gaba cells in the central nucleus of the amygdala control ... · gaba cell activation only promotes...

100
GABA Cells in the Central Nucleus of the Amygdala Control Cataplexy by Matthew Braden Snow A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Cell and Systems Biology University of Toronto © Copyright by Matthew Braden Snow 2016

Upload: others

Post on 10-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

GABA Cells in the Central Nucleus of the Amygdala Control Cataplexy

by

Matthew Braden Snow

A thesis submitted in conformity with the requirements for the degree of Master of Science

Graduate Department of Cell and Systems Biology University of Toronto

© Copyright by Matthew Braden Snow 2016

Page 2: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

ii

GABA Cells in the Central Nucleus of the Amygdala Control Cataplexy

Matthew Braden Snow

Master of Science

Graduate Department of Cell and Systems Biology University of Toronto

2016

Abstract

Cataplexy is a hallmark of narcolepsy characterized by the sudden onset of muscle weakness or

paralysis during wakefulness. It can occur spontaneously but is typically triggered by positive

emotions like laughter. Although cataplexy was identified over 130 years ago, its neural mechanism

remains unclear. Here, we show that a newly identified GABA circuit within the central nucleus

of the amygdala (CeA) may play a causal role in promoting cataplexy. We found that, in

narcoleptic mice, chemogenetic activation of GABA cells within the CeA triggered a 253%

increase in the number of cataplexy attacks without affecting their duration. We also show that

GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding

stimuli such as wheel running, without impacting spontaneous attacks. Our results indicate that

the CeA plays a pivotal role in controlling cataplexy onset, and that emotionally rewarding

stimuli may trigger cataplexy by activating GABA cells in the CeA.

Page 3: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

iii

Acknowledgments

My successful completion of this project would not have been possible without the help and support of many individuals. First and foremost, I would like to sincerely thank my supervisor, Dr. John Peever, for giving me the opportunity to pursue graduate studies in his lab. John, you have been a superb mentor who has cultivated my passion for sleep research. You have challenged me intellectually and contributed immensely to my academic and professional development, enabling me to become the scientist I am today.

I would like to thank my supervisory committee, Dr. Richard Horner and Dr. Sheena Josselyn, whose exceptional guidance of and enthusiasm for my work augmented my experience as a graduate student. I would also like to thank Dr. Horner for directly supporting my research through the Sleep and Biological Rhythms Toronto program. Finally, I would like to thank Dr. Kaori Takehara-Nisiuchi and Dr. Richard Stephenson for participating in my oral examination.

I am particularly grateful to my colleagues in the Peever Lab who have provided me with unequalled support and insight and have created a wonderful environment in which to work. Dr. Jimmy Fraigne, Dr. Jennifer Lapierre, and Dr. Nicole Yee, your unparalleled scientific skill and expertise, and your contributions to the lab as whole and my work specifically, have been tremendously helpful and greatly appreciated. Zoltan Torontali, your knowledge and energy have made my lab experience truly memorable and your friendship is valued beyond measure. Sharshi Bulner and Daniel Li, I sincerely thank you both for your help, understanding, and good humor through all we have experienced and accomplished. Simon Lui, Dillon McKenna, Sara Pintwala, and Gabrielle Thibault-Messier, I am grateful for your encouragement and assistance; it has been a pleasure to work alongside you. I also wish to acknowledge the fantastic team of undergraduate students whose hard work has contributed in various ways to my research and would especially like to recognize Victoria Chuen, Shirin Mollayeva, Hinal Patel, Amanda Stojcevski, Aren Thomasian, and Wendy Xie. Thank you as well to all my friends on the third floor.

I would like to thank my sisters, Lindsay and Kimberly, for their unwavering support and pride in my accomplishments, and my parents, Howard and Emily, whose unique perspectives and continuous guidance have colored my experiences in graduate school and beyond.

I would like to express a special thank you to my dear Hayley and the rest of the Eidelman family. Hayley, you have stood by me and supported me through more than I can express and your interest, encouragement, friendship, and love have helped me in every step of my research career. I could not have done this without you.

This work was supported by the Canadian Institutes of Health Research, the Sleep and Biological Rhythms Toronto program, and the Natural Sciences and Engineering Research Council of Canada. Conference attendance was made possible in part by the Canadian Sleep Society, the School of Graduate Studies, and the Department of Cell and Systems Biology.

Page 4: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

List of Appendices ......................................................................................................................... ix

Chapter 1 – Introduction .................................................................................................................1

1.1 Narcolepsy ...........................................................................................................................1

1.1.1 Overview ..................................................................................................................1

1.1.2 Etiology ....................................................................................................................2

1.1.3 Diagnosis..................................................................................................................4

1.1.4 Treatment .................................................................................................................6

1.2 Cataplexy .............................................................................................................................7

1.2.1 Clinical Manifestation ..............................................................................................8

1.2.2 REM Sleep Intrusion Hypothesis ...........................................................................12

1.2.3 GABA Mechanisms of Cataplexy .........................................................................14

1.2.4 Other Mechanisms of Cataplexy ............................................................................16

1.3 The Amygdala ....................................................................................................................18

1.3.1 Anatomy and Connectivity ....................................................................................18

1.3.2 Functions ................................................................................................................22

1.3.3 The Amygdala and Cataplexy ................................................................................23

1.4 Current Model of Cataplexy ..............................................................................................26

1.5 Methodological Background ..............................................................................................28

1.5.1 DREADDs .............................................................................................................28

Page 5: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

v

1.5.2 Mouse Model .........................................................................................................31

1.6 Specific Aim ......................................................................................................................31

– Methods ....................................................................................................................33Chapter 2

2.1 Animals ..............................................................................................................................33

2.2 Chemogenetic Methods .....................................................................................................33

2.3 Drug Preparation ................................................................................................................34

2.4 Viral Injection Surgery ......................................................................................................34

2.5 EEG and EMG Instrumentation .........................................................................................35

2.6 Experimental Protocols ......................................................................................................35

2.7 Data Acquisition ................................................................................................................36

2.8 Data Analysis .....................................................................................................................36

2.9 Histology and Fluorescence In Situ Hybridization ............................................................37

2.10Statistical Analysis .............................................................................................................38

– Results ......................................................................................................................40Chapter 3

3.1 CeA activation exacerbates cataplexy in narcoleptic mice ................................................40

3.2 CeA activation does not cause cataplexy in wild-type mice ..............................................44

3.3 Orexin-/- mice that express Cre in GABA neurons exhibit typical cataplexy ....................46

3.4 hM3Dq receptors are expressed in GABA cells of the CeA .............................................50

3.5 GABA cells in the CeA promote cataplexy .......................................................................52

3.6 GABA cells in the CeA mediate cataplexy associated with rewarding stimuli .................56

– Discussion ................................................................................................................59Chapter 4

4.1 The amygdala promotes cataplexy associated with positive emotion ...............................59

4.2 GABA cells in the CeA mediate cataplexy ........................................................................60

4.3 GABA cells in the CeA are responsible for initiating cataplexy .......................................64

4.4 Additional circuits involved in cataplexy ..........................................................................65

4.5 Technical considerations ....................................................................................................65

Page 6: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

vi

4.6 Conclusions ........................................................................................................................66

References ......................................................................................................................................68

Appendix 1 – Behavioral State Analysis Macro ............................................................................82

Page 7: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

vii

List of Tables

Table 1 Diagnostic criteria for narcolepsy 5 Table 2 Behavioral state architecture in orexin-/- and orexin-/-,VGAT-Cre mice 48

Page 8: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

viii

List of Figures

Figure 1 Electrophysiological representation of cataplexy and wakefulness in a narcoleptic human

10

Figure 2 Common cataplexy triggers in narcoleptic humans 11 Figure 3 Schematic depicting the major afferent and efferent pathways of the

amygdala 21

Figure 4 Summary of evidence implicating the amygdala in cataplexy 25

Figure 5 Hypothesized circuit underlying cataplexy 27 Figure 6 Cre-dependent hM3Dq constructs permit manipulation of specific cell

populations 30

Figure 7 CeA activation exacerbates cataplexy in narcoleptic mice 42

Figure 8 The behavioral arrests triggered by CeA activation are cataplexy 43 Figure 9 CeA activation does not alter sleep-wake architecture in wild-type mice 45

Figure 10 Orexin-/- mice that express Cre in GABA neurons exhibit typical cataplexy 49 Figure 11 hM3Dq receptors are expressed in GABA cells of the CeA 51

Figure 12 GABA cells in the CeA promote cataplexy 54 Figure 13 Activating GABA CeA cells does not influence sleep-wake behavior 55

Figure 14 GABA cells in the CeA mediate cataplexy associated with rewarding stimuli

58

Figure 15 The amygdala acts as a “relay centre” between the cortex and the pons that enables positive emotions to elicit cataplexy

63

Page 9: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

ix

List of Appendices

Appendix 1 Behavioral State Analysis Macro 82

Page 10: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

1

Chapter 1 – Introduction

1.1 Narcolepsy

Narcolepsy is a serious sleep disorder that affects over 3 million people worldwide

(Overeem et al., 2001; Longstreth et al., 2007). It was first described by Dr. Carl Friedrich Otto

Westphal in 1877, who treated a patient suffering from daytime hypersomnolence and frequent

bouts of muscle weakness or paralysis during wakefulness (Westphal, 1877; Schenck et al.,

2007). Dr. Jean-Baptiste-Édouard Gélineau, who encountered an individual experiencing similar

symptoms, gave the disorder its name three years later, deriving the term “narcolepsy” from the

Greek words for “drowsiness” and “to seize” (Gélineau, 1880; Schenck et al., 2007). These

etymological roots highlight the core problem in narcolepsy: a failure by the brain to effectively

regulate sleep-wake architecture, or a “disease of state boundary control” (Broughton et al.,

1986; Fromherz and Mignot, 2004; Thorpy and Dauvilliers, 2015). This breakdown in control

manifests in the symptomatology of narcolepsy, four of which are classically known as the

“narcoleptic tetrad”: excessive daytime sleepiness (EDS), cataplexy, hypnagogic hallucinations,

and sleep paralysis (Yoss and Daly, 1957; Aldrich, 1993; Overeem et al., 2011).

1.1.1 Overview

EDS, which refers to inescapable feelings of tiredness and/or sleep attacks, and cataplexy,

the sudden and involuntary loss of muscle tone during wakefulness despite preserved

consciousness, are considered the “core” symptoms of narcolepsy and represent the major

complaints of narcoleptic individuals (Overeem et al., 2001; Liblau et al., 2015). These are often

accompanied by hypnagogic (or hypnopompic) hallucinations (vivid dreamlike events) and sleep

paralysis (muscle atonia despite preserved consciousness), both of which occur during transitions

between wakefulness and sleep (Liblau et al., 2015). Many of these symptoms resemble features

of rapid eye movement (REM) sleep, which has led to the hypothesis that cataplexy and other

symptoms of narcolepsy represent intrusions of REM sleep into wakefulness (Dauvilliers et al.,

2014; Fraigne et al., 2015). More recently, disturbed nocturnal sleep has been added as a fifth

Page 11: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

2

symptom of narcolepsy, further underscoring the apparent dysregulation of arousal state that

characterizes the disorder (Overeem et al., 2001).

Narcolepsy is closely linked to the loss of orexin-producing neurons in the lateral

hypothalamus (Peyron et al., 2000; Thannickal et al., 2000), and the disorder most commonly

presents in mid-adolescence or early adulthood (Dauvilliers et al., 2003). The current treatments

available for narcolepsy focus primarily on managing symptoms, which persist for life and

remain debilitating for those who suffer them (Overeem et al., 2001). Narcoleptic individuals

report that their condition substantially limits their performance and progress in work and school,

impacts their ability to drive, decreases the quality of their social interactions, and reduces their

enjoyment of recreational activities (Broughton et al., 1984). Because narcolepsy has such a

pervasive negative influence on patients’ lives, establishing the neurobiological basis of its

symptoms is a crucial step towards preventing disease onset in vulnerable individuals or more

effectively managing symptoms after they have appeared. Therefore, this thesis project

investigated the neural pathway that triggers cataplexy, a major complaint of narcoleptic

individuals that appears in approximately 70% of cases (Overeem et al., 2001).

1.1.2 Etiology

The pathophysiological basis of narcolepsy remained a mystery for over 100 years after

being described by Westphal and Gélineau. The first clue towards the neurobiological cause of

narcolepsy came in 1998 with the discovery of a novel neuropeptide produced in the lateral

hypothalamus (LH). This compound was discovered independently by two different groups and

so had been given two different names: Sakurai et al., who were studying hypothalamic

regulation of feeding behavior, termed the substance “orexin” (from the Greek orexis, “appetite”)

(Sakurai et al., 1998), while de Lecea et al. named the peptide “hypocretin” due to its structural

similarity to the hormone secretin (de Lecea et al., 1998) (for consistency, this peptide will be

referred to as orexin throughout this thesis). Orexin is synthesized exclusively by a population of

neurons in the LH (Peyron et al., 1998) and comes in two forms, orexin A and B, which arise

from alternative splicing of a common precursor known as prepro-orexin (de Lecea et al., 1998;

Sakurai et al., 1998), The orexin peptides bind specifically to two G protein-coupled receptors,

Page 12: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

3

OX1R and OX2R, which activate neurons via coupling to the Gq signaling pathway (Sakurai et

al., 1998).

Soon after the identification of orexin, additional discoveries were made to suggest that the

pathophysiological mechanism responsible for narcolepsy was in fact the loss of an intact orexin

system. For instance, the gene responsible for narcolepsy in canine models, canarc-1, was found

to be a mutant of OX2R in which the mRNA transcript is improperly spliced, ultimately

rendering the receptor constructed non-functional (Lin et al., 1999). Furthermore, disruption of

the orexin system in mice by deleting the genes for the orexin peptides (Chemelli et al., 1999) or

receptors (Willie et al., 2003), or by destroying orexinergic neurons postnatally with a slow-

acting neurotoxin (Hara et al., 2001), produced phenotypes strikingly similar to narcolepsy in

humans. Finally, post-mortem examination of brain tissue derived from human narcoleptics

showed a distinct lack of orexin, but not other, neurons in the hypothalamus (Peyron et al., 2000;

Thannickal et al., 2000), and orexin levels in the cerebrospinal fluid of living narcoleptic patients

is lower than normal or undetectable (Nishino et al., 2000a).

Although these observations strongly suggest the proximal cause of human narcolepsy is the

specific loss orexinergic neurons, the reason for this loss is not well understood. A current theory

is that narcolepsy has an autoimmune origin, as the majority of narcolepsy patients possess

specific genetic markers, such as HLA-DR2 and HLA-DQB1*0602, whose protein products are

involved in antigen presentation (Mignot et al., 1994). Furthermore, the narcolepsy incidence

among individuals possessing these markers tends to increase after flu epidemics (e.g., H1N1) or

widespread vaccinations that activate the immune system (Burgess and Scammell, 2012; Ahmed

et al., 2015). Despite these intriguing trends, a molecular mechanism linking the immune system

to the degradation of orexin neurons has yet to be identified.

In healthy individuals, orexin neurons project diffusely throughout the brain, innervating not

only other areas within the hypothalamus but also the locus coeruleus (LC), thalamus,

periaqueductal grey, raphe nuclei, amygdala, cortical areas, and more (Peyron et al., 1998).

Orexinergic neurons produce other neurotransmitters as well, such as glutamate (Torrealba et al.,

2003), and have been implicated in many functions including stimulating appetite (Sakurai et al.,

1998), promoting motor behavior (Hagan et al., 1999), and, importantly in the context of

narcolepsy, maintaining arousal and wakefulness (Chemelli et al., 1999; Hagan et al., 1999).

Page 13: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

4

While orexin signaling is higher in wakefulness than in sleep, it is maximal in contexts that often

trigger cataplexy in narcoleptic humans or animals such as positive emotion, social interaction,

food seeking, exploratory behaviors, and other goal-oriented actions (Lee et al., 2005;

Mileykovskiy et al., 2005; Blouin et al., 2013). Moreover, orexin cells project to and strongly

excite brainstem areas such as the LC, a region essential for supporting muscle tone during

wakefulness (Mileykovskiy et al., 2000; Peyron et al., 2000). Therefore, since narcolepsy is

characterized by motor dysfunctions such as cataplexy that do not occur in healthy individuals,

orexin may also play a role in coupling arousal with skeletal muscle tone (Mileykovskiy et al.,

2005; Liblau et al., 2015). The orexin system, then, is anatomically and functionally well-

positioned to prevent narcolepsy and particularly cataplexy, although the details surrounding

how orexin normally facilitates arousal and muscle tone are still under study.

1.1.3 Diagnosis

Narcolepsy is generally diagnosed on the basis of EDS and disordered REM sleep

phenomena that are not better explained by another factor, as well as cataplexy and/or orexin

deficiency (American Academy of Sleep Medicine, 2014). The specific diagnostic criteria for

narcolepsy have been revised several times as its symptoms and underlying pathophysiology has

become better understood. Initially, narcolepsy had been divided into two subtypes based on

whether or not cataplexy was exhibited, but once orexin cell loss had been identified as the cause

of narcolepsy, the cataplexy-based categorization became difficult to use in practice since some

narcoleptic patients with orexin deficiency do not display cataplexy (Andlauer et al., 2012).

Therefore, the most recent edition of the International Classification of Sleep Disorders has

redefined the subtypes of narcolepsy to address orexin deficiency rather than the presence of

cataplexy: type 1 narcolepsy (with orexin deficiency) and type 2 narcolepsy (without orexin

deficiency) (American Academy of Sleep Medicine, 2014) (Table 1). However, orexin

deficiency is hard to measure directly since orexin assays are not readily available and require a

sample of cerebrospinal fluid (Sateia, 2014). For this reason, and because most narcoleptic

patients with orexin deficiency do have cataplexy, the presence of cataplexy remains an

important inclusion criterion for type 1 narcolepsy and the main symptom that differentiates it

from type 2 narcolepsy (Nishino et al., 2000a; Sateia, 2014).

Page 14: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

5

Page 15: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

6

1.1.4 Treatment

There is currently no cure for narcolepsy, but there are many treatments available to help

patients manage their symptoms. In many cases, improving “sleep hygiene” behaviorally by

imposing a structured nap schedule can help individuals alleviate their sleepiness (Overeem et

al., 2001). More often, however, sleepiness is controlled medicinally through the use of

stimulants and wake-promoting agents such as amphetamines, methylphenidate, or modafinil,

which elevate levels of arousal-promoting monoamines in the brain (Overeem et al., 2001;

Leschziner, 2014). Modafinil (and its enantiomer armodafinil) in particular is favored because it

is not addictive and has few serious side effects in contrast to the more classical stimulants (De la

Herran-Arita and Garcia-Garcia, 2013).

While certain stimulant drugs have some efficacy in combating cataplexy in addition to

EDS, specific anticataplectic medications are often prescribed, and these often help reduce

hypnagogic hallucinations and sleep paralysis as well (Overeem et al., 2001). Tricyclic

antidepressants (e.g., clomipramine), selective serotonin reuptake inhibitors (SSRIs) (e.g.,

fluoxetine), and serotonin/noradrenaline reuptake inhibitors (SNRIs) (e.g., venlaxafine) elevate

monoamine levels in the brain and are thought to improve cataplexy by inhibiting REM sleep

(Overeem et al., 2001; De la Herran-Arita and Garcia-Garcia, 2013; Leschziner, 2014).

Monoamine oxidase inhibitors (MAOIs) such as selegeline hydrochloride accomplish the same

but have fallen out of favor due to their harsh side effects (Dauvilliers et al., 2014). Sodium

oxybate (or gamma-hydroxybutyrate, GHB) is a hypnotic that functions as a GABAB receptor

agonist and also modulates dopamine signaling (Dauvilliers et al., 2014). It has gained traction as

a holistic treatment for narcolepsy as it targets both EDS and cataplexy in addition to fragmented

nocturnal sleep (De la Herran-Arita and Garcia-Garcia, 2013), and is regarded as a good

alternative to classical stimulants and tricyclic antidepressants due to relatively mild side effects

(Overeem et al., 2001; Dauvilliers et al., 2014). However, it is not always the preferred

medication as it must be taken twice throughout the night due to its short half-life, and its higher

cost combined with its potential for abuse limit its availability in some populations (Leschziner,

2014).

More recently, attention has been focused on novel therapies geared towards preventing

disease progression rather than simply targeting symptoms. For instance, there has been

Page 16: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

7

experimentation with intravenous immunoglobulin or plasmapheresis soon after narcolepsy onset

aimed at counteracting autoimmune destruction of orexin neurons, although results surrounding

the effectiveness of these treatments have been inconsistent (Plazzi et al., 2008; Valko et al.,

2008; Dauvilliers et al., 2014). Enhancing histamine release via inverse agonists of the H3

receptor is another area currently under investigation and recent work indicates this strategy is

useful for treating narcoleptic mice and humans (Lin et al., 2008). Finally, orexin peptide

replacement therapy, orexin gene therapy, and synthetic orexin receptor agonists have also been

the subject of much research. Although intravenous or intraventricular administration of the

orexin peptide or transduction of existing neurons with the orexin gene show promise in animal

models of narcolepsy, there are difficulties with translating this approach to humans (John et al.,

2000; Mieda et al., 2004; Liu et al., 2008).

1.2 Cataplexy

The hypothesis that narcolepsy represents a loss of state boundary control was first put forth

by Broughton et al. in 1986 based on several observations (Broughton et al., 1986).

Electroencephalographic (EEG) recordings of individuals performing a vigilance task showed

that whereas healthy subjects maintained a constant state of wakefulness throughout, narcoleptics

continuously fluctuated between wakefulness and light sleep (Valley and Broughton, 1983).

