= 0.1725.) There were

also no significant group differences seen in the ratio of

open arm entries to closed arm entries.

Sensorimotor behavior. No evidence of sensorimotor

deficits was seen on the Platform Recognition or Rota-

rod tests at any of the ages tested (Fig. 6). There were no

statistical differences between groups, trials or the

interaction in the Platform Recognition test and no

significant differences (p> 0.05) on Rota-rod

performance between the two genotypes at any age. It is

worth noting that these mice clearly had no visual deficits,

despite their C3H background, most likely because they

were out crossed for multiple generations to the C57BL/6

line. A single mouse with acute ophthalmological

problems was excluded from final analysis. Fig. 6A

Platform Recognition: a 2-way mixed-design ANOVA for

repeated measures; genotype: F(1,8) = 1.393,

p= 0.2718; trial: F(3,24) = 1.822, p= 0.1701;

interaction: F(3,24) = 0.7244, p= 0.5473; 6mo NT n= 6

and DBL-KO n= 5. Fig. 6B Rota-rod: Student’s t-test;6mo: NT n= 6 and DBL-KO n= 5, p= 0.6356; 12mo

NT n= 10 and DBL-KO n= 10, p= 0.2396; 15mo NT

n= 6 and DBL-KO n= 4, p= 0.1506.)

Increased locomotor activity in DBL-KO mice. Assess-

ment of locomotor activity beginning at 3 months of age

Serum Melatonin (RIA)

0 6 12 15 18 210

20

40

60

80

100

120

ZT (Hours)

Mel

aton

in (p

g/m

L)

NTDBL-KO

Fig. 3. Radioimmunoassay (RIA) for serum melatonin in 6-month NT

and DBL-KO mice. Commercial radioimmunoassay of blood serum

from both DBL-KO (red) and NT (blue) mice confirmed that endo-

genous melatonin was rhythmic and present in similar levels (n= 3

mice per genotype per time point). Zeitgeber time 0 corresponds to

light onset (a 2-way RM ANOVA, p > 0.05). (For interpretation of the

references to color in this figure legend, the reader is referred to the

web version of this article.)

Elevated Plus Maze

6 mo 12 mo 15 mo0

50

100

150

Tim

e(s

)

DBL-KONT

Fig. 5. Analysis of anxiety in NT and DBL-KO mice at multiple ages.

The elevated plus maze (EPM) revealed no significant difference

between the NT (blue) and DBL-KO (red) mice in the time spent in

open arms at any age (Student t-test, p 6 0.05; error bars represent

SEM). Striped bars indicate nighttime testing. 6mo n= 6 (NT) and 5

(DBL-KO); 12mo n= 11 (NT) and 10 (DBLO); 15mo n= 6 (NT) and

4 (DBL-KO). (For interpretation of the references to color in this figure

legend, the reader is referred to the web version of this article.)

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 513

was performed using both the Y-Maze and the Open Field

tests. Initially, at 3 months of age, a trend toward a lower

total number of entries was seen in the Y-Maze by the

DBL-KO mice as compared to control NT mice,

irrespective of time of day (NT n= 3 and DBL-KO

n= 5; Fig. 7A – 3mo: a 2-way ANOVA using between-

group experimental design; genotype � time; genotype:

F(1,12) = 7.292, p= 0.0193; time: F(1,12) = 0.0582,

p= 0.8135; interaction: F(1,12) = 0.7892, p= 0.3918;

Fig. 7B – 3mo: a 2-way ANOVA using between-group

experimental design; genotype � time; genotype:

F(1,12) = 1.335, p= 0.2705; time: F(1,12) = 0.3202,

p= 0.5819; interaction: F(1,12) = 0.5782, p= 0.4617;).

By 6 months of age, the control mice were making

substantially fewer entries in the Y-Maze than they were

Fig. 4. Circadian behavior in 6-month NT and DBL-KO mice. Representative

mice maintained under 12-h light:12-h dark (LD), then constant darkness (DD

running period in constant darkness was 23.25 h for the NT mice, and 23.73

indicate wheel running and recordings from consecutive days are stacked in

second day of each horizontal bar were re-plotted as those for the first day

when younger such that their performance was then

significantly below that of the DBL-KO mice, also

irrespective of time of day. NT n= 6 and DBL-KO

n= 4; a 2-way ANOVA using between-group

experimental design; genotype � time; genotype:

F(1,13) = 14.55, p= 0.0021; time: F(1,13) = 1.113,

p= 0.3106; interaction: F(1,13) = 0.1394, p= 0.7149;

Bonferroni post hoc analysis revealed a significant

difference between genotypes during the day

(p= 0.0388) and at night (p= 0.0336). The activity

level of juvenile male mice is known to be higher than

that of adult male mice (Weinert and Waterhouse,

1999). At 12-months of age, the DBL-KO mice continued

to make significantly more entries in the Y-Maze than the

controls (NT n= 11 and DBL-KO n= 10; Student’s

t-test, p= 0.0460). Similar to the Y-Maze, a significantly

higher level of activity was exhibited by the DBL-KO

in the Open Field test (Fig. 7B) at 6 months of age

double-plotted wheel-running actograms for NT (A) and DBL-KO (B)

), followed by a repeat of both photoperiods to calculate tau. The free-

h for the DBL-KO mice (Student t-test, p= 0.028). Black markings

rows. Each horizontal bar represents 2 days (48 h). The data for the

of the next horizontal bar. n= 4 (NT) and 5 (DBL-KO).

Platform Recognition

1 2 3 40

10

20

30

40

Trial

Late

ncy

to P

latfo

rm (s

)

NTDBL-KO

Rota-rod Balance

6 mo 12 mo 15 mo0

100

200

300

Bal

ance

Mai

ntai

ned

(s)

DBL-KONT

A

B

Fig. 6. Analysis of sensorimotor behavior in 6-, 12-, or 15-month NT

and DBL-KO mice. (A) A test of visual and motor skills in a water

maze Platform Recognition test revealed no significant differences

between the NT (blue) and DBL-KO (red) mice at 6 months of age.

