PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

258
PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT EXPOSURE DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Laura K. Fonken Graduate Program in Neuroscience The Ohio State University 2013 Dissertation Committee: Dr. Randy J. Nelson, Advisor Dr. A. Courtney DeVries Dr. Jonathan P. Godbout Dr. Dana M. McTigue

Transcript of PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

Page 1: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY

NIGHTTIME LIGHT EXPOSURE

DISSERTATION

Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy

in the Graduate School of The Ohio State University

By

Laura K. Fonken

Graduate Program in Neuroscience

The Ohio State University

2013

Dissertation Committee:

Dr. Randy J. Nelson, Advisor

Dr. A. Courtney DeVries

Dr. Jonathan P. Godbout

Dr. Dana M. McTigue

Page 2: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

Copyright by

Laura K. Fonken

2013

Page 3: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

ii

ABSTRACT

For more than 3 billion years, life outside the highest latitudes has evolved under

brightly illuminated days and dark nights. Most organisms have developed endogenously

driven circadian rhythms which are synchronized to this light/dark cycle. In recent years,

daily light schedules have become artificial and irregular due to the use of electric

lighting. In this dissertation, I propose that exposure to light at night (LAN) disrupts the

circadian system altering metabolic, immunological, and behavioral functions.

The global increase in the prevalence of obesity and metabolic disorders coincides

with increases in exposure to LAN and shift work. Therefore, my first experiments

examined whether exposure to LAN affects metabolism. Mice exposed to dimly lit (5

lux) as compared to dark nights increased body mass and reduced glucose processing

without changing caloric intake or activity. Exposure to dim light at night diminished the

daily rhythm in food intake and restricting food access to the dark phase prevented

weight gain in mice exposed to dimly lit nights (Chapter 2). Furthermore, metabolic

changes associated with exposure to LAN are not permanent; placing mice back in dark

nights partially reversed increases in body mass caused by exposure to dim light at night

(Chapter 3). In Chapters 4 & 5, I investigated the interactions among LAN and more

traditional risk factors for obesity such as high fat diet and lethargy.

Because light is the most potent synchronizing factor for the circadian system and

Page 4: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

iii

disruption in clock genes is associated with significant changes in metabolism, I next

investigated the effects of exposure to LAN on the circadian system (Chapter 6).

Exposure to dimly lit nights attenuated core circadian clock rhythms in both the master

circadian pacemaker and peripheral tissues.

In addition to altering metabolism, exposure to LAN is implicated as a

contributing factor to several diseases involving dysregulation of the immune system.

This led to experiments examining the effects of acute exposure to dim LAN on recovery

following cardiac arrest (Chapter 7). Exposure to dimly lit as compared to dark nights

following global ischemia increased hippocampal inflammation, neuronal cell death, and

mortality. Selectively inhibiting inflammation and altering the spectrum of nighttime

light to which mice were exposed reduced damage among mice exposed to dim LAN.

In the experiments described above, I worked with nocturnal mice in order to

assess the effects of nighttime light exposure independent of changes in sleep

architecture. However, the secretion patterns of many hormones and immune parameters

are different in nocturnal and diurnal species. In the final set of experiments, I

demonstrated that diurnal Nile Grass rats (Arvicanthus Niloticus) exposed to dim LAN

increased immunological measures (Chapter 8) and altered hippocampal connectivity in

addition to changing cognitive and affective behaviors (Chapter 9). Taken together,

these studies indicate that exposure to ecologically relevant levels of dim LAN attenuate

core circadian clock mechanisms in rodents resulting in physiological and behavioral

consequences.

Page 5: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

iv

DEDICATION

To my family – for their love, encouragement, and support.

Page 6: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

v

ACKNOWLEDGEMENTS

First and foremost, I thank my advisor Dr. Randy Nelson for his superb

mentorship. Randy has provided me with abundant opportunities and resources, as well

as much of his own time over the past five years. Randy is a truly dedicated and

supportive mentor who has given me both excellent scientific advice and savvy insight

into what one needs to do to become a successful researcher.

I am grateful for all of the wonderful people with whom I have had the

opportunity to work in the Nelson and DeVries labs. I thank James Walton, Dr. Joanna

Workman, Dr. Kate Weil, and John Morris for their fantastic guidance and patience in

teaching me skills ranging all the way from how to work a pipette to performing

immunohistochemistry and PCR. Dr. Zachary Weil has been an excellent mentor and

friend over the past few years. I thank Zach for his valuable advice and support.

Furthermore, I am grateful to Joanna Workman, Tracy Bedrosian, and Taryn Aubrecht

for their help, advice, and friendship both inside and outside the lab. My lab experience

would not have been the same without the “executive broads,” our international travel

adventures, or the occasional stealth trip to Easton! I thank Greg Norman, Brant Jarrett,

Shannon Chen, Jeremy Borniger, Tomoko Ikeno, Adam Hinzey, Katie Stuller, and Jackie

Thomas for assisting me and for making the Nelson and DeVries labs such a great place

to work. I am also indebted to all of the undergraduate volunteers that helped make this

Page 7: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

vi

work possible including: Rebecca Lieberman, O. Hecmarie Meléndez-Fernández, Emily

Kitsmiller, Dan McCarthy, Heather Michaels, Brittany Jones, Jordan Grier, Amanda

Grunenwald, Natalie Hood, Brian Klein, Kristopher Gaier, Zachary McHenry, and

Joseph Ferraro. I would like to thank Ning Zhang as well, for her remarkable surgical

skills and friendly attitude.

I have had a lot of help from other faculty members and students, both at Ohio

State and other institutions. First, I want to thank Nathaniel Thomas and Dr. Catherine

Cornwell for introducing me to behavioral neuroscience at Syracuse University. I am

grateful to Dr. Courtney DeVries for serving on my candidacy and dissertation

committees, and for her outstanding mentorship on several experiments. Dr. DeVries is a

great role model for any aspiring female scientist and she has provided me with excellent

advice over the years. I also thank Drs. Dana McTigue and Jonathan Godbout for

providing me with insightful feedback while serving on my candidacy and dissertation

committees. I am grateful to Dr. Laura Smale, Dr. Phillip Popovich, Dr. Andrew Gaudet,

Dr. Qinghua Sun, Dr. Sanjay Rajagopalan, Dr. Xiaohua Xu, and Dr. Cuiqing Lui for

providing me the opportunity to collaborate on very interesting projects. Thank you to

Holly Brothers, Roxanne Kaercher, Ashley Fenn, Puneet Sodhi, and Jodie Hall for being

great friends and always having helpful scientific advice. Special thanks to Andrew

Gaudet for being incredibly supportive, and willing to help me in the lab at all hours of

the day and night.

Finally I would like to thank my family. I am very grateful to my parents, Carol

and David Fonken, for providing me with so many opportunities, tireless encouragement,

Page 8: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

vii

and support. I can trace my initial interest in science to conversations I had with my dad

while walking the dog. Dad has an incredible knowledge of many scientific topics, and

the ability to make all science seem both accessible and exciting! I also thank Erin and

Brian for being the best siblings I can imagine and always giving me love, support, and

advice.

Page 9: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

viii

VITA

May 21, 1986 .................................................Born – Austin, TX, USA

2004................................................................L.C. Anderson High School

2008................................................................ B.S. Biology and Psychology, Syracuse

University

2009-present ..................................................Graduate Research Fellow, Department of

Neuroscience, The Ohio State University

PUBLICATIONS

1. Bedrosian, T.A., Herring, K.L., Walton, J.C., Fonken, L.K., Weil, Z.M., &

Nelson, R.J. (2013). Possible feedback control of pineal melatonin secretion.

Neuroscience Letters (In press).

2. Fonken, L.K., Weil, Z.M., & Nelson, R.J. (2013). Dark nights reverse metabolic

disruption caused by dim light at night. Obesity, (In press).

3. Fonken, L.K. & Nelson, R.J. (2013). Dim light at night increases depressive-like

responses in male C3H/HeNHsd mice. Behavioural Brain Research, 243 (3): 74-

78.

4. Fonken, L.K., Kitsmiller, E., Smale, L., & Nelson, R.J. (2012). Dim nighttime

light impairs cognition and provokes depressive-like responses in a diurnal

rodent. Journal of Biological Rhythms, 27 (4): 319-27.

5. Fonken, L.K., Bedrosian, T.A., Michaels, H., Weil, Z.M., & Nelson, R.J. (2012).

Short photoperiods attenuate central responses to an inflammogen. Brain,

Behavior, and Immunity, 26 (4): 617-22.

Page 10: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

ix

6. Bedrosian, T.A., Fonken, L.K., Demas, G.E., & Nelson, R.J. (2012). Photoperiod-

dependent effects of neuronal nitric oxide synthase inhibition on aggression in

Phodopus sungorus. Hormones and Behavior, 61 (2): 176-80.

7. Fonken, L.K., Haim, A., & Nelson, R.J. (2012). Dim light at night increases

immune function in grass rats, a diurnal rodent. Chronobiology International, 29

(1): 26-34.

8. Fonken, L.K. & Nelson, R.J. (2011). Illuminating the deleterious effects of light

at night. F1000 Med Report, 3: 18.

9. Workman, J.L., Fonken, L.K., Gusfa, J., Kassouf, K., & Nelson, R.J. (2011). Post-

weaning environmental enrichment reduces negative affective responses and

interacts with behavioral testing to alter nNOS expression. Pharmacology,

Biochemistry and Behavior, 100 (1): 25-32.

10. Fonken, L.K., Xu, X., Weil, Z.M., Chen, G., Sun, Q., Rajagopalan, S., & Nelson,

R.J. (2011). Inhalation of fine particulates alters hippocampal neuronal

morphology. Molecular Psychiatry, 16 (10): 973.

11. Fonken, L.K., Xu, X., Weil, Z.M., Chen, G., Sun, Q., Rajagopalan, S., & Nelson,

R.J. (2011). Air pollution impairs cognition, provokes depressive-like behaviors,

and alters hippocampal cytokine expression and morphology. Molecular

Psychiatry, 16 (10): 987-995.

12. Fenn, A.M.*, Fonken, L.K.*, & Nelson, R.J. (2011). Sustained melatonin

treatment blocks body mass, pelage, reproductive and fever responses to short day

lengths in female Siberian hamsters. Journal of Pineal Research, 51: 180-186.

*equal contributors

13. Bedrosian, T.A., Fonken, L.K., Walton, J.C., Haim, A., & Nelson, R.J. (2011).

Dim light at night provokes depression-like behaviors and reduces CA1 dendritic

spine density in female hamsters. Psychoneuroendocrinology. 36: 1062-1069.

14. Bedrosian, T.A., Fonken, L.K., Walton, J.C., & Nelson, R.J. (2011). Chronic

exposure to dim light at night suppresses immune responses in Siberian hamsters.

Biology Letters, 7 (3): 468-471.

15. Fonken, L.K., Morris, J.S., & Nelson, R.J. (2011). Early life experiences affect

adult delayed-type hypersensitivity in short- and long-photoperiods.

Chronobiology International, 28 (2): 101-108.

16. Fonken, L.K., Workman, J.L., Walton, J.C., Weil, Z.M., Morris, J.S., Haim, A., &

Nelson, R.J. (2010). Light at night increases body mass by shifting the time of

Page 11: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

x

food intake. Proceedings of the National Academy of Sciences, 107 (43): 18664-

18669.

17. Thomas, N.R., Fonken, L.K., LeBlanc, M., & Cornwell, C.A. (2010). Maternal

separation alters social odor preference development in infant mice (Mus

musculus). Journal of Comparative Psychology, 124 (3): 295-301.

18. Workman, J.L., DeWitt, S.J., Fonken, L.K., & Nelson, R.J. (2010). Environmental

enrichment enhances delayed-type hypersensitivity in both short- and long-day

Siberian hamsters. Physiology and Behavior, 99 (5): 638-643.

19. Fonken, L.K., Finy, M.S., Walton, J.C., Weil, Z.M., Workman, J.L., Ross, J., &

Nelson, R.J. (2009). Influence of light at night on murine anxiety- and depressive-

like responses. Behavioural Brain Research, 205 (2): 349-354.

FIELDS OF STUDY

Major Field: Neuroscience

Page 12: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

xi

TABLE OF CONTENTS

Page

Abstract ............................................................................................................................... ii

Dedication .......................................................................................................................... iv

Acknowledgements ..............................................................................................................v

Vita ................................................................................................................................... viii

List of Tables ................................................................................................................... xiii

List of Figures .................................................................................................................. xiv

Chapters:

1. The “Skinny” on Light at Night, Circadian Clocks, and Metabolism ............................1

2. Light at Night Increases Body Mass through Altered Timing of Food Intake .............29

3. Dark Nights Reverse Metabolic Changes Caused by Dim Light at Night ...................49

4. Dim Light at Nigh Exaggerates Weight Gain and Inflammation Associated with a

High Fat Diet..........................................................................................................66

5. Exercise Attenuates the Metabolic Effects of Dim Light at Night ...............................83

6. Dim Light at Night Disrupts Molecular Circadian Rhythms ........................................94

7. Dim Light at Night Affects Cardiac Arrest Outcome .................................................112

8. Dim Light at Night Elevates Inflammatory Responses in Diurnal Grass Rats ...........140

9. Dim Light at Night Impairs Cognition and Provokes Depressive-like Responses in

Grass Rats ............................................................................................................161

Page 13: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

xii

Page

Conclusions ......................................................................................................................178

List of References ............................................................................................................188

Page 14: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

xiii

LIST OF TABLES

Table Page

2.1 Food intake in food restricted mice exposed to dimly lit or dark nights………...44

3.1 Experimental design……………………………………………………………...54

8.1 Tissue masses in Nile grass rats exposed to dimly lit or dark nights…………...157

9.1 Hippocampal neuron characteristics in grass rats exposed to dark or dimly lit

nights....................................................................................................................174

Page 15: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

xiv

LIST OF FIGURES

Figure Page

2.1. Light at night affects body mass and glucose tolerance ............................................45

2.2. Activity is comparable between mice exposed to dimly lit and dark nights .............47

2.3. Time of food intake but not corticosterone is altered by dim light at night...............48

2.4. Resticting feeding to the dark phase prevents dim light at night induced body mass

gain .........................................................................................................................49

3.1. Return to dark nights affects body mass after exposure to dim light at night ...........63

3.2. Dim light at night affects fat mass and composition..................................................64

3.3. Dark nights reverse changes in feeding behavior and glucose processing ................65

4.1. Light at night exaggerates weight gain on a high fat diet ..........................................79

4.2. High fat diet and light at night alter timing of food intake ........................................80

4.3. High fat diet and light at night increase adipose inflammation .................................81

4.4. Hypothalamic inflammation is elevated by high fat diet but not exposure to dim light

at night ...................................................................................................................82

5.1. Voluntary exercise prevents weight gain induced by dim light at night....................92

5.2. Dim light at night disrupts daily wheel running patterns in a subset of mice ............93

6.1. Mice exposed to dim light at night alter somatic measures .....................................107

6.2. Dim light at night attenuates clock gene expression ................................................108

6.3. Dim light at night suppresses Rev-Erb expression in peripheral tissue ...................109

Page 16: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

xv

Figure Page

6.4. Clock protein expression is reduced in the SCN of mice exposed to dimly lit

nights ....................................................................................................................110

7.1. Ambient lighting in cardiac intensive care unit patient rooms ................................131

7.2. Cardiac arrest and cardiopulmonary resuscitation surgical parameters ...................132

7.3. Dim light at night impairs cardiac arrest recovery...................................................133

7.4. Corticosterone concentrations in the 24 h following cardiac arrest .........................134

7.5. Cytokine expression is elevated in the hippocampus and microglia of mice exposed

to dim light at night following cardiac arrest .......................................................135

7.6. Selective inhibition of specific cytokines attenuates inflammation and neuronal cell

death following cardiac arrest and exposure to dim light at night .......................137

7.7. Manipulation of lighting wavelength minimizes light at night induced damage

following cardiac arrest........................................................................................138

8.1. Nile grass rats exposed to dimly lit nights elevate corticosterone concentrations and

delayed-type hypersensitivity swelling response .................................................158

8.2. Rats exposed to light at night enhance humoral immune function and plasma

bactericidal capacity.............................................................................................159

8.3. Grass rats exposed to dark and dimly lit nights have comparable activity ..............160

9.1. Grass rats exposed to dim light at night show impairments in learning and

memory ................................................................................................................175

9.2. Exposure to dim light at night increases depressive-like responses in grass rats ....176

9.3. Exposure to dim light at night alters hippocampal morphology in grass rats ..........177

Page 17: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

1

CHAPTER 1

THE “SKINNY” ON LIGHT AT NIGHT, CIRCADIAN CLOCKS, AND

METABOLISM

Over the course of the 20th

century the prevalence of obesity and metabolic

disorders rapidly increased worldwide. By the year 2000 the number of adults with

excess adiposity surpassed those who were underweight for the first time in evolutionary

history (Caballero, 2007). The growth in obesity has been exponential in recent decades

particularly for the highest weight categories. From 2000 to 2005 the number of

individuals qualifying as morbidly obese (BMI over 50) increased by 75% in the United

States (Sturm, 2007). Obesity is a pathogenic condition defined by the accumulation of

excess adipose tissue and is associated with serious health complications including

diabetes, cardiovascular disease, hypertension, asthma, and reproductive dysfunction

(Guh et al., 2009). Obesity reduces quality of life, results in significant health related

complications, and more than doubles healthcare costs. In addition to typical obesogenic

factors such as high calorie diet and sedentary lifestyles contributing to obesity, other

environmental factors are likely involved in the development and maintenance of this

condition (Symonds, Sebert, & Budge, 2011).