Additionally, narcoleptics who were administered the multiple sleep latency test entered non-

REM (NREM) sleep with substantially decreased latency or even bypassed NREM sleep entirely

and entered REM sleep directly instead (Broughton et al., 1986). However, while narcolepsy as a

whole may involve inappropriate transitions between all arousal states, emphasis has been placed

on the notion that dissociated REM sleep specifically underlies most symptoms of narcolepsy,

including cataplexy (Dauvilliers et al., 2007; Siegel, 2011; Dauvilliers et al., 2014). As the focus

of this thesis is on cataplexy, the following section will examine the neurobiology of this

symptom in detail, including discussion of its clinical features, the hypothesized involvement of

REM sleep circuitry, and the neuroanatomical and neurochemical substrates that mediate it.

Page 17: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

8

1.2.1 Clinical Manifestation

Cataplexy refers to the abrupt, involuntary loss of muscle tone bilaterally during

wakefulness despite preserved consciousness (Fig. 1). It is a pathognomonic symptom of

narcolepsy and represents part of the diagnostic criteria of the disease (Dauvilliers et al., 2003;

Dauvilliers et al., 2014). Narcolepsy manifests most often in the mid-teens or mid-30s, and

although most patients present first with complaints of EDS (often accompanied by sleep

paralysis and/or hypnagogic hallucination), three-quarters eventually develop cataplexy as well

(Guilleminault et al., 1974; Overeem et al., 2001). Cataplexy sometimes presents at the same

time as EDS, but it more often appears years later, and tends to improve with age (Dauvilliers et

al., 2003; Mattarozzi et al., 2008).

Cataplexy can affect all striated muscles, although those that control eye movements and

respiration tend to be affected least, while those in the face, neck, and legs tend to be affected

most (Overeem et al., 2001; Dauvilliers et al., 2014). Individual attacks can involve a partial loss

of muscle tone leading to slurred speech, head drooping, and weakness in the extremities, or

complete muscle paralysis that causes full postural collapse (Dauvilliers et al., 2014).

Unfortunately, partial cataplexy is often ignored by patients and overlooked by physicians, which

may contribute to a delay in the diagnosis of narcolepsy until cataplexy attacks become more

severe years later (Dauvilliers et al., 2014). Cataplexy episodes typically last for seconds but may

persist for minutes, and may occur several times per month or year or multiple times a day (Gelb

et al., 1994). Rarely, some narcoleptic individuals experience an attack that persists for several

hours, known as status cataplecticus, which most often occurs after withdrawal from

anticataplectic medication (Dauvilliers et al., 2003). Despite its abrupt onset, cataplexy rarely

causes injury to narcoleptics since they often sense an attack before it begins and either resist its

onset or orient themselves into a safer position before losing muscle tone (Overeem et al., 2011;

Dauvilliers et al., 2014). Finally, there are autonomic and spinal reflex changes associated with

cataplexy, such as decreased heart rate, brief periods of apnea, absent sympathetic skin

responses, and suppression of the H-reflex (Dauvilliers et al., 2014).

Cataplexy may occur spontaneously but most often is precipitated by an emotional trigger.

A questionnaire study conducted by Overeem et al. in a population of Dutch narcoleptic patients

identified excited laughter as the most common trigger, followed by making a sharp-minded

Page 18: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

9

remark, telling a joke or anticipating its punch line, and being tickled (Overeem et al., 2011)

(Fig. 2). The observation that the most common triggers are specifically humorous in nature

suggests that cataplexy may be an extreme version of “feeling weak with laughter”, which results

from a suppression of the H-reflex (Overeem et al., 1999). Although the emotions that precipitate

cataplexy are typically positive, negative emotions such as anger, fear, or pain have been

documented as triggers for some cataplexy attacks as well (Overeem et al., 2011). Since strong

emotions so often produce cataplexy, it is hypothesized that brain areas involved in processing

emotions, such as the amygdala, are part of the neural pathway underlying cataplexy (Dauvilliers

et al., 2014; Fraigne et al., 2015). Therefore, the major focus of this thesis is on the role the

amygdala, and particularly its central nucleus (CeA), plays in triggering cataplexy attacks.

Page 19: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

10

Figure 1. Electrophysiological representation of cataplexy and wakefulness in a narcoleptic human. In narcoleptic humans, cataplexy (left) is characterized by the abrupt, involuntary loss of skeletal muscle tone during wakefulness. A, Electroencephalographic recordings during cataplexy are typically rich in theta frequencies (4-8 Hz) and may resemble brain activity during REM sleep, whereas wakefulness (right) is characterized by a mix of different frequencies. B-E, Electromyographic recordings demonstrate dramatic decreases in muscle tone during cataplexy in jaw, shoulder, calf, and thigh muscles that sharply contrast the basal level of muscle tone during wakefulness. F, A unique feature of cataplexy is the loss of the H-reflex, which otherwise only occurs during REM sleep. Modified from Guilleminault et al, 1974.

Page 20: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

11

Figure 2. Common cataplexy triggers in narcoleptic humans. A group of narcoleptic individuals who experience cataplexy (n=109) were surveyed regarding behaviors and emotions that preceded their cataplexy attacks. The chart above depicts the percentage of individuals who responded they always, often, sometimes, or never experienced cataplexy following each of the triggers listed. Note that the most common triggers of cataplexy are associated with positive emotions (green), with excited laughter eliciting cataplexy in over 90% of respondents, while neutral (yellow) and negative (red) triggers occur less often. Modified from Overeem et al, 2011.

Page 21: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

12

1.2.2 REM Sleep Intrusion Hypothesis

Although cataplexy is an especially debilitating symptom of narcolepsy, the brain circuits

that cause this symptom to occur have not yet been identified. The defining feature of cataplexy

is muscle atonia, a phenomenon that otherwise only occurs during REM sleep, and cataplexy,

like REM sleep, is typified by theta-rich EEG activity and loss of the H-reflex (Guilleminault et

al., 1974; Peever, 2011; Dauvilliers et al., 2014). Furthermore, sleep paralysis is similar to REM

sleep atonia, hypnagogic hallucinations may reflect the vivid dreaming associated with REM

sleep, and sleep attacks often progress into REM sleep after substantially reduced latency (sleep-

onset REM periods, SOREMPs) (Vogel, 1960; Rechtschaffen et al., 1963; Aldrich, 1993;

Dauvilliers et al., 2003). Finally, some narcoleptic patients report experiencing hypnagogic

hallucinations during cataplexy, while others transition from cataplexy into REM sleep before

resuming normal wakefulness (Vetrugno et al., 2010). These observations have led to the

hypothesis that many symptoms of narcolepsy, and cataplexy in particular, are a consequence of

the inappropriate intrusion of REM sleep atonia into wakefulness (Dauvilliers et al., 2003;

Peever, 2011; Fraigne et al., 2015).

From a neurocircuitry standpoint, REM sleep is thought to be generated by a “flip-flop

switch” mechanism whereby a series of mutually inhibitory REM-on and REM-off regions in the

brainstem tightly control transitions into and out of this state (Lu et al., 2006). A characteristic

feature of REM sleep is muscle atonia, which occurs due to active inhibition and disfacilitation

of spinal motoneurons (Peever, 2011). A core component of the REM-on nuclei, the

subcoeruleus region (SubC, or sublaterodorsal tegmentum), is important for actively inhibiting

motoneurons (Boissard et al., 2002; Lu et al., 2006). The SubC contains a large population of

glutamatergic cells that send excitatory projections to GABA/glycine neurons in parts of the

ventral medial medulla (VMM) such as the ventral gigantocellular reticular nucleus (GIG),

which directly inhibit spinal motoneurons and so suppress muscle tone (Lu et al., 2006; Clement

et al., 2011; Brooks and Peever, 2012). Disfacilitation simultaneously occurs because spinal

motoneurons lose excitatory inputs originating from monoaminergic regions in the brainstem

such as the noradrenergic LC (Peever, 2011; Burgess and Peever, 2013; Dauvilliers et al., 2014).

During wakefulness, orexin neurons help stabilize REM-off areas such as the ventrolateral

periaqueductal grey (vlPAG, which inhibits the SubC), LC, dorsal raphe nucleus (DRN), and

Page 22: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

13

lateral pontine tegmentum (LPT), and in this way are able to indirectly inhibit REM-on areas and

support muscle tone (Peyron et al., 1998; Boissard et al., 2003; Lu et al., 2006; Burgess and

Peever, 2013). Consequently, the loss of orexin neurons in narcolepsy may weaken the REM-off

half of the flip-flop switch, predisposing the REM-on half to being inappropriately recruited and

producing phenomena such as cataplexy (Dauvilliers et al., 2014; Fraigne et al., 2015).

There is substantial evidence suggesting physiological similarity between REM sleep and

cataplexy. For instance, many brain areas show similar patterns of activity during both these

states in humans (Maquet et al., 1996; Hong et al., 2006). In the narcoleptic dog, atonia-

promoting neurons in the VMM, as well as cells in the CeA, were shown to have maximal

activity during both REM sleep and cataplexy (Siegel et al., 1991; Gulyani et al., 2002). The LC,

which helps maintain muscle tone during wakefulness, is virtually silent during both REM sleep

and cataplexy (Wu et al., 1999). Elevating monoamine availability by pharmacological means

such as amphetamines, tricyclic antidepressants, and MAOIs suppress both REM sleep and

cataplexy (Mayer et al., 1995; Dauvilliers et al., 2007; Burgess et al., 2010). Cataplexy and REM

sleep are also characterized by a predomination of theta wave activity in the EEG that peaks at

~7 Hz (although in cataplexy, there is more “slow” theta activity in the 4-6 Hz range) (Vassalli et

al., 2013).

Notably, however, there have been a number of observations that underscore differences

between cataplexy and REM sleep. For instance, some neurons in the VMM that are active

during REM sleep are silent during cataplexy (Siegel et al., 1991). The SubC is only half as

active during cataplexy as it is during REM sleep (Siegel et al., 1992). Wake-on/REM-off

serotonin neurons in the DRN have an intermediate level of activity during cataplexy that is

similar to that of NREM sleep (Wu et al., 2004). Saporin lesions of REM-off neurons in the

vlPAG of narcoleptic mice increase the number of REM sleep bouts without a concomitant

increase in cataplexy (Kaur et al., 2009). Also, cataplexy is associated with preserved

consciousness, which may be related to sustained histaminergic activity that is absent during

REM sleep (John et al., 2004). Finally, REM sleep is homeostatically regulated and has a defined

ultradian rhythm, even in narcolepsy, whereas cataplexy can be triggered at any time and is not

confined to a specific diurnal pattern (Nishino et al., 2000b).

Page 23: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

14

In summary, it appears that most parallels between cataplexy and REM sleep relate to brain

areas or neurotransmitter systems involved in regulating muscle tone. The differences, in

contrast, seem to be associated with other parameters, such as where these states originate in the

brain or whether environmental awareness is preserved. These mixed observations suggest that

cataplexy, despite being similar in many respects to REM sleep, is not the same state per se.

Rather, the neural circuits responsible for generating cataplexy and REM sleep are likely to be

distinct, but the pathways responsible for certain elements of these states, such as those that

produce muscle atonia, may overlap. Thus, the model of cataplexy examined in this thesis is

based on the hypothesis that cataplexy shares the same circuits that are specifically responsible

for muscle atonia during REM sleep (see Section 1.4).

1.2.3 GABA Mechanisms of Cataplexy

The previous section addressed the hypothesis that cataplexy represents an intrusion of

REM sleep into wakefulness. The flip-flop switch model of REM sleep generation is based

largely on mutually inhibitory GABA signaling between different regions in the brainstem, and a

major source of muscle atonia during REM sleep is the GABA-mediated inhibition of

motoneurons (Brooks and Peever, 2008, 2011; Peever, 2011; Brooks and Peever, 2012).

Furthermore, REM-off regions of the brainstem are themselves under GABAergic regulation

from more rostral areas such as the CeA (Burgess et al., 2013). Although the GABAergic

regulation of REM sleep has been well-characterized thus far, little work has been done to date

on GABA mechanisms in cataplexy. However, if cataplexy shares neural circuits with REM

sleep, GABA is likely to play an important role in cataplexy as well. Therefore, studying

GABAergic signaling in the context of cataplexy is not only likely to provide additional insights

into the pathophysiology of this symptom, but also permit additional comparisons between

cataplexy and REM sleep. This section will focus on GABA mechanisms of cataplexy as the cell

population of interest in this study is GABAergic, with reference to REM sleep circuitry as well.

Several GABAergic brain areas important in REM sleep control have also been implicated

in cataplexy. For example, GABAergic neurons in the VMM are highly active during both REM

sleep and cataplexy (Lai and Siegel, 1988; Siegel et al., 1991). Since excitotoxic lesions of the

Page 24: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

15

VMM in healthy cats produce REM sleep without atonia (Schenkel and Siegel, 1989), inhibition

of this region may help rescue muscle tone during cataplexy if it shares the same atonia-

generating pathways as REM sleep. The vlPAG is another brainstem area involved in REM sleep

and potentially cataplexy. It contains a significant number of GABA cells, some of which are

REM-on and may inhibit areas that promote arousal and muscle tone, and others which are

REM-off and thought to inhibit other REM-on nuclei, such as the SubC, outside of REM sleep

(Lu et al., 2006; Sapin et al., 2009; Luppi et al., 2012; Fraigne et al., 2015). REM-on GABA

cells of the vlPAG strongly inhibit the LC, which normally supports muscle tone and is silent

during REM sleep and cataplexy (Gervasoni et al., 1998; Wu et al., 1999; Mileykovskiy et al.,

2000). Moreover, administration of the GABAA receptor agonist muscimol increases REM sleep

and produces SOREMPs reminiscent of those seen in narcolepsy (Sapin et al., 2009). However,

saporin lesions of the vlPAG in narcoleptic mice increase REM sleep without altering cataplexy,

but this may be due to the activity-independent nature of the lesioning (i.e., vlPAG cells were

ablated regardless of whether they were REM-on or REM-off) and that lesions were not

restricted exclusively to GABA cells (Kaur et al., 2009).

Moreover, GABAergic forebrain areas such as the hypothalamus and amygdala may be

involved in the control of cataplexy. Recent evidence suggests that melanin-concentrating

hormone (MCH) neurons in the hypothalamus, which are active during REM sleep, also co-

release GABA onto targets in the brainstem that include the REM-off vlPAG, GABAergic LPT

(which inhibits the SubC), and the monoaminergic nuclei (Boissard et al., 2003; Luppi et al.,

2011; Jego et al., 2013). It has been hypothesized that because MCH neurons promote REM

sleep, they may contribute to the symptoms seen in narcolepsy by inhibiting REM-off circuitry,

particularly because, in narcolepsy, they are unopposed by the wake-promoting actions of orexin

neurons (Hassani et al., 2009). Like hypothalamic MCH neurons, the amygdala is naturally

active during REM sleep (Maquet et al., 1996; Nofzinger et al., 1997), and unit recordings in the

CeA, which is largely GABAergic, have identified cells that are active during cataplexy and

REM sleep (Gulyani et al., 2002). Importantly, it has recently been shown that lesions of the

CeA alleviate cataplexy in narcoleptic mice (Burgess et al., 2013). Because CeA neurons also

project to the REM-off regions referred to above, they are positioned to produce muscle atonia,

especially if they are strongly activated by emotions (Dauvilliers et al., 2014).

Page 25: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

16

Finally, GHB, the most effective drug currently available to treat cataplexy and other

aspects of narcolepsy in humans, may act through a GABA mechanism. It is thought that GHB

weakly agonizes GABAB receptors on interneurons in dopaminergic regions, causing

disinhibition of dopamine cells and facilitating both muscle tone and arousal (Mamelak, 2009;

Kohlmeier et al., 2013a). Furthermore, a specific GHB receptor has been identified that may be

related to GABAA receptors, and activation of the GHB receptor is thought to reduce the

incidence of cataplexy by dampening hypersensitive autoinhibition in the LC (Szabadi, 2015).

Administering GHB to narcoleptic mice in a regimen similar to that in humans produced a

significant decrease in the amount of cataplexy they experienced, including a decrease in

cataplexy density (episodes per hour of wakefulness) (Black et al., 2014). Baclofen, a specific

antagonist of the GABAB receptor, produces even greater reductions in cataplexy, although its

particular influence on the circuits that produce cataplexy has yet to be elucidated (Cruz et al.,

2004; Black et al., 2014).

1.2.4 Other Mechanisms of Cataplexy

Many other neurotransmitter systems have been studied in the context of cataplexy,

particularly those already linked to arousal such as noradrenaline, dopamine, serotonin, and

acetylcholine. The noradrenergic locus coeruleus has received a great deal of attention in the

study of cataplexy because it facilitates muscle tone during wakefulness (via an α1-receptor

mechanism) but is silent during REM sleep (Fung et al., 1991; Takahashi et al., 2010).

Interestingly, the locus coeruleus ceases firing with the start of a cataplexy attack and resumes

firing at its end, suggesting that loss of muscle tone may be due, at least in part, to a removal of

the LC-mediated noradrenergic drive onto motoneurons (Wu et al., 1999). The importance of the

noradrenergic system in cataplexy is underscored by past work wherein α1-receptor agonism

decreased the number of cataplexy episodes while α1-receptor antagonism exacerbated them in

narcoleptic dogs and mice, and that application of α1-receptor agonists in the middle of a

cataplexy episode is capable of rescuing muscle tone (Mignot et al., 1988; Burgess and Peever,

2013). Because the LC is a target of dense excitatory orexinergic innervation, the loss of orexin

cells in narcolepsy may underlie LC hypoactivity during cataplexy (Burgess and Peever, 2013).

Page 26: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

17

These observations may also explain why some of the most effective anticataplectic medications

are also SNRIs.

Dopamine is another neurotransmitter positioned to mediate cataplexy and other elements of

narcolepsy, particularly because it already has established roles in modulating emotion and

reward (Kvetnansky et al., 2009). The dopaminergic system is perturbed in narcolepsy as

evidenced by D2 receptor expression and activity, which is elevated in narcoleptic canines and

humans (Bowersox et al., 1987; Eisensehr et al., 2003). Drugs used to combat cataplexy and

EDS (e.g., modafinil and GHB) are thought to act via dopamine mechanisms (Schmidt-Mutter et

al., 1999; Wisor et al., 2001; Wisor and Eriksson, 2005). Past studies have also demonstrated that

modulating activity of D1 receptors specifically influences sleep attacks, while altering activity

of D2 receptors specifically affects cataplexy (Burgess et al., 2010). More recently, work from

our lab has identified the dopaminergic A11 region of the hypothalamus as important in coupling

arousal and muscle tone, as semi-chronic optogenetic stimulation of this region reduces

cataplexy, while specific stimulation of this region during cataplexy rescues muscle tone within

seconds (Dr. Jimmy Fraigne, unpublished observations). These studies also revealed that the A11

projects to motoneurons (e.g., the trigeminal motor pool) and the CeA, and therefore possesses

requisite anatomical connections to control cataplexy.

Serotonin has been studied in cataplexy due to its relationship with promoting arousal like

other monoamines. In contrast to noradrenaline and dopamine, however, work done on

serotonergic signaling in cataplexy has yielded mixed results. A major source of serotonin in the

brainstem, the DRN, decreases its firing activity during cataplexy, but not to the same extent as

REM sleep (Wu et al., 2004). In narcoleptic canines, serotonin receptor agonists greatly reduce

cataplexy and influence behavior, but serotonin receptor antagonists do not exacerbate cataplexy

(Nishino et al., 1995). These results led the authors to conclude that the anticataplectic effects

observed were more likely due to the changes in behavior induced by serotonin activity, rather

than mediated through the serotonin receptor itself. Despite this, SSRIs are used to manage

cataplexy, even though their specific method of action in narcolepsy is still unknown (Godbout

and Montplaisir, 1986; Lopez and Dauvilliers, 2013).

The cholinergic system has had longstanding role in facilitating REM sleep (McCarley,

2004; Lu et al., 2006), and work over the last few decades have provided evidence of its

Page 27: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

18

involvement of cataplexy as well. For example, the food-elicited cataplexy test produces pontine

spikes in extracellular acetylcholine in narcoleptic canines (Reid et al., 1994a).

Acetylcholinesterase inhibitors and muscarinic receptor agonists increase the frequency of

cataplexy episodes in narcoleptic mice and canines (Reid et al., 1994b; Kalogiannis et al., 2011).

These effects may arise due to glutamatergic potentiation at or direct activation of the SubC by

nearby cholinergic nuclei such as the pedunculopontine tegmentum (PPT) and laterodorsal

tegmentum (LDT), since optogenetic activation of the PPT/LDT induces REM sleep, and the

SubC is activated by cholinergic agonists such as carbachol (Torontali et al., 2014; Weng et al.,

2014; Van Dort et al., 2015).

1.3 The Amygdala

The amygdala refers to a structurally and functionally diverse group of nuclei in the

temporal lobe that receive inputs from and send outputs to an extensive variety of brain regions

(Swanson and Petrovich, 1998). The amygdala is classically associated with emotion processing

and emotional behavior, particularly in the context of fear, although over the last few decades it

has become increasingly apparent that the amygdala processes many other emotions and is an

important component of many other brain circuits (LeDoux, 2007; Murray, 2007; Janak and Tye,

2015). In narcoleptic individuals, strong positive emotions most commonly precipitate cataplexy,

and the brain areas responsible for this are now being elucidated. The amygdala in particular has

become a target of study in this context due to its role in processing emotional stimuli, and there

is now evidence to suggest that one of its nuclei, the CeA, is especially important in triggering

cataplexy (Burgess et al., 2013). The following sections will give a brief overview of amygdala

anatomy and function, followed by its association with cataplexy, with special reference made to

the CeA, the brain area of interest in this thesis project.

1.3.1 Anatomy and Connectivity

Although the specific divisions of the amygdala and their nomenclature have historically

been a point of contention, the region can be subdivided into three sets of nuclei based broadly

Page 28: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

19

on similarities in their histological features and developmental origins: the basolateral group, the

cortical-like group, and the centromedial group (Sah et al., 2003; LeDoux, 2007). The basolateral

and centromedial groups have been studied in the most detail and the former typically refers to

the lateral, basolateral, and basomedial nuclei, whereas the latter is commonly associated with

the central and medial nuclei (Sah et al., 2003).