Animals in both groups were able to see, swim to, and climb onto the

platform and trended toward improvement between the first and last

trials. (B) The Rota-rod revealed no significant difference at any age

between the NT (blue) and DBL-KO (red) mice in the length of time

balance was maintained on a rotating horizontal rod that accelerated

from 4 to 40 rpm (a 2-way RM ANOVA for A and Student t-test for B;p 6 0.05; error bars represent SEM). Striped bars indicate nighttime

testing. Platform Recognition and Rota-rod 6mo n= 6 (NT) and 5

(DBL-KO); Rota-rod 12mo n= 10 (NT) and 10 (DBL-KO); Rota-rod

15mo n= 6 (NT) and 4 (DBL-KO). (For interpretation of the

references to color in this figure legend, the reader is referred to

the web version of this article.)

514 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

(NT n = 6 and DBL-KO n = 4; Student’s t-test, p =

0.0284) and 12 months of age (NT n= 11 and DBL-KO

n= 10; Student’s t-test, p= 0.0238). By 15-months of

age these differences are attenuated in both testing mea-

sures (15mo NT n= 6 and DBL-KO n= 4; Student’s

t-test, p= 0.3112 for Y-Maze and p= 0.2444 for Open

Field).

Increased water consumption in DBL-KO mice. In

accordance with the increased free-running period

(Fig. 4) and increased locomotor activity (Fig. 7) noted

in the DBL-KO mice, daily water consumption was also

found to be significantly higher in the DBL-KO mice as

compared to control NT which consumed close to the

daily averages reported for mice (5.8 ± 0.2 mL/day)

(Bachmanov et al., 2002). Six-month old NT mice con-

sumed 5.77 ± 0.10, mL/day (n= 10), while the DBL-

KO mice consumed 7.29 ± 0.15 mL/day (n= 10;

Student’s t-test, p< 0.0001).

Cognitive enhancement in male DBL-KO mice. Mice

were tested for cognitive performance with a standard

behavioral test for spatial/reference learning (Barnes

Maze) and basic working memory (Y-Maze). While both

groups of mice showed learning in the Barnes Maze (day

4 was significantly different than day 1), the DBL-KO

mice had a significantly lower latency to find the target

escape hole on days two and three compared to their NT

counterparts (Fig. 8A Barnes Maze: a 2-way mixed-

design ANOVA for repeated measures; NT n= 6 and

DBL-KO n= 4; genotype: F(1,8) = 0.6253, p= 0.4519;

days: F(3,24) = 22.20, p< 0.0001; interaction: F(3,24) =

4.781, p= 0.0095). For the DBL-KO, Bonferroni post hoc

analysis revealed significant differences between day 1

vs. 2 (p< 0.0001), day 1 vs. 3 (p< 0.0001), and day 1

vs. 4 (p< 0.0001). For the NT, Bonferroni post hoc

analysis revealed no significant differences between day

1 vs. 2 (p= 0.1663) or between day 1 vs. 3

(p= 0.0595), but a significant difference between day 1

vs. 4 (p= 0.0008). DBL-KO mice also showed a

statistically significant elevation in the number of arm

alternations in the Y-Maze when assessed during the day

and a similar trend when assessed at night (Fig. 8B

Y-Maze: a 2-way ANOVA using between-group

experimental design; genotype � percent alternation; NT

n= 7 and DBL-KO n= 9; genotype: F(1,24) = 12.81,

p= 0.0015; percent alternation: F(1,24) = 1.868,

p= 0.1844; interaction: F(1,24) = 0.1719, p= 0.6821;

Bonferroni post hoc analysis between genotypes during

the day (p= 0.0143) and at night (p= 0.0829).

LTP heightened in male DBL-KO mice

Behavioral data from both the Barnes Maze and the

Y-Maze alternation task revealed enhanced cognition in

the DBL-KO mice compared to the NT mice. Hence, in

order to determine whether a neurobiological correlate

might exist, we assayed for alterations in synaptic

efficacy following tetanization. Tetani were applied to

the Schaffer collateral pathway in the HPC while

recording from pyramidal neurons in the CA1 (Fig. 9A).

Changes in amplitude of the minimally evoked EPSPs

were monitored before and after tetanization. We found

EPSPs in both groups to be significantly potentiated, but

potentiation in DBL-KO mice was greater compared to

the NT mice (p 6 0.001, t-test) (Fig. 9B). At �5 min

post-tetanization NT and DBL-KO EPSP amplitudes

averaged 195 ± 29% and 287 ± 25% of their pre-

tetanus baseline values respectively (100%, n= 9, 8

cells; p 6 0.05, paired t-test) (Fig. 9C). Potentiation was

observed in �90% of all trials. The potentiation in both

groups was long lasting (�45 min) and series

resistance, which was monitored periodically, remained

unchanged throughout the course of the experiments.

The averaged paired-pulse ratio (PPR), defined as the

Y-Maze Day v Night - 3mo

NT Day

NT Night

DBL-KO D

ay

DBL-KO Night0

20

40

60

Tota

l Ent

ries ns

Y-Maze Day v Night - 6mo

NT Day

NT Night

DBL-KO D

ay

DBL-KO Night0

20

40

60

Y-Maze Day - 12mo

NT

DBL-KO

0

20

40

60 p=.0460

Y-Maze Day - 15mo

NT

DBL-KO

0

20

40

60 ns

ns

Open Field Day v Night - 3mo

NT Day

NT Night

DBL-KO D

ay

DBL-KO Night0

50

100

150

200

Line

s C

ross

ed

ns

Open Field Night - 6mo

NT

DBL-KO

0

50

100

150

200 p=.0284

Open Field Day - 12mo

NT

DBL-KO

0

50

100

150

200 p=.0238

Open Field Day - 15mo

NT

DBL-KO

0

50

100

150

200

A

B

* *

Fig. 7. Analysis of locomotor activity in NT and DBL-KO mice at multiple ages. Locomotor activity was assessed by Y-Maze (A) and Open Field (B)

tests at four different ages. Juvenile NT mice (3mo) showed a trend toward higher activity levels as compared to DBL-KO mice in both tests. From

6mo onward, the DBL-KO (red) were significantly more active than the NT (blue) in both tests (a 2-way ANOVA or Student t-test, p 6 0.05; error

bars represent SEM). Where evaluated, striped bars indicate nighttime testing. Y-Maze and Open Field: 3mo = 3 (NT) and 5 (DBL-KO); 6mo n= 6

(NT) and 4 (DBL-KO); 12mo n= 11 (NT) and 10 (DBL-KO); 15mo n= 6 (NT) and 4 (DBL-KO). (For interpretation of the references to color in this

figure legend, the reader is referred to the web version of this article.)