Page 18: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

2

Unprecedented transitions in human lifestyle occurred during the past century,

such as advances in travel and communication, greater urbanization, and the eradication

of multiple diseases (Engineering, 2000). One environmental change that had a mostly

unappreciated, yet dramatic, effect on human lifestyle was the widespread adoption of

electric lighting. Electric lights have provided many societal advances. Brightening the

night has shed the negative stigma of nighttime as a time solely for crime, sickness, and

death (Ekirch, 2005). The use of electric light at night played a large role in the industrial

revolution allowing for the creation of shift work. Furthermore, electric lighting has given

individuals the freedom to function on a self-selected sleep/wake schedule. Because the

invention of electric lighting occurred prior to an understanding of circadian biology,

little concern was given to the potential effects exposure to unnatural light schedules may

have on human health. It is becoming apparent, however, that there are significant

physiological repercussions associated with exposure to light at night (Fonken & Nelson,

2011; Navara & Nelson, 2007). In a sense, shift workers, who are exposed to high levels

of light at night in the workplace, have served as society‟s „canaries in the coal mine‟ for

maladaptive consequences of nighttime light exposure. Epidemiological evidence from

shift workers demonstrates that prolonged exposure to light at night increases the risk of

developing cancer (Stevens, 2009b), sleep disturbances (Kohyama, 2009), mood

disorders (Driesen, Jansen, Kant, Mohren, & van Amelsvoort, 2010), metabolic

dysfunction (B. H. Karlsson, Knutsson, Lindahl, & Alfredsson, 2003; Knutsson, 2003;

Obayashi et al., 2013; Parkes, 2002; Puttonen, Viitasalo, & Harma, 2011; van

Page 19: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

3

Amelsvoort, Schouten, & Kok, 1999), and cognitive impairments (K. Cho, 2001; Vetter,

Juda, & Roenneberg, 2012).

The reason that exposure to electric lights likely affects physiology is because

many organism have developed endogenously driven 24 h rhythms, termed circadian

rhythms, that are most potently synchronized by light information. The rotation of the

Earth about its axis produces a highly consistent cycle of light and dark that varies

latitudinally and on a seasonal basis. For more than 3 billion years, life on Earth has

evolved under bright days and dark nights. In order to optimally time physiological,

behavioral, and metabolic functions many organisms developed circadian rhythms.

Circadian rhythms allow organisms to anticipate predictable daily events such as food

availability and rest. Here I propose that increases in exposure to light at night during the

20th

century and concomitant changes in lifestyle are associated with alterations in

metabolism. In this introductory chapter, I will first provide an introduction to the

circadian system, with a specific emphasis on the effects of light on circadian rhythms.

Next I address interactions between the circadian system and metabolism. Animal models

have provided a vast amount of knowledge about the effects of circadian rhythm

disruption on metabolism, as well as the effects of disrupted metabolism on the circadian

system. Finally, I will tie in current experimental and epidemiological work associating

exposure to light at night and metabolism.

2. Circadian clock work

Most organisms, from unicellular cyanobacterium, to fruit flies and humans, have

developed an endogenous timekeeping system that synchronizes physiological and

Page 20: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

4

behavioral processes to the external solar cycle (Bell-Pedersen et al., 2005). Biological

clocks have the ability to both coordinate interactions among animals (e.g., knowing the

time of day can help animals avoid predation or engage in mating) and synchronize

internal physiological and biochemical processes within an individual (e.g., rhythmic

hormone release in anticipation of food availability and sleep). Circadian rhythms are

defined specifically as internally driven oscillation that meet several characteristics

including: (1) the period of the rhythm is about 24 h in the absence of environmental

cues, (2) rhythms are buffered against changes in environment such as temperature

fluctuations and behavioral feedback, and (3) rhythms can shift under the influence of

certain factors but entrainment is limited to a specific range (Dunlap, Loros, &

DeCoursey, 2004).

One common example of a circadian rhythm in mammals is the presence of a

sleep/wake cycle. In diurnal species, such as humans, sleep typically occurs during the

dark portion of the day, whereas nocturnal animals, such as house mice (Mus musculus),

generally sleep during the light. The propensity for sleep and activity is influenced by

endogenously controlled rhythmic hormone release. For example, cortisol secretion

spikes in the early morning directly prior to awakening in humans and then drops

throughout the day, reaching its nadir around the time of sleep onset (S. L. Bailey &

Heitkemper, 2001). Overall, circadian rhythms exist in many facets of physiology and the

importance of the circadian system is clearly demonstrated by considering pathogenic

conditions that result from altering circadian physiology (Takahashi, Hong, Ko, &

McDearmon, 2008). Circadian rhythm disruptions contribute to a wide range of disorders

Page 21: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

5

including cognitive impairments, mood disturbances, and increased risk of

cardiometabolic disorders (Zee, Attarian, & Videnovic, 2013). In order to understand

why these pathogenic conditions arise from disruption of the circadian clock it is first

important to understand where circadian oscillations originate.

2.1. The suprachiasmatic nucleus is the master circadian oscillator

The suprachiasmatic nuclei (SCN) of the hypothalamus comprise the master

circadian clock in mammals, at the top of a hierarchy of independent self sustaining

oscillators. The SCN is located in the anterior hypothalamus directly above the optic

chiasm and is composed of approximately 50,000 densely packed small neurons in

humans and 10-20,000 neurons in rodents (Dunlap, Loros, & DeCoursey, 2004). There

are several lines of evidence confirming that the SCN is the master circadian oscillator in

mammals: (1) SCN lesions abolish circadian rhythms (Stephan & Zucker, 1972), (2)

electrical and chemical stimulation of the SCN induce phase shifts (Michel et al., 2013;

Rusak & Groos, 1982), (3) transplanting an SCN into an animal whose own SCN has

been ablated restores circadian activity (R. Silver, LeSauter, Tresco, & Lehman, 1996),

and (4) individual neurons dissociated from the SCN display long term self-sustaining

oscillations (Welsh, Logothetis, Meister, & Reppert, 1995). Cellular synchrony within the

SCN is established through multiple mechanisms such as sodium dependent action

potentials (Yamaguchi et al., 2003) and humoral signals (R. Silver, LeSauter, Tresco, &

Lehman, 1996).

2.2. Molecular mechanisms of the circadian clock

Page 22: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

6

The circadian clock in mammals is driven by an autoregulatory feedback loop of

transcriptional activators and repressors (reviewed in (Mohawk, Green, & Takahashi,

2012; Reppert & Weaver, 2002). CLOCK and BMAL1 form heterodimers that induce

expression of Period (Per1, Per2, and Per3) and Cryptochrome (Cry1 and Cry2) through

E-box enhancers (Gekakis et al., 1998). PER and CRY proteins accumulate in the

cytoplasm throughout the circadian day. Upon reaching a critical amount, PER and CRY

form a complex that translocates back to the nucleus to associate with CLOCK and

BMAL and repress their own transcription (Mohawk, Green, & Takahashi, 2012; Reppert

& Weaver, 2002). This process takes approximately 24 h to complete a full cycle. In

addition to the primary feedback loop, other regulatory loops influence the circadian

clockwork. For example, the CLOCK:BMAL1 heterodimer also activates transcription of

retinoic acid-related orphan nuclear receptors, Rev-erbα and Rora, which have feedback

effects primarily on Bmal1 (Preitner et al., 2002).

Core clock components are defined as genes with protein products that are

essential for the generation and regulation of circadian rhythms (Takahashi, 2004).

Ablation of the core clock genes Clock, Bmal1 (Bunger et al., 2000), Per1, Per2 (Bae et

al., 2001), Cry1, and Cry2 (van der Horst et al., 1999) all disrupt circadian physiology

(see Table 1 in (Ko & Takahashi, 2006). The line between what constitutes a core clock

gene is constantly evolving. Rev-Erb and Per3 were not initially considered critical for

maintaining clock function; however, the importance of these genes for circadian

regulation is now widely accepted (H. Cho et al., 2012; Pendergast, Niswender, &

Yamazaki, 2012).

Page 23: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

7

2.3. Additional clocks persist outside the SCN

In multicellular organisms, circadian oscillators are present in most if not all

tissues. The SCN serves as the master circadian clock at the top of a hierarchically

organized system (Mohawk, Green, & Takahashi, 2012). Tissue specific clocks contain

the molecular machinery necessary for self sustaining oscillations (King et al., 1997) and

have virtually the same molecular makeup as circadian oscillators in the SCN. Peripheral

clocks are entrained by the SCN through both neural and hormonal signals (Guo, Brewer,

Champhekar, Harris, & Bittman, 2005; McNamara et al., 2001; Reddy et al., 2007), as

well as local factors such as nutritional signals (Vollmers et al., 2009). Peripheral clocks

do not appear to communicate with each other but they are coupled to the SCN (A. C. Liu

et al., 2007). Ablation of the SCN in vivo has profound effects on peripheral oscillators

(Akhtar et al., 2002) and peripheral oscillators show more rapid dampening of circadian

rhythms in vitro (Balsalobre, Damiola, & Schibler, 1998). Whereas SCN rhythms can

persist for more than one month in vitro, peripheral rhythms are not as robust and

diminish within two to seven cycles (Yamazaki et al., 2000).

2.4. Light entrains the circadian system

In most organisms, the circadian system functions at approximately but not

exactly 24 h. Therefore, circadian clocks require external input to entrain them to the

environment (Golombek & Rosenstein, 2010). Light is the most potent synchronizing

factor for the circadian system. Light information travels directly from intrinsically

photo-sensitive melanopsin containing retinal ganglion cells (ipRGCs), through the

retinohypothalmic tract, to the SCN (Hattar, Liao, Takao, Berson, & Yau, 2002). Within

Page 24: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

8

the SCN, light induces rapid changes in cellular activity that have been extensively

characterized by examining the expression of immediate early genes (Rusak, Robertson,

Wisden, & Hunt, 1990).

At the molecular level, exposure to light results in rapid induction of Per1

(Albrecht, Sun, Eichele, & Lee, 1997; Shigeyoshi et al., 1997). A pulse of light during the

night can phase advance or delay the circadian clock depending on the strength and time

of the light signal (Miyake et al., 2000). For example, light at dawn advances the clock

through advancing the onset of the Per1 rhythm and acutely increasing mRNA

transcription, whereas light at dusk delays the clock through delaying the offset of Per2

(Schwartz, Tavakoli-Nezhad, Lambert, Weaver, & de la Iglesia, 2012). The SCN can

rapidly adjust to light shifts, whereas peripheral tissues shift more slowly and in different

ways (Yamazaki et al., 2000). Although light is the most potent signal for the mammalian

circadian system, other factors such as food availability and locomotor activity can

feedback and influence circadian clock function (Fuller, Lu, & Saper, 2008; Mistlberger

& Antle, 2011). These types of stimuli can leave SCN rhythms intact, specifically

altering clock gene expression in peripheral tissues (Damiola et al., 2000; Vollmers et al.,

2009).

Multiple characteristics of the circadian system are conserved between rodents

and humans. In humans, specifically-timed light pulses can also shift the circadian clock

(Czeisler et al., 1989; Smith, Revell, & Eastman, 2009). Moreover, both humans and

rodents are most responsive to 460 nm nighttime light exposure (Brainard, Richardson,

Petterborg, & Reiter, 1982; Ruger et al., 2013). Longer wavelengths of lighting, such as

Page 25: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

9

red light, do not activate the melanopsin containing retinal ganglion cells that project to

the SCN and therefore minimally influence the circadian system (Brainard et al., 2008;

Figueiro & Rea, 2010).

Exposure to constant dim light or total darkness results in a free-running circadian

system. This knowledge has been used to test the effects of different factors, such as

melatonin, on synchronizing circadian activity (Redman, Armstrong, & Ng, 1983). In

contrast, the effects of dim and bright light at night on the circadian system are less well

documented. In Chapter 2 of this dissertation I demonstrate that exposure to constant

light alters activity rhythms and flattens circadian rhythms in glucocorticoids (Coomans

et al., 2013; Fonken et al., 2010), two principle outputs of the circadian system.

There is evidence that very dim levels of light at night can influence circadian

rhythms in both rodents and humans (Evans, Elliott, & Gorman, 2005; Jasser, Hanifin,

Rollag, & Brainard, 2006). However, the effects of physiologically relevant levels of

light exposure on the circadian system are not well characterized. Therefore, in this

dissertation I evaluate the effects of chronic exposure to dim light at night (~5 lux) on the

murine circadian system. The level of dim light used in my studies is ecologically

relevant and comparable to levels of light pollution found in urban areas (Gaston, Davies,

Bennie, & Hopkins, 2012; Kloog, Haim, & Portnov, 2009) and sleeping environments

(Obayashi et al., 2013). In Chapter 5, I document that exposure to chronic low levels of

light at night alters circadian clock genes in both the SCN and peripheral tissue (Fonken,

L.K., Aubrecht, T.G., Meléndez-Fernández, O.H., Weil, Z.M., & Nelson, R.J., unpublished

observations). Exposure to dim light at night specifically attenuates the rhythm in Per1

Page 26: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

10

and Per2 gene and protein expression in the SCN around the light/dark transition.

Furthermore, expression of Bmal1, Per1, Per2, Cry1, Cry2, and Rev-Erb are all

suppressed in the liver. Nocturnal light may affect the liver through both autonomic and

hormonal pathways (Cailotto et al., 2009). The changes in clock gene expression

associated with exposure to dim light at night do not result in disruption of either the

glucocorticoid rhythm or locomotor activity rhythm (Chapter 2). Similar changes in

circadian clock function are also apparent in the SCN of hamsters exposed to low levels

of light at night (Bedrosian, T.A., Galan, A., Vaughn, C.A., Weil, Z.M., & Nelson, R.J.,

unpublished observations). Hamsters exposed to dim light suppress PER1 and PER2

protein rhythms in the SCN independent of changes in activity rhythm. Overall, these

results indicate that exposure to levels of nighttime lighting that are commonly found in

urban settings can affect the circadian system.

3. The circadian system regulates metabolism and vice versa

Approximately 10% of the mammalian transcriptome displays circadian

regulation (K. L. Eckel-Mahan et al., 2012; Panda et al., 2002; Storch et al., 2002).

Among the rhythmic genes identified, many have a specific role in coordinating nutrient

metabolism (K. Eckel-Mahan & Sassone-Corsi, 2009). For example, there is circadian

expression of glucose transporters and the glucagon receptor (Panda et al., 2002),

multiple enzymes involved in the metabolism of sugars and the biosynthesis of

cholesterols display circadian oscillation (la Fleur, Kalsbeek, Wortel, Fekkes, & Buijs,

2001; Panda et al., 2002), and PCG-1α, an essential activator of gluconeogensis, has a

key role in regulating circadian rhythms (C. Liu, Li, Liu, Borjigin, & Lin, 2007).

Page 27: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

11

Metabolically related hormones such as glucagon, insulin, ghrelin, leptin, and

corticosterone also oscillate in a circadian fashion (Kalsbeek et al., 2001; Ruiter et al.,

2003; Sinha et al., 1996). There is rhythmic expression of orexigenic signals including

neuropeptide Y, galanin, and pre-opiomelanocortin within the hypothalamus, an area

critical for coordinating metabolic signals (Jhanwar-Uniyal, Beck, Burlet, & Leibowitz,

1990; B. Xu, Kalra, Farmerie, & Kalra, 1999). Moreover, neuroanatomical organization

provides evidence of interactions between metabolism and the circadian system;

hypothalamic nuclei such as the paraventricular nucleus receive direct neuronal input

from the SCN (reviewed in (Kalra et al., 1999).