Despite the vast array of intrinsic and extrinsic projections the amygdala possesses, some

general statements can be made about its anatomical connectivity with other brain areas and

within its nuclei, in addition to the functional importance of these connections. The basolateral

group is regarded as the input region of the amygdala, with the lateral nucleus specifically

functioning as a “gatekeeper” where most sensory information from the cortex and thalamus

relating to vision, audition, olfaction, gustation, and somatosensation arrive (Pitkanen et al.,

1997; Sah et al., 2003; LeDoux, 2007) (Fig. 3). Nuclei in the basolateral group are densely

interconnected and many neurons within these nuclei ultimately send projections to the

centromedial group (Pitkanen et al., 1995; Savander et al., 1995, 1996; Janak and Tye, 2015).

Although the centromedial group is considered the primary output region of the amygdala, it also

receives inputs from the hypothalamus and brainstem that relay autonomic and behavioral

information (Sah et al., 2003).

Most amygdala nuclei send reciprocal projections to areas where their afferent inputs

originate, although there are several salient connections that should be noted. The lateral nucleus

has some direct connections with the centromedial group, but it more often communicates via a

polysynaptic pathway that involves either the basolateral nucleus or the intercalated cell group, a

population of inhibitory interneurons that lie in the fibrous zone separating the basolateral

nucleus from the CeA (LeDoux, 2007). The basolateral group extensively innervates areas

associated with memory, including the hippocampus and perirhinal cortex, as well as the

prefrontal cortex and nucleus accumbens, through predominantly glutamatergic projections (Sah

et al., 2003).

The centromedial group, and particularly the CeA, densely innervates the hypothalamus,

bed nucleus of the stria terminalis, and brainstem regions that regulate the autonomic system and

muscle tone, which collectively produce the physiological and behavioral responses to various

emotions (Davis, 1992; Sah et al., 2003) (Fig. 3). The CeA also projects to areas that mediate

Page 29: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

20

arousal and muscle tone such as the LC and DRN (Sah et al., 2003). In contrast to the basolateral

group, extrinsic projections of the centromedial group are predominantly GABAergic in nature

(Nitecka and Ben-Ari, 1987; McDonald and Augustine, 1993; Pitkanen and Amaral, 1994). On

the basis of cytoarchitecture, the CeA has been further subdivided into as many as four

interconnected parts, although in practice, reference is made primarily to lateral and medial cell

groups (McDonald, 1982; Jolkkonen and Pitkanen, 1998). Neurons in the CeA are chiefly

medium spiny neurons, which form some local synapses but eventually project elsewhere in the

brain (Hall, 1972; McDonald, 1982). Immunohistochemical evidence indicates that other

peptides (e.g., enkephalin, neurotensin, somatostatin, and corticotropin releasing hormone) are

differentially co-expressed in the GABA cells of the CeA (Moga and Gray, 1985; Cassell et al.,

1986; Cassell and Gray, 1989), and the various combinations of peptides expressed in each

neuron may point to distinct functions those neurons subserve (Davis, 1992; Sah et al., 2003).

Moreover, the results of some studies point to a separate, albeit small, population of

glutamatergic CeA neurons (Boissard et al., 2003; Xi et al., 2011).

Page 30: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

21

Figure 3. Schematic depicting the major afferent and efferent pathways of the amygdala. Most sensory information from the cortex and thalamus enter the amygdala through nuclei in the basolateral group, although axons originating in the brainstem and hypothalamus more often terminate in the centromedial group. The basolateral group possesses reciprocal connections with many areas from which it receives afferents. However, most information leaves the amygdala via the central nucleus of the centromedial group, which coordinates physiological and behavioral responses to various stimuli. B, basolateral nucleus; Ce, central nucleus; itc, intercalated cell group; La, lateral nucleus; M, medial nucleus. Modified from LeDoux, 2007.

Page 31: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

22

1.3.2 Functions

The amygdala is part of a network of brain structures that roughly encircle the thalamus that

are collectively called the limbic system (Nakano, 1998). This network is important for the

processing of and reactions to emotional stimuli, as well as related functions including emotional

memory, arousal, and motivation (Janak and Tye, 2015). To help facilitate these roles, the

amygdala in particular is inhibited by local circuits to keep it relatively quiescent so that it

responds only to strong, novel stimuli (LeDoux, 2007). The amygdala has historically been

associated with fear specifically since initial studies of mice, monkeys, and humans with

lesioned amygdalae revealed reduced aggression, fear, and anger, and an inability to associate

stimuli with their emotional content (Sah et al., 2003; Janak and Tye, 2015). Furthermore, fear

conditioning paradigms provide a relatively uncomplicated and direct way of examining the

amygdala in vivo because fear produces a variety of quantifiable changes in behavior and

physiology (e.g., elevated blood pressure and heart rate, release of adrenaline and corticosterone,

freezing behavior, etc.) (Davis, 1992; Sah et al., 2003). Importantly, however, the amygdala

plays a significant role in processing many emotions in addition to fear, including reward and

anxiety, and its influence extends beyond emotion into memory, addiction, olfaction and other

senses, learning-related plasticity, autonomic motor behavior, and more (Swanson and Petrovich,

1998; Murray, 2007; Janak and Tye, 2015). Strong positive emotions represent the most common

trigger for cataplexy, so the role the amygdala plays in processing emotions is emphasized here.

Based on fear conditioning studies, the amygdala has been modeled as an essential structure

for quickly detecting aversive external stimuli and producing adaptive responses to them (Davis,

1992). Indeed, electrically stimulating the amygdala produces fear responses in humans and

animals (Kaada, 1951; Chapman et al., 1954; Davis, 1992). Detailed examinations of the lateral

nucleus revealed many cells that were strongly activated by negative conditioned stimuli, and

optogenetic activation of these neurons can take the place of either a conditioned or

unconditioned stimulus and produce freezing behavior in rats that typifies the rodent fear

response (Quirk et al., 1995; Johansen et al., 2010; Nabavi et al., 2014). Moreover, lesioning all

or part of the amygdala attenuates fear-induced behaviors and produces deficits in fear-motivated

learning (Blanchard and Blanchard, 1972; Hitchcock and Davis, 1986; Vazdarjanova et al.,

2001).

Page 32: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

23

However, there is growing appreciation that the amygdala is important for processing and

acting on positive emotions as well. For example, neuroimaging studies in humans have shown

that the amygdala is activated by positive emotions in addition to negative emotions (Breiter et

al., 1996; Garavan et al., 2001). Neurons in the rat amygdala become excited during anticipation

of reward (Schoenbaum et al., 1998; Tye and Janak, 2007), and neurons within the monkey

amygdala fire in response to positive stimuli of various modalities (Nishijo et al., 1988; Paton et

al., 2006). Lesions within the amygdala weaken reward-based behavior such as conditioned

place preference using amphetamine or food-based positive reinforcement (Gallagher et al.,

1990; Hiroi and White, 1991). Because the amygdala reacts to both subjectively positive and

subjectively negative stimuli, some have hypothesized that this region is important in processing

the intensity of certain emotions, rather than exclusively their affective valence (Breiter et al.,

1996; Garavan et al., 2001; Costa et al., 2010; Bonnet et al., 2015).

1.3.3 The Amygdala and Cataplexy

As mentioned previously, cataplexy is often precipitated by strong positive emotions such as

laughter and joking (Overeem et al., 2011). The amygdala, by virtue of its role in processing

emotion, is therefore an important target of cataplexy research, and there is already much

evidence linking this brain region to the symptom. The amygdala is anatomically well-positioned

to produce muscle paralysis during cataplexy as its main output nucleus, the CeA, has extensive

inhibitory projections to brainstem regions that promote muscle tone such as the LC and vlPAG

(Burgess et al., 2013; Dauvilliers et al., 2014; Fraigne et al., 2015). The CeA may also possess

direct excitatory projections to the SubC, which is important for generating muscle atonia during

REM sleep (Hopkins and Holstege, 1978; Boissard et al., 2003; Lu et al., 2006; Xi et al., 2011).

Brain imaging studies conducted in narcoleptic individuals have found that the amygdala

response to humorous or rewarding stimuli is higher than that of healthy controls (Schwartz et

al., 2008; Ponz et al., 2010). Measurements of cerebral perfusion changes during cataplexy

reveal hyperperfusion in the amygdala for the duration of cataplexy attacks (Hong et al., 2006;

Meletti et al., 2015) (Fig. 4A). Cell recordings of narcoleptic dogs reveal that many neurons in

the CeA specifically have activity that is closely associated with cataplexy attacks (Gulyani et

Page 33: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

24

al., 2002) (Fig. 4B). More recently, the first functional evidence to link the amygdala and

cataplexy came with the observation that in narcoleptic mice, lesions of the CeA significantly

reduced the amount of cataplexy they experienced (Burgess et al., 2013) (Fig. 4C). This same

study identified strong GABAergic projections from the CeA to REM-off areas in the brainstem

stem, suggesting that activation of the CeA may be able to inhibit these regions and so produce

muscle atonia by disinhibiting REM sleep circuitry (Figs. 4D-I).

Moreover, if cataplexy and REM sleep share the same circuits, one would expect the

amygdala to influence the expression of REM sleep or its features. In fact, not only does

amygdala activity naturally increase during REM sleep in healthy individuals (Maquet et al.,

1996; Nofzinger et al., 1997), but manipulations of the amygdala in general and the CeA

specifically alter REM sleep architecture (Fraigne et al., 2015). For example, pharmacological

inhibition of the CeA using TTX reduces REM sleep frequency and duration (Tang et al., 2005;

Sanford et al., 2006), while electrical stimulation of the amygdala increases overall REM sleep

amounts (Smith and Miskiman, 1975). Activation or inhibition of GABAA receptors in the CeA

result in decreases or increases respectively in REM sleep amounts (Sanford et al., 2002).

Injection of 5-HT into of the amygdala during NREM sleep leads to transitions to REM sleep

(Sanford et al., 1995), while injection of carbachol into the CeA, but not other amygdala nuclei,

produces more frequent REM sleep episodes (Calvo et al., 1996). Taken together, these results

point to a putative role of the amygdala and specifically the CeA in regulating REM sleep in

addition to cataplexy, and also underscore the notion that cataplexy and REM sleep share similar

brain circuits.

Page 34: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

25

Figure 4. Summary of evidence implicating the amygdala in cataplexy. Existing evidence identifies the amygdala as being functionally and anatomically well-positioned to promote emotionally-induced cataplexy. A, Functional magnetic resonance imaging demonstrated that the amygdala is highly active during cataplexy attacks in narcoleptic humans. B, Unit recordings in narcoleptic dogs identified many neurons in the CeA with activity tightly correlated to cataplexy attacks. C, Bilateral excitotoxic lesions of the CeA significantly reduced the amount of cataplexy narcoleptic mice experienced. D-F, A retrograde tracer (CTb) injected into the vlPAG/LPT labeled many neurons in the CeA, indicating the presence of CeA-vlPAG/LPT projections. Colocalization of vesicular GABA transporter (VGAT) mRNA in these cells with in situ hybridization implies these CeA projections are GABAergic. G-I, An anterograde tracer (AAV-ChR2-mCherry) injected into the CeA identified strong projections to the vlPAG and LPT, as well as the locus coeruleus and dorsal raphe nucleus. These labeling studies point towards a possible mechanism by which the CeA promotes cataplexy. Amy, amygdala; Aq, aqueduct; BLA, basolateral nucleus of the amygdala; CeA, central nucleus of the amygdala; LPT, lateral pontine tegmentum; scp, superior cerebral peduncle; vlPAG, ventrolateral periaqueductal grey. Scale bars, 250 µm. Modified from Meletti et al, 2015; Gulyani et al, 2002; Burgess et al, 2013.

Page 35: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

26

1.4 Current Model of Cataplexy

The current model of cataplexy is based on the hypothesis that in narcoleptic individuals,

the loss of orexin neurons destabilizes the brainstem circuitry that control muscle atonia during

REM sleep, enabling losses of muscle tone to inappropriately occur during wakefulness

(Dauvilliers et al., 2014; Fraigne et al., 2015) (Fig. 5). Both orexin neurons of the LH and GABA

neurons of the CeA project to muscle tone-promoting regions in the brainstem such as the

vlPAG, LPT, and LC. Although both the orexin and CeA GABA cells are activated by positive

emotions, the excitatory projections of orexin cells to the vlPAG, LPT, and LC are thought to

normally override inhibitory GABA projections from the CeA, and in this way maintain muscle

tone in healthy individuals. In narcolepsy, however, orexin neurons are absent, leaving the

vlPAG, LPT, and LC subject only to GABAergic inputs of the CeA. As a result, when positive

emotions are experienced, GABA CeA cells become activated and inhibit the vlPAG, LPT, and

LC. This leads to a disfacilitation of motoneurons, as well as direct inhibition of motoneurons via

recruitment of the SubC-VMM circuit, similar to what is seen in physiological REM sleep. The

combined effect of these processes is to suppress muscle tone and produce cataplexy.

Page 36: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

27

Figure 5. Hypothesized circuit underlying cataplexy. Cataplexy is thought to occur when the brainstem circuitry that produces REM sleep atonia is activated during wakefulness. During REM sleep, glutamatergic cells of the SubC activate GABA/glycine cells in the VMM, which project to and inhibit spinal motoneurons to produce muscle atonia. During wakefulness, however, this SubC-VMM circuit is inhibited by brainstem regions such as the vlPAG, and muscle tone is further promoted by excitatory noradrenergic projections from the LC. In healthy individuals, the activity of the vlPAG and LC are supported by excitatory orexin inputs from the LH, which override inhibitory GABA inputs originating in the CeA, even when positive emotions are experienced. In narcolepsy, however, the orexin neurons are absent, leaving nothing to balance inputs from GABA CeA cells. As a result, when positive emotions are experienced and GABA CeA cells are strongly activated, they inhibit the vlPAG and LC, leading to the disfacilitation of motoneurons and the recruitment of the SubC-VMM circuit. The combined effect of these processes is to suppress muscle tone and produce cataplexy. CeA, central nucleus of the amygdala; LC, locus coeruleus; LH, lateral hypothalamus; MNs, motoneurons; SubC, subcoeruleus; vlPAG, ventrolateral periaqueductal grey; VMM, ventral medial medulla. From Fraigne, Torontali, Snow, and Peever, 2015.

Page 37: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

28

1.5 Methodological Background

1.5.1 DREADDs

This research project involves studying the role of a discrete population of neurons in

cataplexy, and so a technique with high cellular specificity must be used. Classical techniques

used to study neural circuits such as electrical stimulation, pharmacological manipulation, and

lesioning lack the resolution required to undertake such a study. For this reason, we have chosen

to use a chemogenetic technique known as Designer Receptors Exclusively Activated by

Designer Drugs (DREADDs), which refers to a family of engineered G protein-coupled

receptors that permit manipulation of neurons in a spatially-restricted and phenotype-specific

manner (Rogan and Roth, 2011).

DREADDs, which were first developed by Bryan Roth’s group, are derived from

endogenous human M3 and M4 muscarinic receptors and include the hM3Dq and hM4Di

receptors (Armbruster et al., 2007; Wess et al., 2013). Directed molecular evolution of the

endogenous receptors introduced two point mutations at conserved residues, resulting in a loss of

affinity for their native ligand, acetylcholine, and instead rendering them responsive exclusively

to the synthetic drug known as clozapine-N-oxide (CNO) (Rogan and Roth, 2011). Although

these mutated receptors now interact with a different ligand, they remain coupled to the same

intracellular signaling cascades as the endogenous receptors (Armbruster et al., 2007). The

excitatory DREADD, hM3Dq, is coupled to the Gq signaling pathway, which activates cells via

intracellular calcium release and ERK1/2 activation. The inhibitory DREADD, hM4Di, is

coupled to the Gi signaling pathway, which inactivates cells by inhibiting adenylyl cyclase and

opening potassium channels to cause hyperpolarization (Rogan and Roth, 2011).

The effects of CNO-mediated DREADD activation persist on the order of hours, making

possible the examination of how persistent manipulation of a certain brain region or neuron

population affects behaviors of interest (Krashes et al., 2011). Furthermore, the reversibility of

DREADD-related interventions allows the brain and its circuits to remain fully intact throughout

an experiment while avoiding confounding factors associated with permanent manipulations

such as lesions. The genes encoding the DREADDs can be packaged into an adeno-associated

virus (AAV), which can subsequently be delivered to target brain areas via stereotaxic injection.

Page 38: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

29

Cellular specificity can be achieved using a Cre-Lox system in which the DREADD gene is

double-floxed and inverted (DIO), and Cre recombinase is expressed in neurons of interest

(Rogan and Roth, 2011) (Fig. 6). In this project, the excitatory DREADD, hM3Dq, was used to

induce long-lasting activation of all neurons, or just GABA cells, in the CeA so that the role of

this brain region in producing cataplexy could be examined.

Page 39: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

30

Figure 6. Cre-dependent hM3Dq constructs permit manipulation of specific cell populations. The expression of genes carried by an adeno-associated virus (AAV) can be restricted to certain neuronal types (e.g., GABA cells) using a Cre-Lox system. The AAV contains a genetic construct consisting of a neuron-specific promoter (hSyn), an effector gene (i.e., hM3Dq), and a reporter gene (i.e., mCherry). However, the effector and reporter genes are in an antisense orientation relative to the promoter, such that they cannot be transcribed, and are flanked by 2 sets of Lox sites (double-floxed). Cre recombinase recognizes the Lox sequences and inverts the double-floxed genes, putting them in the same orientation as the promoter and enabling them to be transcribed. Specific expression of Cre-dependent genes becomes possible when Cre recombinase expression is localized to certain cell types (e.g., GABA cells), permitting the study of how those cells in particular influence behaviors of interest (e.g., cataplexy). To achieve expression in all cells, regardless of phenotype, a construct in which the effector and reporter genes are already in the same orientation as the promoter (i.e., they are not double-floxed) can be used instead. Such a construct would be expressed in all successfully transduced cells, regardless of phenotype and independent of Cre recombinase.

Page 40: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

31

1.5.2 Mouse Model

Since human narcolepsy is associated with the loss of a functional orexin system, the animal

model chosen for use in this experiment is the prepro-orexin knockout (orexin-/-) mouse. This

mouse model displays the main symptoms of human narcolepsy such as disturbed REM sleep,

sleep attacks, and cataplexy (Chemelli et al., 1999; Mochizuki et al., 2004; Scammell et al.,

2009). Since the focus of this project is cataplexy, it is important that the mouse model used

replicates this symptom as closely as possible. Cataplexy in orexin-/- mice is characterized by the

abrupt loss of skeletal muscle tone during wakefulness, a theta-rich EEG, and possibly the

maintenance of consciousness (Mochizuki et al., 2004; Scammell et al., 2009; Vassalli et al.,

2013). As in human cataplexy, attacks usually last under 2 min, and end with a rapid restoration

of muscle tone and a return to the activity being performed prior to the episode (Chemelli et al.,

1999; Scammell et al., 2009). Importantly, just as cataplexy is triggered by strong positive

emotions in humans, cataplexy in mice can be triggered by rewarding stimuli such as palatable

food and running wheels (Espana et al., 2007; Clark et al., 2009; Burgess et al., 2013).

In order to manipulate specific types of neurons (i.e., GABA neurons), orexin-/- mice were

crossed with VGAT-Cre mice to produce a new mouse model of narcolepsy, the orexin-/-,VGAT-

Cre mouse. VGAT-Cre mice express Cre recombinase downstream of the gene for the vesicular

GABA transporter (VGAT), which is a marker of GABA cells (Wojcik et al., 2006; Vong et al.,

2011). In this way, Cre recombinase is expressed only in cells where the VGAT gene is

transcribed (i.e., GABA cells). Thus, using Cre-dependent AAV constructs in conjunction with

orexin-/-,VGAT-Cre mice restricts the expression of DREADDs to GABA neurons, thereby

enabling targeted manipulation of these cells in the context of cataplexy.

1.6 Specific Aim

The model outlined in Section 1.4 is consistent with many independent observations, but

there are still components of the cataplexy circuit whose functions are not fully characterized.

Existing evidence suggests that the CeA is an important component of the cataplexy circuit,

especially in the context of emotion-triggered cataplexy attacks. However, the specific role the

Page 41: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

32

CeA plays in generating cataplexy, and the cellular mechanism by which it acts, is not well

understood. We tested the specific hypothesis that GABA neurons in the CeA are the mechanism

through which positive emotions trigger cataplexy in narcolepsy. To do this, we used

chemogenetic, electrophysiological, and behavioral techniques to examine how persistent

manipulations of GABA CeA cells influence cataplexy in narcoleptic mice.

Page 42: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

33

– Methods Chapter 2

2.1 Animals

We used three different lines of mice in these experiments. Male wild-type (i.e., C57BL/6;

age: 19 ± 2 weeks; mass: 25 ± 2 g) and male orexin knockout (orexin-/-; age: 13 ± 2 weeks; mass:

23 ± 1 g) were used to determine how CeA activation impacts behavior. We also used a new

mouse line, which we developed over the last 2 years. This line was developed so that GABA

cells in the CeA could be selectively targeted using either chemogenetic (or optogenetic)

methods. We call these mice orexin-/-,VGAT-Cre (age: 13 ± 2 weeks; mass: 24 ± 2 g) because

they were generated by crossing orexin-/- mice (Chemelli et al., 1999) with VGAT-Cre mice

(Vong et al., 2011). Throughout experiments, mice were housed individually on a 12:12

light/dark cycle (lights off at 1900h) in a temperature- and humidity-controlled environment, had

food and water available ad libitum, and had access to a running wheel. All procedures and

experimental protocols were approved by the University of Toronto’s Local Animal Care

Committee and were in accordance with the Canadian Council on Animal Care.