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 515

ratio of the second to the first response, (50-ms interval)

remained invariant following tetanus in DBL-KO mice

(1.7 ± 0.1 vs. 1.4 ± 0.1, n= 6; p= 0.1, paired t-test)and in NT mice (1.4 ± 0.2 vs. 1.2 ± 0.1, n= 10;

p= 0.3, paired t-test), suggests that the potentiation in

both groups is mediated post-synaptically (Fig. 9D).

Protein analysis by immunoblotting

In view of our behavioral and electrophysiological data

pointing to neurobiological differences between control

and melatonin receptor-deficient mice (see above), we

decided to perform quantitative infrared fluorescence

immunoblotting for several key signaling and synaptic

markers in the brain. Levels of phosphoCREB

(normalized to constitutive CREB (cAMP response

element-binding protein) levels) were significantly

elevated in the frontal cortices of DBL-KO mice with a

similar non-significant trend in the HPC (n= 4–6 for all

analyses; Fig. 10A: t-test, p= 0.0056; Fig. 10G: t-test,p= 0.4325). Conversely, expression of phosphoERK1/2

(normalized to ERK1/2) was significantly lower in the

DBL-KO brain frontal cortex with a similar trend in the

HPC (Fig. 10B: t-test, p= 0.0404; Fig. 10H: t-test,p= 0.2574). PhosphoSynapsin levels (normalized to

synapsin) were also significantly lower in the DBL-KO

frontal cortex but not in the HPC (Fig. 10C: t-test,p= 0.0226; Fig. 10I: t-test, p= 0.4834). Protein

expression levels for the dendritic spine marker,

spinophilin was statistically less in both the frontal cortex

and HPC of DBL-KO mice (Figs. 10D: t-test, p= 0.0403

and 10J: t-test, p= 0.0007), as was the key enzyme of

GABA synthesis, glutamic acid decarboxylase 1 (GAD1;

Figs. 10E: t-test, p= 0.0464 and 10K: t-test,p= 0.0494). A significant elevation of the ionotropic

glutamate AMPA receptor subunit 1 (GluR1) was also

noted in the frontal cortex (Fig. 10F: t-test, p= 0.0417)

with a similar trend in the HPC (Fig. 10L: p= 0.4204).

DISCUSSION

The results of the present investigations demonstrate

molecular, electrophysiological, and behavioral

alterations in mice genetically lacking all known

G-protein-coupled membrane melatonin receptors. The

behavioral phenotype includes mild hyperactivity in DBL-

KO mice, a longer free-running circadian clock, and

enhanced cognition. No evidence of altered anxiety or

changes in sensorimotor performance was apparent.

Hippocampal function at the level of LTP was found to

be significantly enhanced in the DBL-KO animals.

Alterations in several key signaling and synaptic

markers also confirm the reorganizational outcome in

the brain of mice lacking melatonin receptors.

The two known mammalian melatonin receptors

(MTNRs) were cloned in the mid-1990s (Reppert et al.,

1994, 1995, 1996) and subsequently shown to be unique

G-protein-coupled membrane receptors based on the

molecular structure and chromosomal localization (cf.

Dubocovich et al., 2010). Both receptors signal via multi-

ple intracellular pathways, including inhibition of cyclic

AMP and cyclic GMP, activation of protein kinase C and

Barnes Maze

1 2 3 40

200

400

600

Days

Late

ncy

to E

scap

e H

ole

(s)

NTDBL-KO

Y-Maze Alternations Day v Night

NT Day

NT Night

DBL-KO D

ay

DBL-KO Night

0

20

40

60

80

Alte

rnat

ion

%

A

B

*

* ***

Fig. 8. Cognitive performance of 6-month NT and DBL-KO mice.

Barnes Maze results (A) revealed that the DBL-KO (red) mice learned

the testing paradigm by day two as compared to NT (blue) which

required until day four to demonstrate significant learning. Results

from the Y-Maze (B) revealed a greater percent alternation in the

DBL-KO mice as compared to NT in the day (p= 0.024) but not at

night. A 2-way RM ANOVA for A and a 2-way ANOVA for B; error

bars represent SEM; Barnes n= 6 (NT) and 4 (DBL-KO); Y-Maze

n= 7 (NT) and 9 (DBL-KO). (For interpretation of the references to

color in this figure legend, the reader is referred to the web version of

this article.)

516 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

extracellular signal-regulated protein kinase (ERK1/2),

and many other pathways (cf. Dubocovich et al., 2010).

Among the numerous downstream targets, we demon-

strate here (Fig. 2) that components of the neuronal anti-

oxidant response, i.e. Nrf2, SOD1, GPx-1 and CAT,

respond to melatonin treatment in an MTNR-dependent

manner.

Melatonin MT1 and MT2 receptors are expressed at

high levels in the embryonic brain (Rivkees and

Reppert, 1991; Seron-Ferre et al., 2007) and are presum-

ably activated by circulating maternal melatonin, which

crosses the placenta to affect the developing fetus

(Klein, 1972). For example, administration of melatonin

to pregnant, suprachiasmatic nuclei (SCN)-lesioned

female Syrian hamsters entrains circadian activity

rhythms in fetal and neonatal pups (Davis and Mannion,

1988; Grosse et al., 1996). However, only a very few

investigations have explored the role for MTNRs in neuro-

development. Melatonin treatment of mammalian neural

stem cells promotes their differentiation (Moriya et al.,

2007; Kong et al., 2008), while treatment of zebrafish

embryos with melatonin or MTNR antagonists modulates

the timing of neuronal differentiation (Danilova et al.,

2004; de Borsetti et al., 2011).

Although MTNRs are expressed in many areas of the

adult brain, most attention over the past years has been

aimed at MTNR expression by the cells of the

hypothalamic SCN, which serve to mediate the

feedback effects of circulating melatonin, whose

secretory rhythm is generated by the circadian activity

of the central clock in the SCN (cf. Dubocovich et al.,

2010). Melatonin acts on SCN neurons via MT1 receptors

to acutely hyperpolarize them (Liu et al., 1997) and via

MT2 (and possibly MT1) receptors to induce phase shifts

in both molecular and electrophysiological rhythms

(Sugden, 1983; Dubocovich et al., 2010). By inhibiting

SCN neurons, and probably other target neurons else-

where in the brain, melatonin is thought to modulate the

sensitivity of these cells to other inputs, such as activity-

dependent neurotransmitter secretion.