3.1. Knocking out the circadian clock and obesity

In addition to fluctuations in metabolic processes suggesting an association

between the circadian and metabolic systems, disrupting the clock network through

genetic manipulations has provided insight into the role of the circadian system in

maintaining metabolic homeostasis. Mice harboring mutations in various components of

the circadian clock are susceptible to obesity and metabolic syndrome. Turek and

colleagues were the first to report that Clock mutant mice on a BALB/c and C57BL/6J

background are susceptible to diet induced obesity. Clock mutants show marked changes

in circadian rhythmicity, as well as disruptions in diurnal food intake and increased body

mass (Turek et al., 2005). This phenotype may partially result from changes in endocrine

regulation as serum leptin, glucose, cholesterol, and triglyceride levels are all increased in

Clock mutants compared to wild type mice. Deletion of Clock on an ICR background also

results in metabolic alterations. In contrast to Clock deletion in a BALB/c and C57BL/6J

Page 28: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

12

background, ICR Clock deficient mice are protected against weight gain due to

impairments in dietary fat absorption (Oishi et al., 2006).

Disruption of other core circadian clock genes similarly affects metabolism. Mice

deficient in Bmal1 alter insulin and glucose secretion (Marcheva et al., 2010) and

rescuing Bmal1 in the central nervous system restores activity rhythms but not changes in

metabolism (McDearmon et al., 2006). Mutation of Cry1 produces symptoms of diabetes

mellitus in mice (Okano, Akashi, Hayasaka, & Nakajima, 2009). Furthermore, mice

deficient in mPer1/2/3 increased weight gain on a high fat diet (Dallmann & Weaver,

2010) and single disruption of the Per2 gene alters glucose homeostasis (Carvas et al.,

2012).

Loss of clock function in peripheral tissues also affects metabolism. Deletion of

pancreatic Clock or Bmal1 reduces glucose and insulin processing abilities, independent

of changes in activity or feeding rhythms in mice (Marcheva et al., 2010). Mice with

liver-specific deletion of Bmal1 exhibit hypoglycemia during the fasting phase,

exaggerated glucose clearance, and loss of rhythmic expression of hepatic glucose

regulatory genes (Kornmann, Schaad, Bujard, Takahashi, & Schibler, 2007; Lamia,

Storch, & Weitz, 2008). Changes in glucose processing with liver specific Bmal1 deletion

occur independently of alterations in feeding behavior or locomotor activity, indicating a

primary defect in metabolic responses (Lamia, Storch, & Weitz, 2008). In contrast,

deletion of Bmal1 in adipocytes alters daily feeding rhythms and results in obesity

(Paschos et al., 2012). Finally, Bmal1 deletion in the central nervous system produces

Page 29: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

13

deficits in locomotor activity entrainment by periodic feeding, reductions in food intake,

and subsequent loss of body weight (Mieda & Sakurai, 2011).

Alterations in secondary clock genes are also associated with metabolic changes

in mice. Modulation of Per3 may be responsible for weight gain in Per1/2/3-/- mice, as

single deletion of Per3 results in significant weight gain (Dallmann & Weaver, 2010).

Mice lacking the VIP-VPAC2 pathway, which plays an important role in SCN

communication (Reppert & Weaver, 2001), dampen feeding rhythms and show

reductions in metabolic rate (Bechtold, Brown, Luckman, & Piggins, 2008). Furthermore,

mice lacking Nocturnin, a gene involved in the posttranscriptional regulation of rhythmic

gene expression (Baggs & Green, 2003), remain lean on a high fat diet. This appears to

be due to either changes in lipid uptake or utilization as genes important for lipid

pathways lose rhythmicity in Nocturnin-/- mice (Douris et al., 2011; Green et al., 2007).

Recently, Rev-Erb has been implicated in the modulation of both metabolism and the

circadian system (Yin et al., 2007). Dual deletion of Reb-Erbα and Rev-Erbβ disrupts

gene networks involved in lipid metabolism (H. Cho et al., 2012) and markedly affects

circadian rhythms. Rev-Erb likely regulates hepatic lipid homeostasis through the

recruitment of histone deacetylase 3 (Feng et al., 2011). Treatment with a Rev-Erb

agonist increases energy expenditure and promotes weight loss in mice fed a high fat diet

(Solt et al., 2012).

Associations between changes in clock genes and metabolism are also apparent in

humans. Body mass index correlates with clock gene expression in peripheral adipose

tissue depots (Zanquetta et al., 2012). Per2 expression levels in visceral adipose tissue

Page 30: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

14

inversely correlate with waist circumference (Gomez-Abellan, Hernandez-Morante,

Lujan, Madrid, & Garaulet, 2008) and Bmal, Per2, and Cry1 levels negatively correlate

with total cholesterol and LDL concentrations. The methylation pattern of difference

CpG sites of Clock, Bmal, and Per1, are significantly associated with metabolic

parameters including body mass index, adiposity, and metabolic syndrome score

(Milagro et al., 2012). Moreover, a common clock polymorphism is coupled to the

presence of metabolic syndrome in humans (E. M. Scott, Carter, & Grant, 2008).

3.2. Metabolism and the circadian clock are reciprocally related

The relationship between the circadian system and metabolism appears to be bi-

directional. In addition to circadian system disruptions causing obesity, metabolic

abnormalities alter circadian rhythms. For example, in humans, metabolically related

diseases such as obesity and anorexia nervosa are associated with altered hormone and

body temperature rhythms (Ferrari, Fraschini, & Brambilla, 1990). Daily rhythms in

glucose and insulin sensitivity are apparent in non-obese but not obese subjects (Lee,

Ader, Bray, & Bergman, 1992). Moreover, obese women display significantly lower

wrist temperature and a more flattened temperature rhythm compared to normal-weight

control women (Corbalan-Tutau et al., 2011).

An intriguing interaction between high fat diets and clock genes has been

discovered. Placing mice on a high fat diet lengthens the circadian period of activity and

attenuates the diurnal pattern of feeding (Kohsaka et al., 2007; Stucchi et al., 2012). Mice

fed a high fat diet dampen circadian rhythms in clock gene expression, specifically in

peripheral metabolically related tissues. These changes appear to occur rapidly, as liver

Page 31: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

15

rhythms are phase advanced 5 h, within one week of initiating a high fat diet (Pendergast

et al., 2013). Furthermore, mice fed a high fat diet show a slower rate of re-entrainment

of behavioral and physiological rhythms after a 6 h phase shift (Mendoza, Pevet, &

Challet, 2008).

Modulation of leptin in mice and rats provides further support for the hypothesis

that disrupting metabolism affects the circadian system. Zucker obese rats, a widely used

obesity model produced by a single mutation in the gene encoding the leptin receptor

(Phillips et al., 1996), exhibit phase advanced circadian rhythms and an attenuated

amplitude of body temperature, activity, and sleep (Mistlberger, Lukman, & Nadeau,

1998; Murakami, Horwitz, & Fuller, 1995). Zucker rats increase daytime food

consumption when compared to lean controls (Becker & Grinker, 1977; Wangsness,

Dilettuso, & Martin, 1978) and preventing food intake during the light phase ameliorates

weight gain in obese rats (Mistlberger, Lukman, & Nadeau, 1998). Furthermore, db/db

mice, which lack the leptin receptor, show changes in the diurnal rhythms of multiple

metabolically related hormones (Roesler, Helgason, Gulka, & Khandelwal, 1985) and

clock genes in peripheral tissues (Su et al., 2012). ob/ob leptin deficient mice attenuate

rhythms in activity, heat production, and clock gene expression (Dauncey & Brown,

1987). Disruptions in circadian rhythms, however, may occur prior to the onset of obesity

in ob/ob mice (Ando et al., 2011).

Other obesity models display similar changes in circadian rhythms. Rats with

ventromedial hypothalamic lesions become obese and alter the daily pattern of food

intake and circadian gene expression (Balagura & Devenport, 1970). KK-Ay mice, a

Page 32: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

16

mouse model of obesity and type II diabetes, attenuate clock gene rhythms in adipose

tissue and the liver (Ando et al., 2005; Hashinaga et al., 2012). Moreover, a subset of

Volcano mice are naturally susceptible to obesity and display phase advanced activity-

onset and attenuated locomotor activity rhythms compared to non-obese Volcano mice

(Carmona-Alcocer et al., 2012). Of note, one common feature in many of the obesity

models discussed above is that rodents shift diurnal rhythms in food intake.

3.3. You are what when you eat?

Food is an entraining signal for the circadian system. Restricting feeding to

certain times of day can lead to anticipatory increases in wakefulness, locomotor activity,

body temperature, and glucocorticoid secretion (Krieger, 1974). Food entrainable

oscillators appear dependent on extra-SCN signaling and likely involve the dorsomedial

hypothalamus (Gooley, Schomer, & Saper, 2006). Indeed, while the SCN remains phase

locked to light/dark cues, restricting feeding to certain times of day rapidly entrains

circadian rhythms in the liver (Stokkan, Yamazaki, Tei, Sakaki, & Menaker, 2001). In the

absence of light cues, however, restricted feeding is capable of affecting the SCN. For

example, in mice made arrhythmic by constant light exposure, restricted feeding rescues

both activity rhythms and Per2 rhythms in the SCN (Lamont, Diaz, Barry-Shaw, Stewart,

& Amir, 2005).

Timing of food intake is now recognized as a critical factor in energy acquisition,

storage, and expenditure. Mice fed a high fat diet only during the 12 h light (resting)

phase gain significantly more weight than mice fed only during the dark (active) phase

(Arble, Bass, Laposky, Vitaterna, & Turek, 2009). This change in body mass may be

Page 33: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

17

dependent on leptin signaling (Arble, Vitaterna, & Turek, 2011). Importantly, in this

model total daily caloric intake and activity do not differ between groups, indicating

changes in body mass can occur independently of changes in energy intake (Arble, Bass,

Laposky, Vitaterna, & Turek, 2009). A more thorough examination of metabolic

characteristics in food restricted mice revealed light-phase fed mice exhibit a higher

respiratory exchange ratio (indicating decreased reliance on fat oxidation), tissue-specific

alterations in metabolically-related genes and circadian clock genes, changes in the

diurnal variations in humoral factors (i.e. corticosterone), and increased weight gain

within 9 days of restricting feeding (Bray et al., 2012). In the study by Bray and

coauthors, mice did show changes in food intake; mice fed during the light phase had

increases in food intake as well as a larger meal on presentation of food (Bray et al.,

2012). The time of day at which dietary fat is consumed also influences multiple

cardiometric parameters (Bray et al., 2010). Consumption of a high fat meal at the end, as

compared to the beginning of the active phase, leads to features of metabolic syndrome

including increased weight gain, elevated adiposity, reductions in glucose processing,

hyperinsulemia, hyperleptinemia, and hypertrigluceridemia (Bray et al., 2010).

As discussed above, multiple obesity models show increases in rest-phase food

intake. This has led to recent research assessing whether preventing rest-phase feeding

can ameliorate weight gain. Mice fed a high fat diet that are food restricted to either 4

(Sherman et al., 2012) or 8 (Hatori et al., 2012) h per day during the active phase are

protected against diet induced obesity. Food restricted mice consume equivalent calories

as mice with ad libitum food access and yet are buffered against obesity, inflammation,

Page 34: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

18

hepatic steatosis, hypercholesterolemia, and hyperinsulinemia. Changes in metabolism in

food restricted mice are associated with improved intracellular signaling and nutrient

utilization as well as greater circadian clock oscillations (Hatori et al., 2012; Sherman et

al., 2012). Similarly, restricting food access to the 14 h dark phase reduces weight gain in

Zucker obese rats (Mistlberger, Lukman, & Nadeau, 1998).

Accumulating epidemiological and experimental evidence in humans, supports

the hypothesis that when energy intake occurs is important in determining energy

utilization. Timing of food intake predicts body mass index in humans, even when

controlling for variables such as sleep timing and duration. People who consume more

food after 2000 h tend to have higher body mass index (Baron, Reid, Horn, & Zee, 2013;

Baron, Reid, Kern, & Zee, 2011). Moreover, weight loss therapy is more effective for

individuals who eat early as compared to late eaters (Garaulet et al., 2013). Short duration

sleepers also have increased risk for obesity, weight gain over time, and higher body fat

composition (Weiss et al., 2010). Controlling for factors such as preference for fatty food,

skipping breakfast, snacking, and eating out only partially accounts for the effects of

short duration sleep on obesity, suggesting that changes in metabolic homeostasis at

different times of day may partially account for different body weight regulation

(Nishiura, Noguchi, & Hashimoto, 2010). Indeed, people with Night Eating Syndrome, a

disorder characterized by evening hyperphasia and nocturnal awakenings accompanied

by food intake (Allison et al., 2010), show alterations in metabolically related hormones

and are more likely to be obese (Birketvedt et al., 1999; Goel et al., 2009).

4. Exposure to light at night and obesity

Page 35: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

19

Given the well established role of light in modulating the circadian system, and

the relationship between circadian and metabolic functions, it is not surprising that

exposure to light at unnatural times affects metabolism. Increases in nocturnal

illumination parallel increases in obesity and metabolic syndrome worldwide. Here I

propose that exposure to light at night may be contributing to increasing rates of obesity.

In this section I will discuss evidence from animal models and epidemiological studies

implicating exposure to light at night in the growing obesity epidemic.

4.1. Light at night and obesity: evidence from animal models

Exposure to continuous light, non-24 h light schedules, and dim light at night are

all associated with metabolic changes in rodents. First, exposure to constant light

desynchronizes circadian activity in rodents (Coomans et al., 2013). In Chapter 2 of this

dissertation I describe the effects of exposure to constant light on metabolism in mice.

Swiss Webster mice exposed to constant light as compared to a standard light/dark cycle

show increases in body mass and reductions in glucose processing without altering total

daily activity or food intake (Fonken et al., 2010). C57/Bl/6J mice show similar

metabolic changes in constant light, with immediate body weight gain upon placement in

constant light and reductions in insulin sensitivity. Following 4 weeks of exposure to

constant light, mice lack a circadian rhythm in both food intake and energy expenditure.

These changes are associated with reductions in circadian rhythm amplitude as measured

by in vivo electrophysiological recordings of the SCN (Coomans et al., 2013). Rats also

change metabolism with continuous light exposure. Rats exposed to constant light

Page 36: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

20

increase visceral adiposity and demonstrate higher feed efficiency than rats exposed to

either a standard light/dark cycle or constant dim light (Wideman & Murphy, 2009).

One limitation to studying the effects of constant light exposure on metabolism is

that the circadian system either free-runs or becomes arrhythmic under constant light

conditions (Fonken et al., 2010; Lamont, Diaz, Barry-Shaw, Stewart, & Amir, 2005).

Furthermore, with the exception of high latitudes, exposure to constant light in natural

settings is rare. For this reason, in Chapters 2-6 of this dissertation I evaluate the effects

of exposure to ecologically relevant levels of dim nighttime light (~5 lux) exposure on

metabolic function. Mice exposed to dimly lit, as compared to dark nights, impair glucose

processing, increase white adipose tissue, and elevate body mass gain (Chapter 2,

(Fonken et al., 2010)). Changes in metabolism occur independently of changes in total

daily food intake or locomotor activity. However, mice exposed to dim light at night shift

timing of food intake, consuming more during the light phase. Much as other obesity

models, restricting food intake to the dark phase prevents weight gain in mice exposed to

dim light at night.

Metabolic changes associated with exposure to dim light at night are not

permanent; placing mice back in dark nights partially reverses increases in body mass

caused by dim light at night exposure (Chapter 3 (Fonken, Weil, & Nelson, 2013)).

Moreover, exposure to dim light at night interacts with more traditional obesogenic risk

factors to affect weight gain. Exposure to dim nights exaggerates weight gain on a high

fat diet, increasing peripheral but not hypothalamic inflammation (Chapter 4). Increases

in weight gain associated with exposure to light at night are also reduced by providing a

Page 37: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

21

running wheel for voluntary exercise (Chapter 5). Providing mice a running wheel in

dim light at night prevents increases in weight gain without restoring feeding rhythms.