2.2 Chemogenetic Methods

To activate CeA cells we drove expression of hM3Dq receptors, which when exposed to

clozapine-N-oxide (CNO) cause neuronal activation (Armbruster et al., 2007; Rogan and Roth,

2011). We virally delivered hM3Dq receptors using constructs obtained from the University of

North Carolina Vector Core (Chapel Hill, NC). Constructs were packaged into adeno-associated

viruses with serotype 8 (AAV8), expressed downstream of a neuron-specific promoter (hSyn),

and used mCherry as a reporter gene. For experiments involving orexin-/- mice and wild-type

mice, the construct contained an excitatory hM3Dq receptor (AAV8/hSyn-hM3Dq-mCherry, 4.5

× 1012 particles/mL). For experiments involving orexin-/-,VGAT-Cre mice, the construct

contained either hM3Dq (excitation experiments) or no hM3Dq (empty vector; control

experiments). To ensure cellular specificity in these mice, viral constructs were in a double-

floxed inverted orientation (DIO) such that the genes would be expressed only in cells containing

Page 43: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

34

Cre recombinase (i.e., GABA cells) (excitation: AAV8/hSyn-DIO-hM3Dq-mCherry, 5.7 × 1012

particles/mL; control: AAV8/hSyn-DIO-mCherry, 8.0 × 1012 particles/mL).

2.3 Drug Preparation

The hM3Dq receptor is exclusively activated by the biologically inert ligand CNO. CNO

injections were prepared by dissolving CNO powder (a generous donation from Dr. Bryan Roth)

in a sterile solution of 0.9% saline and 0.05% dimethyl sulfoxide (DMSO) (Sigma-Aldrich). We

administered CNO i.p. in doses of 1 mg/kg for wild-type mice, or 2.5 mg/kg and 5 mg/kg for

orexin-/- and orexin-/-,VGAT-Cre mice, in order to activate hM3Dq receptors on CeA cells; these

doses have been previously shown to activate hM3Dq receptors (Farrell and Roth, 2013; Weber

et al., 2015). Because we found no response difference for doses in orexin-/- mice (i.e., 2.5 mg/kg

and 5 mg/kg CNO all increased cataplexy by the same magnitude), we therefore combined these

doses into a single CNO data set (“CeA Activation”) (see Fig. 7). In all cases, we compared

effects of CNO injection to vehicle injections (“Baseline”) that contained only 0.9% saline and

0.05% DMSO, but no CNO.

2.4 Viral Injection Surgery

Mice were anesthetized using isoflurane (2-5%) and secured in a stereotaxic frame (model

902; David Kopf Instruments). 200 nL of virus was slowly (50 nL/min) infused into the left and

right CeA (1.35 mm posterior to bregma, ±2.75 mm lateral, 4.5 mm ventral). Infusions were

performed through a 28-gauge cannula connected to a digital microinjection syringe pump

(Pump 11 Elite; Harvard Apparatus). Mice were administered ketoprofen (5 mg/kg, s.c.) up to 48

hours after surgery and allowed to recover for at least 14 days.

Page 44: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

35

2.5 EEG and EMG Instrumentation

At the end of this 14-day recovery period, mice underwent a second surgery during which

electroencephalogram (EEG) and electromyogram (EMG) electrodes were implanted in order to

identify sleep states and cataplexy. Electrodes were constructed from multi-stranded stainless

steel wire (AS 632; Cooner Wire). For EEG recordings, we bilaterally implanted stainless steel

screws (P0090CE125; J.I. Morris) into the frontal and parietal bones (1.5 mm anterior and ± 1.5

mm lateral to bregma; 2 mm posterior and ± 2.75 mm lateral to bregma). For EMG recordings,

we sutured two electrodes into the neck extensor muscles and two electrodes into the right

masseter muscle. All EEG and EMG electrodes were soldered to a micro-strip connector (CLP-

105-02-L-D; Electrosonic) that we affixed to the skull using dental cement (Ketac-cem and C&B

Metabond Cement System; K-dental). Mice were administered ketoprofen (5 mg/kg, s.c.) up to

48 hours after surgery and allowed to recover for at least 14 days before experiments began.

2.6 Experimental Protocols

One week after the second surgery, mice were transferred into a plexi-glass recording

chamber containing a running wheel (Bio-Serv). As mice require 10-12 days to maximize their

wheel running (Espana et al., 2007), this provided sufficient time for them to habituate to the

wheel prior to experiments, which began 14 days after being placed into the recording chamber.

Orexin-/- mice experience more cataplexy when they have access to running wheels presumably

because running wheels represent a rewarding stimulus (Lett et al., 2002; Espana et al., 2007;

Burgess et al., 2013). Therefore, we provided mice running wheels in order to study how

rewarding conditions impact cataplexy.

After being transferred to the recording chamber, we gently handled mice at the start of the

dark phase (1900h) on three separate occasions to habituate them to human contact. One week

after being transferred to the recording chamber, we fastened a lightweight recording cable

(CW7117; Cooner Wire) to the micro-strip connector, and gave mice an additional week to

habituate to this cable. During this habituation period mice were given 3 i.p. injections of 0.3 mL

of lactated ringer solution (B. Braun Medical Inc.) to habituate them to i.p. injections.

Page 45: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

36

At the end of this week, we recorded mice up to four consecutive nights for 12 hours each

(beginning at 1900h). No intervention was performed on the first night of recording so that we

could observe the natural behavior of the mice. On the subsequent nights, we randomly gave

mice i.p. injections of saline or CNO at the start of the dark phase (1900h). EEG, EMG, and

video were recorded each night to observe changes in cataplexy and sleep-wake behaviors.

2.7 Data Acquisition

EEG and EMG signals were passed through a Super-Z Head-Stage and a BMA-400

Bioamplifier (CWE Inc.), which amplified signals by a factor of 500. All signals were digitized

at 1000 Hz (Spike 2 Software, 1401 Interface, Cambridge Electronic Design Ltd.), had a DC

offset applied with a time constant of 0.4 s, and were digitally filtered (EEG, 1-100 Hz; EMG,

30-1000 Hz). In signals where electrical noise was detected, a 60 Hz Notch filter was applied in

all recordings for that mouse. EMG signals were rectified. We simultaneously captured and

synchronized videos to electrophysiological recordings to assist in identifying behaviors of

interest.

2.8 Data Analysis

We analyzed the first 6 hours of recording for orexin-/- mice to determine the time course of

any CNO-induced effects on cataplexy and behavior (see Fig. 7E). Because we found CNO

effects persisted for only 3 hours after injection, all subsequent analyses were performed only on

the first 3 hours of the recording period. We scored behavioral states in 5-second epochs based

on EEG, EMG, and video using a custom-made script for Spike 2 (sleepscore v1.01). We

identified wakefulness, non-rapid eye movement (NREM) sleep, and REM sleep using standard

criteria described previously (Burgess et al., 2010; Brooks and Peever, 2012; Burgess and

Peever, 2013). Cataplexy was scored according to the consensus definition criteria: (1) an abrupt

episode of nuchal atonia lasting at least 10 seconds, (2) the mouse is immobile during the

episode, (3) theta activity dominates the EEG during the episode, and (4) at least 40 seconds of

wakefulness precedes the episode (Scammell et al., 2009). Both the electrophysiological signals

Page 46: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

37

and video recording were used to determine the activity the mouse was engaged in immediately

before the attack began. Almost all cataplexy attacks were triggered by wheel running,

grooming, exploring, eating, or drinking. We considered cataplexy episodes without a clear

preceding behavior spontaneous attacks and had the trigger classified as “other”. We identified

sleep attacks on the basis of a gradual loss of neck muscle tone and the appearance of EEG slow

waves, in addition to automatic behaviors such as chewing that were evident in the masseter

EMG (Burgess et al., 2010). Sleep attacks progressed into either wakefulness, NREM sleep, or

REM sleep, and the subsequent state was scored as such.

We quantified the number and length of cataplexy attacks during both CNO and saline (i.e.,

baseline) treatments. We calculated percent time in cataplexy by summing the total time spent in

cataplexy and dividing this value by the length of the recording period. The number of cataplexy

episodes recorded was used to divide the total time spent in cataplexy to determine the average

duration of episodes. We quantified muscle tone during cataplexy in each mouse by normalizing

the average integrated EMG activity of the neck and masseter muscles during cataplexy to that

muscle’s activity during NREM sleep during the saline treatment. EMG activity was quantified

for each muscle using a custom-made Spike 2 script (5s epoch analysis). All calculations

involving behavioral state architecture and muscle tone were done using a custom-made Excel

VBA macro (see Appendix 1). We generated EEG power spectra during cataplexy by performing

a fast Fourier transform of the EEG signal during cataplexy using Spike 2 software. Relative

power was achieved by summing the absolute power of 1-20 Hz and dividing the absolute power

of each intermediate frequency by that sum.

2.9 Histology and Fluorescence In Situ Hybridization

After experiments, mice were deeply anesthetized with Avertin (250 mg/kg, i.p.), produced

from 2,2,2-tribromoethanol and 2-methyl-2-butanol (Sigma-Aldrich), and isoflurane. Upon loss

of the foot withdrawal and righting reflexes, mice were immobilized and transcardially perfused

with 0.1 M phosphate-buffered saline (PBS) and 4% paraformaldehyde (PFA). The brain was

isolated and immersed in 4% PFA for 24 hours, then transferred to a 30% sucrose solution for 48

hours for cryoprotection. Cryoprotected brains were frozen in Tissue-Tek OCT Compound

Page 47: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

38

(Electron Microscopy Sciences) and coronally sectioned at 40 µm using a cryostat (CM3050 S;

Leica). Slices were subsequently mounted on glass slides with Permafluor and expression of

mCherry was visually confirmed with fluorescence microscopy. Only mice in which expression

encompassed the CeA bilaterally were analyzed.

For fluorescence in situ hybridization, after mice were anesthetized, brains were

immediately frozen in –30°C isopentane and stored at –80°C. Brains were coronally sectioned at

16 µm and mounted on glass slides, which were stored at –80°C. Sections containing the CeA

were immersed in 4% PFA for 20 min, then incubated in 0.1 M PBS containing 0.3% H2O2 for

10 min at room temperature. Slices were acetylated for 10 min using 0.1 M TEA buffer

containing 0.25% acetic anhydride, dehydrated with ethanol, then transferred to a humid

chamber saturated with formamide and incubated in a hybridization buffer (40% formamide, 10

mM Tris-HCl, pH 8.0, 200 µg/mL yeast tRNA, 10% dextran sulfate, 1× Denhardt’s solution, 600

mM NaCl, 1 mM EDTA, pH 8.0) for 2 hours at 57.5°C. Sections were next transferred to a

hybridization buffer containing the antisense GAD67 riboprobe (1:1000) and incubated

overnight at 57.5°C, then washed with serial SSC buffers under gentle agitation. Sections were

next incubated in a blocking solution containing 4% goat serum and 0.5% blocking reagent

(Roche) for 1 hour at room temperature, then incubated in a polyclonal mouse antibody to

mCherry (1:1000, T513, Signalway Antibody) for 36 hours at room temperature. At the end of

this period, sections were incubated in sheep anti-DIG-POD (1:500, 11207733910, Roche)

overnight at 4°C, then washed five times for 5 min washes in 0.1 M PBS containing 0.1%

Triton-X (PBS-T) and transferred to a solution containing the TSA Plus Cyanine 5 System

(1:100, NEL745001KT, PerkinElmer) for 10 min. Slices were next incubated in Cy3-conjugated

goat anti-mouse antibodies (1:200, NB7607, Cedarlane) for 3 hours at room temperature then

stained with 4’,6-diamidino-2-phenylindole (DAPI, 1:1000) for 5 min. Slices were left to dry

overnight, then coverslipped with Permafluor and examined with a fluorescent microscope.

2.10 Statistical Analysis

We performed between-group comparisons between using unpaired two-tailed t-tests and

within-group comparisons using paired two-tailed t-tests. For data that was not normally

Page 48: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

39

distributed, we instead used Mann-Whitney U or Wilcoxon signed-rank tests. When more than

two interventions within a group were compared, a one-way repeated measures ANOVA was

used followed by Newman-Keuls multiple comparison post hoc tests. We compared EEG power

spectra using two-way repeated measures ANOVA within groups, or two-way ANOVA without

repeated measures between groups. All statistical analyses were performed using GraphPad

Prism (La Jolla, CA) and had a critical alpha value of p < 0.05 applied. All data are presented as

mean + SEM unless otherwise indicated.

Page 49: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

40

– Results Chapter 3

3.1 CeA activation exacerbates cataplexy in narcoleptic mice

The CeA may have a causal role in triggering cataplexy. For example, CeA cells increase

and decrease their discharge activity at the beginning and end of cataplexy in narcoleptic dogs

(Gulyani et al., 2002), and CeA lesions suppress cataplexy amounts in orexin-/- mice (Burgess et

al., 2013). Therefore, our first goal was to determine if activation of CeA neurons would promote

cataplexy. The CeA is composed of at least two distinct cell populations. A minority of CeA

cells are glutamatergic and have relatively sparse projections beyond the amygdala (Boissard et

al., 2003; Xi et al., 2011), while the majority of CeA cells contain GABA and densely innervate

multiple other brain regions, including those that control muscle tone (Swanson and Petrovich,

1998; Sah et al., 2003; Burgess et al., 2013). Instead of selectively targeting either of these cell

groups we decided to first use a Cre-independent strategy to activate all CeA cells so that we

could establish a functional role for the CeA in mediating cataplexy. We did this by using AAVs

to drive bilateral hM3Dq receptor expression in CeA neurons of orexin-/- mice and then

determined how CNO-induced activation of these receptors influenced cataplexy, sleep attacks,

and general sleep-wake behavior.

We successfully targeted hM3Dq receptors to cells in the left and right CeA in 5 orexin-/-

mice (Figs. 7A-B). CNO-induced activation of CeA cells had no observable impact on typical

waking behaviors such as eating, drinking, exploring, grooming, or wheel running, but it induced

an intense increase in the number of cataplexy attacks relative to baseline conditions (Figs. 7C-

D). Cataplexy amounts were elevated for 3 hours after CNO administration, and returned to

baseline levels thereafter, suggesting the stimulatory effects of CNO on hM3Dq receptors last for

about 3 hours (Fig. 7E). Analysis of group data showed that CeA activation triggered up to 3.5

times more cataplexy during the 3-hour period (1-way RM ANOVA, F=6.288, p=0.0083; Fig.

7F). Importantly, we determined that the changes in cataplexy we observed after CeA activation

were a consequence of CNO because saline injections alone did not alter the levels of cataplexy

mice experienced (compared to when no intervention was given). We also found that both 2.5

mg/kg CNO and 5 mg/kg CNO produced identical changes in cataplexy, and so combined these

doses into a single CNO data set for all subsequent comparisons. CeA activation exacerbated

Page 50: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

41

cataplexy by markedly increasing the number of episodes (paired t-test, t(4)=2.811, p=0.0483;

Fig. 7G), but had no impact on the length of cataplexy events (paired t-test, t(4)=0.2547,

p=0.8115; Fig. 7H). This observation suggests that cells in the CeA play a functional role in

initiating, but not maintaining, cataplexy.

From a behavioral perspective, attacks were indistinguishable from typical baseline

cataplexy in orexin-/- mice. They featured the standard hallmarks of cataplexy in that they were

characterized by an abrupt postural collapse and a sudden loss of EMG tone, and the appearance

of theta-rich EEG activity, both of which punctuated normal waking behavior (see Figs. 10A-B).

Like typical baseline cataplexy, attacks lasted about 50 s and ended with an abrupt and rapid

return to active wakefulness (Fig. 7H). However, we nonetheless wanted to verify these events

were indeed cataplexy and not some other form of behavioral arrest such as sleep attacks. To do

this, we compared EEG power spectral profiles as well as levels of EMG tone during baseline

cataplexy with that elicited during CeA activation. We quantified these two particular variables

because murine cataplexy is characterized by both muscle paralysis (Burgess and Peever, 2013)

and a wake-like, theta-rich cortical EEG (Vassalli et al., 2013). We found that CeA activation

produced cataplexy episodes with levels of muscle tone that were comparable to those during

typical baseline cataplexy (masseter tone: paired t-test, t(4)=0.9472, p=0.3972; neck tone: paired

t-test, t(4)=0.1637, p=0.8779; Figs. 8A-B). We also found that EEG activity peaked in the theta

band during baseline and CeA activation-induced cataplexy, and overall power spectral profiles

were identical in both conditions (2-way RM ANOVA, F=3.458 × 10-4, p=0.9852; Fig. 8C).

Together, these data suggest that the cataplexy produced by CeA activation is physiologically

and behaviorally identical to attacks that occur during baseline conditions in orexin-/- mice.

Because the amygdala has been implicated in sleep regulation (Smith and Miskiman, 1975;

Calvo et al., 1996; Sanford et al., 2006), we wanted to determine if CeA activation would affect

either sleep-wake behavior or sleep attacks, which are common in orexin-/- mice. Although CNO-

induced CeA activation triggered marked increases in cataplexy, this same intervention had no

measureable effect on amounts of wakefulness (paired t-test, t(4)=2.315, p=0.0816), NREM

sleep (paired t-test, t(4)=1.1319, p=0.2576), or REM sleep (paired t-test, t(4)=0.6870, p=0.5299),

nor did it affect sleep attacks (paired t-test, t(4)=2.497, p=0.0670; Fig. 8D). These findings

indicate that CeA activation does not have broad effects on behavior; rather, its activation

selectively targets the mechanisms mediating cataplexy.

Page 51: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

42

Figure 7. CeA activation exacerbates cataplexy in narcoleptic mice. A, Stereotaxic map displaying the location of expression of the Cre-independent hM3Dq construct in the CeA of 5 orexin-/- mice. B, The extent of expression was determined on the basis of mCherry fluorescence. A representative sample of mCherry-positive neurons is shown, as is a single neuron (inset). C-D, Hypnograms displaying behavioral states of orexin-/- mice during baseline conditions (saline) or CeA activation (CNO). E, Compared to baseline, CNO produced increases in cataplexy that persisted for the first 3 hours after injection, but amounts of cataplexy during the next 3 hours were identical to that seen during baseline conditions. For this reason, only the first 3 hours after CNO injection were examined in detail. F, Following global CeA activation, a large increase in the total time spent in cataplexy was observed following CNO, but not saline, injections. There was no difference in the effects produced by 2.5 mg/kg CNO or 5 mg/kg CNO, so they were combined into a single CNO dataset for all subsequent comparisons. G, The increase in cataplexy arose from an increase in the number of cataplexy episodes mice experienced. H, The average duration of cataplexy episodes did not change. BLA, basolateral nucleus of the amygdala; ic, internal capsule; W, wakefulness; NREM, NREM sleep; REM, REM sleep; SA, sleep attacks; CAT, cataplexy. *, p < 0.05 compared to no intervention and saline or baseline as appropriate.

Page 52: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

43

Figure 8. The behavioral arrests triggered by CeA activation are cataplexy. A-B, In 5 orexin-/-

mice, activation of all CeA cells produced behavioral arrests characterized by similar levels of muscle tone in the masseter and neck muscles compared to typical baseline cataplexy. C, These arrests also had similar EEG spectral profiles to baseline cataplexy (data presented as mean ± SEM). D, CeA activation did not produce any changes in the overall amounts of wakefulness (W), NREM sleep (NREM), or REM sleep (REM), nor did it alter sleep attacks (inset).

Page 53: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

44

3.2 CeA activation does not cause cataplexy in wild-type mice

Next, we wanted to verify that cataplexy was only triggered by CeA activation in orexin-/-

mice, and that it could not be triggered by CeA stimulation in wild-type mice with an intact

orexin system. Therefore, we used a Cre-independent strategy to drive hM3Dq expression in

CeA cells of wild-type mice (n=4), then examined how CNO-induced activation of these cells

affected behavior. Using EEG, EMG and videography, we found no evidence indicating that

CeA activation causes cataplexy in wild-type mice. Unlike orexin-/- mice that experienced as

many as 12 cataplexy attacks during the 3-hour recording period (see Fig. 7G), we saw no

evidence of postural collapses or intermittent losses of EMG tone during waking behaviors in

wild-type mice after CeA stimulation. We also found no evidence that CeA activation influenced

the overall amount or architecture of sleep-wake behavior (Figs. 9A-C). These findings suggest

that the orexin system protects against cataplexy, and that CeA activation only promotes

cataplexy in narcoleptic mice that already lack an intact orexin signaling system (see Fig. 15).

Although previous studies show that the amygdala can promote REM sleep (Calvo et al., 1996;

Sanford et al., 2006), our results indicate that CeA activation does not affect sleep-wake

behavior.

Page 54: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

45

Figure 9. CeA activation does not alter sleep-wake architecture in wild-type mice. A, Bilateral activation of all CeA cells in wild-type mice (n=4) did not influence the amounts of time they spent in wakefulness (W), NREM sleep (NREM), or REM sleep (REM). CeA activation produced no differences in the B, frequency or C, duration of W, NREM, or REM, nor was there evidence of overt behaviors resembling cataplexy.

Page 55: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

46

3.3 Orexin-/- mice that express Cre in GABA neurons exhibit

typical cataplexy

Having shown that general CeA stimulation promotes cataplexy, our next step was to

identify which type of CeA cell is responsible for this effect. Here, we test the role for GABA

neurons because they are the primary extrinsic pathway from the CeA and they are anatomically

connected to nuclei that regulate skeletal muscle tone (Swanson and Petrovich, 1998; Sah et al.,

2003; Burgess et al., 2013) (see Fig. 15). However, to understand how GABA cells in the CeA

regulate cataplexy, we needed to develop a new narcoleptic mouse line in which we could

selectively target and manipulate these cells exclusively. To do this, we crossed orexin-/- mice

with VGAT-Cre mice (Chemelli et al., 1999; Vong et al., 2011) to produce so-called orexin-/-

,VGAT-Cre mice so that Cre-dependent AAVs could be used to deliver hM3Dq receptors

directly to GABA cells in the CeA. However, before using these new mice we needed to

determine whether they behave like typical orexin-/- mice, and we did this by comparing amounts

of cataplexy, sleep attacks, NREM/REM sleep and wakefulness in orexin-/- (n=5) and orexin-/-

,VGAT-Cre (n=13) mice.