In the present study, we determined that animals

lacking MTNRs throughout development and into

adulthood do not show locomotor arrhythmia, i.e. the

basic mechanisms of circadian oscillation in the SCN

remains intact. This is also evident at the level of

melatonin secretory rhythms, which are similar in both

NT and DBL-KO mice (Fig. 3). However, the

endogenous free-running circadian period of the DBL-

KO mice is significantly lengthened as compared to NT

mice expressing both MTNRs (Fig. 4). We interpret

these results to indicate that circadian melatonin signals

from the maternal system, either during embryonic/fetal

neurodevelopment and/or during early postnatal life

(e.g. maternal milk) may play an important role in

shaping the molecular and cellular maturation of the

SCN clockwork and its output. Numerous mechanisms

could be envisioned, such as altered gene expression in

developing SCN neurons, or modulation of innervation

patterns; however, this remains to be explored more

fully. A limitation of the current investigation is the global

and irreversible nature of the melatonin receptor

deletion. In this regard, future studies with conditional

and tissue-targeted melatonin receptor silencing would

overcome this hurdle.

Melatonin, when administered acutely, has been

demonstrated to have sedative/hypnotic properties in

rodents (Sugden, 1983, 1995) and humans (Lieberman

et al., 1984; Dollins et al., 1993; Tzischinsky and Lavie,

1994). Acute administration to mice also decreases spon-

taneous motor activity and at very high doses produces

motor incoordination (Sugden, 1995). Our present find-

ings of significantly enhanced locomotor activity in both

the Y-Maze and Open Field tests (Fig. 7) is consistent

with the view that melatonin, even though it is released at

night during the mouse’s active phase, contributes net

Fig. 9. Long-term potentiation in hippocampi of 6-month NT and DBL-KO mice. (A) Hippocampal slice preparation showing placement of

stimulating electrode (S) on the Schaffer collaterals and recording electrode (R) in the CA1 region. (B) Plot of the ensemble data showing time

course of changes in EPSP amplitude (percentage of baseline, 100%= average of all control values) following tetanus (indicated by an arrow) of

DBL-KO (red) and NT (blue) mice. DBL-KO mice had significantly elevated EPSP compared to NT. For both, each point (closed circles) representsthe ensemble average across the indicated number of experiments of 5–7 consecutive measurements of EPSP amplitude. Error bars represent

SEM, where this is greater than the size of the symbols. Traces (B, inset) are averaged consecutive records (P15) of minimally evoked EPSPs

taken from 20 min post-tetanus region of the time course. Time course of changes in Rs (± SEM) (closed diamonds), a measure of series

resistance (% baseline) is also shown. (C) Bar plots representing the mean (±SEM) percentage potentiation of NT and DBL-KO EPSPs following

tetanus. (D) Traces are averaged consecutive records of EPSPs evoked during paired stimulation of NT and DBL-KO mice before and after tetanus.

Bar plots showing no significant changes in the mean (± SEM) paired-pulse ratio following tetanus for both NT (n= 10) and DBL-KO (n= 6) mice

measured across experiments. Statistically significant change (⁄, p< 0.05; ⁄⁄⁄, p< 0.001) with the t-test when comparing between NT and

DBL-KO mice or with the paired t-test when comparing before and after effects within groups; ns (no significant difference) p> 0.05. (For

interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 517

inhibitory signals to the brain’s motor system via binding to

the MTNRs. When these receptors are absent, such as in

the current studies, the animals show meaningfully higher

levels of locomotor activity. Consistent with our findings,

DBL-KO mice were reported to spend more of the 24-h

cycle awake as compared to mice with both MT1 and

MT2 receptors (Comai et al., 2013), which the authors pro-

pose is due to a reduction of REM sleep that accompanies

deletion of the MT1 receptor in combination with reduction

of non-REM sleep arising from loss of the MT2 receptor.

Recently, in a mutation screening of humans with attention

deficit/hyperactivity disorder (ADHD) a loss-of-function

mutation of MT1 was reported (Chaste et al., 2011). Addi-

tionally, damped rhythms of serum melatonin have also

been associated repeatedly with both pediatric and adult

ADHD (Van der Heijden et al., 2005; Van Veen et al.,

2010; Baird et al., 2012), and the clinical data thus far –

while still limited (cf. Bendz and Scates, 2010) – suggests

that melatonin administration may be effective in the treat-

ment of ADHD-associated insomnia, consistent with the

hypothesis that melatonin generally serves to inhibit motor

activity. Based on our results presented here, the DBL-KO

mouse may provide a useful model for understanding how

melatonin regulates locomotor activity.

Acute melatonin administration has been reported to

have anxiolytic effects in both animal models (Papp

et al., 2006; Crupi et al., 2010) and in human trials

(Srinivasan et al., 2006; Caumo et al., 2009). Recently,

Ochoa-Sanchez and colleagues (2012) reported that the

novel MT2-selective agonist, UCM765, showed anxiolytic

properties in the Open Field test and EPM. However, in

contrast to the prediction that DBL-KO mice might be

subject to a general anxiety disorder, our results find no

significant change – either during the day time (data not

shown) or night time – in the time spent in open arms of

the EPM or in the center portion of an Open Field test

(data not shown) when compared to the control NT mice.