Models of circadian desynchrony provide further support for an association

between exposure to altered light schedules and changes in metabolism. Placing mice in a

20 h light/dark cycles incongruous with their endogenous ~24 h circadian period,

accelerates weight gain and alters metabolically related hormones (Karatsoreos, Bhagat,

Bloss, Morrison, & McEwen, 2011). Circadian desynchrony in mouse models of shift

work, demonstrate altered light or activity patterns, can disrupt liver transcriptome

rhythms (Barclay et al., 2012), flatten glucose rhythms, increase abdominal fat (Salgado-

Delgado, Angeles-Castellanos, Saderi, Buijs, & Escobar, 2010), and cause reductions in

body mass when shift work is combined with other environmental challenges (Preuss et

al., 2008). Notably, preventing daytime food intake may rescue metabolic changes in

mice undergoing a shift work paradigm (Salgado-Delgado, Angeles-Castellanos, Saderi,

Buijs, & Escobar, 2010).

4.2. Exposure to light at night and obesity: Evidence in humans

Multiple epidemiological studies in shift working populations have linked

exposure to light at night to altered metabolism (X. S. Wang, Armstrong, Cairns, Key, &

Travis, 2011). Health care personnel that work night, as compared to day shifts, show

elevated risk for metabolic syndrome (Pietroiusti et al., 2010). Shift work is associated

with increased blood pressure, cholesterol, obesity, and hypertriglyceridemia in males

(Ha & Park, 2005; B. Karlsson, Knutsson, & Lindahl, 2001) and increased risk for

obesity, hypertension, and hypertriglyceridemia in females (B. Karlsson, Knutsson, &

Page 38: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

22

Lindahl, 2001). A study of offshore personnel either chronically working day shifts or

transitioning between day and nights shifts, demonstrated that the number of years of

shift work is positively associated with body mass index in the day/night workers. In

contrast, age is the greatest predictor for body mass index in the day shift population

(Parkes, 2002). This suggests that chronic exposure to light at night can affect body mass

index to a more appreciable extent than typical predictors of body mass, such as age.

Several other studies indicate that working rotating shift schedules as compared to day

schedules increases risk for developing metabolic syndrome (De Bacquer et al., 2009;

Esquirol et al., 2009; Lin, Hsiao, & Chen, 2009a, , 2009b; Sookoian et al., 2007).

Importantly, the effects of shift work on metabolism may be long lasting as former shift

workers show increases in obesity (Puttonen, Viitasalo, & Harma, 2011). Changes in

metabolic signals may contribute to increased body mass in shift workers. For example,

brief behavioral and endogenous circadian misalignment in a controlled laboratory setting

alters leptin, insulin, and cortisol secretion and elevates blood pressure (Scheer, Hilton,

Mantzoros, & Shea, 2009). Moreover, forced desynchrony can produce postprandial

glucose responses comparable to a prediabetic state (Scheer, Hilton, Mantzoros, & Shea,

2009).

In industrialized economies, approximately 20% of the population are shift

workers (Monk, 2000; Rajaratnam & Arendt, 2001). However, exposure to light at night

occurs beyond the scope of shift work. Over 99% of the population in US and Europe

experiences nighttime light exposure (Cinzano, Falchi, & Elvidge, 2001). Associations

between exposure to light at night and increases in body mass index are also apparent

Page 39: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

23

outside the shift working population. A recent study by Obayashi et al reported that

increased light exposure in an uncontrolled home setting is associated with obesity and

other metabolic consequences. People with nocturnal light levels above 3 lux display

significantly higher body weight, elevated body mass index, increased waist

circumference, and elevated triglyceride and low-density lipoprotein cholesterol levels

(Obayashi et al., 2013). Moreover, a large scale epidemiological study demonstrated that

“social jetlag” is associated with increased BMI, even when controlling for factors such

as sleep duration (Roenneberg, Allebrandt, Merrow, & Vetter, 2012).

One final piece of evidence linking exposure to light at night and obesity comes

from a population where nighttime light exposure is minimal, the Amish. The Old Order

Amish abstain from using public power, and therefore, are not exposed to common

sources of light at night such as televisions, computers, and electric lights (S. Scott &

Pellman, 1990). The prevalence of obesity among the Amish is far lower than the general

population in the United States (Tremblay, Esliger, Copeland, Barnes, & Bassett, 2008).

Lower obesity rates among the Amish are for the most part attributed to changes in

activity and diet (Esliger et al., 2010; Tremblay, Esliger, Copeland, Barnes, & Bassett,

2008). However, other environmental factors are likely involved in limiting disease rates

among the Amish. For example, the Amish also suffer disproportionately lower rates of

breast and prostate cancer (Westman et al., 2010). Controlling for known carcinogenic

variables such as tobacco use does not completely account for the disparate rates of

cancer. Importantly, exposure to light at night is associated with increased risk for

Page 40: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

24

developing both breast and prostate cancer (Kloog, Haim, Stevens, Barachana, &

Portnov, 2008; Kloog, Haim, Stevens, & Portnov, 2009).

4. Conclusions

In this introduction, I specifically focused on how light at night may influence

metabolism through disruption of the circadian clock genes. There are, however, several

additional pathways through which light can affect metabolism, including disruptions in

melatonin production, alterations in glucocorticoids, and sleep disturbances (Reiter, Tan,

Korkmaz, & Ma, 2011). Exposure to sufficient levels and duration of nighttime lighting

can suppress pineal melatonin secretion (Brainard, Rollag, & Hanifin, 1997). Melatonin

is an endogenously synthesized molecule that is secreted by the pineal gland during the

night in both nocturnal and diurnal mammals (Reiter, 1991). Melatonin has recently been

reported to have anti-obesity effects (Mantele et al., 2012; Tan, Manchester, Fuentes-

Broto, Paredes, & Reiter, 2011). Although there is compelling evidence that suppression

of melatonin secretion can contribute to weight gain, I specifically did not focus on

melatonin for several reasons: (1) pineal melatonin suppression requires high and

sustained levels of nighttime light exposure (Brainard, Rollag, & Hanifin, 1997), (2)

nighttime light exposure below the threshold for melatonin suppression is associated with

changes in metabolism (Obayashi et al., 2013), and (3) multiple strains of laboratory mice

that lack pineal melatonin demonstrate changes in metabolism with nighttime light

exposure (Coomans et al., 2013; Fonken et al., 2010). Additionally, I did not focus on the

effects of exposure to light at night on sleep or glucocorticoids because nighttime light

Page 41: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

25

exposure affects metabolism in mice independently of changes in sleep architecture or

corticosterone release.

Chapters 2-7 of this dissertation are all conducted in nocturnal laboratory mice.

A nocturnal rodent was selected for the studies in order to separate the effects of

exposure to light at night from sleep disruption. Indeed, exposure to dimly lit nights does

not interrupt sleep in Swiss Webster mice (Borniger, J., Weil, Z.M., & Nelson, R.J.,

unpublished observations). However, both circadian influences on behavior and the

masking effects of light are very different in diurnal as compared to nocturnal species (R.

Cohen, Kronfeld-Schor, Ramanathan, Baumgras, & Smale, 2010; Shuboni, Cramm, Yan,

Nunez, & Smale, 2012). Rhythmic release of multiple hormones also occurs 180° degrees

out of phase between diurnal and nocturnal rodents. Thus, in the final two chapters of this

dissertation (Chapters 8 & 9), I consider the physiological implications of exposing

diurnal Nile grass rats (Arvicanthis niloticus) to dim light at night. Grass rats exposed to

dimly lit as compared to dark nights, enhance cell-mediated immune function as

measured by delayed-type hypersensitivity, elevate antibody production following

inoculation with keyhole lymphocyte hemocyanin, and increase bactericidal capacity

(Chapter 8 (Fonken, Haim, & Nelson, 2011)). Furthermore, Grass rats exposed to dim

light at night display cognitive impairments in a Barnes maze test and increases in

depressive-like behavior (Chapter 9 (Fonken, Haim, & Nelson, 2011)). Changes in

cognitive and affective responses are associated with altered hippocampal connectivity in

grass rats exposed to dimly lit as compared to dark nights. In contrast to nocturnal rodents

exposed to dim light at night (Fonken et al., 2010), grass rats chronically exposed to

Page 42: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

26

dimly lit nights elevate corticosterone concentrations compared to rats exposed to dark

nights.

One important population that is often neglected when considering light at night is

patients in hospitals. Although multiple epidemiological studies have been conducted on

nurses, there are no studies on the affects of light at night on the patients with whom they

work. Many in-patients are already at high risk of increased inflammation and disrupted

physiology, which may be exacerbated by light at night. For example, the circadian

system regulates multiple immune related functions (Lange, Dimitrov, & Born, 2010).

Antigen presentation, toll-like receptor function, cytokine gene expression, and

lymphocyte proliferation all occur in a circadian pattern (Arjona & Sarkar, 2006; A. C.

Silver, Arjona, Walker, & Fikrig, 2012). Furthermore, circadian clock proteins directly

regulate the expression of pro-inflammatory cytokines (Narasimamurthy et al., 2012).

Immune cells such as natural killer cells, macrophages, dendritic cells, and B cells

possess molecular clock mechanisms necessary for self-sustaining oscillations (Arjona &

Sarkar, 2005; Keller et al., 2009; A. C. Silver, Arjona, Hughes, Nitabach, & Fikrig,

2012). Because recent research has demonstrated that light at night may detrimentally

affect the immune system (Bedrosian, Fonken, Walton, & Nelson, 2011a) in Chapter 7

of this dissertation I investigate the effects of exposure to light at night on recovery from

global cerebral ischemia.

Most permanent damage to the central nervous system that occurs post-ischemia

is mediated by endogenous secondary processes, typically involving inflammation. The

delay in damage following cerebral ischemia suggests that the immediate post-recovery

Page 43: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

27

environment may affect the trajectory of recovery. Because the circadian system is

controlled by an endogenous biological clock, and physiological processes such as

inflammation become dysregulated in disruptive lighting conditions, I hypothesized that

cardiac arrest outcome may be negatively affected by light at night. In agreement with the

hypothesis, mice exposed to dim light at night in the week following a cardiac arrest and

cardiopulmonary resuscitation procedure increase mortality compared to mice exposed to

dark nights. Moreover, mice exposed to dim light at night increase hippocampal cell

death, microglia activation, and pro-inflammatory cytokine expression. Selectively

inhibiting IL-1β or TNFα ameliorate damage in mice exposed to dim light at night,

suggesting increases in inflammation are critical for light-associated damage. Restricting

the wavelength of the nighttime light exposure to ~640 nm also reduces

neuroinflammation and eliminates the detrimental effects of light at night on CA

outcome. These findings implicate the involvement of the circadian system in dim light at

night-associated damage.

Modern society now functions on a 24 h schedule. Although there are many

economic and other societal benefits to such a schedule, there is converging evidence

from epidemiological and experimental work that light at night has unintended,

maladaptive consequences. In many ways, this field of study is just beginning; further

characterization of the biological and psychological effects of light at night is needed

along with effective interventions to ameliorate the unintended negative effects of light at

night on health.

Page 44: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

28

Overall, preventing the general population from excessive exposure to light at

night can be achieved with relatively low-cost manipulations, such as using curtains to

block out street lights, turning off hallway lights, and removing all light sources,

including televisions and computers, from bedrooms. However, these methods do not

prevent the social jet lag that many of us experience and exposure to light at night is often

unavoidable in shift-working populations. To that end, there are studies currently

comparing visual aids that may alleviate some of the maladaptive effects of exposure to

light at night in shift workers. Specifically, manipulation of wavelength may prove

effective in blocking out some of the light-induced physiological changes.

Page 45: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

29

CHAPTER 2

LIGHT AT NIGHT INCREASES BODY MASS THROUGH ALTERED TIMING

OF FOOD INTAKE

During the past two decades obesity has shifted from a US-centered epidemic to a

global issue. Although well-documented factors such as caloric intake, dietary choices,

and lack of exercise are known to contribute to the prevalence of obesity and metabolic

disorders, additional environmental factors are now considered critical in the

development and maintenance of obesity (Hill, Wyatt, Reed, & Peters, 2003). The

increase of light at night (LAN) during the 20th

century coincides with increasing rates of

obesity and metabolic disorders throughout the world. Artificial lighting allows people to

extend daytime activities into the night, but as a consequence produces significant

environmental light pollution caused by light straying into the atmosphere and

brightening the nighttime sky.

Circadian regulation of energy homeostasis is controlled by an endogenous

biological clock, located in the suprachiasmatic nuclei (SCN) of the hypothalamus that

are synchronized by photic information that travels directly from light-sensitive ganglion

cells in the retina to the SCN, thereby entraining individuals‟ physiology and behavior to

Page 46: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

30

the external day-night cycle (Golombek & Rosenstein, 2010). Importantly, light is the

most potent entraining signal for the circadian clock, although other factors such as food

consumption influence clock signaling (Fuller, Lu, & Saper, 2008). To promote optimal

adaptive functioning, the circadian clock prepares individuals for predictable events such

as food availability and sleep. Shift work disrupts clock function and is linked to

circadian and metabolic consequences including sleep disturbances (Kohyama, 2009),

elevated body mass index (BMI) (Parkes, 2002; van Amelsvoort, Schouten, & Kok,

1999), altered plasma lipid metabolism and adiposity (B. H. Karlsson, Knutsson, Lindahl,

& Alfredsson, 2003), and increased risk for cardiovascular disease (Ha & Park, 2005).

Multiple studies suggest a link between the molecular circadian clock and

metabolism (for review see (Bray & Young, 2007). Mice harboring a mutation in their

clock genes are susceptible to obesity and metabolic syndrome (Turek et al., 2005).

Clock mutants show profound changes in circadian rhythmicity, as well as disrupted

diurnal food intake and increased body mass. Serum leptin, glucose, cholesterol, and

triglyceride levels are also increased in Clock mutants compared to wild type mice. Mice

lacking the VIP-VPAC2 pathway which plays an important role in SCN communication

(Reppert & Weaver, 2001) have metabolic abnormalities similar to those in Clock mutant

mice (Bechtold, Brown, Luckman, & Piggins, 2008). Furthermore, consumption of a

high-fat diet alters circadian rhythmicity and the cycling of circadian clock genes in mice

(Kohsaka et al., 2007).

Because multiple studies have linked disruption of the molecular circadian clock

and metabolic disorders (Bechtold, Brown, Luckman, & Piggins, 2008; Rudic et al.,

Page 47: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

31

2004; Turek et al., 2005), I hypothesized that exposure to light at night alters circadian

organization and affects metabolic parameters. I investigated the possibility of a direct

link between altered light cycles and metabolic disorder by housing mice in either a

standard light/dark cycle (LD; 16 h light at ~150 lux/8 h dark at ~0 lux), a light/dim light

cycle (dLAN; 16h light at ~150 lux/ 8h dim light at ~5 lux), or 24 h of continuous

lighting (LL; constant ~150 lux) and assessed metabolic parameters. I included a dLAN

group in addition to LL because mice in constant lighting have no temporal cue to

distinguish time of day, and their biological clocks free-run. The dLAN group may more

directly model environmental light pollution experienced in industrialized nations;

however, I predicted circadian alterations in daily activity would not be as profound in a

dLAN environment. By including the LL group, this study not only focused on the

effects of light pollution, but also on the effects of a desynchronized circadian system on

metabolism. I hypothesized that mice housed in dLAN and LL conditions would alter

metabolic parameters compared to mice housed in LD. More specifically, I hypothesized

that housing mice in dLAN and LL would result in reduced glucose tolerance and

increased body mass in comparison to LD-housed mice. I also hypothesized that mice

housed in LL and dLAN would alter stress levels as evaluated by circulating

corticosterone and that LL would induce locomotor arrhythmicity indicative of circadian

disruption.

Methods

Animals

Page 48: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

32

Eighty male Swiss Webster mice (~8 weeks of age) were obtained from Charles

River Labs (Kingston, NY) for use in these studies. The mice were individually housed in

propylene cages (30 x 15 x 14 cm) at an ambient temperature of 22 ±2°C and provided

food (D12450B: 10% kcal% fat, 70% kcal% carbohydrate, 20% kcal protein; Research

Diets Inc., New Brunswick, NJ, USA) and water ad libutum. Upon arrival, all mice were

maintained under a 16:8 light/dark (lights on at 23:00 Eastern Standard Time [EST] ~130

lux) cycle for one week to allow them to entrain to local conditions and recover from the

effects of shipping. A 16:8 light dark cycle was used rather than 12:12 to avoid providing

a seasonally ambiguous signal. Because Swiss Websters are an outbred species and may

retain some responsiveness to photoperiod, mice might interpret a 12:12 light cycle as

either a long or short photoperiod, and thus impose higher variability in phenotype

(Nelson, 1990). All experimental procedures in this dissertation were approved by The

Ohio State University Institutional Animal Care and Use Committee, and animals were

maintained in accordance with the recommendations of the National Institutes of Health

and the Guide for the Care and Use of Laboratory Animals.