First, we show that general sleep-wake behavior is unaffected in orexin-/-,VGAT-Cre mice.

Specifically, we found that orexin-/- and orexin-/-,VGAT-Cre mice spent the same amount of time

in wakefulness, NREM sleep, and REM sleep, and that the length and number of each behavioral

state was comparable (Table 2). We also found that the number and length of sleep attacks was

similar between genotypes (Table 2). Together, these results suggests that orexin-/-,VGAT-Cre

mice are phenotypically indistinguishable from orexin-/-mice.

Next, we showed that cataplexy was behaviorally and electrophysiologically identical in

orexin-/-,VGAT-Cre and orexin-/- mice. In both mouse lines we found that cataplexy occurred

during active wakefulness when mice were engaged in purposeful activities such as wheel

running, grooming, exploring, eating, and drinking. Attacks were characterized by abrupt

postural collapse and rapid loss of EMG tone (Figs. 10A-B). Mice remained immobile

throughout attacks, which were terminated by a swift return of skeletal muscle tone and

resumption of prior waking activities (Figs. 10A-B). Levels of EMG tone and cortical EEG

activity during cataplexy were similar in both orexin-/-,VGAT-Cre and orexin-/- mice (masseter

Page 56: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

47

EMG: t-test, t(11)=1.525, p=0.1554; neck EMG: t-test, t(13)=0.4438; p=0.6645; EEG: 2-way

ANOVA, F=2.756 × 10-3, p=0.9582; Figs. 10C-E).

Both mouse lines also spent comparable amounts of time in cataplexy. During the 3-hour

recording period orexin-/- and orexin-/-,VGAT-Cre mice spent 2.3 ± 0.5% and 1.7 ± 0.5% of their

time in cataplexy (Mann-Whitney U test, U(16)=22.50, p=0.3488; Fig. 10F). They also had the

same number of attacks that lasted for the same amount of time. Orexin-/- mice experienced 5 ± 1

attacks that lasted for 53 ± 8s and orexin-/-,VGAT-Cre mice experienced 3 ± 1 attacks that lasted

for 49 ± 7s (number: Mann-Whitney U test, U(16)=16.50, p=0.1248; duration: t-test,

t(16)=0.3527, p=0.7289; Figs. 10G-H). Together, these data indicate that orexin-/-,VGAT-Cre

mice exhibit cataplexy that is behaviorally and electrophysiologically indistinguishable from that

of orexin-/- mice, making this new mouse line useful for determining how GABA cell

manipulation affects cataplexy.

Page 57: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

48

Page 58: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

49

Figure 10. Orexin-/- mice that express Cre in GABA neurons exhibit typical cataplexy. A, In orexin-/- mice, cataplexy is characterized by the abrupt loss of skeletal muscle tone during wakefulness and a theta-rich electroencephalogram (EEG). B, Magnification of boxed area in A. Orexin-/-,VGAT-Cre mice express cataplexy electrophysiologically indistinguishable from that seen in conventional orexin-/- mice as measured by C, masseter electromyogram (EMG), D, neck EMG, and E, EEG spectral power. F-H, There are no significant differences in the time spent in, number of episodes, or average duration of cataplexy attacks of orexin-/-,VGAT-Cre mice compared to orexin-/- mice. A.U., arbitrary units.

Page 59: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

50

3.4 hM3Dq receptors are expressed in GABA cells of the CeA

Our next step was to determine whether Cre-dependent AAV delivery targeted GABA cells

in the CeA. In 13 orexin-/-,VGAT-Cre mice, we successfully targeted and bilaterally delivered

hM3Dq receptors to GABA cells in the left and right CeA (Fig. 11A). To verify the specificity of

hM3Dq expression, we used fluorescence in situ hybridization to examine the distribution of

GAD67 (a specific marker of GABA cells) in the CeA. We found that not only was the CeA

densely populated by GAD67-positive neurons, but that hM3Dq receptors extensively co-

localized with these cells (Fig. 11B), suggesting that our subsequent CNO manipulations would

primarily target GABA cells in the CeA.

Page 60: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

51

Figure 11. hM3Dq receptors are expressed in GABA cells of the CeA. A, Stereotaxic map displaying the location of expression of the Cre-dependent hM3Dq construct in the CeA of 13 orexin-/-,VGAT-Cre mice. B, The extent of expression was determined on the basis of mCherry fluorescence (red, top left panel; single mCherry-positive neuron showed in inset), which encompassed the CeA where GAD67-positive cells predominate (green, top right panel; single GAD67-positive neuron shown in inset). Most cells (identified by DAPI, blue) expressing mCherry also expressed GAD67 (bottom left, middle, and right panels), while cells that did not express mCherry rarely expressed GAD67.

Page 61: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

52

3.5 GABA cells in the CeA promote cataplexy

Our last aim was to determine whether GABA cells in the CeA are responsible for

promoting cataplexy. In 13 orexin-/-,VGAT-Cre mice, we successfully delivered hM3Dq

receptors to GABA cells in both the left and right CeA (see Figs. 11A-B), and found that CNO-

induced activation of these cells triggered a robust increase in cataplexy that was identical in

effect to that produced by activating the entire CeA cell population. Cataplexy occupied 3.7

times more of the 3-hour recording period after GABA cell activation than it did before (paired t-

test, t(12)=4.573; p=0.0006; Figs. 12A-C). Increases in cataplexy were due to an increase in the

number of attacks, rather than the duration of attacks. Before CeA activation mice experienced 5

± 1 episodes in 3 hours, but during GABA cell stimulation they experienced 16 ± 3 arrests during

the same time period (Wilcoxon signed-rank test, W(12)=-89; p=0.0021; Fig. 12D). In contrast,

CeA stimulation had no effect on the length of cataplexy episodes (baseline vs. CNO: 43 ± 6s

and 42 ± 4s; paired t-test, t(12)=0.1341, p=0.8956; Fig. 12E), suggesting that GABA CeA cells

gate the entrance into, but not the exit from, cataplexy.

Once again, we wanted to confirm that the behavioral arrests produced by specifically

activating GABA CeA cells were in fact cataplexy. While these events were behaviorally

identical to typical baseline cataplexy, we again performed a rigorous comparison of the

electrophysiological features that characterized these attacks. We found that activation of GABA

CeA cells produced cataplexy attacks with levels of muscle tone (masseter EMG: paired t-test,

t(9)=1.056 × 10-7, p=1.0000; neck EMG: paired t-test t(11)=0.2364, p=0.8175; Figs. 13A & B)

and EEG activity (2-way RM ANOVA, F=0.001451, p=0.9697; Fig. 13C) indistinguishable

from baseline cataplexy. These observations suggest that activating GABA CeA cells is capable

of evoking cataplexy attacks.

Next, we wanted to verify that activation of GABA CeA cells did not affect sleep-wake

behavior. We found that CNO-induced CeA stimulation had no effect on overall amounts of

NREM sleep (paired t-test, t(12)=1.526; p=0.1529), REM sleep (Wilcoxon signed-rank test,

W(12)=24; p=0.3804) or sleep attacks (paired t-test, t(12)=0.03691; p=0.9712), but GABA cell

activation did decrease amounts of wakefulness by ~7% (paired t-test, t(12)=3.437; p=0.0049;

Fig. 13D). However, this change was actually due to the substantial increase in the amount of

time spent in cataplexy, and this apparent decrease in wakefulness disappeared when time awake

Page 62: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

53

and time in cataplexy were combined (paired t-test, t(12)=0.8996; p=0.3861; Fig. 13E).

Together, these results indicate that activation of GABA CeA neurons promotes cataplexy, but

does not influence sleep-wake activity.

Lastly, we needed to demonstrate that increases in cataplexy were in fact mediated by CNO-

induced activation of hM3Dq receptors on CeA cells. To do this we drove bilateral expression of

the physiologically inert mCherry protein in GABA CeA cells in orexin-/-,VGAT-Cre mice

(n=6). As expected, we found that CNO application had no effect on cataplexy amounts in these

“hM3Dq-negative” mice (paired t-test, t(5)=0.8153, p=0.4520; Fig. 12C). CNO injection in

hM3Dq-negative mice had no impact on either the number (paired t-test, t(5)=1.360, p=0.2321)

or duration (paired t-test, t(5)=1.530, p=0.1867) of cataplexy attacks (Fig. 12D-E). These

findings demonstrate that CNO administration triggers cataplexy by activating GABA CeA cells

through an hM3Dq-dependent mechanism. They also suggest that neither CNO application nor

mCherry expression within the CeA impacts cataplexy.

Page 63: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

54

Figure 12. GABA cells in the CeA promote cataplexy. A-B, Hypnograms displaying behavioral states of one orexin-/-

,VGAT-Cre mouse during baseline conditions and activation of GABA CeA cells. C, During activation of GABA CeA cells only in 13 orexin-/-,VGAT-Cre mice (denoted as “hM3Dq”), a large increase in the total time spent in cataplexy was observed. This same increase was not apparent in a group of 6 hM3Dq-negative orexin-/-,VGAT-Cre mice (denoted as “mCherry”). D, This cataplexy change arose from an increase in the number of cataplexy episodes hM3Dq-expressing mice experienced. Mice lacking the hM3Dq receptor experienced similar numbers of cataplexy episodes after CNO injection. E, The average duration of cataplexy episodes did not change following CNO injection in hM3dq-positive and hM3Dq-negative mice. W, wakefulness; NREM, NREM sleep; REM, REM sleep; SA, sleep attacks; CAT, cataplexy. BLA, basolateral nucleus of the amygdala; ic, internal capsule. ***, p < 0.001; **, p < 0.01.

Page 64: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

55

Figure 13. Activating GABA CeA cells does not influence sleep-wake behavior.A-B, In 13 orexin-/-,VGAT-Cre mice, activation of GABA CeA cells produced behavioral arrests characterized by similar levels of muscle tone in the masseter and neck muscles compared to typical baseline cataplexy. C, These arrests also had similar EEG spectral profiles to baseline cataplexy (data presented as mean ± SEM), suggesting they were indeed cataplexy. D, GABA CeA activation did not produce any changes in the overall amounts NREM sleep (NREM), or REM sleep (REM), nor did it alter sleep attacks (inset), but it did decrease the amount of wakefulness (W) mice experienced. E, However, this decrease in wakefulness was an artifact resulting from mice spending significantly more of their waking time in cataplexy, and was nullified when time awake and time in cataplexy were combined. **, p < 0.01

Page 65: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

56

3.6 GABA cells in the CeA mediate cataplexy associated with

rewarding stimuli

In narcoleptic mice cataplexy can occur spontaneously, but it is typically associated with

positive or rewarding stimuli such as wheel running, grooming, and palatable food (Chemelli et

al., 1999; Espana et al., 2007; Clark et al., 2009; Burgess et al., 2013). However, the neural

mechanism by which such stimuli elicit cataplexy remains unidentified. Because the CeA is

involved in processing positive emotions and rewarding stimuli and is anatomically connected to

structures that mediate muscle tone (Murray, 2007; Burgess et al., 2013), we hypothesize that

GABA CeA cells could be a neural substrate through which these stimuli trigger cataplexy. To

test this hypothesis, we documented the types of behaviors that triggered cataplexy under

baseline conditions and how they changed during GABA CeA activation in 13 orexin-/-,VGAT-

Cre mice.

We found that virtually all cataplexy attacks occurred when mice were engaged in

purposeful behaviors such as wheel running, grooming, exploring, eating, or drinking, and rarely

occurred during non-purposeful behaviors such as sitting stationary (Fig. 14A). During baseline

recordings, 77% of cataplexy events occurred when mice were either wheel running or

grooming, and only 23% of events occurred spontaneously or when mice were exploring, eating,

or drinking (Fig. 14A). Remarkably, CeA activation almost exclusively triggered cataplexy

attacks associated with either wheel running or grooming, increasing cataplexy triggered during

wheel running by 385% (Wilcoxon signed-rank test, W(12)=-78; p=0.0025) and during grooming

by 182% (paired t-test, t(12)=2.878; p=0.0139) (Fig. 14B). However, there were proportionally

more attacks triggered by wheel running after GABA CeA activation at the expense of attacks

triggered by other activities (Fig. 14A). Cataplexy associated with exploring (paired t-test,

t(12)=0, p=1.0000), eating (Wilcoxon signed-rank test, W(12)=-34, p=0.0843), drinking

(Wilcoxon signed-rank test, W(12)=-20, p=0.1725), and other behaviors (Wilcoxon signed-rank

test, W(12)=-1, p=1.0000), did not change during CeA activation (Fig. 14B), suggesting that

GABA CeA cells selectively facilitate cataplexy associated with rewarding stimuli such as wheel

running.

Page 66: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

57

Because CeA stimulation proportionally increased cataplexy triggered by wheel running

and produced the largest increase in running-triggered cataplexy attacks, we wanted to determine

whether this intervention impacted cataplexy expression by changing levels of wheel running

activity. Although CeA activation did not change overall amounts of wheel running (paired t-

test, t(12)=0.5476, p=0.5940) or the number of running bouts (W(12)=-54, p=0.0639), it

significantly shortened running bout length (t(12)=2.366, p=0.0356; Figs. 14C-E). Running

bouts were shortened because they were punctuated by more frequent cataplexy attacks during

CeA activation. During baseline conditions, cataplexy occurred every 43 ± 13 min of wheel

running, but during CeA activation it occurred every 10 ± 3 min of wheel running, thereby

increasing the likelihood of running-induced cataplexy by 77% (paired t-test, t(9)=3.138;

p=0.0120; Fig. 14F). We therefore suggest that activation of GABA CeA neurons lowers the

threshold for triggering cataplexy associated rewarding stimuli such as wheel running.

Page 67: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

58

Figure 14. GABA cells in the CeA mediate cataplexy associated with rewarding stimuli. A, In orexin-/-

,VGAT-Cre mice (n=13), cataplexy episodes were most often triggered by rewarding behaviors such as wheel running and grooming under baseline conditions. During GABA CeA activation, proportionally more cataplexy attacks were triggered by wheel running. B, GABA CeA activation significantly increased in the number of cataplexy episodes triggered by wheel running and grooming, without influencing cataplexy triggered by other behaviors. C, The increase in running-triggered cataplexy episodes was not due to more wheel running, since mice spent the same fraction of their time awake engaged in this activity. D-E, The number of wheel running bouts did not change, but the length of running bouts significantly decreased, as cataplexy more often interrupted wheel running. F, GABA CeA activation caused mice to experience cataplexy after significantly less wheel running compared to baseline conditions, indicating a reduction in the stimulus threshold required to trigger cataplexy. *, p < 0.05; **, p < 0.01.

Page 68: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

59

– Discussion Chapter 4

Our current results indicate that positive stimuli may trigger cataplexy in orexin-/- mice by

recruiting GABA neurons in the CeA. We found that activation of GABA CeA cells potently

increased the frequency of cataplexy attacks without influencing their length, demonstrating that

GABA cells play a role in triggering, but not maintaining, cataplexy. We also found that GABA

cell activation only promotes cataplexy associated with rewarding conditions, suggesting that

positive stimuli trigger cataplexy by activating GABA cells in the CeA.

4.1 The amygdala promotes cataplexy associated with positive

emotion

Although the link between positive emotions and cataplexy was identified over 130 years

ago, the pathways by which emotions trigger cataplexy remains speculative. A longstanding

hypothesis in sleep medicine is that the amygdala plays a causal role in mediating cataplexy

(Luppi et al., 2012; Dauvilliers et al., 2014; Fraigne et al., 2015). This idea is based on the fact

that the amygdala is involved in processing emotional stimuli (Murray, 2007; Bonnet et al.,

2015; Janak and Tye, 2015), and that cataplexy is typically elicited by positive stimuli (Nishino

and Mignot, 1997; Espana et al., 2007; Clark et al., 2009; Overeem et al., 2011). Therefore, in

the present study, we set out to test the role the amygdala plays in producing cataplexy

associated with positive emotional stimuli.

Evidence indicates that the amygdala is functionally capable of mediating cataplexy, and

may be a key component of the pathway by which positive stimuli elicit cataplexy. For example,

imaging studies indicate that amygdala activity increases during cataplexy associated with

emotional stimuli in humans (Hong et al., 2006; Meletti et al., 2015), and that the intensity of the

amygdala response to positive stimuli is abnormally high in narcoleptics (Schwartz et al., 2008;

Ponz et al., 2010). While these observations suggest that amygdala activity in response to

positive stimuli may trigger cataplexy, it is unclear whether these changes in amygdala activity

are a cause or consequence of cataplexy. Our current results support the idea that the amygdala,

and particularly the CeA, plays a functional role in mediating emotion-triggered cataplexy,

Page 69: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

60

because, in examining the behaviors that immediately preceded cataplexy attacks, we found that

CeA activation preferentially increased cataplexy attacks associated with rewarding stimuli such

as wheel running. These findings are consistent with the hypothesis that the amygdala, by virtue

of its role in processing emotions, is important in initiating emotionally-induced cataplexy

attacks, and also complement work demonstrating that CeA lesions reduce cataplexy associated

with rewarding stimuli in orexin-/- mice (Burgess et al., 2013).

The amygdala also influences generalized arousal and motivated behaviors (Davis and

Whalen, 2001; Sah et al., 2003; Tye and Janak, 2007; Robinson et al., 2014; Janak and Tye,

2015), and may promote REM sleep (Sanford et al., 1995; Calvo et al., 1996; Maquet et al.,

1996; Sanford et al., 2002; Sanford et al., 2006). However, we found that CeA stimulation had

no effect on either sleep-wake activity or the time mice spent running on wheels (a potential

index of motivated behaviors), suggesting that changes in cataplexy are specific and primarily

associated with augmented amygdala activity rather than secondary effects arising from

behavioral changes associated with CeA activation. Furthermore, the fact that CeA activation

increased cataplexy without concurrently influencing REM sleep may imply that these two

states, while similar behaviorally, are ultimately governed by separate executive mechanisms.

However, we also performed our experiments during the dark phase when mice spend less time

asleep overall, and mice were given access to a running wheel which increases and consolidates

wakefulness at the expense of sleep (Espana et al., 2007). As these conditions are not permissive

to REM sleep, this too may explain why we did not observe substantial changes in this sleep

state. Finally, we did not detect any changes in the time mice experienced sleep attacks, the other

major symptom of narcolepsy, suggesting that cataplexy and sleep attacks are distinct symptoms

controlled by different neural mechanisms.

4.2 GABA cells in the CeA mediate cataplexy

Despite evidence suggesting the amygdala is the neural substrate through which emotions

trigger cataplexy, the specific cellular mechanism by which the amygdala promotes cataplexy

had, until now, remained unidentified. Our current data suggest that GABA cells in the CeA play

a permissive role in controlling cataplexy. We show that targeted stimulation of GABA CeA

Page 70: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

61

cells triggered a 3.5-fold increase in cataplexy attacks, most of which were associated with

rewarding conditions like wheel running, without affecting the length of cataplexy attacks.

Moreover, the magnitude of this increase was comparable to that seen when CeA cells were

activated independent of their neurochemical phenotype, suggesting that the influence of this

brain area on cataplexy is principally or perhaps exclusively mediated by GABA cells in the

CeA. This finding is particularly interesting because it suggests that previously identified non-

GABAergic, and possibly glutamatergic, neurons originating in the CeA and projecting directly

to the SubC may be less important for cataplexy, despite potentially playing a role in other

functions (e.g., REM sleep) (Boissard et al., 2003; Xi et al., 2011). We therefore suggest that

GABA neurons represent the main cellular mechanism in the CeA through which positive

stimuli trigger cataplexy.

Our results are consistent with the current mechanistic model of cataplexy, which posits that

cataplexy occurs when positive emotions lead to the untimely recruitment of brainstem circuits that

inhibit muscle tone (Dauvilliers et al., 2014; Fraigne et al., 2015). We hypothesize that GABA cells

in the CeA function as a “relay centre” between the cortical structures that interpret emotional

stimuli and the brainstem circuits that generate motor paralysis during cataplexy (Fig. 15). This

hypothesis is based on data showing that rewarding conditions activate the medial prefrontal

cortex (mPFC), which innervates circuits within the amygdala (Vertes, 2004; Etkin et al., 2011;

Oishi et al., 2013). Connections between the mPFC and CeA are integral in promoting cataplexy

because removing either of them suppresses cataplexy in orexin-/- mice (Burgess et al., 2013;

Oishi et al., 2013). GABA cells, which form the primary extrinsic pathway from the CeA (Nitecka

and Ben-Ari, 1987; Pitkanen and Amaral, 1994; Sah et al., 2003), innervate the LC, LPT, and

vlPAG, which collectively function to facilitate waking muscle tone by silencing atonia-generating

regions in the dorsal pons (e.g., the SubC). We hypothesize that positive emotions elicit cataplexy by

initially activating the mPFC, which subsequently activates GABA CeA cells that inhibit the LC,

LPT, and vlPAG and thereby generate muscle paralysis (Fig. 15).