These results largely conform to those recently published

by Comai and Gobbi (2014) using the MT2 knockout

mouse model, but they would appear to contrast with

the results of Larson et al. (2006) who reported signifi-

cantly greater time spent in the center of the Open Field

test in mice lacking only the MT2 receptor. Possibly the

complete lack of MT receptors in our DBL-KO mice

eliminated this MT2-specific effect. Alternatively, the time

spent in the center of the Open Field test arena may sim-

ply reflect altered decision-making functions rather than

anxiety per se (Rodgers and Johnson, 1995). Though

p-CREB / CREB

0.0

0.2

0.4

0.6

% R

elat

ive

IR F

luor

esce

nce p=.0056

p-ERK / ERK

0.00

0.05

0.10

0.15

% R

elat

ive

I R F

luo r

esce

n ce p=.0404

p-Synapsin / Synapsin

0.00

0.02

0.04

0.06

0.08

0.10

% R

elat

ive

IR F

luor

esce

nce p=.0226

Spinophilin / Actin

0.00

0.02

0.04

0.06

0.08

% R

elat

ive

IR F

luor

esce

nce p=.0403

GAD1 / Actin

% R

ela t

ive

IR F

luor

e sce

nce

0.00

0.02

0.04

0.06

0.08

0.10 p=.0464

GluR1/Actin

% R

ela t

i ve

IR F

luo r

esce

nce

0.0

0.2

0.4

0.6

0.8 p=.0417

Frontal Cortex Hippocampus

p-CREB / CREB

% R

elat

ive

IR F

luor

e sce

nce

0.0

0.5

1.0

1.5

2.0

p-ERK / ERK

0.0

0.2

0.4

0.6

0.8

% R

elat

ive

IR F

luor

esce

nce

p-Synapsin / Synapsin

0.000

0.002

0.004

0.006

0.008

0.010

% R

elat

ive

IR F

luor

esce

nce

Spinophilin / Actin

0.0

0.1

0.2

0.3

0.4

0.5

% R

elat

ive

IR F

luor

e sce

nce p=.0007

GAD1 / Actin

0.00

0.05

0.10

0.15

0.20

% R

elat

ive

IR F

luor

esce

nce p=.0494

GluR1 / Actin

0

1

2

3

4

% R

elat

ive

IR F

luo r

esce

nce

A B G H

I J

K L

C D

E F*

Representative dual-labeled fluorescent Western

(Green GluR1/Red Actin)

* NT DBL-KO100 kDa

42 kDa

DBL-KONT

Fig. 10. Immunoblotting results from frontal cortex and hippocampus of 6-month NT and DBL-KO mice using quantitative infrared

immunofluorescence. (A, G) Ratio of phosphoCREB to CREB signal. (B, H) Ratio of phosphoERK1/2 to ERK1/2 signal. (C, I) Ratio of

phosphoSynapsin to Synapsin signal. (D, J) GAD1 expression normalized to b-actin. (E, K) Spinophilin expression normalized to b-actin. (F⁄, L)GluR1 expression normalized to b-actin. ⁄Representative dual-labeled LI-COR fluorescent Western blot for GluR1/b-actin in frontal cortex. Student

t-test, p 6 0.05 for all comparisons; error bars represent SEM; n= 4–6 for all analyses.

518 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

previous reports show melatonin administration to have

anxiolytic effects, it should be remembered that compar-

ing acute hormonal effects in an intact animal is not inver-

sely equivalent with the consequences of genetic deletion

of that hormone’s receptor. The latter is most likely asso-

ciated with significant alterations in neurodevelopment

(see Discussion above).

Numerous previous studies have documented

melatonin effects on hippocampal functions (Collins and

Davies, 1997; Hogan et al., 2001; El-Sherif et al., 2002,

2003) including a concentration-dependent inhibition of

LTP, which could be blocked by selective melatonin

receptor antagonists (Wang et al., 2005) supporting

involvement of melatonin receptor signaling in the HPC.

Larson and colleagues (Larson et al., 2006) subsequently

reported that mice deficient in only the MT2 melatonin

receptor have significantly attenuated LTP responses as

compared to control animals. Additionally, this group

found that whereas control mice quickly developed a pref-

erence for the closed arms of an EPM, the MT2-deficient

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 519

mice showed significantly less preference for the closed

arms. Our results with the EPM (Fig. 5) are in fact consis-

tent with the findings of Larson et al. [ibid] although we

interpret the data as a trend toward less anxiety in the

DBL-KO mice. On most measures of cognitive perfor-

mance in the present investigation (Fig. 8) mice lacking

all melatonin receptors performed significantly better than

the intact NT mice. The Barnes Maze data are consistent

with the strong differences in hippocampal LTP (Fig. 9)

such that the DBL-KO mice showed enhanced LTP. Indi-

rect support of our data may be found in the results of

O’Leary et al. (2011) who examined learning performance

on the Barnes Maze in 13 mouse strains and reported that

among the highest performing strains was the C57BL/6 J

mouse, which is genetically compromised in its ability to

synthesize melatonin (Roseboom et al., 1998; Kasahara

et al., 2010), i.e. they develop in uterowithout the maternal

melatonin signal. Overall, our data support the hypothesis

that neurodevelopment is significantly influenced by

chronic input signaling of the melatonin receptors. This

likely involves site-specific alterations in gene expression,

intracellular signaling, and synaptic function consistent

with the recent work of Vilches and colleagues (2014),

who suppressed maternal melatonin in pregnant rats and

demonstrated impaired hippocampal gene expression

and spatial memory in their offspring, which could be res-

cued by melatonin supplementation of the pregnant dams.

To more fully characterize the molecular

consequences of MNTR deficient mice in light of the

behavioral results, we examined protein expression in

the frontal cortex and HPC of NT and DBL-KO mice

(Fig. 10). Signaling through cyclic AMP/protein kinase A

and cyclic GMP/protein kinase G activates constitutive

CREB via phosphorylation and has been implicated in

both locomotor activity and cognition (Bliss and

Collingridge, 1993; Lu et al., 1999). CREB activates

numerous target genes through cyclic AMP response ele-

ments, thus mediating signals from numerous physiologi-

cal stimuli, resulting in regulation of a broad array of

cellular responses. It plays a large regulatory role in the

nervous system and is believed to promote neuronal sur-

vival, precursor proliferation, neurite outgrowth, and neu-

ronal differentiation in certain neuronal populations.

Additionally, CREB signaling is involved in learning and

memory in several organisms. Melatonin’s classic signal-

ing signature – via both MT1 and MT2 – is to inhibit cAMP

generation and thereby protein kinase activation in target

tissues (Reppert et al., 1996; cf. Dubocovich et al., 2010).

Our data indicate significantly increased levels of cortical

phosphoCREB expression relative to CREB levels in the

DBL-KO (Fig. 10A) implying enhanced signaling through

this pathway. Similar results were found in hippocampal

tissue (although the latter did not reach statistical signifi-

cance), consistent with enhanced LTP in the DBL-KO

brain (Fig. 9).

Mitogen-activated protein kinases (MAPKs) are a

widely conserved family of serine/threonine protein

kinases involved in many cellular programs, such as cell

proliferation, differentiation, motility, and death. The p44/

p42 MAPK (ERK1/2) signaling pathway can be activated

in response to a diverse range of extracellular stimuli

including mitogens, growth factors, and cytokines.