Experiment 1

Following the habituation period, mice were pseudo-randomly assigned to one of

three groups (n=10/group); mice were housed in either a standard light/dark cycle,

constant light, or a light/dim light cycle. Mice were weighed during group assignment to

establish that all groups had a similar baseline body mass. All mice weighing >35 grams

were excluded from the experiments. After group assignment, the LL mice were placed

in a constant light room (LL) where they were exposed to a constant amount of

Page 49: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

33

continuous light (~150 lux). The dLAN mice were placed in a room with a 16:8 light/dim

cycle; during the light period they were exposed to ~150 lux of light and during the dim

period they were exposed to ~5 lux of light at cage level.

Food intake was measured daily immediately before the onset of the dark period

(15:00 EST) throughout the study and body mass was measured weekly. After 6 weeks in

light conditions food intake was measure twice daily, at the onset of the dark period

(15:00 EST) and onset of the light period (23:00 EST) in order to quantify timing of food

consumption (expressed as percentage: 100 x consumption light/(consumption light +

consumption dark)). Home cage activity was monitored during the 5th

and 6th

weeks in

light conditions; each room was monitored for 4 consecutive days including a weekend.

At week 4 mice underwent an intra-peritoneal glucose tolerance test (GTT). After 8

weeks in light conditions mice were killed by cervical dislocation at one of two time

points, either directly after the onset of darkness (between 15:00 and 17:00 EST) or

during the middle of the light phase (5:00 to 7:00 EST), in order to collect blood samples

at the peak and nadir of locomotor activity. Epididymal fat pads were also collected as an

index of white adipose tissue.

Experiment 2

Following the habituation period, mice were pseudo-randomly assigned a number

and divided into one of six groups (n=8-9/group); mice were housed in either LD or

dLAN and had either 24 h/day food access (FA; food weighed twice daily at 9:00 and

19:00 EST), food access during the dark phase (FD; food in: 9:00 EST, food out: 19:00

EST), or food access during the light phase (FL; food in 19:00 EST, food out: 9:00 EST).

Page 50: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

34

After 7 weeks in light conditions mice underwent a series of three retro-orbital blood

collections with at least 48 h between collection points to assess serum corticosterone

concentrations. After 8 weeks in light conditions mice were killed by cervical dislocation

at one of two time points, either directly after the onset of darkness (10:00-12:00 EST) or

during the middle of the light phase (23:00-1:00 EST). Epididymal fat pads were

collected. All entrances into the animal rooms and collections after lights off were made

under dim red illumination.

Glucose tolerance test

The glucose tolerance test was given after four weeks in experimental light

conditions. Mice were administered a 1.5 g/kg body mass intra-peritoneal glucose bolus

at 10:00 EST after an 18 h fast. Blood samples of ~5 µL were collected via

submandibular bleed before injection, and at 15, 30, 60, 90, and 120 min following

injection. Blood glucose was immediately measured with the Contour blood glucose

monitoring system and corresponding test strips (Bayer HealthCare, Mishawaka, IN).

Activity analyses

Locomotor activity was tracked in 8 mice per group using OPTO M3 animal

activity monitors (Columbus Instruments, Columbus, OH) that continuously compile data

using MDI software system. Results from locomotor activity were used to determine

whether lack of physical activity or altered rhythmicity may have affected metabolism

(Laposky, Bass, Kohsaka, & Turek, 2008).

Blood collection and hormone analyses

Page 51: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

35

Blood samples were then allowed to clot, had the clot removed, and were

centrifuged at 4°C for 30 min at 3300 g. Serum aliquots were aspirated and stored in

sealable polypropylene microcentrifuge tubes at -80°C for subsequent analysis. Total

serum corticosterone concentrations, for mice, was determined in duplicate in an assay

using an ICN Diagnostics 125

I double antibody kit (Costa Mesa, CA USA). The high and

low limits of detectability of the assay were 1200 and 3 ng/ml, respectively. The intra-

assay coefficient of variation was 7.00% for experiment 1 and 16.83% for experiment 2.

Statistical analyses

Hormone concentrations, total daily food consumption and total daily activity

were analyzed using a one-way analysis of variance (ANOVA). Following a significant

F score, multiple comparisons were conducted with Tukey‟s HSD tests. Glucose

tolerance test results were analyzed with a repeated measures ANOVA with lighting

condition as the within-subject factor and time as the between subject variable. A

repeated measures ANOVA was also used to analyze change in body mass over time.

Following a significant result on repeated measures ANOVA, single time point

comparisons were made using Student‟s t-tests. The above statistical analyses were

conducted with StatView software (v. 5.0.1, Cary, NC). Fourier analysis was used to

determine whether locomotor activity was rhythmic and followed 24 h periodicity using

Clocklab software from Actimetrics (Wilmette, IL). Mice were considered rhythmic

when the highest peak occurred at ~1 cycle per day with an absolute power of at least

0.005 mV/Hz as previously described (Kriegsfeld et al., 2008). Nonlinear regression

analysis was used in GraphPad Prism software (v. 4 La Jolla, CA). In all cases,

Page 52: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

36

differences between group means and correlation coefficients were considered

statistically significant if p ≤ 0.05.

Results

Body mass

Body mass was differentially affected by light condition over the 8 experimental

weeks (F24,192 = 3.457; p < 0.0001; Fig. 1a). A significant increase in body mass among

LL and dLAM mice, relative to the LD control group, was evident beginning one week

after onset of light treatment and continuing throughout the 8 week study (F2,24 = 4.441,

10.187, 12.660, 12.232, 6.561, 4.568, 4.293 respectively, p ≤ 0.01). The elevated body

mass among LL and dLAN groups was due to increased body mass gain (F2,24 = 4.291; p

< 0.05; Fig 1b), rather than initial differences in body mass among groups. Furthermore,

epididymal fat pad mass was significantly greater at the end of the study in mice with

light at night suggesting increased body mass reflected increases in white adipose tissue

(F2,24 = 4.767; p < 0.05; Fig. 1b) .

Glucose tolerance test

After four weeks in experimental light conditions, LL and dLAN mice displayed

impaired glucose tolerance during an intra-peritoneal glucose tolerance test (GTT).

Injection of glucose increased blood glucose levels in all groups following an 18 hour fast

(F5,23 = 151.015; p < 0.0001; Fig. 1c). LL and dLAN mice failed to recover glucose

levels as rapidly as the LD group (F10,115 = 2.514; p < 0.01). dLAN mice significantly

elevated glucose levels after 60 min and glucose levels remained elevated after 90 and

120 min in dLAN and LL mice compared to the LD group (post hoc; p < 0.05).

Page 53: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

37

Furthermore, final glucose levels in the GTT positively correlated with body mass (R =

0.5236; p < 0.05; Fig. 1d).

Locomotor Activity

In contrast to the LD and dLAN groups, which displayed the typical circadian

rhythm in locomotor activity, as measured in the home cage via an infrared beam

crossing system, the LL mice were arrhythmic (7 out of 8 arrhythmic as measured by

Fourier analysis; Fig. 2a-c). However, total daily locomotor activity was similar for all

groups (F2,153= 0.0002; p > 0.95; Fig. 2d).

Glucocorticoids

Corticosterone was decreased in the LL (p < 0.05; Fig. 3a), but not dLAN mice;

these results suggest that changes in glucocorticoid concentrations were not necessary for

altered metabolism.

Energy Intake

Total 24 h food consumption did not differ between groups (F2,24 = 0.107; p >

0.85). Although no differences in total food consumption and home cage locomotor

activity were detected between groups, feeding behavior was altered in the dLAN group

(F1,38 = 29.315; p ≤ 0.05; Fig. 3b). The dLAN mice consumed 55.5% of their food during

the light phase as compared to 36.5% in LD mice, indicating that mice exposed to LAN

ate more food during the day, rather than at night. LL mice were not considered in this

comparison because they had no temporal signal to distinguish the light and dark phase.

Furthermore, correlation analyses confirmed that percentage of daytime food

Page 54: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

38

consumption was positively related to final body mass and final glucose levels in the

GTT (R = 0.5058 and R = 0.6066 respectively; p < 0.01; Fig. 3c/d).

Timed feeding

Because altered timing of food consumption may mediate changes in body mass

in the dLAN mice, I performed an additional experiment with mice housed in either LD

or dLAN with either continuous access to food (FA) or with food availability limited to

either the light (FL) or dark (FD) phase, respectively. Timed feeding affected body and

epididymal fat pad mass gain (F2,44 = 5.392 and 4.372 respectively; p < 0.05; Fig. 4a/b).

Within dLAN mice limiting food access to the dark phase prevented weight and fat gain

(weight: t14=1.940, fat: t14=2.526, p≤0.05). dLAN-FD mice gained an equivalent amount

of weight and fat as LD-FD mice and LD mice with 24h food access (weight: t14=-.250,

t14=-1.176 fat: t14=-.076, t14=-1.004; p > 0.05). Furthermore, FL mice increased body and

fat pad masses (post hoc, p < 0.05); this increase was not dependent on light/dark cycle

and LD- and dLAN-FL had comparable weight gain. There was no effect of light on

corticosterone concentrations at the 6 time points measured (p > 0.05). However, timed

feeding altered corticosterone concentrations at ZT 16 (F2,24 = 15.316; p < 0.05; Fig. 4c),

such that timed feeding during the dark phase increased corticosterone concentrations

(post hoc, p < 0.05). Food consumption changed over time such that consumption

decreased throughout the study (F6,258=664.474; p < 0.05; Table 1). Furthermore, there

was an interaction between timed feeding and food consumption over time (F12,258 =

98.051; p < 0.05), with timed feeding groups initially consuming more than mice with

food access all the time.

Page 55: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

39

Discussion

Swiss-Webster mice were housed in either standard light/dark cycles, 24 h of

continuous lighting, or in light/dim cycles. A significant increase in body mass among

LL and dLAN mice, relative to the LD control group, was evident beginning one week

after onset of light treatment and continuing throughout the 8-week study. After four

weeks, LL and dLAN mice displayed impaired glucose tolerance during an intra-

peritoneal glucose tolerance test; the mice with dLAN failed to recover glucose levels as

effectively as the LD group. Increased body mass and reduced glucose tolerance are

indicative of a pre-diabetic-like state (Kahn, Hull, & Utzschneider, 2006). Thus, as little

as 5 lux of light exposure during the typical dark period is sufficient to increase body

mass and compromise glucose regulation.

Total daily locomotor activity, as measured in the home cage via an infrared beam

crossing system, was similar for all groups. However, in contrast to the LD and dLAN

groups, that displayed the typical circadian rhythm in locomotor activity, the LL mice

were arrhythmic. Although no differences in total daily food consumption were detected

between groups, feeding behavior was altered in the dLAN group. The dLAN mice

consumed 55.5% of their food during the light phase as compared to 36.5% in LD mice,

indicating that mice exposed to dLAN ate more food during the day, rather than at night.

LL mice were not considered in this comparison because they had no temporal signal to

distinguish the light and dark phase. Again, total 24 h consumption did not differ among

groups, but food intake is substantially higher at night among nocturnal rodents (Zucker,

1971) and altered timing of food consumption has been associated with metabolic

Page 56: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

40

syndrome in other animal models (Arble, Bass, Laposky, Vitaterna, & Turek, 2009;

Turek et al., 2005). Correlation analyses confirmed that percentage of daytime food

consumption was positively related to final body mass and final glucose levels in the

GTT.

To establish whether the altered timing of food intake contributed to the increased

weight gain, I performed an additional study with a timed feeding schedule. Because

mice housed in LL would likely entrain to the time of food access, they were omitted

from the food access iteration (Mistlberger, 2009). Mice were housed in either LD or

dLAN with either 24 h ad libitum access to food, food access only during the light phase,

or food access only during the dark/dim phase. Restricting food availability to the dark

phase prevented weight and fat gain among dLAN mice. dLAN-FD mice gained an

equivalent amount of weight and fat as LD-FD mice and LD mice with no food

restriction. This further suggests that altered timing of food consumption in the dLAN

mice leads to increased body mass gain. As previously reported for rats and mice, FL

mice increased body mass; this increase was not dependent on light/dark cycle and LD-

and dLAN-FL had comparable weight gain. Mice with limited access to food displayed

an initial spike in consumption that decreased over time. When food access was limited

to the light phase, mice displayed an increased elevation in food consumption during

week one; however, mice with access to food limited to the dark phase consumed more

food in subsequent weeks. Again, overall food intake for the study was comparable

among all groups suggesting that the timing of food intake is a critical factor mediating

increased weight gain.

Page 57: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

41

Alterations in light cycles are typically considered to be stressful; the results of

previous research on the effects of LAN on glucocorticoid concentrations, however, are

equivocal (Abilio, Freitas, Dolnikoff, Castrucci, & Frussa-Filho, 1999; Fonken et al.,

2009). Because high glucocorticoid concentrations can alter metabolism resulting in

obesity I measured circulating glucocorticoids (Dallman et al., 2004). Contrary to my

predictions, corticosterone was decreased in the LL, but not dLAN mice, which suggests

that changes in glucocorticoid concentrations were unnecessary for altered metabolism.

The decreased corticosterone concentrations in the LL mice likely reflect masking of the

glucocorticoid rhythm. Glucocorticoid concentrations were affected by food restriction.

Mice with food access during the dark phase had elevated peak glucocorticoid

concentrations. There were no differences in glucocorticoid concentrations between

dLAN and LD mice that were on the FA or FL feeding schedules.

These results establish that night-time illumination at a level as low as 5 lux is

sufficient to uncouple timing of food consumption and locomotor activity resulting in

metabolic abnormalities. dLAN mice display desynchrony between internal metabolic

activity and food intake, as demonstrated by altered timing of food consumption; this

may be the primary factor leading to increased weight gain. Similarly, in LL mice the

arrhythmic home cage activity suggests that there may be desynchrony between food

intake and metabolic parameters leading to increased weight gain by a similar but distinct

mechanism. Mice exposed to LAN may have disrupted melatonin signaling leading to a

misalignment of food intake and activity resulting in altered fuel metabolism. Melatonin

concentrations have been relatively unexplored in Swiss-Webster mice, however, retinal

Page 58: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

42

melatonin levels were undetectable in one previous study in common with melatonin

values in common strains of laboratory mice such as C57Bl/6 (Tosini & Menaker, 1998).

Although previous research failed to detect melatonin in many strains of mice (Goto,

Oshima, Tomita, & Ebihara, 1989), more recent studies report attenuated, but rhythmic,

melatonin expression in strains such as C57Bl/6 which were previously thought void of

melatonin (Kennaway, Voultsios, Varcoe, & Moyer, 2002). Melatonin rhythmicity, rather

than absolute quantities of nightly melatonin secretion, plays a crucial role in metabolic

function (Korkmaz, Topal, Tan, & Reiter, 2009). For example blunted nighttime

melatonin rhythms due to LL increased visceral adiposity in rats (Wideman & Murphy,

2009) and daily administration of melatonin suppressed abdominal fat and plasma leptin

levels (Rasmussen, Boldt, Wilkinson, Yellon, & Matsumoto, 1999). Furthermore,

melatonin influences clock gene expression in peripheral tissues such as the heart

(Zeman, Szantoova, Stebelova, Mravec, & Herichova, 2009) and may similarly modulate

clock gene expression in peripheral tissue involved in metabolism.

Mice exposed to LAN may also have disrupted clock expression leading to altered

metabolism. The SCN are the primary pacemaker at the top of a hierarchy of temporal

regulatory systems wherein multiple peripheral tissues contain molecular machinery

necessary for self-sustaining circadian oscillation (Kohsaka & Bass, 2007). In addition to

becoming obese, mice fed a high fat diet have disrupted mClock expression in the liver

(Kohsaka et al., 2007) and mice with mutations in clock genes have altered energy

homeostasis (Turek et al., 2005). Moreover, mice with mutations in either Clock or Bmal1

show impaired glucose tolerance, reduced insulin secretion, and defects in the

Page 59: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

43

proliferation and size of pancreatic islets (Marcheva et al., 2010). Several models of

obesity have reported attenuated amplitude of circadian clock gene expression and

changes in the phase and daily rhythm of clock genes may cause obesity (Barnea, Madar,

& Froy, 2009).