However, positive emotions do not trigger cataplexy in humans or animals that do not have

narcolepsy. We hypothesize that the functional orexin system healthy individuals possess is

necessary to protect against CeA-mediated cataplexy. Specifically, GABA-mediated inhibition

originating from the CeA is offset by excitatory signaling from orexin neurons, which are also active

during rewarding conditions (e.g., grooming and social reunion) (Lee et al., 2005; Mileykovskiy et

Page 71: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

62

al., 2005; Siegel and Boehmer, 2006; Blouin et al., 2013; Giardino and de Lecea, 2014), and

innervate the same structures as GABA CeA cells (e.g., LC, LPT, and vlPAG) (Peyron et al.,

1998; Kiyashchenko et al., 2001; Mieda and Sakurai, 2012; Hasegawa et al., 2014). The loss of

orexin signaling in narcolepsy upsets this balance, enabling GABA CeA cells to inhibit the LC, LPT,

and vlPAG without opposition and thereby create an environment conducive to cataplexy (Fig. 9).

Furthermore, orexin neurons innervate the amygdala and may help control its activity, as orexin

levels in the amygdala increase during positive emotions and are closely correlated with

hypothalamic orexin activity (Blouin et al., 2013). Finally, orexin signaling may also stabilize

other monoaminergic or cholinergic cell groups which themselves influence muscle tone and

arousal, thereby preventing dissociated states such as cataplexy from occurring (see Section

1.2.4).

Page 72: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

63

Figure 15. The amygdala acts as a “relay centre” between the cortex and the pons that enables positive emotions to elicit cataplexy. Positive emotions activate cortical areas such as the mPFC, which in turn activates the CeA. GABA cells in the CeA then inhibit midbrain regions including the LC, LPT, and vlPAG, which inhibit the atonia-generating network in the brainstem or facilitate muscle tone directly. Inhibition of these midbrain regions disinhibits the SubC-GIG circuit, which produces muscle paralysis during cataplexy by inhibiting skeletal motoneurons. In healthy individuals with an intact orexin system, CeA-mediated inhibition of the LC, LPT, and vlPAG is counterbalanced by excitatory orexin inputs, which prevent positive emotions from triggering muscle atonia and cataplexy. CeA, central nucleus of the amygdala; GIG, gigantocellular nucleus; LC, locus coeruleus; LPT, lateral pontine tegmentum; mPFC, medial prefrontal cortex; SubC, subcoeruleus; vlPAG, ventrolateral periaqueductal grey.

Page 73: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

64

4.3 GABA cells in the CeA are responsible for initiating cataplexy

Our current results indicate that the CeA plays a role in triggering, but not maintaining,

cataplexy because we show that CeA activation increases the frequency, but not the duration, of

cataplexy attacks. If CeA activity contributed to the length of cataplexy, its activation should

have produced status cataplecticus (prolonged periods of cataplexy) (Overeem et al., 2001;

Dauvilliers et al., 2007; Dauvilliers et al., 2014). Since we found no change in the length of

cataplexy episodes, our results suggest that the CeA functions primarily as a cataplexy “on

switch” by gating the entrance into cataplexy, while other brain areas are responsible for

maintaining and/or terminating cataplexy attacks. The circuit mechanisms that function to switch

off cataplexy remain unidentified.

The CeA may increase the incidence of cataplexy by lowering the stimulus threshold for

attack induction. To test this hypothesis, we examined wheel-running behavior in our orexin-/-

mice as an index of this threshold. Although CeA activation did not change overall amounts of

wheel running in our mice, it significantly shortened the length of individual running bouts

because they were punctuated by more frequent cataplexy attacks. Moreover, CeA stimulation

caused mice to experience cataplexy attacks after 77% less wheel running compared to control

conditions, suggesting that CeA activation lowered the threshold stimulus required to trigger

cataplexy. These findings are consistent with the idea that the amygdala acts as a cataplexy “on

switch” during positive emotions, particularly in light of the observation that cataplexy occurs

most often in response to positive emotions in humans (Overeem et al., 2011), and that the

probability of an attack is significantly elevated in narcoleptic animals exposed to positive

stimuli such as palatable food and running wheels (Nishino and Mignot, 1997; Espana et al.,

2007; Clark et al., 2009). These findings are also consistent with studies demonstrating that the

amygdala response to positive emotions is heightened in human narcolepsy (Schwartz et al.,

2008; Ponz et al., 2010), which may indicate that affected individuals are “primed” to experience

cataplexy.

Page 74: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

65

4.4 Additional circuits involved in cataplexy

Additional brain circuits may be involved in producing cataplexy. For instance,

neuroimaging studies of narcoleptic humans have identified that, like the amygdala, other

emotion-processing centres such as the ventral tegmental area and nucleus accumbens are highly

active during cataplexy and may be anatomically positioned to influence muscle tone

independent of the CeA (Groenewegen and Russchen, 1984; Usuda et al., 1998; Meletti et al.,

2015). Moreover, recent work in narcoleptic mice provides evidence for a cataplexy-promoting

circuit originating in the mPFC but parallel to the CeA that involves MCH neurons of the

hypothalamus (Oishi et al., 2013). Like the CeA, these neurons project to brainstem areas that

regulate muscle tone and appear to be active during cataplexy. Cells in the zona incerta,

laterodorsal tegmentum, and dorsal pons have been shown to suppress cataplexy (Burlet et al.,

2002; Liu et al., 2011; Blanco-Centurion et al., 2013; Oishi et al., 2013). Drugs that affect

cholinergic tone (e.g., cholinesterase inhibitors) can also worsen cataplexy, suggesting

involvement of the cholinergic system in the cataplexy circuit (Reid et al., 1994a; Kalogiannis et

al., 2011; Kohlmeier et al., 2013b), while the noradrenergic, serotonergic, and dopaminergic

systems may also contribute to cataplexy since their activation can influence cataplexy (Mignot

et al., 1988; Mieda et al., 2004; Liu et al., 2008; Burgess et al., 2010; Burgess and Peever, 2013;

Hasegawa et al., 2014). Our current results indicate that GABA cells in the CeA play a major role

initiating cataplexy attacks, but it is likely that other brain regions and neurotransmitter systems, such

as those reviewed here, contribute to this in addition to other features of cataplexy attacks, such as

their length. Although multiple brain circuits, including GABA cells in the CeA, are clearly

involved in controlling cataplexy, it remains unclear how they communicate with and influence

one another, and understanding how these systems function together represents a major

challenge in identifying circuit mechanisms of cataplexy.

4.5 Technical considerations

While our study provides strong functional evidence that GABA cells in the CeA mediate

cataplexy triggered by positive emotions, it cannot identify the specific extra-amygdalar circuits

and brain targets by which these cells exert their effects. However, most extrinsic projections of

Page 75: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

66

the CeA are descending GABAergic projections that innervate brainstem regions associated with

various autonomic functions, with dense projections to those that regulate muscle tone such as

the LC, LPT, and vlPAG (Nitecka and Ben-Ari, 1987; Pitkanen and Amaral, 1994; Burgess et

al., 2013). The CeA is therefore anatomically positioned to influence cataplexy through one or

more of these nuclei, and although the contribution of CeA inputs to each of these regions in the

context of cataplexy is beyond the scope of this study, it can be evaluated with other more

spatially-restricted technologies currently available (e.g., optogenetics). Additionally, though we

could not identify the activation profile of CeA neurons in relation to cataplexy, a great deal of

evidence suggests that amygdala activity is tightly correlated to cataplexy attacks. For example,

recordings of individual CeA neurons in narcoleptic dogs demonstrate a close association of

firing activity to the onset and offset of cataplexy attacks, while cataplexy in humans is

characterized by hyperperfusion within the amygdala (Gulyani et al., 2002; Hong et al., 2006).

To identify the functional role of GABA CeA cells in regulating cataplexy, we developed a

new transgenic orexin-/- mouse line that enabled us to specifically target and manipulate GABA

cells. After detailed analysis we showed that orexin-/-,VGAT-Cre mice were phenotypically

indistinguishable from orexin-/- mice, because they experience the same quality and quantity of

cataplexy, as well as comparable amounts of NREM sleep, REM sleep, wakefulness, and sleep

attacks as conventional orexin-/- mice. We also showed that hM3Dq receptors could be virally

delivered to GABA cells, and that mass activation of GABA cells in the CeA produced robust

and consistent increases in cataplexy incidence. These mice could be a valuable resource for

dissecting the roles that other GABA circuits (e.g., GABA vlPAG cells) play in controlling

cataplexy and other features of narcolepsy such as excessive sleepiness. They will also be useful

for determining how GABA mechanisms influence other behaviors that are orexin-dependent

(e.g., addiction).

4.6 Conclusions

Our current data provide the first evidence to show that positive or rewarding stimuli may

trigger cataplexy by recruiting GABA CeA cells in orexin-/- mice. We propose that GABA CeA

cells function as a “relay center” between the cortical structures that interpret emotional stimuli

Page 76: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

67

and the brainstem circuits that trigger motor paralysis during cataplexy. Our findings are

consistent with studies showing amygdala activation during cataplexy in narcoleptic humans and

dogs, and lesion studies in narcoleptic mice demonstrating that the amygdala promotes

cataplexy. The CeA is a primary output nucleus of the amygdala and is especially important in

coordinating the body’s physiological response to emotion (LeDoux, 2007). Part of this response

may include transient losses of muscle tone, as healthy individuals often experience feeling

“weak with laughter” which is associated with a diminished monosynaptic H-reflex (Overeem et

al., 1999). In narcolepsy, cataplexy may represent an extreme motor response of the body to

emotion in which not just the H-reflex but all postural muscle tone is completely lost

(Guilleminault et al., 1974), and the neural origin of cataplexy may involve unfettered CeA

output that arises with the loss of orexin neurons. Our results also point to the GABA CeA cells

as a possible target of sodium oxybate, the most effective anti-cataplectic medication to date,

which is thought to inhibit neurons via a GABAB receptor-mediated pathway (Black et al.,

2014), suggesting that targeted reductions in amygdala activity could be used to alleviate

cataplexy in narcolepsy. In addition to identifying a novel amygdala circuit involved in

generating cataplexy, our work provides a foundation with which to examine the interactions

amygdala cells have with other brain areas in the context of cataplexy, and underscores the

potential in developing novel cataplexy therapies aimed at mitigating amygdala activity.

Page 77: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

68

References

Ahmed SS, Volkmuth W, Duca J, Corti L, Pallaoro M, Pezzicoli A, Karle A, Rigat F, Rappuoli R, Narasimhan V, Julkunen I, Vuorela A, Vaarala O, Nohynek H, Pasini FL, Montomoli E, Trombetta C, Adams CM, Rothbard J, Steinman L (2015) Antibodies to influenza nucleoprotein cross-react with human hypocretin receptor 2. Sci Transl Med 7:294ra105.

Aldrich MS (1993) The neurobiology of narcolepsy-cataplexy. Prog Neurobiol 41:533-541.

American Academy of Sleep Medicine (2014) International Classification of Sleep Disorders, 3rd Edition. Darien, IL: American Academy of Sleep Medicine.

Andlauer O, Moore Ht, Hong SC, Dauvilliers Y, Kanbayashi T, Nishino S, Han F, Silber MH, Rico T, Einen M, Kornum BR, Jennum P, Knudsen S, Nevsimalova S, Poli F, Plazzi G, Mignot E (2012) Predictors of hypocretin (orexin) deficiency in narcolepsy without cataplexy. Sleep 35:1247-1255F.

Armbruster BN, Li X, Pausch MH, Herlitze S, Roth BL (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A 104:5163-5168.

Black SW, Morairty SR, Chen TM, Leung AK, Wisor JP, Yamanaka A, Kilduff TS (2014) GABAB agonism promotes sleep and reduces cataplexy in murine narcolepsy. J Neurosci 34:6485-6494.

Blanchard DC, Blanchard RJ (1972) Innate and conditioned reactions to threat in rats with amygdaloid lesions. J Comp Physiol Psychol 81:281-290.

Blanco-Centurion C, Liu M, Konadhode R, Pelluru D, Shiromani PJ (2013) Effects of orexin gene transfer in the dorsolateral pons in orexin knockout mice. Sleep 36:31-40.

Blouin AM, Fried I, Wilson CL, Staba RJ, Behnke EJ, Lam HA, Maidment NT, Karlsson KAE, Lapierre JL, Siegel JM (2013) Human hypocretin and melanin-concentrating hormone levels are linked to emotion and social interaction. Nat Commun 4:1547.

Boissard R, Fort P, Gervasoni D, Barbagli B, Luppi PH (2003) Localization of the GABAergic and non-GABAergic neurons projecting to the sublaterodorsal nucleus and potentially gating paradoxical sleep onset. Eur J Neurosci 18:1627-1639.

Boissard R, Gervasoni D, Schmidt MH, Barbagli B, Fort P, Luppi PH (2002) The rat ponto-medullary network responsible for paradoxical sleep onset and maintenance: a combined microinjection and functional neuroanatomical study. Eur J Neurosci 16:1959-1973.

Bonnet L, Comte A, Tatu L, Millot JL, Moulin T, Medeiros de Bustos E (2015) The role of the amygdala in the perception of positive emotions: an "intensity detector". Front Behav Neurosci 9:178.

Bowersox SS, Kilduff TS, Faull KF, Zeller-DeAmicis L, Dement WC, Ciaranello RD (1987) Brain dopamine receptor levels elevated in canine narcolepsy. Brain Res 402:44-48.

Page 78: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

69

Breiter HC, Etcoff NL, Whalen PJ, Kennedy WA, Rauch SL, Buckner RL, Strauss MM, Hyman SE, Rosen BR (1996) Response and habituation of the human amygdala during visual processing of facial expression. Neuron 17:875-887.

Brooks PL, Peever JH (2008) Unraveling the mechanisms of REM sleep atonia. Sleep 31:1492-1497.

Brooks PL, Peever JH (2011) Impaired GABA and glycine transmission triggers cardinal features of rapid eye movement sleep behavior disorder in mice. J Neurosci 31:7111-7121.

Brooks PL, Peever JH (2012) Identification of the transmitter and receptor mechanisms responsible for REM sleep paralysis. J Neurosci 32:9785-9795.

Broughton R, Valley V, Aguirre M, Roberts J, Suwalski W, Dunham W (1986) Excessive daytime sleepiness and the pathophysiology of narcolepsy-cataplexy: a laboratory perspective. Sleep 9:205-215.

Broughton RJ, Guberman A, Roberts J (1984) Comparison of the psychosocial effects of epilepsy and narcolepsy/cataplexy: a controlled study. Epilepsia 25:423-433.

Burgess CR, Scammell TE (2012) Narcolepsy: neural mechanisms of sleepiness and cataplexy. J Neurosci 32:12305-12311.

Burgess CR, Peever JH (2013) A noradrenergic mechanism functions to couple motor behavior with arousal state. Curr Biol 23:1719-1725.

Burgess CR, Tse G, Gillis L, Peever JH (2010) Dopaminergic regulation of sleep and cataplexy in a murine model of narcolepsy. Sleep 33:1295-1304.

Burgess CR, Oishi Y, Mochizuki T, Peever JH, Scammell TE (2013) Amygdala lesions reduce cataplexy in orexin knock-out mice. J Neurosci 33:9734-9742.

Burlet S, Tyler CJ, Leonard CS (2002) Direct and indirect excitation of laterodorsal tegmental neurons by Hypocretin/Orexin peptides: implications for wakefulness and narcolepsy. J Neurosci 22:2862-2872.

Calvo JM, Simon-Arceo K, Fernandez-Mas R (1996) Prolonged enhancement of REM sleep produced by carbachol microinjection into the amygdala. Neuroreport 7:577-580.

Cassell MD, Gray TS (1989) Morphology of peptide-immunoreactive neurons in the rat central nucleus of the amygdala. J Comp Neurol 281:320-333.

Cassell MD, Gray TS, Kiss JZ (1986) Neuronal architecture in the rat central nucleus of the amygdala: a cytological, hodological, and immunocytochemical study. J Comp Neurol 246:478-499.

Chapman WP, Schroeder HR, Geyer G, Brazier MA, Fager C, Poppen JL, Solomon HC, Yakovlev PI (1954) Physiological evidence concerning importance of the amygdaloid

Page 79: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

70

nuclear region in the integration of circulatory function and emotion in man. Science 120:949-950.

Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T, Lee C, Richardson JA, Williams SC, Xiong Y, Kisanuki Y, Fitch TE, Nakazato M, Hammer RE, Saper CB, Yanagisawa M (1999) Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell 98:437-451.

Clark EL, Baumann CR, Cano G, Scammell TE, Mochizuki T (2009) Feeding-elicited cataplexy in orexin knockout mice. Neuroscience 161:970-977.

Clement O, Sapin E, Berod A, Fort P, Luppi PH (2011) Evidence that Neurons of the Sublaterodorsal Tegmental Nucleus Triggering Paradoxical (REM) Sleep Are Glutamatergic. Sleep 34:419-423.

Costa VD, Lang PJ, Sabatinelli D, Versace F, Bradley MM (2010) Emotional imagery: assessing pleasure and arousal in the brain's reward circuitry. Hum Brain Mapp 31:1446-1457.

Cruz HG, Ivanova T, Lunn ML, Stoffel M, Slesinger PA, Luscher C (2004) Bi-directional effects of GABA(B) receptor agonists on the mesolimbic dopamine system. Nat Neurosci 7:153-159.

Dauvilliers Y, Billiard M, Montplaisir J (2003) Clinical aspects and pathophysiology of narcolepsy. Clin Neurophysiol 114:2000-2017.

Dauvilliers Y, Arnulf I, Mignot E (2007) Narcolepsy with cataplexy. Lancet 369:499-511.

Dauvilliers Y, Siegel JM, Lopez R, Torontali ZA, Peever JH (2014) Cataplexy--clinical aspects, pathophysiology and management strategy. Nat Rev Neurol 10:386-395.

Davis M (1992) The role of the amygdala in fear and anxiety. Annu Rev Neurosci 15:353-375.

Davis M, Whalen PJ (2001) The amygdala: vigilance and emotion. Mol Psychiatry 6:13-34.

De la Herran-Arita AK, Garcia-Garcia F (2013) Current and emerging options for the drug treatment of narcolepsy. Drugs 73:1771-1781.

de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS, 2nd, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM, Sutcliffe JG (1998) The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci U S A 95:322-327.

Eisensehr I, Linke R, Tatsch K, von Lindeiner H, Kharraz B, Gildehaus FJ, Eberle R, Pollmacher T, Schuld A, Noachtar S (2003) Alteration of the striatal dopaminergic system in human narcolepsy. Neurology 60:1817-1819.

Espana RA, McCormack SL, Mochizuki T, Scammell TE (2007) Running promotes wakefulness and increases cataplexy in orexin knockout mice. Sleep 30:1417-1425.

Page 80: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

71

Etkin A, Egner T, Kalisch R (2011) Emotional processing in anterior cingulate and medial prefrontal cortex. Trends Cogn Sci 15:85-93.

Farrell MS, Roth BL (2013) Pharmacosynthetics: Reimagining the pharmacogenetic approach. Brain Res 1511:6-20.

Fraigne JJ, Torontali ZA, Snow MB, Peever JH (2015) REM Sleep at its Core - Circuits, Neurotransmitters, and Pathophysiology. Front Neurol 6:123.

Fromherz S, Mignot E (2004) Narcolepsy research: past, present, and future perspectives. Arch Ital Biol 142:479-486.

Fung SJ, Manzoni D, Chan JY, Pompeiano O, Barnes CD (1991) Locus coeruleus control of spinal motor output. Prog Brain Res 88:395-409.

Gallagher M, Graham PW, Holland PC (1990) The amygdala central nucleus and appetitive Pavlovian conditioning: lesions impair one class of conditioned behavior. J Neurosci 10:1906-1911.

Garavan H, Pendergrass JC, Ross TJ, Stein EA, Risinger RC (2001) Amygdala response to both positively and negatively valenced stimuli. Neuroreport 12:2779-2783.

Gelb M, Guilleminault C, Kraemer H, Lin S, Moon S, Dement WC, Mignot E (1994) Stability of cataplexy over several months--information for the design of therapeutic trials. Sleep 17:265-273.

Gélineau J (1880) De la narcolepsie. Gazette des Hôpitaux 53:626-628.

Gervasoni D, Darracq L, Fort P, Souliere F, Chouvet G, Luppi PH (1998) Electrophysiological evidence that noradrenergic neurons of the rat locus coeruleus are tonically inhibited by GABA during sleep. Eur J Neurosci 10:964-970.

Giardino WJ, de Lecea L (2014) Hypocretin (orexin) neuromodulation of stress and reward pathways. Curr Opin Neurobiol 29:103-108.

Godbout R, Montplaisir J (1986) The effect of zimelidine, a serotonin-reuptake blocker, on cataplexy and daytime sleepiness of narcoleptic patients. Clin Neuropharmacol 9:46-51.

Groenewegen HJ, Russchen FT (1984) Organization of the efferent projections of the nucleus accumbens to pallidal, hypothalamic, and mesencephalic structures: a tracing and immunohistochemical study in the cat. J Comp Neurol 223:347-367.

Guilleminault C, Wilson RA, Dement WC (1974) A study on cataplexy. Arch Neurol 31:255-261.

Gulyani S, Wu MF, Nienhuis R, John J, Siegel JM (2002) Cataplexy-related neurons in the amygdala of the narcoleptic dog. Neuroscience 112:355-365.

Page 81: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

72

Hagan JJ et al. (1999) Orexin A activates locus coeruleus cell firing and increases arousal in the rat. Proc Natl Acad Sci U S A 96:10911-10916.

Hall E (1972) The amygdala of the cat: a Golgi study. Z Zellforsch Mikrosk Anat 134:439-458.

Hara J, Beuckmann CT, Nambu T, Willie JT, Chemelli RM, Sinton CM, Sugiyama F, Yagami K, Goto K, Yanagisawa M, Sakurai T (2001) Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30:345-354.

Hasegawa E, Yanagisawa M, Sakurai T, Mieda M (2014) Orexin neurons suppress narcolepsy via 2 distinct efferent pathways. J Clin Invest 124:604-616.

Hassani OK, Lee MG, Jones BE (2009) Melanin-concentrating hormone neurons discharge in a reciprocal manner to orexin neurons across the sleep-wake cycle. Proc Natl Acad Sci U S A 106:2418-2422.