Melatonin has generally been reported to activate

ERK1/2 signaling mechanisms (cf. Dubocovich et al.,

2010). The decreased levels of pERK1/2 expression rela-

tive to ERK1/2 levels in the frontal cortex (and a similar

trend in the HPC) of DBL-KO mice (Fig. 10B) are consis-

tent with this and may account for the presumptive differ-

ences in the neurodevelopmental program in the brains of

these mice as compared to NT control mice that devel-

oped in the presence of functional MTNRs.

Synapsin is a synaptic vesicle-associated protein,

participating in synapse formation, regulation of the

synthesis of other synaptic vesicle proteins, and

promotion of neurotransmitter release. Phosphorylation

of the synapsin by protein kinase A (PKA) or calcium

calmodulin-dependent protein kinases (CaMK) inhibits

its binding to phospholipids and dissociates synapsin

protein from synaptic vesicles, ultimately leading to

more efficient neurotransmitter release at the

presynaptic terminal (Bonanomi et al., 2005). Expression

levels of phosphoSynapsin were reduced in the DBL-KO

mouse as compared to control animals (Fig. 10C).

Although this suggests reduced synaptic activity (at least

for the frontal cortex) it remains unclear whether the effect

is a global one, i.e. affecting excitatory and inhibitory neu-

rotransmission equally. Consistent with the synapsin

data, spinophilin expression data reveal significantly

lower levels in the DBL-KO mice (Fig. 10D). Spinophilin

is an actin-associated scaffolding protein mainly localized

to the heads of dendritic spines and these spines are

known to receive excitatory input (Harnett et al., 2012).

To better understand the increased activity seen in the

DBL-KO mice, we also examined the expression levels of

the key GABA-synthesizing enzyme, GAD1. As shown in

Fig. 10E, GAD1 protein levels were significantly reduced

in the DBL-KO frontal cortex, consistent with the overall

trend toward hyperactivity in the DBL-KO mice (Fig. 7).

Conversely, expression of the post-synaptic

glutamate AMPA receptor subunit 1 (GluR1) was

significantly elevated in the frontal cortex of DBL-KO

mice as compared to NT mice (Fig. 10F) with a similar

trend in the HPC (Fig. 10L). This is consistent with

higher levels of LTP in the DBL-KO mice.

CONCLUSIONS

The finding that genetic deletion of melatonin receptors

produces mice with enhanced cognitive and motor

performance supports the emerging view that these

receptors play an important role in neurobehavioral

development. Future investigations on innate melatonin

receptor dysfunctions in the context of human cognitive

and motor development will likely provide new horizons

for treating a diverse variety of behavioral disorders.

AUTHOR CONTRIBUTIONS

G. O’Neal-Moffitt, J. Olcese, J. Pilli, and S.S. Kumar

designed the experiments; G. O’Neal-Moffitt and J. Pilli

performed the experiments, analyzed the data

and performed statistical analysis; G. O’Neal-Moffitt,

520 G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521

J. Olcese, J. Pilli, and S.S. Kumar wrote and approved the

final manuscript.

Acknowledgments—The authors thank the following for their

respective contributions: M. D’Alessandro and Dr. S. Beesley

for circadian protocols and data analysis; Dr. D. Weaver for the

mice; Drs. Y. Zhou and C. Ouimet for antibodies; Drs. Y. Zhou

and J. Rodefer for use of behavioral equipment and constructive

advice on testing; J. Wolfson and M. de Jesus for assistance with

behavioral testing; C. Badland for assistance in generating fig-

ures. This work was supported by grants to J. Olcese from the

Florida State University Council on Faculty Research & Creativ-

ity, and the FSU College of Medicine (Division of Research).

REFERENCES

Allen PB, Ouimet CC, Greengard P (1997) Spinophilin, a novel

protein phosphatase 1 binding protein localized to dendritic

spines. Proc Natl Acad Sci U S A 94:9956–9961.

Bachmanov AA, Reed DR, Beauchamp GK, Tordoff MG (2002) Food

intake, water intake, and drinking spout side preference of 28

mouse strains. Behav Genet 32:435–443.

Baird AL, Coogan AN, Siddigui A, Donev RM, Thorne J (2012) Adult

attention-deficit hyperactivity disorder is associated with

alterations in circadian rhythms at the behavioural, endocrine

and molecular levels. Mol Psychiatry 17:988–995.

Bendz LM, Scates AC (2010) Melatonin treatment for insomnia in

pediatric patients with attention-deficit/hyperactivity disorder. Ann

Pharmacother 44:185–191.

Bliss TV, Collingridge GL (1993) A synaptic model of memory: long-

term potentiation in the hippocampus. Nature 361:31–39.

Bonanomi D, Menegon A, Miccio A, Ferrari G, Corradi A, Kao HT,

Benfenati F, Valtorta F (2005) Phosphorylation of synapsin I by

cAMP-dependent protein kinase controls synaptic vesicle

dynamics in developing neurons. J Neurosci 25:7299–7308.

Bonnefond A, Clement N, Fawcett K, Yengo L, Vaillant E, Guillaume

JL, Dechaume A, Payne F, Roussel R, Czernichow S, Hercberg

S, Hadjadj S, Balkau B, Marre M, Lantieri O, Langenberg C,

Bouatia-Naji N, Meta-Analysis of Glucose and Insulin-Related

Traits Consortium (MAGIC), Charpentier G, Vaxillaire M,

Rocheleau G, Wareham NJ, Sladek R, McCarthy MI, Dina C,

Barroso I, Jockers R, Froguel P (2012) Rare MTNR1B variants

impairing melatonin receptor 1B function contribute to type 2

diabetes. Nat Genet 44:297–301.

Caumo W, Levandovski R, Hidalgo MP (2009) Preoperative anxiolytic

effect of melatonin and clonidine on postoperative pain and

morphine consumption in patients undergoing abdominal

hysterectomy: a double-blind, randomized, placebo-controlled

study. J Pain 10:100–108.

Chang C, Lee SO, Wang RS, Yeh S, Chang TM (2013) Androgen

receptor (AR) physiological roles in male and female reproductive

systems: lessons learned from ARKO mice lacking AR in

selective cells. Biol Reprod 89:21.

Chaste P, Clement N, Botros HG, Guillaume JL, Konyukh M, Pagan

C, Scheid I, Nygren G, Anckarsater H, Rastam M, Stahlberg O,

Gillberg IC, Melke J, Delorme R, Leblond C, Toro R, Huguet G,

Fauchereau F, Durand C, Boudarene L, Serrano E, Lemiere N,

Launay JM, Leboyer M, Jockers R, Gillberg C, Bourgeron T

(2011) Genetic variations of the melatonin pathway in patients

with attention-deficit and hyperactivity disorders. J Pineal Res

51:394–399.