Metabolism and the circadian clock are intrinsically related (K. Eckel-Mahan &

Sassone-Corsi, 2009) with desynchrony of feeding and activity causing metabolic

alterations (Arble, Bass, Laposky, Vitaterna, & Turek, 2009; Salgado-Delgado, Angeles-

Castellanos, Saderi, Buijs, & Escobar, 2010). In humans even brief circadian

misalignment results in adverse metabolic and cardiovascular consequences (Scheer,

Hilton, Mantzoros, & Shea, 2009). The seemingly innocuous environmental light

manipulation used in this study that changed feeding behavior resulting in obesity may

have important implications for humans. Patients with night-eating syndrome are obese

and appear to display circadian rhythm disruption (Benca et al., 2009; Stunkard, Grace, &

Wolff, 1955). More generally, prolonged computer use and television viewing have been

identified as risk factors for obesity, diabetes, and metabolic disorders (Fung et al., 2000).

For the most part, researchers considering this correlation have focused on the lack of

physical activity associated with television and computer use; however the results from

the current study suggest that exposure to nighttime lighting and the resulting changes in

the daily pattern of food intake and activity may also be contributing factors.

Page 60: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

44

Mean ± SEM daily food intake (g)

Week 1 Week 2 Week 3 Week 4 Week 5 Week 6 Week 7

LD-FA 4.7 ± 0.2 4.5 ± 0.1 4.2 ± 0.1 4.0 ± 0.1 3.9 ± 0.1 3.4 ± 0.1 3.7 ± 0.2

LD-FD 5.1 ± 0.2 4.6 ± 0.1 4.2 ± 0.2 4.3 ± 0.2 4.1 ± 0.1 3.7 ± 0.1 3.8 ± 0.1

LD-FL 5.7 ± 0.2 4.9 ± 0.1 4.1 ± 0.2 3.8 ± 0.1 3.8 ± 0.1 3.8 ± 0.1 3.8 ± 0.1

DM-FA 4.7 ± 0.1 4.6 ± 0.1 4.5 ± 0.2 4.1 ± 0.1 3.8 ± 0.1 3.6 ± 0.2 3.8 ± 0.2

DM-FD 5.4 ± 0.2 4.5 ± 0.2 4.2 ± 0.1 4.2 ± 0.1 4.0 ± 0.1 4.2 ± 0.1 4.0 ± 0.1

DM-FL 5.8 ± 0.3 4.8 ± 0.1 4.3 ± 0.1 3.9 ± 0.1 3.7 ± 0.2 3.7 ± 0.2 3.5 ± 0.2

...... Table 2.1. Food intake in food restricted mice exposed to dimly lit or dark nights.

44

Page 61: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

45

Figures

0 30 60 90 120

0

100

200

300

400

LDdLANLL

+

* *

Time (min)

Glu

co

se (

% b

aseli

ne)

0 1 2 3 4 5 6 7 825

30

35

40

45

LDdLAN

*

***

*

**

LL

&

Weeks

Bo

dy m

ass (

g)

LD dLAN LL0

4

8

12*

*

Bo

dy m

ass g

ain

(g

)

LD dLAN LL0

1

2

3

.06 *

Ep

idid

ym

al

fat

(g)

32 34 36 38 40 42 44 46 48

100

200

300

400

500

R=0.5236p=0.0036

Body mass (g)

Fin

al

glu

co

se (

mg

/dl)

A

C

B

D

Figure 2.1. Light at night affects body mass and glucose tolerance.

Body mass, fat pad mass, and glucose tolerance in mice exposed dark or lit nights. (A)

Mice with light at night elevated body mass beginning one week after placement in light

conditions and continuing throughout the remainder of the study. (B) Body mass gain and

epididymal fat pad mass differed among groups at the conclusion of the study suggesting

increases in body mass may be due to changes in body fat composition (C) Mice exposed

to either dim or bright light at night had reduced glucose tolerance; dLAN and LL mice

failed to recover blood glucose as rapidly as LD mice (D) Furthermore, body mass at the

time of the glucose tolerance test positively correlated with final blood glucose levels.

*p≤0.05 when LD differs from both groups, &p≤0.05 between all groups.

Page 62: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

46

Figure 2.2. Activity is comparable between mice exposed to dimly lit and dark nights.

Representative activity records from mice held in (A) a standard light/dark cycle (B) a

light/dim cycle and (C) constant lighting. Homecage locomotor activity was measured in

an infrared beam crossing system and is shown in a double plotted actogram. LL mice

became arrhythmic in constant light, however dLAN mice maintained rhythmicity. (D)

All mice had equivalent total 24 h activity (p>0.05).

Page 63: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

47

LD dLAN LL0

25

50

75

100

125Light

Dark/Dim

*

Seru

m c

ort

ico

ste

ron

e

(n

g/m

l)

20 30 40 50 60 70 80 9030

35

40

45

50

R=0.5058p=0.006

% Daytime consumption

Fin

al

bo

dy m

ass (

g)

LD dLAN LL0

10

20

30

40

50

60

70

80

*

*

% D

ayti

me f

oo

d

co

nsu

mp

tio

n20 30 40 50 60 70 80 90

100

200

300

400

500R=0.6066p=0.0005

% Daytime consumptionF

inal

glu

co

se (

mg

/dl)

A

C D

B

Figure 2.3. Time of food intake but not corticosterone is altered by dim light at night.

(A) Serum corticosterone concentrations were reduced in the LL but not dLAN mice at

the two time points measured suggesting that glucocorticoids did not mediate the changes

in body mass (*p≤0.05 from LL and dLAN). (B) Mice exposed to bright and dim light at

night ate more food during the light phase than in the dark phase which is atypical in

nocturnal animals (*p≤0.05). (C) Body mass and (D) blood glucose levels are associated

with percentage of daytime food consumption.

Page 64: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

48

FA FD FL FA FD FL30

35

40

45

LD dLAN

#

Bo

dy m

ass g

ain

(g

)

0 4 8 12 16 20 240

50

100

150

200

250

300 FA

FD

FL

*

LD

Time (hrs)

Seru

m c

ort

ico

ste

ron

e (

ng

/mL

)

FA FD FL FA FD FL0

1

2

3#

Ep

idid

ym

al

fat

(g)

0 4 8 12 16 20 240

50

100

150

200

250

300*dLAN

Time (hrs)

A B

C

Figure 2.4. Restricting feeding to the dark phase prevents dim light at night induced

body mass gain.

Body mass gain and epididymal fat pad mass differed between groups at the conclusion

of the study (A) Total body mass gain and (B) epididymal fat pad mass (*p≤0.05 when

group differs from LD, †p≤0.05 when FD differs from both). Timed feeding during the

dark in mice with light at night prevented increased weight gain and elevated epididymal

fat pad mass. (C) Serum corticosterone concentrations are altered by timed feeding.

Feeding mice during the dark resulted in increased corticosterone concentrations at

Zeitgeber Time (ZT) 16 irrespective of light condition (*p≤0.05 FD differs from FA and

FL).

Page 65: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

49

CHAPTER 3

DARK NIGHTS REVERSE METABOLIC CHANGES CAUSED BY DIM LIGHT

AT NIGHT

Metabolic disorders are increasing in prevalence worldwide and represent a major

global health threat. The metabolic syndrome is categorized by the development of

several metabolic abnormalities that increase the risk of coronary artery disease, stroke,

and diabetes. Hypercaloric food intake and physical lethargy are known to underlie the

development of metabolic syndrome and obesity. However, additional nontraditional

factors are likely involved. The increasing prevalence of metabolic disorders coincides

with increasing exposure to light at night (Fonken & Nelson, 2011; Reiter, Tan,

Korkmaz, & Ma, 2011; Wyse, Selman, Page, Coogan, & Hazlerigg, 2011). Recent

epidemiological and experimental studies implicate the introduction of artificial light in

the development of metabolic syndrome (Maury, Ramsey, & Bass, 2010). Indeed, shift-

workers who experience high levels of light at night are at increased risk for

cardiovascular disease (Ha & Park, 2005; Knutsson, 2003) and elevated body mass index

(Parkes, 2002). Even brief behavioral and circadian misalignment alters metabolic

homeostasis in humans, resulting in hyperglycemia, hyperinsulinemia, and postprandial

Page 66: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

50

glucose levels comparable to a pre-diabetic state (Scheer, Hilton, Mantzoros, & Shea,

2009). Moreover, in rodent models exposure to light at night produces changes in

metabolism (Fonken et al., 2010; Vinogradova, Anisimov, Bukalev, Semenchenko, &

Zabezhinski, 2009; Wideman & Murphy, 2009).

Metabolic processes fluctuate throughout the day. The suprachiasmatic nuclei

(SCN) of the hypothalamus comprise the master circadian clock in mammals and control

physiological and behavioral circadian rhythms. Photic input to the SCN is the dominant

cue for entraining the circadian clock. Light travels directly from intrinsically

photosensitive retinal ganglion cells (ipRGCs) to the SCN via the retino-hypothalamic

tract (RHT). Prior to the wide-spread adoption of electric lighting the circadian system

was principally synchronized to the solar cycle. In contrast, modern light exposure occurs

in a variety of patterns. Because of the importance of light in synchronizing the circadian

system, exposure to aberrant light schedules disrupt circadian activity. Disruption in the

clock gene network is linked to changes in sleep, body mass, locomotor activity, and food

intake. Homozygous Clock mutant mice have significant increases in energy intake and

body weight, and total arrhythmicity when housed in constant darkness (Turek et al.,

2005). These mutants also showed dyslipidemia, hyperglycemia, and hypoinsulinemia-

all markers of metabolic dysregulation (Turek et al., 2005). Manipulation of other genes

in the clock gene family similarly cause metabolic abnormalities (Marcheva et al., 2010).

Interactions between metabolism and the circadian system appear to be reciprocal as diet

induced obesity alters the period of the central clock and dampens diurnal rhythm in

locomotor activity (K. Eckel-Mahan & Sassone-Corsi, 2009; Kohsaka et al., 2007). Both

Page 67: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

51

short and long duration exposure to light at night or constant light also produce symptoms

of metabolic syndrome (Fonken et al., 2010; Vinogradova, 2007; Vinogradova,

Anisimov, Bukalev, Semenchenko, & Zabezhinski, 2009).

Although the SCN is the dominant brain region involved in driving circadian

activity, peripheral clock mechanisms are present throughout the body. Metabolic tissues

such as liver, adipose, pancreas, and muscle all display independent rhythmic clock gene

expression. The SCN principally regulates peripheral clock activity through neural and

endocrine signaling pathways (Guo, Brewer, Champhekar, Harris, & Bittman, 2005;

McNamara et al., 2001). Extra-SCN clock activity occurs in a tissue-specific manner

which enables organs to cope with local physiological demands and respond to local

factors. Multiple signals related to feeding and fasting entrain clock activity in metabolic

tissue. Environmental cues that occur at aberrant times may lead to asynchronous activity

within or between tissues which can lead to organ dysfunction (Vollmers et al., 2009).

For example, changes in peripheral clock function contribute to symptoms of metabolic

syndrome such as body weight gain and reduced glucose tolerance (Carvas et al., 2012;

Kennaway, Owens, Voultsios, Boden, & Varcoe, 2007; Lamia, Storch, & Weitz, 2008).

We previously reported that constant light (LL) and a bright/dim light cycles

(dLAN) alter metabolic parameters in mice (Fonken et al., 2010). Mice housed in LL and

dLAN increase body mass and white adipose tissue, impair glucose processing, and alter

food intake patterns compared to mice housed in LD. dLAN mice consume more food

during the light period than at night which is atypical in nocturnal rodents (Fonken et al.,

2010). Altered timing of food intake could be the mechanism by which light at night

Page 68: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

52

induces weight gain as it has previously been shown to induce metabolic disorder (Arble,

Bass, Laposky, Vitaterna, & Turek, 2009) and uncouple central and peripheral clock gene

expression (Damiola et al., 2000). The goal of the present experiment was to determine

whether or not changes in metabolism dissipate after removal of the aberrant light

schedule. Studies in shift-workers offer contrasting views about whether removing

circadian disruption can produce a return to baseline state. For example, current shift-

workers increase systemic markers of inflammation, but former shift workers do not

differ from day shift controls (Puttonen, Viitasalo, & Harma, 2011). However, former

shift workers have increased risk for obesity (Puttonen, Viitasalo, & Harma, 2011). Thus,

I tested whether metabolic disruption that occurs with light at night is an enduring effect

following placement back in a standard light dark cycle. Mice were housed under dim

light at night for 4 weeks and then transferred back to a standard light dark cycle.

Placement back into dark nights ameliorated the effects of exposure to dim light at night.

This suggests that changes in metabolism that occur with nighttime light exposure are not

necessarily permanent.

Methods

Animals

Sixty male Swiss-Webster mice (~8 weeks of age) from Charles River Kingston

were used in this study. Nocturnal rodents were used in this study to investigate the

effects of nighttime light exposure independent of sleep disruption. The mice were

individually housed in propylene cages (dimensions: 27.8 x 7.5 x 13 cm) at an ambient

temperature of 22 ±2°C and provided with Harlan Teklad 8640 food (Madison, WI) and

Page 69: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

53

filtered tap water ad libitum. All mice appeared healthy throughout the study and showed

no signs of sickness behavior.

Mice were assigned to one of four groups: (1) a control group that remained in

standard light-dark conditions [14h light (150 lux): 10h dark (0 lux); LD/LD] for the 8

weeks study, (2) a group housed in dim light at night for 8 weeks [14h light (150 lux):

10h dim (5 lux); dLAN/dLAN], (3) a group housed in LD for 4 weeks, then 4 weeks of

dLAN (LD/dLAN), and (4) a group housed in dLAN for 4 weeks then 4 weeks of LD

(dLAN/LD). All mice were housed in LD for one week to entrain to the local light-dark

cycle and recover from the effects of shipping prior to entering the study. On day 1 of the

experiment mice were weighed and transferred from an LD room to a cabinet with either

an LD or dLAN light cycle. Body mass was measured weekly and glucose tolerance was

evaluated after 7 weeks in experimental conditions. After 6 weeks in lighting conditions

timing of food intake was measured for 4 consecutive days. At the conclusions of the

study mice were anesthetized with isoflurane vapors and rapidly decapitated. Epididymal

fat pads of mice that underwent the glucose tolerance test were collected and weighed

and then flash frozen for qPCR analyses.

Page 70: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

54

Table 3.1. Experimental design for Chapter 3.

Quantitative PCR (qPCR)

The mRNA levels of MAC1, IL6, and TNFα were assayed in epididymal fat pads

as an index of peripheral inflammation (Reed et al., 2010). A small portion of the distal

epididymal fat pad was collected and flash frozen at the conclusion of the study. Total

RNA was extracted using a homogenizer (Ultra-Turrax T8, IKAWorks, Wilmington, NC)

and an RNeasy Mini Kit (Qiagen, Austin, TX). RNA was reverse transcribed into cDNA

with M-MLV Reverse Transcriptase enzyme (Invitrogen, Carlsbad, CA) according to the

manufacturer‟s protocol. Gene expression for MAC1 was determined using inventoried

primer and probe assays (Applied Biosystems, Foster City, CA) on an ABI 7500 Fast

Real Time PCR System using Taqman® Universal PCR Master Mix. The universal two-

step RT-PCR cycling conditions used were: 50 °C for 2 min, 95 °C for 10 min, followed

by 40 cycles of 95 °C for 15 s and 60 °C for 1 min. Relative gene expression of

Page 71: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

55

individual samples run in duplicate was calculated by comparison to a relative standard

curve and standardized by comparison to 18S rRNA signal.

Intra-peritoneal glucose tolerance test (GTT)

After 7 weeks in experimental light conditions a subset of each group of mice was

administered an intra-peritoneal glucose bolus (1.5 g/kg body mass) after an 18 h fast.

Blood samples of 5 µL were collected via submandibular bleed before injection and at

15, 30, 60, 120, and 180 min following injection. Blood glucose was measured

immediately with a Contour blood glucose monitoring system and corresponding test

strips (Bayer HealthCare, Mishawaka, IN).

Statistical analyses

Body mass and glucose tolerance test were compared between groups using

repeated measures analysis of variance (ANOVA). Body mass gain, fat pad mass, gene

expression, and food intake comparisons were analyzed using one-way ANOVA.

Following a significant F score, multiple comparisons were conducted with Tukey‟s HSD

tests. The above statistical analyses were conducted with StatView software (v. 5.0.1,

Cary, NC). In all cases, differences between group means were considered statistically

significant if p ≤ 0.05.