Hiroi N, White NM (1991) The lateral nucleus of the amygdala mediates expression of the amphetamine-produced conditioned place preference. J Neurosci 11:2107-2116.

Hitchcock J, Davis M (1986) Lesions of the amygdala, but not of the cerebellum or red nucleus, block conditioned fear as measured with the potentiated startle paradigm. Behav Neurosci 100:11-22.

Hong SB, Tae WS, Joo EY (2006) Cerebral perfusion changes during cataplexy in narcolepsy patients. Neurology 66:1747-1749.

Hopkins DA, Holstege G (1978) Amygdaloid projections to the mesencephalon, pons and medulla oblongata in the cat. Exp Brain Res 32:529-547.

Janak PH, Tye KM (2015) From circuits to behaviour in the amygdala. Nature 517:284-292.

Jego S, Glasgow SD, Herrera CG, Ekstrand M, Reed SJ, Boyce R, Friedman J, Burdakov D, Adamantidis AR (2013) Optogenetic identification of a rapid eye movement sleep modulatory circuit in the hypothalamus. Nat Neurosci 16:1637-1643.

Johansen JP, Hamanaka H, Monfils MH, Behnia R, Deisseroth K, Blair HT, LeDoux JE (2010) Optical activation of lateral amygdala pyramidal cells instructs associative fear learning. Proc Natl Acad Sci U S A 107:12692-12697.

John J, Wu MF, Siegel JM (2000) Systemic administration of hypocretin-1 reduces cataplexy and normalizes sleep and waking durations in narcoleptic dogs. Sleep Res Online 3:23-28.

John J, Wu MF, Boehmer LN, Siegel JM (2004) Cataplexy-active neurons in the hypothalamus: implications for the role of histamine in sleep and waking behavior. Neuron 42:619-634.

Jolkkonen E, Pitkanen A (1998) Intrinsic connections of the rat amygdaloid complex: projections originating in the central nucleus. J Comp Neurol 395:53-72.

Page 82: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

73

Kaada BR (1951) Somato-motor, autonomic and electrocorticographic responses to electrical stimulation of rhinencephalic and other structures in primates, cat, and dog; a study of responses from the limbic, subcallosal, orbito-insular, piriform and temporal cortex, hippocampus-fornix and amygdala. Acta Physiol Scand Suppl 24:1-262.

Kalogiannis M, Hsu E, Willie JT, Chemelli RM, Kisanuki YY, Yanagisawa M, Leonard CS (2011) Cholinergic modulation of narcoleptic attacks in double orexin receptor knockout mice. PLoS One 6:e18697.

Kaur S, Thankachan S, Begum S, Liu M, Blanco-Centurion C, Shiromani PJ (2009) Hypocretin-2 saporin lesions of the ventrolateral periaquaductal gray (vlPAG) increase REM sleep in hypocretin knockout mice. PLoS One 4:e6346.

Kiyashchenko LI, Mileykovskiy BY, Lai YY, Siegel JM (2001) Increased and decreased muscle tone with orexin (hypocretin) microinjections in the locus coeruleus and pontine inhibitory area. J Neurophysiol 85:2008-2016.

Kohlmeier KA, Vardar B, Christensen MH (2013a) gamma-Hydroxybutyric acid induces actions via the GABAB receptor in arousal and motor control-related nuclei: implications for therapeutic actions in behavioral state disorders. Neuroscience 248:261-277.

Kohlmeier KA, Tyler CJ, Kalogiannis M, Ishibashi M, Kristensen MP, Gumenchuk I, Chemelli RM, Kisanuki YY, Yanagisawa M, Leonard CS (2013b) Differential actions of orexin receptors in brainstem cholinergic and monoaminergic neurons revealed by receptor knockouts: implications for orexinergic signaling in arousal and narcolepsy. Front Neurosci 7:246.

Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, Maratos-Flier E, Roth BL, Lowell BB (2011) Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest 121:1424-1428.

Kvetnansky R, Sabban EL, Palkovits M (2009) Catecholaminergic systems in stress: structural and molecular genetic approaches. Physiol Rev 89:535-606.

Lai YY, Siegel JM (1988) Medullary regions mediating atonia. J Neurosci 8:4790-4796.

LeDoux J (2007) The amygdala. Curr Biol 17:R868-874.

Lee MG, Hassani OK, Jones BE (2005) Discharge of identified orexin/hypocretin neurons across the sleep-waking cycle. J Neurosci 25:6716-6720.

Leschziner G (2014) Narcolepsy: a clinical review. Pract Neurol 14:323-331.

Lett BT, Grant VL, Koh MT, Flynn G (2002) Prior experience with wheel running produces cross-tolerance to the rewarding effect of morphine. Pharmacol Biochem Behav 72:101-105.

Liblau RS, Vassalli A, Seifinejad A, Tafti M (2015) Hypocretin (orexin) biology and the pathophysiology of narcolepsy with cataplexy. Lancet Neurol 14:318-328.

Page 83: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

74

Lin JS, Dauvilliers Y, Arnulf I, Bastuji H, Anaclet C, Parmentier R, Kocher L, Yanagisawa M, Lehert P, Ligneau X, Perrin D, Robert P, Roux M, Lecomte JM, Schwartz JC (2008) An inverse agonist of the histamine H(3) receptor improves wakefulness in narcolepsy: studies in orexin-/- mice and patients. Neurobiol Dis 30:74-83.

Lin L, Faraco J, Li R, Kadotani H, Rogers W, Lin X, Qiu X, de Jong PJ, Nishino S, Mignot E (1999) The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell 98:365-376.

Liu M, Thankachan S, Kaur S, Begum S, Blanco-Centurion C, Sakurai T, Yanagisawa M, Neve R, Shiromani PJ (2008) Orexin (hypocretin) gene transfer diminishes narcoleptic sleep behavior in mice. Eur J Neurosci 28:1382-1393.

Liu M, Blanco-Centurion C, Konadhode R, Begum S, Pelluru D, Gerashchenko D, Sakurai T, Yanagisawa M, van den Pol AN, Shiromani PJ (2011) Orexin gene transfer into zona incerta neurons suppresses muscle paralysis in narcoleptic mice. J Neurosci 31:6028-6040.

Longstreth WT, Jr., Koepsell TD, Ton TG, Hendrickson AF, van Belle G (2007) The epidemiology of narcolepsy. Sleep 30:13-26.

Lopez R, Dauvilliers Y (2013) Pharmacotherapy options for cataplexy. Expert Opin Pharmacother 14:895-903.

Lu J, Sherman D, Devor M, Saper CB (2006) A putative flip-flop switch for control of REM sleep. Nature 441:589-594.

Luppi PH, Clement O, Sapin E, Peyron C, Gervasoni D, Leger L, Fort P (2012) Brainstem mechanisms of paradoxical (REM) sleep generation. Pflugers Arch 463:43-52.

Luppi PH, Clement O, Sapin E, Gervasoni D, Peyron C, Leger L, Salvert D, Fort P (2011) The neuronal network responsible for paradoxical sleep and its dysfunctions causing narcolepsy and rapid eye movement (REM) behavior disorder. Sleep Med Rev 15:153-163.

Mamelak M (2009) Narcolepsy and depression and the neurobiology of gammahydroxybutyrate. Prog Neurobiol 89:193-219.

Maquet P, Peters J, Aerts J, Delfiore G, Degueldre C, Luxen A, Franck G (1996) Functional neuroanatomy of human rapid-eye-movement sleep and dreaming. Nature 383:163-166.

Mattarozzi K, Bellucci C, Campi C, Cipolli C, Ferri R, Franceschini C, Mazzetti M, Russo PM, Vandi S, Vignatelli L, Plazzi G (2008) Clinical, behavioural and polysomnographic correlates of cataplexy in patients with narcolepsy/cataplexy. Sleep Med 9:425-433.

Mayer G, Ewert Meier K, Hephata K (1995) Selegeline hydrochloride treatment in narcolepsy. A double-blind, placebo-controlled study. Clin Neuropharmacol 18:306-319.

Page 84: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

75

McCarley RW (2004) Mechanisms and models of REM sleep control. Arch Ital Biol 142:429-467.

McDonald AJ (1982) Cytoarchitecture of the central amygdaloid nucleus of the rat. J Comp Neurol 208:401-418.

McDonald AJ, Augustine JR (1993) Localization of GABA-like immunoreactivity in the monkey amygdala. Neuroscience 52:281-294.

Meletti S, Vaudano AE, Pizza F, Ruggieri A, Vandi S, Teggi A, Franceschini C, Benuzzi F, Nichelli PF, Plazzi G (2015) The Brain Correlates of Laugh and Cataplexy in Childhood Narcolepsy. J Neurosci 35:11583-11594.

Mieda M, Sakurai T (2012) Overview of orexin/hypocretin system. Prog Brain Res 198:5-14.

Mieda M, Willie JT, Hara J, Sinton CM, Sakurai T, Yanagisawa M (2004) Orexin peptides prevent cataplexy and improve wakefulness in an orexin neuron-ablated model of narcolepsy in mice. Proc Natl Acad Sci U S A 101:4649-4654.

Mignot E, Guilleminault C, Bowersox S, Rappaport A, Dement WC (1988) Role of central alpha-1 adrenoceptors in canine narcolepsy. J Clin Invest 82:885-894.

Mignot E, Lin X, Arrigoni J, Macaubas C, Olive F, Hallmayer J, Underhill P, Guilleminault C, Dement WC, Grumet FC (1994) DQB1*0602 and DQA1*0102 (DQ1) are better markers than DR2 for narcolepsy in Caucasian and black Americans. Sleep 17:S60-67.

Mileykovskiy BY, Kiyashchenko LI, Siegel JM (2005) Behavioral correlates of activity in identified hypocretin/orexin neurons. Neuron 46:787-798.

Mileykovskiy BY, Kiyashchenko LI, Kodama T, Lai YY, Siegel JM (2000) Activation of pontine and medullary motor inhibitory regions reduces discharge in neurons located in the locus coeruleus and the anatomical equivalent of the midbrain locomotor region. J Neurosci 20:8551-8558.

Mochizuki T, Crocker A, McCormack S, Yanagisawa M, Sakurai T, Scammell TE (2004) Behavioral state instability in orexin knock-out mice. J Neurosci 24:6291-6300.

Moga MM, Gray TS (1985) Evidence for corticotropin-releasing factor, neurotensin, and somatostatin in the neural pathway from the central nucleus of the amygdala to the parabrachial nucleus. J Comp Neurol 241:275-284.

Murray EA (2007) The amygdala, reward and emotion. Trends Cogn Sci 11:489-497.

Nabavi S, Fox R, Proulx CD, Lin JY, Tsien RY, Malinow R (2014) Engineering a memory with LTD and LTP. Nature 511:348-352.

Nakano I (1998) The limbic system: An outline and brief history of its concept. Neuropathology 18:211-214.

Page 85: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

76

Nishijo H, Ono T, Nishino H (1988) Single neuron responses in amygdala of alert monkey during complex sensory stimulation with affective significance. J Neurosci 8:3570-3583.

Nishino S, Mignot E (1997) Pharmacological aspects of human and canine narcolepsy. Prog Neurobiol 52:27-78.

Nishino S, Shelton J, Renaud A, Dement WC, Mignot E (1995) Effect of 5-HT1A receptor agonists and antagonists on canine cataplexy. J Pharmacol Exp Ther 272:1170-1175.

Nishino S, Ripley B, Overeem S, Lammers GJ, Mignot E (2000a) Hypocretin (orexin) deficiency in human narcolepsy. Lancet 355:39-40.

Nishino S, Riehl J, Hong J, Kwan M, Reid M, Mignot E (2000b) Is narcolepsy a REM sleep disorder? Analysis of sleep abnormalities in narcoleptic Dobermans. Neurosci Res 38:437-446.

Nitecka L, Ben-Ari Y (1987) Distribution of GABA-like immunoreactivity in the rat amygdaloid complex. J Comp Neurol 266:45-55.

Nofzinger EA, Mintun MA, Wiseman M, Kupfer DJ, Moore RY (1997) Forebrain activation in REM sleep: an FDG PET study. Brain Res 770:192-201.

Oishi Y, Williams RH, Agostinelli L, Arrigoni E, Fuller PM, Mochizuki T, Saper CB, Scammell TE (2013) Role of the medial prefrontal cortex in cataplexy. J Neurosci 33:9743-9751.

Overeem S, Lammers GJ, van Dijk JG (1999) Weak with laughter. Lancet 354:838.

Overeem S, Mignot E, van Dijk JG, Lammers GJ (2001) Narcolepsy: clinical features, new pathophysiologic insights, and future perspectives. J Clin Neurophysiol 18:78-105.

Overeem S, van Nues SJ, van der Zande WL, Donjacour CE, van Mierlo P, Lammers GJ (2011) The clinical features of cataplexy: a questionnaire study in narcolepsy patients with and without hypocretin-1 deficiency. Sleep Med 12:12-18.

Paton JJ, Belova MA, Morrison SE, Salzman CD (2006) The primate amygdala represents the positive and negative value of visual stimuli during learning. Nature 439:865-870.

Peever J (2011) Control of motoneuron function and muscle tone during REM sleep, REM sleep behavior disorder and cataplexy/narcolepsy. Arch Ital Biol 149:454-466.

Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, Kilduff TS (1998) Neurons containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci 18:9996-10015.

Peyron C et al. (2000) A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains. Nat Med 6:991-997.

Page 86: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

77

Pitkanen A, Amaral DG (1994) The distribution of GABAergic cells, fibers, and terminals in the monkey amygdaloid complex: an immunohistochemical and in situ hybridization study. J Neurosci 14:2200-2224.

Pitkanen A, Savander V, LeDoux JE (1997) Organization of intra-amygdaloid circuitries in the rat: an emerging framework for understanding functions of the amygdala. Trends Neurosci 20:517-523.

Pitkanen A, Stefanacci L, Farb CR, Go GG, LeDoux JE, Amaral DG (1995) Intrinsic connections of the rat amygdaloid complex: projections originating in the lateral nucleus. J Comp Neurol 356:288-310.

Plazzi G, Poli F, Franceschini C, Parmeggiani A, Pirazzoli P, Bernardi F, Mignot E, Cicognani A, Montagna P (2008) Intravenous high-dose immunoglobulin treatment in recent onset childhood narcolepsy with cataplexy. J Neurol 255:1549-1554.

Ponz A, Khatami R, Poryazova R, Werth E, Boesiger P, Bassetti CL, Schwartz S (2010) Abnormal activity in reward brain circuits in human narcolepsy with cataplexy. Ann Neurol 67:190-200.

Quirk GJ, Repa C, LeDoux JE (1995) Fear conditioning enhances short-latency auditory responses of lateral amygdala neurons: parallel recordings in the freely behaving rat. Neuron 15:1029-1039.

Rechtschaffen A, Wolpert EA, Dement WC, Mitchell SA, Fisher C (1963) Nocturnal Sleep of Narcoleptics. Electroencephalogr Clin Neurophysiol 15:599-609.

Reid MS, Siegel JM, Dement WC, Mignot E (1994a) Cholinergic mechanisms in canine narcolepsy--II. Acetylcholine release in the pontine reticular formation is enhanced during cataplexy. Neuroscience 59:523-530.

Reid MS, Tafti M, Geary JN, Nishino S, Siegel JM, Dement WC, Mignot E (1994b) Cholinergic mechanisms in canine narcolepsy--I. Modulation of cataplexy via local drug administration into the pontine reticular formation. Neuroscience 59:511-522.

Robinson MJ, Warlow SM, Berridge KC (2014) Optogenetic excitation of central amygdala amplifies and narrows incentive motivation to pursue one reward above another. J Neurosci 34:16567-16580.

Rogan SC, Roth BL (2011) Remote control of neuronal signaling. Pharmacol Rev 63:291-315.

Sah P, Faber ES, Lopez De Armentia M, Power J (2003) The amygdaloid complex: anatomy and physiology. Physiol Rev 83:803-834.

Sakurai T et al. (1998) Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573-585.

Sanford LD, Parris B, Tang X (2002) GABAergic regulation of the central nucleus of the amygdala: implications for sleep control. Brain Res 956:276-284.

Page 87: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

78

Sanford LD, Tejani-Butt SM, Ross RJ, Morrison AR (1995) Amygdaloid control of alerting and behavioral arousal in rats: involvement of serotonergic mechanisms. Arch Ital Biol 134:81-99.

Sanford LD, Yang L, Liu X, Tang X (2006) Effects of tetrodotoxin (TTX) inactivation of the central nucleus of the amygdala (CNA) on dark period sleep and activity. Brain Res 1084:80-88.

Sapin E, Lapray D, Berod A, Goutagny R, Leger L, Ravassard P, Clement O, Hanriot L, Fort P, Luppi PH (2009) Localization of the brainstem GABAergic neurons controlling paradoxical (REM) sleep. PLoS One 4:e4272.

Sateia MJ (2014) International classification of sleep disorders-third edition: highlights and modifications. Chest 146:1387-1394.

Savander V, Go CG, LeDoux JE, Pitkanen A (1995) Intrinsic connections of the rat amygdaloid complex: projections originating in the basal nucleus. J Comp Neurol 361:345-368.

Savander V, Go CG, Ledoux JE, Pitkanen A (1996) Intrinsic connections of the rat amygdaloid complex: projections originating in the accessory basal nucleus. J Comp Neurol 374:291-313.

Scammell TE, Willie JT, Guilleminault C, Siegel JM, International Working Group on Rodent Models of N (2009) A consensus definition of cataplexy in mouse models of narcolepsy. Sleep 32:111-116.

Schenck CH, Bassetti CL, Arnulf I, Mignot E (2007) English translations of the first clinical reports on narcolepsy and cataplexy by Westphal and Gelineau in the late 19th century, with commentary. J Clin Sleep Med 3:301-311.

Schenkel E, Siegel JM (1989) REM sleep without atonia after lesions of the medial medulla. Neurosci Lett 98:159-165.

Schmidt-Mutter C, Muller C, Zwiller J, Gobaille S, Maitre M (1999) Gamma-hydroxybutyrate and cocaine administration increases mRNA expression of dopamine D1 and D2 receptors in rat brain. Neuropsychopharmacology 21:662-669.

Schoenbaum G, Chiba AA, Gallagher M (1998) Orbitofrontal cortex and basolateral amygdala encode expected outcomes during learning. Nat Neurosci 1:155-159.

Schwartz S, Ponz A, Poryazova R, Werth E, Boesiger P, Khatami R, Bassetti CL (2008) Abnormal activity in hypothalamus and amygdala during humour processing in human narcolepsy with cataplexy. Brain 131:514-522.

Siegel JM (2011) REM sleep: a biological and psychological paradox. Sleep Med Rev 15:139-142.

Siegel JM, Boehmer LN (2006) Narcolepsy and the hypocretin system--where motion meets emotion. Nat Clin Pract Neurol 2:548-556.

Page 88: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

79

Siegel JM, Nienhuis R, Fahringer HM, Chiu C, Dement WC, Mignot E, Lufkin R (1992) Activity of medial mesopontine units during cataplexy and sleep-waking states in the narcoleptic dog. J Neurosci 12:1640-1646.

Siegel JM, Nienhuis R, Fahringer HM, Paul R, Shiromani P, Dement WC, Mignot E, Chiu C (1991) Neuronal activity in narcolepsy: identification of cataplexy-related cells in the medial medulla. Science 252:1315-1318.

Smith CT, Miskiman DE (1975) Increases in Paradoxical Sleep as a Result of Amygdaloid Stimulation. Physiology & Behavior 15:17-19.

Swanson LW, Petrovich GD (1998) What is the amygdala? Trends Neurosci 21:323-331.

Szabadi E (2015) GHB for cataplexy: Possible mode of action. J Psychopharmacol 29:744-749.

Takahashi K, Kayama Y, Lin JS, Sakai K (2010) Locus coeruleus neuronal activity during the sleep-waking cycle in mice. Neuroscience 169:1115-1126.

Tang X, Yang L, Liu X, Sanford LD (2005) Influence of tetrodotoxin inactivation of the central nucleus of the amygdala on sleep and arousal. Sleep 28:923-930.

Thannickal TC, Moore RY, Nienhuis R, Ramanathan L, Gulyani S, Aldrich M, Cornford M, Siegel JM (2000) Reduced number of hypocretin neurons in human narcolepsy. Neuron 27:469-474.

Thorpy MJ, Dauvilliers Y (2015) Clinical and practical considerations in the pharmacologic management of narcolepsy. Sleep Med 16:9-18.

Torontali ZA, Grace KP, Horner RL, Peever JH (2014) Cholinergic involvement in control of REM sleep paralysis. J Physiol 592:1425-1426.

Torrealba F, Yanagisawa M, Saper CB (2003) Colocalization of orexin a and glutamate immunoreactivity in axon terminals in the tuberomammillary nucleus in rats. Neuroscience 119:1033-1044.

Tye KM, Janak PH (2007) Amygdala neurons differentially encode motivation and reinforcement. J Neurosci 27:3937-3945.

Usuda I, Tanaka K, Chiba T (1998) Efferent projections of the nucleus accumbens in the rat with special reference to subdivision of the nucleus: biotinylated dextran amine study. Brain Res 797:73-93.

Valko PO, Khatami R, Baumann CR, Bassetti CL (2008) No persistent effect of intravenous immunoglobulins in patients with narcolepsy with cataplexy. J Neurol 255:1900-1903.

Valley V, Broughton R (1983) The physiological (EEG) nature of drowsiness and its relation to performance deficits in narcoleptics. Electroencephalogr Clin Neurophysiol 55:243-251.

Page 89: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

80

Van Dort CJ, Zachs DP, Kenny JD, Zheng S, Goldblum RR, Gelwan NA, Ramos DM, Nolan MA, Wang K, Weng FJ, Lin Y, Wilson MA, Brown EN (2015) Optogenetic activation of cholinergic neurons in the PPT or LDT induces REM sleep. Proc Natl Acad Sci U S A 112:584-589.