Collins DR, Davies SN (1997) Melatonin blocks the induction of long-

term potentiation in an N-methyl-D-aspartate independent

manner. Brain Res 767:162–165.

Comai S,GobbiG (2014)Unveiling the role ofmelatoninMT2 receptors

in sleep, anxiety and other neuropsychiatric diseases: a novel

target in psychopharmacology. J Psychiatry Neurosci 39:6–21.

Comai S, Ochoa-Sanchez R, Gobbi G (2013) Sleep–wake

characterization of double MT1/MT2 receptor knockout mice

and comparison with MT1 and MT2 receptor knockout mice.

Behav Brain Res 243:231–238.

Crupi R, Mazzon E, Marino A, La Spada G, Bramanti P, Cuzzocrea S,

Spina E (2010) Melatonin treatment mimics the antidepressant

action in chronic corticosterone-treated mice. J Pineal Res

49:123–129.

Danilova N, Krupnik VE, Sugden D, Zhdanova IV (2004) Melatonin

stimulates cell proliferation in zebrafish embryo and accelerates

its development. FASEB J 18:751–753.

Davis FC, Mannion J (1988) Entrainment of hamster pup circadian

rhythms by prenatal melatonin injections to the mother. Am J

Physiol 255:R439–R448.

de Borsetti NH, Dean BJ, Bain EJ, Clanton JA, Taylor RW, Gamse JT

(2011) Light and melatonin schedule neuronal differentiation in

the habenular nuclei. Dev Biol 358:251–261.

Deming SL, Lu W, Beeghly-Fadiel A, Zheng Y, Cai Q, Long J, Shu

XO, Gao YT, Zheng W (2012) Melatonin pathway genes and

breast cancer risk among Chinese women. Breast Cancer Res

Treat 132:1–7.

Dollins AB, Lynch HJ, Wurtman RJ, Deng MH, Kischka KU, Gleason

RE, Lieberman HR (1993) Effect of pharmacological daytime

doses of melatonin on human mood and performance.

Psychopharmacology (Berl) 112:490–496.

Donaldson ZR, Young LJ (2008) Oxytocin, vasopressin and the

neurogenetics of sociality. Nature 322:900–904.

Dubocovich ML, Delagrange P, Krause DN, Sugden D, Cardinali DP,

Olcese J (2010) International Union of Basic and Clinical

Pharmacology. LXXV. Nomenclature, classification, and

pharmacology of G protein coupled melatonin receptors.

Pharmacol Rev 62:343–380.

Ebihara S, Marks T, Hudson DJ, Menaker M (1986) Genetic control of

melatonin synthesis in the pineal gland of the mouse. Science

23:491–493.

El-Sherif Y, Hogan MV, Tesoriero J, Wieraszko A (2002) Factors

regulating the influence of melatonin on hippocampal evoked

potentials: comparative studies on different strains of mice. Brain

Res 945:191–201.

El-Sherif Y, Tesoriero J, Hogan MV, Wieraszko A (2003) Melatonin

regulates neuronal plasticity in the hippocampus. J Neurosci Res

72:454–460.

Golde WT, Gollobin P, Rodriguez LL (2005) A rapid, simple, and

humane method for submandibular bleeding of mice using a

lancet. Lab Anim 34:39–43.

Grosse J, Velickovic A, Davis FC (1996) Entrainment of Syrian

hamster circadian activity rhythms by neonatal melatonin

injections. Am J Physiol 270:R533–R540.

Harmar AJ, Marston MH, Shen S, Spratt C, West KM, Sheward WJ,

Morrison CF, Dorin JR, Piggins HD, Reubi JC, Kelly JS, Maywood

ES, Hastings MH (2002) The VPAC(2) receptor is essential for

circadian function in the mouse suprachiasmatic nuclei. Cell

109:497–508.

Harnett MT, Makara JK, Spruston N, Kath WL, Magee JC (2012)

Synaptic amplification by dendritic spines enhances input

cooperativity. Nature 491:599–602.

Hogan MV, El-Sherif Y, Wieraszko A (2001) The modulation of

neuronal activity by melatonin: in vitro studies on mouse

hippocampal slices. J Pineal Res 30:87–96.

Hutchinson AJ, Hudson RL, Dubocovich ML (2012) Genetic deletion

of MT1 and MT2 melatonin receptors differentially abrogates the

development and expression of methamphetamine-induced

locomotor sensitization during the day and the night in C3H/

HeN mice. J Pineal Res 53:399–409.

Jin X, von Gall C, Pieschl RL, Gribkoff VK, Stehle JH, Reppert SM,

Weaver DR (2003) Targeted disruption of the mouse Mel1b

melatonin receptor. Mol Cell Biol 23:1054–1060.

Kasahara T, Abe K, Mekada K, Yoshiki A, Kato T (2010) Genetic

variation of melatonin productivity in laboratory mice under

domestication. Proc Natl Acad Sci U S A 107:6412–6417.

G. O’Neal-Moffitt et al. / Neuroscience 277 (2014) 506–521 521

Kennaway DJ, Voultsios A, Varcoe TJ, Moyer RW (2002) Melatonin

in mice: rhythms, response to light, adrenergic stimulation, and

metabolism. Am J Physiol Reg 282:R358–R365.

Kilic U, Yilmaz B, Ugur M, Yuksel A, Reiter RJ, Hermann DM, Kilic E

(2012) Evidence that membrane-bound G protein-coupled

melatonin receptors MT1 and MT2 are not involved in the

neuroprotective effects of melatonin in focal cerebral ischemia. J

Pineal Res 52:228–235.

Klein DC (1972) Evidence for the placental transfer of 3 H-acetyl-

melatonin. Nat New Biol 237:117–118.

Kong X, Li X, Cai Z, Yang N, Liu Y, Shu J, Pan L, Zuo P (2008)

Melatonin regulates the viability and differentiation of rat midbrain

neural stem cells. Cell Mol Neurobiol 28:569–579.

Larson J, Jessen RE, Uz T, Arslan AD, Kurtuncu M, Imbesi M, Manev

H (2006) Impaired hippocampal long-term potentiation in

melatonin MT2 receptor-deficient mice. Neurosci Lett 393:23–26.