Results

Body mass

Body mass was significantly affected by light conditions over the 8 experimental

weeks (F21,336 = 3.537; p < 0.0001; Fig. 1A). After 4 weeks in their respective lighting

conditions, and prior to the light schedule transfer, significant differences in weight gain

Page 72: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

56

were observed (F3,50 = 3.710; p < 0.05). Both groups that were initially housed under

dLAN showed elevated body mass gain compared to those housed in standard lighting

conditions (post hoc analyses; p < 0.05; Fig 1B). Directly following collection of week 4

body weights mice in the dLAN/LD and LD/dLAN groups were transferred to the new

light schedules. After 3 weeks of the new lighting schedules body mass gain was also

significantly affected by lighting conditions (F3,50 = 3.906 from beginning of study; Fig

1C; F3,50 = 8.862 from transfer; Fig. 1D; p < 0.05 respectively). LD/dLAN and

dLAN/dLAN mice gained significantly more weight throughout the study as compared to

LD/LD mice (post hoc; p < 0.05; Fig 1C). dLAN/LD mice did not differ from any group

with respect to weight gain when evaluating the entire 7 week period. When evaluating

body mass gain following the transfer however, dLAN/LD mice significantly reduced

weigh gain compared to dLAN/dLAN and LD/dLAN mice (post hoc; p < 0.05; Fig 1D).

Relative epididymal fat pad mass (corrected for total body mass), a representative

measure of white adipose tissue, significantly differed by the conclusion of the study

(F3,31 = 4.089; p < 0.05; Fig. 2A). Mice that were housed with dLAN for the entirety of

the study showed significantly elevated relative epididymal fat pad mass as compared to

LD/LD and dLAN/LD mice. LD/dLAN mice also had significantly elevated fat pad mass

as compared to LD/LD mice (post hoc; p < 0.05). These results are particularly important

because they suggest that increases in body mass among dLAN mice reflect increases in

white adipose tissue (Rogers & Webb, 1980). Furthermore, these results demonstrate that

a switch back to a standard light dark cycle after 4 weeks of housing in dLAN allows

Page 73: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

57

restoration of fat levels to those of LD/LD mice. Lighting conditions did not affect paired

testes, epididymides, spleen, or adrenal mass (p > 0.05 in each case).

Gene expression

MAC1 mRNA expression in epididymal fat pads was significantly affected by

lighting conditions (F3,31 = 2.948; p < 0.05; Fig. 2B). dLAN/dLAN mice elevated MAC1

expression as compared to both LD/LD and dLAN/LD mice (post hoc; p < 0.05). In

contrast, expression of IL6 and TNFα in the fat pads did not significantly differ between

groups (data not shown, p > 0.05).

Glucose tolerance test

Three weeks after the transfer to the new lighting schedules (7 experimental

weeks) mice underwent a GTT. Injection of glucose led to a rapid increase in blood

glucose levels in all groups (F5,155 = 267.514; p < 0.0001; Fig 3A). Furthermore, there

was a significant interaction between lighting conditions and blood glucose levels over

time (F15,155 = 2.427; p < 0.005), such that dLAN/dLAN mice failed to recover glucose

levels as effectively as all other groups. Single time point comparisons revealed that

glucose levels were significantly elevated in dLAN/dLAN mice as compared to LD/LD

mice at T60 and as compared to all other groups at T120 and T180 (F3,31 = 3.285, 4.879,

4.622, respectively; p < 0.05). These results suggest that dysregulation in glucose

processing is secondary to increased weight gain as the LD/dLAN groups does not yet

show impairments in glucose tolerance. Furthermore, the improvement in glucose

processing in the dLAN/LD group suggests that impairments in glucose regulation are

reversible among dLAN mice by a transfer back to standard lighting conditions.

Page 74: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

58

Energy intake

Total 24 h food intake did not differ among groups (p > 0.05). There was a main

effect of light cycle on timing of food intake (F3,31 = 3.912; p < 0.05; Fig. 3B);

dLAN/dLAN and LD/dLAN mice consumed significantly more of their food during the

light phase as compared LD/LD mice (post hoc; p < 0.05). LD/dLAN also consumed

significantly more food during the light phase than dLAN/LD mice (post hoc; p < 0.05).

Discussion

The goal of the current study was to determine whether mice returned to dark

nights after dLAN exposure recover metabolic function. This study replicates previous

results demonstrating that mice housed with dim light at night develop symptoms of

metabolic syndrome (Fonken et al., 2010). As expected, both groups of mice housed in

dLAN for the initial segment of the experiment (dLAN/dLAN and dLAN/LD)

significantly increased body mass gain compared to LD mice. Half of the dLAN mice

(dLAN/LD) were then transferred to LD and vice versa (LD/dLAN). Following the

transfer dLAN/dLAN and LD/dLAN mice gained significantly more weight than LD/LD

and dLAN/LD mice. At the conclusion of the study dLAN/LD mice did not differ from

either LD/LD or dLAN/dLAN mice with respect to body mass gain. The intermediary

results of the dLAN/LD mice may reflect an inability to completely recover body mass

after the 4 week exposure to LAN. This would indicate permanent changes in metabolism

occur after nighttime light exposure. Former shift workers show symptoms of metabolic

dysfunction after return to a day shift schedule which indicates that this may be the case

in humans (Puttonen, Viitasalo, & Harma, 2011). However, in the study on shift workers

Page 75: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

59

there was no specified duration of time workers were on the non-shifting schedule. An

alternative explanation to the findings is that 3 weeks in LD may be insufficient to

completely recover body mass to LD levels. For example, recovery of other metabolic

markers can occur prior to reduction in body mass (Poudyal, Panchal, Ward, Waanders,

& Brown, 2012). Food intake patterns, epididymal fat pad mass, as well as the GTT

results support the latter hypothesis.

In agreement with previous findings, dLAN/dLAN mice shifted the timing of

food intake compared to LD/LD controls without changing the amount consumed.

dLAN/dLAN mice ate a significantly higher percentage of food during the light phase

which is atypical for nocturnal rodents and may contribute to weight gain. Consuming

higher amounts of food during the light phase is associated with increased weight gain in

rodents (Arble, Bass, Laposky, Vitaterna, & Turek, 2009). Moreover, restricting food

intake to the dark phase can prevent weight gain in mice fed a high fat diet (Hatori et al.,

2012; Sherman et al., 2012). Three weeks after dLAN/LD mice were moved back to LD,

food intake no longer differed from LD/LD controls. In contrast, LD/dLAN mice shifted

food intake to the light phase, with similar levels of daytime consumption as

dLAN/dLAN mice. These results suggest that 3 weeks of dLAN is sufficient to induce

altered timing of food intake.

Reduced glucose tolerance is a key symptom of metabolic syndrome (Kahn, Hull,

& Utzschneider, 2006). Here I show that impaired glucose clearance abilities associated

with dLAN are recovered by 3 weeks of exposure to LD. After 7 weeks in lighting

conditions mice underwent a glucose tolerance test. dLAN/dLAN mice showed

Page 76: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

60

significantly reduced glucose tolerance compared to all other groups. Importantly, the

dLAN/LD group was comparable to LD/LD controls in the GTT. This suggests that

dLAN/LD mice recover glucose processing abilities. Although dLAN/LD mice did not

show a complete reduction in body mass, the GTT results suggest that metabolic function

is restored.

Increases in body mass in dLAN are associated with increased white adipose

tissue (Fonken et al., 2010). dLAN/dLAN mice displayed significantly elevated

epididymal fat pad masses compared to LD/LD mice indicating increases in body mass in

dLAN are due to increased fat depots. At the conclusion of the study, dLAN/LD mice

had significantly reduced white adipose tissue compared to dLAN/dLAN mice.

Furthermore, LD/dLAN mice had intermediary fat pad mass compared to the LD/LD and

dLAN/dLAN groups. Elevated fat mass is associated with widespread chronic low-grade

inflammation in peripheral metabolic tissue which led us to evaluate levels of

macrophage expression in the epididymal fat pads (Marceau et al., 1999; Plomgaard et

al., 2005). dLAN/dLAN mice significantly elevated expression of MAC1, a marker for

macrophages, in the epididymal fat pads. This indicates that there is increased

macrophage infiltration into peripheral fat tissue. Peripheral inflammation can lead to

disrupted insulin and leptin signaling further propagating fat accumulation (Hotamisligil,

2006). LD/dLAN mice had intermediary level of MAC1 expression compared to LD/LD

and dLAN/dLAN mice whereas dLAN/LD mice had comparable MAC1 expression to

LD/LD controls. This suggests that increases in peripheral inflammation are associated

metabolic dysfunction following exposure to dLAN.

Page 77: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

61

This study also provides important insight into the time-course of development of

metabolic syndrome in dLAN. For example, altered timing of feeding likely precede

changes in glucose clearance as LD/dLAN mice did not differ in the GTT compared to

LD/LD controls although they already showed a pattern of food intake similar to

dLAN/dLAN mice. As discussed above, timing of food intake is a critical factor in the

development of metabolic disease (Arble, Bass, Laposky, Vitaterna, & Turek, 2009;

Hatori et al., 2012) and light at night likely elevates body mass gain through altering time

of food intake (Fonken et al., 2010). Additionally, changes in weight gain appear to occur

prior to the development of glucose intolerance because LD/dLAN mice show elevated

weight gain but do not have impairments in the GTT. This would be consistent with other

models of obesity in which adipose tissue releases factors (such as non-esterified fatty

acids, glycerol, pro-inflammatory cytokines, etc.) that can contribute to the development

of insulin resistance and β-cell dysfunction (Kahn, Hull, & Utzschneider, 2006).

Overall, these results demonstrate that re-exposure to dark nights ameliorates

metabolic disruption caused by dim light at night. dLAN appears as an innocuous

environmental manipulation, which may be why it was overlooked as a significant risk

factor for health and disease for many years. However, because of the profound affect

light has on the circadian system and upon downstream outputs such as hormone

secretion, dLAN likely exerts a significant effect on many physiological processes. The

circadian clock and metabolic pathways are intrinsically linked (Dallmann, Viola,

Tarokh, Cajochen, & Brown, 2012; K. Eckel-Mahan & Sassone-Corsi, 2009) with

desynchrony of feeding and activity causing metabolic alterations (Arble, Bass, Laposky,

Page 78: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

62

Vitaterna, & Turek, 2009; Salgado-Delgado, Angeles-Castellanos, Saderi, Buijs, &

Escobar, 2010). In humans even brief circadian misalignment results in adverse

metabolic and cardiovascular consequences (Scheer, Hilton, Mantzoros, & Shea, 2009).

The findings presented here suggest that exposure to nighttime lighting and the resulting

changes in the daily pattern of food intake may be contributing factors in the current

obesity epidemic. If these results apply to humans, then humans who experience weight

gain in response to exposure to dim light at night may be able to help manage body

weight by adjusting when they eat or by using low cost light blocking interventions such

as sleep masks.

Page 79: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

63

Figures

0 1 2 3 4 5 6 730

33

36

39

42

45

LD/LDdLAN/dLANdLAN/LDLD/dLAN

Weeks

Bo

dy m

ass (

g)

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0

2

4

6

8

10

A A

B

B

Bo

dy m

ass g

ain

befo

re t

ran

sft

er

(g-g

)

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0

2

4

6

8

10

12

A

B

ABB

Fin

al

bo

dy m

ass

gain

(g

-g)

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0

1

2

3

4

5

B

B

A AB

od

y m

ass g

ain

fro

m t

ran

sfe

r (g

-g)

A B

C D

Figure 3.1. Return to dark nights affects body mass after exposure to dim light at night.

Body mass was differentially affected by the lighting conditions throughout the study.

(A) Weekly body mass for mice over the course of the study. (B) Body mass gain after 4

weeks of lighting conditions and prior to the transfer. (C) Body mass gain at the

conclusion of the study. (D) Body mass gain from the point of the transfer at

experimental week 4 (different letters denote differences between groups, p≤0.05).

Page 80: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

64

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0

1

2

3

4

5

6

A

BBC

ACR

ela

tive e

pid

idym

al

fat

mass (

% b

od

y m

ass)

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0.0

0.3

0.6

0.9

1.2

1.5

1.8

A

A

B

AB

Rela

tive M

AC

1 m

RN

A

exp

ressio

n i

n f

at

A B

Figure 3.2. Dim light at night affects fat mass and composition.

dLAN increased relative fat pad mass and macrophage gene expression in fat pads. (A)

Relative epididymal fat pad mass at the conclusion of the study. (B) Relative MAC1

mRNA expression from epididymal fat pads (different letters denote differences between

groups, p≤0.05).

Page 81: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

65

0 30 60 90 120 150 1800

100

200

300

400

500

600LD/LDdLAN/dLANLD/dLANdLAN/LD

*

*

Time

Glu

co

se

(m

g/d

L)

LD/L

D

dLAN/d

LAN

LD/d

LAN

dLAN/L

D0

10

20

30

40

50

60

70

A

BCC

AB

% D

ay

tim

e c

on

su

mp

tio

n

A B

Figure 3.3. Dark nights reverse changes in feeding behavior and glucose processing

(A) Glucose tolerance was evaluated after 7 weeks in lighting conditions (*indicates

LD/LD and dLAN/dLAN differ, †indicates dLAN/dLAN differs from LD/LD,

LD/dLAN, and dLAN/LD). (B) Percentage of food consumed during the light phase

(different letters denote differences between groups, p≤0.05).

Page 82: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

66

CHAPTER 4

DIM LIGHT AT NIGHT EXAGGERATES WEIGHT GAIN AND

INFLAMMATION ASSOCIATED WITH A HIGH FAT DIET

Obesity is a significant public health problem that reduces quality of life,

increases mortality risk, and is a financial burden on society (Y. Wang, Beydoun, Liang,

Caballero, & Kumanyika, 2008). Once termed a disease of western societies it is now

clear that prevalence rates are increasing on a global scale (WHO, 2000). Health

problems associated with obesity are replacing concerns such as under nutrition and

infectious disease as the most significant contributors to global ill health (Antipatis &

Gill, 2001). Although well-documented factors such as dietary choices and lethargy are

known to contribute to the prevalence of obesity and metabolic disorders, additional

environmental factors are now considered critical in the development and maintenance of

obesity (Hill, Wyatt, Reed, & Peters, 2003).

The worldwide increase in obesity and metabolic disorders correlates with

increased exposure to artificial light at night (LAN) during the 20th

century.

Approximately 99% of the US and Europe currently experiences light pollution that alters

the natural cycle of light and dark (Navara & Nelson, 2007). Artificial lighting allows

Page 83: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

67

people to extend daytime activities into the night and engage in countercyclical night

time shift work. This type of aberrant light exposure can disrupt the circadian system

because light is the most potent entraining signal for the mammalian biological clock

(Reppert & Weaver, 2002). Many homeostatic processes are regulated by the circadian

system including metabolism (Lowrey & Takahashi, 2004). For example, there are 24-

hour variations in the expression of genes involved in gluconeogenesis, lipogenesis, and

lipid catabolism, among others (Oishi et al., 2003; Yang et al., 2006). Disruption of both

primary and secondary clock genes cause profound changes in metabolism (Marcheva et

al., 2010; Paschos et al., 2012; Solt et al., 2012; Turek et al., 2005). Similarly, shifting the

timing of food intake can alter weight gain independently of changes in total caloric

intake (Hatori et al., 2012; Sherman et al., 2012). Even brief circadian misalignment can

results in adverse metabolic and cardiovascular consequences in humans (Scheer, Hilton,

Mantzoros, & Shea, 2009).

Communication between the circadian clock and metabolic system appears bi-

directional as diet induced obesity is associated with behavioral and molecular circadian

rhythm disturbances (Hsieh et al., 2010; Kohsaka et al., 2007). I have previously

demonstrated that mice exposed to dim light at night (dLAN) significantly increase body

mass and reduced glucose processing compared with mice in a standard light-dark cycle

(LD), despite equivalent caloric intake and total daily activity. Nocturnal rodents

typically eat substantially more food at night; however, dLAN mice consume more than

half of their food during the light phase. Restricting food intake to the active phase in

dLAN mice prevents body mass gain. These results suggest that low levels of light at

Page 84: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

68

night disrupt the timing of food intake and other metabolic signals, leading to excess

weight gain (Fonken, Kitsmiller, Smale, & Nelson, 2012; Fonken et al., 2010).

In addition to altering metabolism, disruption of circadian clock through activities such

as shift work has serious health consequences including increased risk for cancer

(Stevens, 2009a), tissue damage (Tunez et al., 2003), heart disease (Morris, Yang, &

Scheer, 2012), and stroke (Vyas et al., 2012). A feature common to all of these

pathologies is elevated inflammation. Inflammation is integrally associated with obesity,

likely contributing to both the development and maintenance of metabolic syndrome

(Hotamisligil, 2006). Elevated expression of tumor necrosis factor-α (TNF-α) in adipose

tissue of obese mice was the first indication of inflammatory dysregulation in obesity

(Hotamisligil, Shargill, & Spiegelman, 1993). Elevated fat mass is associated with

widespread chronic low-grade inflammation in adipose tissue, liver, and skeletal muscle

(Marceau et al., 1999; Plomgaard et al., 2005). This inflammatory response is

characterized by increased levels of circulating pro-inflammatory cytokines, as well as

infiltration of the tissue by immune cells such as macrophages, neutrophils, and

eosinophils (Weisberg et al., 2003; H. Xu et al., 2003). Peripheral inflammation leads to

disruption in insulin signaling further propagating fat accumulation. Peripheral

inflammation described above is both a cause and consequence of obesity.