Vassalli A, Dellepiane JM, Emmenegger Y, Jimenez S, Vandi S, Plazzi G, Franken P, Tafti M (2013) Electroencephalogram paroxysmal theta characterizes cataplexy in mice and children. Brain 136:1592-1608.

Vazdarjanova A, Cahill L, McGaugh JL (2001) Disrupting basolateral amygdala function impairs unconditioned freezing and avoidance in rats. Eur J Neurosci 14:709-718.

Vertes RP (2004) Differential projections of the infralimbic and prelimbic cortex in the rat. Synapse 51:32-58.

Vetrugno R, D'Angelo R, Moghadam KK, Vandi S, Franceschini C, Mignot E, Montagna P, Plazzi G (2010) Behavioural and neurophysiological correlates of human cataplexy: a video-polygraphic study. Clin Neurophysiol 121:153-162.

Vogel G (1960) Studies in psychophysiology of dreams. III. The dream of narcolepsy. Arch Gen Psychiatry 3:421-428.

Vong L, Ye C, Yang Z, Choi B, Chua S, Jr., Lowell BB (2011) Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 71:142-154.

Weber F, Chung S, Beier KT, Xu M, Luo L, Dan Y (2015) Control of REM sleep by ventral medulla GABAergic neurons. Nature 526:435-438.

Weng FJ, Williams RH, Hawryluk JM, Lu J, Scammell TE, Saper CB, Arrigoni E (2014) Carbachol excites sublaterodorsal nucleus neurons projecting to the spinal cord. J Physiol 592:1601-1617.

Wess J, Nakajima K, Jain S (2013) Novel designer receptors to probe GPCR signaling and physiology. Trends Pharmacol Sci 34:385-392.

Westphal C (1877) Eigentümliche mit Einschlafen verbundene Anfälle. Archiv für Psychiatrie und Nervenkrankheiten 7:631-635.

Willie JT, Chemelli RM, Sinton CM, Tokita S, Williams SC, Kisanuki YY, Marcus JN, Lee C, Elmquist JK, Kohlmeier KA, Leonard CS, Richardson JA, Hammer RE, Yanagisawa M (2003) Distinct narcolepsy syndromes in Orexin receptor-2 and Orexin null mice: molecular genetic dissection of Non-REM and REM sleep regulatory processes. Neuron 38:715-730.

Wisor JP, Eriksson KS (2005) Dopaminergic-adrenergic interactions in the wake promoting mechanism of modafinil. Neuroscience 132:1027-1034.

Page 90: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

81

Wisor JP, Nishino S, Sora I, Uhl GH, Mignot E, Edgar DM (2001) Dopaminergic role in stimulant-induced wakefulness. J Neurosci 21:1787-1794.

Wojcik SM, Katsurabayashi S, Guillemin I, Friauf E, Rosenmund C, Brose N, Rhee JS (2006) A shared vesicular carrier allows synaptic corelease of GABA and glycine. Neuron 50:575-587.

Wu MF, Gulyani SA, Yau E, Mignot E, Phan B, Siegel JM (1999) Locus coeruleus neurons: cessation of activity during cataplexy. Neuroscience 91:1389-1399.

Wu MF, John J, Boehmer LN, Yau D, Nguyen GB, Siegel JM (2004) Activity of dorsal raphe cells across the sleep-waking cycle and during cataplexy in narcoleptic dogs. J Physiol 554:202-215.

Xi M, Fung SJ, Sampogna S, Chase MH (2011) Excitatory projections from the amygdala to neurons in the nucleus pontis oralis in the rat: an intracellular study. Neuroscience 197:181-190.

Yoss RE, Daly DD (1957) Criteria for the diagnosis of the narcoleptic syndrome. Proc Staff Meet Mayo Clin 32:320-328.

Page 91: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

82

Appendix 1 – Behavioral State Analysis Macro

The following is an Excel VBA macro (version date: March 17, 2015) coded by Matthew

Snow that was used to automatically and accurately calculate parameters relating to behavioral

state architecture and muscle tone using output generated by the “5s epoch analysis” Spike 2

script. In a dedicated Microsoft Excel file, users are prompted for the states they wish to analyze

and the time intervals in which they wish to bin their data, and the macro then performs all

relevant calculations based on this input. Private Sub AnalyzeButton_Click() Dim StateCombo() As String Dim StateName() As String Dim TreatmentName() As String Dim MasseterTone() As Single Dim NeckTone() As Single Dim PercentOfTime As Single Dim Continue As Boolean Dim BinNumber As Integer Dim DataColumn As Integer Dim DataRow As Integer Dim i As Integer Dim MasseterCounter() As Integer Dim NeckCounter() As Integer Dim NumberOfBins As Integer Dim NumberOfEpochs As Integer Dim NumberOfStates As Integer Dim StateNamePos1 As Integer Dim StateNamePos2 As Integer Dim TableColumn As Integer Dim TableRow As Integer Dim Transitions() As Integer Dim TransitionsPos As Integer Dim TreatmentNamePos As Integer Dim variable As String Sheets(2).Cells.Clear 'NumberOfStates = UBound(Split(StateInput, "|")) - LBound(Split(StateInput, "|")) + 1 'Defines value of NumberOfStates StateName() = Split(StateInput, "|") 'Populates StateName() NumberOfStates = UBound(StateName) + 1

Page 92: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

83

ReDim StateCombo(NumberOfStates ^ 2 - 1) As String 'Defines size of StateCombo() DataColumn = 1 TreatmentNamePos = 0 Sheets(1).Activate Do While Cells(1, DataColumn) <> "" 'Counts the number of treatments listed on Raw Data sheet TreatmentNamePos = TreatmentNamePos + 1 DataColumn = DataColumn + 6 Loop ReDim TreatmentName(TreatmentNamePos - 1) As String DataColumn = 1 For TreatmentNamePos = 0 To UBound(TreatmentName) 'Populates TreatmentName() TreatmentName(TreatmentNamePos) = Cells(1, DataColumn) DataColumn = DataColumn + 6 Next TreatmentNamePos TableRow = 1 TableColumn = 1 DataColumn = 1 For TreatmentNamePos = 0 To UBound(TreatmentName) 'Repeats all calculations for each treatment Sheets(1).Activate NumberOfEpochs = WorksheetFunction.CountA(Range(Cells(5, DataColumn), Cells(130000, DataColumn))) 'Defines value of NumberOfEpochs. 130000 is an arbitrary limit and can be changed depending on need. Chosen to be suitable for up to 1 week nonstop recording (720 epochs/hour * 24 hours/day * 7 days/week = 120960) NumberOfBins = (NumberOfEpochs * 5 / 3600) / Val(TimeInput) 'Defines value of NumberOfBins If NumberOfEpochs > NumberOfBins * Val(TimeInput) * 720 Then NumberOfBins = NumberOfBins + 1 'Ensures the last bin is counted when it contains less than the maximum number of epochs (e.g., recording stopped in middle of bin) Sheets(2).Activate Columns("B").ColumnWidth = 5 With Range(Cells(TableRow, TableColumn), Cells(TableRow + 2 * NumberOfBins + 10, TableColumn)) 'Creates/formats title cell for treatment on State Analysis sheet .MergeCells = True .Value = TreatmentName(TreatmentNamePos) .Orientation = 90 .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Font.Size = 30 .Interior.Color = RGB(204, 255, 204) .Borders(xlEdgeBottom).LineStyle = xlContinuous

Page 93: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

84

.Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With For i = 1 To 6 'Creates/formats tables for non-transitions variables to display result outputs If i < 6 Then With Range(Cells(TableRow, TableColumn + 2), Cells(TableRow + 1, TableColumn + NumberOfStates + 3)) 'Creates/formats table title cells for non-transitions variables .MergeCells = True If i = 1 Then variable = "Time Spent in State (%)" If i = 2 Then variable = "Number of Episodes" If i = 3 Then variable = "Average Duration of Episodes (s)" If i = 4 Then variable = "Average Muscle Activity - Masseter" If i = 5 Then variable = "Average Muscle Activity - Neck" .Value = variable .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Font.Size = 18 .Interior.Color = RGB(136, 192, 254) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With With Range(Cells(TableRow + 3, TableColumn + 4), Cells(TableRow + 3, TableColumn + NumberOfStates + 3)) 'Creates/formats state name title cell for non-transitions variables .MergeCells = True .Value = "State" .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Interior.Color = RGB(254, 195, 10) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With With Range(Cells(TableRow + 4, TableColumn + 4), Cells(TableRow + 4, TableColumn + NumberOfStates + 3)) 'Creates/formats/populates state name column titles for non-transitions variables .Value = StateName() .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Borders(xlEdgeBottom).LineStyle = xlContinuous

Page 94: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

85

.Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlMedium End With With Range(Cells(TableRow + 5, TableColumn + 2), Cells(TableRow + NumberOfBins + 4, TableColumn + 2)) 'Creates/formats bin number title cell for non-transitions variables .MergeCells = True .Value = "Bin # (" & Val(TimeInput) & "-hour bins)" .WrapText = True .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Interior.Color = RGB(254, 195, 10) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With With Range(Cells(TableRow + 5, TableColumn + 3), Cells(TableRow + NumberOfBins + 4, TableColumn + 3)) 'Creates/formats cells for row titles for non-transitions variables .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlMedium End With With Range(Cells(TableRow + 5, TableColumn + 4), Cells(TableRow + NumberOfBins + 4, TableColumn + NumberOfStates + 3)) 'Creates/formats table cells that will contain results outputs .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Interior.Color = RGB(255, 255, 135) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThin End With For BinNumber = 0 To NumberOfBins - 1 'Numbers bins in all tables Cells(TableRow + 5, TableColumn + 3) = BinNumber + 1 If i = 1 Then Cells(TableRow + NumberOfBins + 11, TableColumn + 3) = BinNumber + 1 'Numbers bins in transitions table during first iteration TableRow = TableRow + 1 Next BinNumber

Page 95: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

86

TableRow = TableRow - NumberOfBins 'Resets value of TableRow TableColumn = TableColumn + NumberOfStates + 3 'Sets TableColumn value to the start of the next non-transitions variable table Else TableColumn = 1 With Range(Cells(TableRow + NumberOfBins + 7, TableColumn + 2), Cells(TableRow + NumberOfBins + 8, TableColumn + NumberOfStates ^ 2 + 3)) 'Creates/formats table title cell for transitions .MergeCells = True .Value = "Transitions" .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Font.Size = 18 .Interior.Color = RGB(136, 192, 254) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With With Range(Cells(TableRow + NumberOfBins + 11, TableColumn + 2), Cells(TableRow + 2 * NumberOfBins + 10, TableColumn + 2)) 'Creates/formats bin number title cell for transitions .MergeCells = True .Value = "Bin # (" & Val(TimeInput) & "-hour bins)" .WrapText = True .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Interior.Color = RGB(254, 195, 10) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThick End With With Range(Cells(TableRow + NumberOfBins + 10, TableColumn + 4), Cells(TableRow + NumberOfBins + 10, TableColumn + NumberOfStates ^ 2 + 3)) 'Creates/formats column title cells for transitions .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Font.Bold = True .Interior.Color = RGB(254, 195, 10) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlMedium End With With Range(Cells(TableRow + NumberOfBins + 11, TableColumn + 3), Cells(TableRow + 2 * NumberOfBins + 10, TableColumn + 3)) 'Creates/formats cells for row titles in transitions tables .HorizontalAlignment = xlCenter

Page 96: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

87

.VerticalAlignment = xlCenter .Font.Bold = True .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlMedium End With With Range(Cells(TableRow + NumberOfBins + 11, TableColumn + 4), Cells(TableRow + 2 * NumberOfBins + 10, TableColumn + NumberOfStates ^ 2 + 3)) 'Creates/formats cells for column titles in transitions tables .HorizontalAlignment = xlCenter .VerticalAlignment = xlCenter .Interior.Color = RGB(255, 255, 135) .Borders(xlEdgeBottom).LineStyle = xlContinuous .Borders(xlEdgeTop).LineStyle = xlContinuous .Borders(xlEdgeLeft).LineStyle = xlContinuous .Borders(xlEdgeRight).LineStyle = xlContinuous .Borders.Weight = xlThin End With End If Next i For BinNumber = 0 To NumberOfBins - 1 'Calculates data for each variable for each bin Sheets(1).Activate ReDim Transitions(NumberOfStates ^ 2 - 1) As Integer 'Defines size of Transitions() For StateNamePos1 = 0 To NumberOfStates - 1 'Counts the number of transitions between states. Transitions are in the form of State1 to State2. This loop changes State1 to the next state to create a new search combination. For StateNamePos2 = 0 To NumberOfStates - 1 'Transitions are in the form of State1 to State2. This loop changes State2 to the next state to create a new seacrh combination. Continue = True For DataRow = Val(TimeInput) * 720 * BinNumber + 4 To Val(TimeInput) * 720 * (BinNumber + 1) + 3 'Searches for target transition in the current bin If BinNumber = 0 And Continue = True Then 'Populates StateCombo() and transitions table column titles during the first iteration of this loop only (i.e., the first bin of the first treatment) to be used for all remaining calculations. StateCombo(TransitionsPos) = StateName(StateNamePos1) & " to " & StateName(StateNamePos2) Sheets(2).Activate Cells(TableRow + NumberOfBins + 10, TableColumn + 4) = StateCombo(TransitionsPos) Sheets(1).Activate Continue = False End If If Cells(DataRow, DataColumn) = StateName(StateNamePos1) And Cells(DataRow + 1, DataColumn) = StateName(StateNamePos2) Then 'Counts the number of target transitions by

Page 97: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

88

adding 1 each time a target transition is found. Sum is stored in Transitions() at the location corresponding to the transition name in StateCombo() Transitions(TransitionsPos) = Transitions(TransitionsPos) + 1 End If Next DataRow Sheets(2).Activate Cells(TableRow + NumberOfBins + 11, TableColumn + 4) = Transitions(TransitionsPos) 'Fills the appropriate cell in the transitions table with the transitions number Sheets(1).Activate DataRow = 5 + Val(TimeInput) * 720 * BinNumber 'Resets value of DataRow to the start of the current bin TableColumn = TableColumn + 1 TransitionsPos = TransitionsPos + 1 Next StateNamePos2 DataRow = 5 + Val(TimeInput) * 720 * BinNumber 'Resets value of DataRow to the start of the current bin Next StateNamePos1 DataRow = 5 TableColumn = 1 TransitionsPos = 0 StateNamePos2 = 0 Sheets(2).Activate For StateNamePos1 = 0 To UBound(StateName) 'Calculates number of episodes for each state for the current bin For TransitionsPos = TransitionsPos To StateNamePos1 + (NumberOfStates - 1) * (StateNamePos1 + 1) 'Transitions are in the form of State1 to State2. Repeats for as many times as there are combinations beginning with the current State1. Cells(TableRow + 5, TableColumn + NumberOfStates + 7) = Cells(TableRow + 5, TableColumn + NumberOfStates + 7) + Transitions(TransitionsPos) 'Transitions are in the form of State1 to State2. Sums the all the values in Transitions() corresponding to the same State1. If TransitionsPos = StateNamePos1 * (NumberOfStates + 1) Then 'Subtracts the current value of Transitions() if State1=State2 (i.e., state has not changed) Cells(TableRow + 5, TableColumn + NumberOfStates + 7) = Cells(TableRow + 5, TableColumn + NumberOfStates + 7) - Transitions(TransitionsPos) End If Sheets(1).Activate If Cells(4 + Val(TimeInput) * 720 * BinNumber, DataColumn) = StateName(StateNamePos1) And Cells(5 + Val(TimeInput) * 720 * BinNumber, DataColumn) = StateName(StateNamePos2) And StateNamePos1 <> StateNamePos2 Then 'Subtracts 1 from current number of episodes count for the current State1 if it occurs in the epoch preceding the first epoch of the current bin (to account for the one more transition than epoch for any bin) Sheets(2).Activate

Page 98: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

89

Cells(TableRow + 5, TableColumn + NumberOfStates + 7) = Cells(TableRow + 5, TableColumn + NumberOfStates + 7) - 1 Else Sheets(2).Activate End If StateNamePos2 = StateNamePos2 + 1 Next TransitionsPos StateNamePos2 = 0 Sheets(1).Activate If WorksheetFunction.Index(Range(Cells(5, DataColumn), Cells(4 + Val(TimeInput) * 720 * (BinNumber + 1), DataColumn)), WorksheetFunction.CountA(Range(Cells(5, DataColumn), Cells(4 + Val(TimeInput) * 720 * (BinNumber + 1), DataColumn))), 1) = StateName(StateNamePos1) Then 'Adds 1 to the current number of episodes for the current State1 if the current State1 is the final epoch of the current bin Sheets(2).Activate Cells(TableRow + 5, TableColumn + NumberOfStates + 7) = Cells(TableRow + 5, TableColumn + NumberOfStates + 7) + 1 Else Sheets(2).Activate End If TableColumn = TableColumn + 1 Next StateNamePos1 TableColumn = 1 StateNamePos1 = 0 For StateNamePos1 = 0 To UBound(StateName) 'Calculates the percent time spent in and average durations of each state for the current bin Sheets(1).Activate PercentOfTime = WorksheetFunction.CountIf(Range(Cells(Val(TimeInput) * 720 * BinNumber + 5, DataColumn), Cells(Val(TimeInput) * 720 * (BinNumber + 1) + 4, DataColumn)), StateName(StateNamePos1)) / WorksheetFunction.CountA(Range(Cells(Val(TimeInput) * 720 * BinNumber + 5, DataColumn), Cells(Val(TimeInput) * 720 * (BinNumber + 1) + 4, DataColumn))) 'Calculates percent of time in state by dividing the number of epochs of that state by the total number of epochs in that bin Sheets(2).Activate With Cells(TableRow + 5, TableColumn + 4) 'Fills the corresponding cell in the percent of time table with the current value of PercentOfTime .Value = PercentOfTime .NumberFormat = "0.00%" End With If Cells(TableRow + 5, TableColumn + NumberOfStates + 7) <> 0 Then 'Calculates the average duration of episodes of the corresponding state as long as the state occurred (i.e., the number of episodes is not 0) With Cells(TableRow + 5, TableColumn + 2 * NumberOfStates + 10) 'Fills the corresponding cell in the average duration of episodes table Sheets(1).Activate

Page 99: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

90

.Value = WorksheetFunction.CountIf(Range(Cells(DataRow + Val(TimeInput) * 720 * BinNumber, DataColumn), Cells(DataRow + Val(TimeInput) * 720 * (BinNumber + 1) - 1, DataColumn)), StateName(StateNamePos1)) * 5 / Sheets(2).Cells(TableRow + 5, TableColumn + NumberOfStates + 7) 'Calculates the average duration by dividing the total time spent in the state (for the current bin) by the number of episodes of that state (for the current bin) Sheets(2).Activate .NumberFormat = "0.00" End With Else Cells(TableRow + 5, TableColumn + 2 * NumberOfStates + 10) = 0 'If there are no episodes of the corresponding state in the current bin, the average duration is set to 0 End If TableColumn = TableColumn + 1 Next StateNamePos1 Sheets(1).Activate ReDim MasseterTone(NumberOfStates) As Single ReDim MasseterCounter(NumberOfStates) As Integer ReDim NeckTone(NumberOfStates) As Single ReDim NeckCounter(NumberOfStates) As Integer For DataRow = Val(TimeInput) * 720 * BinNumber + 5 To Val(TimeInput) * 720 * (BinNumber + 1) + 4 'Calculates average muscle tone for masseter and neck For StateNamePos2 = 0 To UBound(StateName) If Cells(DataRow, DataColumn) = StateName(StateNamePos2) Then If Cells(DataRow, DataColumn + 3) <> "" Then MasseterTone(StateNamePos2) = MasseterTone(StateNamePos2) + Cells(DataRow, DataColumn + 3) MasseterCounter(StateNamePos2) = MasseterCounter(StateNamePos2) + 1 End If If Cells(DataRow, DataColumn) = StateName(StateNamePos2) And Cells(DataRow, DataColumn + 4) <> "" Then NeckTone(StateNamePos2) = NeckTone(StateNamePos2) + Cells(DataRow, DataColumn + 4) NeckCounter(StateNamePos2) = NeckCounter(StateNamePos2) + 1 End If Exit For End If Next StateNamePos2 Next DataRow Sheets(2).Activate TableColumn = 1 For StateNamePos2 = 0 To UBound(StateName) If MasseterCounter(StateNamePos2) <> 0 Then With Cells(TableRow + 5, TableColumn + NumberOfStates * 3 + 13) 'Fills masseter activity table

Page 100: GABA Cells in the Central Nucleus of the Amygdala Control ... · GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli such as wheel running,

91

.Value = MasseterTone(StateNamePos2) / MasseterCounter(StateNamePos2) .NumberFormat = "0.000000" End With Else With Cells(TableRow + 5, TableColumn + NumberOfStates * 3 + 13) 'Fills masseter activity table .Value = "no data" End With End If If NeckCounter(StateNamePos2) <> 0 Then With Cells(TableRow + 5, TableColumn + NumberOfStates * 4 + 16) 'Fills neck activity table .Value = NeckTone(StateNamePos2) / NeckCounter(StateNamePos2) .NumberFormat = "0.000000" End With Else With Cells(TableRow + 5, TableColumn + NumberOfStates * 4 + 16) 'Fills neck activity table .Value = "no data" End With End If TableColumn = TableColumn + 1 Next StateNamePos2 TableRow = TableRow + 1 TableColumn = 1 TransitionsPos = 0 Next BinNumber DataColumn = DataColumn + 6 'Sets DataColumn to the column of the next treatment TableRow = TableRow + NumberOfBins + 15 'Sets TableRow to the next available space to output results for the next treatment Next TreatmentNamePos Unload Me End Sub