Li DY, Smith DG, Hardeland R, Yang MY, Zu HL, Zhang L, Yin HD,

Zhu Q (2013) Melatonin receptor genes in vertebrates. Int J Mol

Sci 14:11208–11223.

Lieberman HR, Waldhauser F, Garfield G, Lynch HJ, Wurtman RJ

(1984) Effects of melatonin on human mood and performance.

Brain Res 323:201–207.

Liu C, Weaver DR, Jin X, Shearman LP, Pieschi RL, Gribkoff VK,

Reppert SM (1997) Molecular dissection of two distinct actions of

melatonin on the suprachiasmatic circadian clock. Neuron

19:91–102.

Lu YF, Kandel ER, Hawkins RD (1999) Nitric oxide signaling

contributes to late-phase LTP and CREB phosphorylation in the

hippocampus. J Neurosci 19:10250–10261.

Mexal S, Horton WJ, Crouch EL, Maier SI, Wilkinson AL, Marsolek M,

Stitzel JA (2012) Diurnal variation in nicotine sensitivity in mice:

role of genetic background and melatonin. Neuropharmacology

63:966–973.

Moriya T, Horie N, Mitome M, Shinohara K (2007) Melatonin

influences the proliferative and differentiative activity of neural

stem cells. J Pineal Res 42:411–418.

Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008)

Mapping and quantifying mammalian transcriptomes by RNA-

Seq. Nat Methods 5:621–628.

Muhlbauer E, Gross E, Labucay K, Wolgast S, Peschke E (2009)

Loss of melatonin signalling and its impact on circadian rhythms in

mouse organs regulating blood glucose. Eur J Pharmacol

606:61–71.

Natarajan R, Einarsdottir E, Riutta A, Hagman S, Raunio M, Mononen

N, Lehtimaki T, Elovaara I (2012) Melatonin pathway genes are

associated with progressive subtypes and disability status in

multiple sclerosis among Finnish patients. J Neuroimmunol

250:106–110.

Ochoa-Sanchez R, Rainer Q, Comai S, Spadoni G, Gedini A, Rivara

S, Fraschini F, Mor M, Tarzia G, Gobbi G (2012) Anxiolytic effects

of the melatonin MT(2) receptor partial agonist UCM765:

comparison with melatonin and diazepam. Prog

Neuropsychopharmacol Biol Psychiatry 39:318–325.

O’Leary TP, Savoie V, Brown RE (2011) Learning, memory and

search strategies of inbred mouse strains with different visual

abilities in the Barnes maze. Behav Brain Res 216:531–542.

Papp M, Litwa E, Gruca P, Mocaer E (2006) Anxiolytic-like activity of

agomelatine and melatonin in three animal models of anxiety.

Behav Pharmacol 17:9–18.

Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA,

Guselia JF (1995) Molecular characterization of a second

melatonin receptor expressed in human retina and brain: the

Mel1b melatonin receptor. Proc Natl Acad Sci U S A

92:8734–8738.

Reppert SM, Weaver DR, Ebisawa T (1994) Cloning and

characterization of a mammalian melatonin receptor that

mediates reproductive and circadian responses. Neuron

13:1177–1185.

Reppert SM, Weaver DR, Godson C (1996) Melatonin receptors step

into the light: cloning and classification of subtypes. Trends

Pharmacol Sci 17:100–102.

Rivkees SA, Reppert SM (1991) Appearance of melatonin receptors

during embryonic life in Siberian hamsters (Phodopus

sungorous). Brain Res 568:345–349.

Rodgers RJ, Johnson NJ (1995) Factor analysis of spatiotemporal

and ethological measures in the murine elevated plus-maze test

of anxiety. Pharmacol Biochem Behav 52:297–303.

Roseboom PH, Namboodiri MA, Zimonjic DB, Popescu NC,

Rodriguez IR, Gastel JA, Klein DC (1998) Natural melatonin

‘knockdown’ in C57BL/6J mice: rare mechanism truncates

serotonin N-acetyltransferase. Brain Res Mol Brain Res

63:189–197.

Seron-Ferre M, Valenzuela GJ, Torres-Farfan C (2007) Circadian

clocks during embryonic and fetal development. Birth Defects Res

C Embryo Today 81:204–214.

Srinivasan V, Smits M, Spence W, Lowe AD, Kayumov L, Pandi-

Perumal SR, Parry B, Cardinali DP (2006) Melatonin in mood

disorders. World J Biol Psychiatry 7:138–151.

Sugden D (1983) Psychopharmacological effects of melatonin in

mouse and rat. J Pharmacol Exp Ther 227:587–591.

Sugden D (1995) Sedative potency and 2-[125]iodomelatonin binding

affinity of melatonin analogues. Psychopharmacology (Berl)

117:364–370.

Tzischinsky O, Lavie P (1994) Melatonin possesses time-dependent

hypnotic effects. Sleep 17:638–645.

Van der Heijden KB, Smits MG, Van Someren EJ, Gunning WB

(2005) Idiopathic chronic sleep onset insomnia in attention-deficit/

hyperactivity disorder; a circadian rhythm sleep disorder.

Chronobiol Int 22:559–570.

Van Veen MM, Kooij JJ, Boonstra AM, Gordijn MC, Van Someren EJ

(2010) Delayed circadian rhythm in adults with attention-deficit/

hyperactivity disorder and chronic sleep-onset insomnia. Biol

Psychiatry 67:1091–1096.

Vilches N, Spichiger C, Mendez N, Abarzua-Catalan L, Galdames

HA, Hazlerigg DG, Richter HG, Torres-Farfan C (2014)

Gestational chronodisruption impairs hippocampal expression of

NMDA receptor subunits Grin 1b/Grin 3a and spatial memory in

the adult offspring. PLOS One 9:e91313.

Vivien-Roels B, Malan A, Rettori MC, Delagrange P, Jeanniot JP,

Pevet P (1998) Daily variations in pineal melatonin concentrations

in inbred and outbred mice. J Biol Rhythms 13:403–409.

Wang LM, Suthana NA, Chaudhury D, Weaver DR, Colwell CS

(2005) Melatonin inhibits hippocampal long-term potentiation. Eur

J Neurosci 22:2231–2237.

Weinert D, Waterhouse J (1999) Daily activity and body temperature

rhythms do not change simultaneously with age in laboratory

mice. Physiol Behav 66:605–612.

(Accepted 12 July 2014)(Available online 18 July 2014)