In contrast to the peripheral response, accumulating evidence suggests that

hypothalamic inflammation resulting from a high fat diet (HFD) may occur prior to

development of obesity through central leptin and insulin resistance. Acute glucose

overload significantly increases NF-κB activity in the hypothalamus, but not peripheral

Page 85: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

69

tissue (Zhang et al., 2008). Furthermore, hypothalamic insulin resistance occurs prior to

insulin resistance in peripheral tissue (Purkayastha et al., 2010). Whereas peripheral

inflammation can take weeks to develop after beginning a HFD (Kim et al., 2008),

changes in hypothalamic inflammation can occur within hours of consuming high fat

food (Thaler et al., 2012).

In developed and developing countries exposure to LAN and high fat diets often

occur in tandem and may contribute to the increasing obesity epidemic. Thus, I

hypothesized that dLAN would exagerate metabolic dysfunction produced by a HFD in

mice. Because dLAN is associated with changes in immune function (Bedrosian, Fonken,

Walton, & Nelson, 2011b; Fonken, Haim, & Nelson, 2011), I also investigated whether

dLAN works through a similar mechanism as HFD producing additional increases in

peripheral and hypothalamic inflammation. Overall, I hypothesized that these two

variables would synergistically affect metabolism through a similar mechanism.

Methods

Animals

Fifty four male Swiss–Webster mice (~8 wk of age) were obtained from Charles

River Laboratories for use in this study. The mice were housed individually in propylene

cages (30 x 15 x 14 cm) at an ambient temperature of 22 ± 2 °C and provided with

filtered tap water ad libitum. Mice were housed in a standard light/dark cycle [14h light

(150 lux): 10h dark (0 lux); LD] and provided basic rodent diet (chow; Harlan Teklad

8640, Madison, WI, USA) for one week after arrival at our facility. After the one week

acclimation period, mice were randomly assigned a number and placed in one of four

Page 86: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

70

groups (n=13-14 per group); mice were housed in either LD or dim light at night [14h

light (150 lux): 10h dim (5 lux); dLAN] and received either a high fat diet (HFD;

Research Diets D12451, New Brunswick, NJ, USA) or chow. On day 1 of the experiment

mice were weighed and transferred from an LD room to a cabinet with either an LD or

dLAN light cycle. At this time mice were also either maintained on chow or switched to a

HFD for the duration of the study. Body mass was measured weekly at ~ZT8 and timing

of food intake was measured twice daily at the onset of the light (ZT 0) and dark (ZT 14)

phase, respectively, for 4 consecutive days during the final experimental week. The

percentage of food consumed during the light phase was calculated for each day and

averaged for each mouse to generate a single percentage value. At the conclusion of the

study 6-7 mice per group were used for quantitative PCR (qPCR) and the remaining mice

per group were used for immunohistochemistry.

qPCR

Between ZT 8 and 10 mice were brought into a procedure room, anesthetized with

isoflurane vapors, a blood sample was collected from the retro-orbital sinus, and mice

were rapidly decapitated. Brains were removed, placed in RNAlater overnight and then

the hypothalamus was dissected. Epididymal fat pads and liver were removed, weighed,

and flash frozen. Total RNA was extracted from hypothalamic, fat, and liver tissue using

a homogenizer (Ultra-Turrax T8, IKAWorks, Wilmington, NC) and an RNeasy Mini Kit

(Qiagen, Austin, TX) according to manufacturer instructions. For fat extractions an

additional chloroform separation step was added prior to using the kit to prevent excess

fat from clogging the spin columns. RNA was then reverse transcribed into cDNA with

Page 87: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

71

M-MLV Reverse Transcriptase enzyme (Invitrogen, Carlsbad, CA). Gene expression for

MAC1, TNFα, and POMC were determined using inventoried primer and probe assays

(Applied Biosystems, Foster City, CA) on an ABI 7500 Fast Real Time PCR System

using Taqman® Universal PCR Master Mix. The universal two-step RT-PCR cycling

conditions used were: 50 °C for 2 min, 95 °C for 10 min, followed by 40 cycles of 95 °C

for 15 s and 60 °C for 1 min. Relative gene expression of individual samples run in

duplicate was calculated by comparison to a relative standard curve and standardized by

comparison to 18S rRNA signal.

Immunohistochemistry

A subset of mice was used to assess histological evidence of microglia infiltration

to the hypothalamus. Between ZT 8 and 10 mice were deeply anesthetized with

isoflurane vapors, a blood sample was collected from the retro-orbital sinus, and mice

were given a sodium pentobarbital overdose. Mice were then perfused transcardially with

ice-cold 0.1M PBS followed by 50 mL of 4% paraformaldehyde. Brains were removed,

post-fixed overnight, cyroprotected in 30% sucrose, and frozen in isopentane with dry

ice. Brains were stored at -80°C and then sectioned on a cryostat at 40 μm into

cryoprotectant. Sections were stored at -20°C until further processing. The sections were

rinsed in phosphate buffered saline (PBS) and blocked with 4% BSA in PBS + Triton-X

(TX) for 1 h with constant agitation. Sections were incubated overnight with primary

rabbit-anti-Iba-1 (Wako Chemicals, Richmond, VA) diluted 1:1000 in PBS + TX. After

PBS rinses the sections were subsequently incubated for 1 h at room temperature with

biotinylated goat-anti-rabbit 1:1000 in PBS + TX (Vector Laboratories, Burligame, CA,

Page 88: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

72

USA). Sections were then quenched for 20 min in methanol containing 0.3% hydrogen

peroxide. After washing with PBS, sections were incubated for 1 hour with avidin-biotin

complex (ABC Elite kit, Vector laboratories). After rinses, the sections were developed

in diaminobenzidine for 2 min (Sigma, D4168). Sections were mounted on gel-coated

slides, dehydrated, and coverslipped with Permount. Images were captured on a Nikon

E800 microscope at 20X and analyzed using Image J software (NIH) to determine

immunoreactive regions. Both sides of bilateral structures were counted in duplicate per

animal.

Statistical Analyses

Comparisons between groups were conducted using a two-way analysis of

variance (ANOVA) with lighting condition and diet as the between subject factors.

Change in body mass over time was analyzed using a repeated measures ANOVA.

Following a significant F score, multiple comparisons were conducted with Tukey‟s HSD

test. The above statistical analyses were conducted with StatView software (v.5.0.1,

Cary, NC). In all cases, differences between group means were considered statistically

significant if p ≤ 0.05.

Results

Somatic measures

To determine whether dLAN exacerbates diet induced weight gain, mice were

exposed to either dLAN or LD while fed HFD or chow. There was an overall increase in

body mass over the 4 experimental weeks (F4,196 = 367.202; p < 0.0001). Both lighting

and dietary conditions affected body mass over time (Light: F4,196 = 15.525, Diet: F4,196 =

Page 89: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

73

150.178; p < 0.0001; Fig. 1A). Average body mass was comparable between all groups at

the start of the study (p > 0.05), but within 1 week of experimental onset both dLAN and

HFD elevated body mass (Light: F1,49 = 17.250, Diet: F1,49 = 50.218; p < 0.0001).

Furthermore, at the conclusion of the study relative body mass gain was increased among

both dLAN and HFD groups ( Light: F1,49 = 15.779; Diet: F1,49 = 136.447; p < 0.001; Fig.

1B). Increases in body mass likely reflected increases in fat mass as both dLAN and HFD

increased relative epididymal fat pad mass, a reliable index of overall adiposity (Light:

F1,21 = 9.785, Diet: F1,21 = 103.282; p < 0.01; Fig. 1C). Moreover, both dLAN and HFD

increased blood glucose levels (Light F1,49 = 4.148, Diet: F1,49 = 5.214; p < 0.05; Fig.

1A). There were no interactions between lighting condition and diet with respect to body

mass, fat pad mass, or blood glucose levels.

Food Intake

Despite increases in body mass among mice housed with dLAN, there were no

differences in total daily food intake between lighting conditions (p > 0.05; Fig. 2A).

Mice fed a high fat diet decreased food intake compared to the mice fed standard chow

(F1,49 = 32.860; P<.0001; Fig. 2A). Decreased food intake among HFD mice was

expected because the high fat food is much more calorie dense than standard chow.

Although there were no differences in total food intake, dLAN and HFD both altered

timing of food intake. dLAN mice increased daytime food intake as compared to mice

exposed to dark nights (Light: F1,49 = 42.649; p < 0.0001, Fig. 2B). High daytime food

intake is atypical for nocturnal rodents and changes in timing of food intake have

previously been associated with changes in metabolism (Arble, Bass, Laposky, Vitaterna,

Page 90: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

74

& Turek, 2009; Sherman et al., 2012; Tsai et al., 2012). HFD also caused a shift toward

daytime food intake which has previously been described in (Kohsaka et al., 2007) (Diet:

F1,49 = 5.509; p < 0.05).

Peripheral Inflammation

In metabolically related peripheral tissues (i.e., white adipose tissue (WAT), liver,

and pancreas) obesity promotes a state of chronic low-level inflammation. This in turn

contributes to insulin resistance, further propagating metabolic syndrome (Hotamisligil,

2006; Myers, Leibel, Seeley, & Schwartz, 2010). For this reason, I evaluated the

expression of pro-inflammatory cytokines in both white adipose tissue (WAT) and liver.

dLAN and HFD both elevated gene expression of MAC1, a marker for macrophages, in

WAT (Light: F1,20 = 9.304, Diet: F1,20 = 25.442; p < 0.01; Fig. 3A). Additionally, dLAN

and a HFD elevated expression of the pro-inflammatory cytokine TNFα in WAT (Light:

F1,20 = 4.649, Diet: F1,20 = 4.979; p < 0.05; Fig. 3B). There were no differences in TNFα

or MAC1 gene expression in the liver (data not shown). This is consistent with previous

research as it is not uncommon for changes in peripheral inflammation to take > 4 weeks

to develop in response to HFD (Kim et al., 2008).

Hypothalamic Inflammation

Previous research indicates that hypothalamic inflammation may occur with a

HFD prior to the onset of inflammation in peripheral tissue and contribute to obesity

development (Thaler et al., 2012). Because light at night is associated with changes in

immune function (Bedrosian, Fonken, Walton, & Nelson, 2011b; Fonken, Haim, &

Nelson, 2011), I hypothesized that hypothalamic inflammation may also be contributing

Page 91: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

75

to changes in weight gain in dLAN mice. Consistent with previous reports (De Souza et

al., 2005; Thaler et al., 2012; Zhang et al., 2008), HFD elevated hypothalamic TNFα

expression relative to standard chow (F1,21 = 4.433; p < 0.05; Fig. 4A). However, no

differences in MAC1 expression were apparent between light or dietary conditions (p >

0.05). This may reflect the distribution of hypothalamic microglia; with

immunohistochemistry I established that the concentration of Iba1 positive cells was

elevated in the arcuate nucleus of the hypothalamus in mice fed a HFD (F1,22 = 9.612; p <

0.01; Fig 3C,E,F), but not other hypothalamic nuclei such as the DMH (p > 0.05; Fig.

3D). There was no main effect of dLAN or interaction between lighting condition and

diet with respect to Iba1 immunoreactivity. However, there was a simple effect of

lighting condition within mice fed chow diet, such that dLAN increased the number of

Iba1 positive cells within the chow group (F1,12 = 4.855; p < 0.05). Neither diet nor

lighting condition affected POMC gene expression (data not shown, p > 0.05).

Discussion

I hypothesized that housing mice in dLAN would exaggerate metabolic changes

induced by a HFD. Moreover, I predicted that dLAN and HFD would both result in

elevated peripheral inflammation and upregulated inflammatory responses in the

hypothalamus, indicating the manipulations worked through a similar mechanism.

Although dLAN and HFD increased weight gain and peripheral inflammation, results in

the central nervous system were less clear. dLAN did not alter hypothalamic TNFα,

MAC1, or POMC gene expression. As anticipated, microglia staining was increased in

the arcuate nucleus of mice fed a high fat diet. However, dLAN only resulted in increased

Page 92: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

76

microglia staining among mice fed the chow diet. Lack of hypothalamic inflammation

among both dLAN groups suggests dLAN induces weigh gain through an alternative

mechanism.

Both diet and lighting condition elevated body mass over the course of the study.

Body mass was elevated by dLAN and HFD within the first week of experimental

conditions. Among HFD mice, dLAN potentiated increases in body and fat pad mass

compared to LD demonstrating that changes in environmental lighting can exacerbate the

adverse affects of a HFD. HFD and dLAN also both altered timing of food intake. As

previously reported, dLAN mice increase daytime food intake (Fonken et al., 2010).

Whereas LD mice consume the majority of their food during the dark phase (~70%),

dLAN mice show no preference for nighttime food intake. Daytime food intake is

atypical for nocturnal rodents and is associated with the development of metabolic

syndrome in animal models (Arble, Bass, Laposky, Vitaterna, & Turek, 2009). In

addition to dLAN shifting the daily pattern of food intake, HFD also caused subtle

changes in timing of food intake. Previous work demonstrates that blocking access to

food during the light phase can prevent increases in weight gain due to a HFD or dLAN

(Fonken et al., 2010; Hatori et al., 2012; Sherman et al., 2012; Tsai et al., 2012). This

suggests that weight gain due to both manipulations may partially result from the shift in

timing of food intake.

Light at night is associated with changes in immune function in rodents

(Bedrosian, Fonken, Walton, & Nelson, 2011b; Fonken, Haim, & Nelson, 2011). The

effects of light at night on metabolic inflammation have not been previously

Page 93: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

77

characterized. Here, I report that dLAN increases inflammation in white adipose tissue.

Both TNFα and MAC1 gene expression were upregulated by dLAN and HFD. Elevated

MAC1 expression is indicative of increased macrophage infiltration into fat tissue and

has previously been described in models of obesity (Weisberg et al., 2003). Macrophage

infiltration can increase pro-inflammatory cytokine release and result in the widespread,

chronic, low-grade peripheral inflammation typical of high fat accumulation (Marceau et

al., 1999; Plomgaard et al., 2005). The development of peripheral inflammation, although

it propagates obesity through disrupting insulin and leptin signaling (Kim et al., 2008)

generally follows the onset of obesity suggesting it is not the primary mechanism driving

weight gain.

In contrast, hypothalamic inflammation may precede and contribute to the

development of obesity as changes in the hypothalamic milieu are apparent within 24 h of

the induction of high fat feeding (Thaler et al., 2012). Therefore, I hypothesized that

hypothalamic inflammation may contribute to changes in weight gain in dLAN mice.

Although HFD elevated hypothalamic TNFα expression relative to standard chow, there

were no differences in TNFα expression between lighting conditions. Moreover, no

differences in MAC1 expression were apparent between light or dietary conditions. Lack

of significant changes in hypothalamic expression of MAC1 may be due to the specificity

of the changes in hypothalamic inflammation. For example, there is a regionally specific

enrichment of IKKβ and NF-κB in the mediobasal hypothalamus (includes the arcuate)

that becomes potently upregulated with HFD (Zhang et al., 2008). This suggests elevated

inflammation in the arcuate nucleus may be masked by the lack of change in

Page 94: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

78

inflammation in other hypothalamic nuclei. In support of this hypothesis, the

concentration of Iba1 positive cells was elevated in the arcuate of mice fed a HFD, but

not in the dorsal medial hypothalamus. Although dLAN did not affect Iba1 positive cells

among HFD mice, dLAN increased the number of Iba1 positive cells within the chow

group. This suggests that hypothalamic inflammation is not essential for weight gain in

dLAN as dLAN-HFD mice elevated weight gain compared to LD-HFD mice without

increasing hypothalamic microglia. Continued elevation of CNS inflammatory responses

combined with a long term HFD leads to gliosis and damage to proopiomelanocortin

neurons (Parton et al., 2007). However, neither diet nor lighting condition affected

POMC gene expression.

These results indicate that dLAN exacerbates weight gain and peripheral

inflammation associated with HFD. Lack of elevated hypothalamic inflammation among

dLAN mice suggests central inflammation is not the primary mechanism for light

induced weight gain. Overall, these results have important implications for industrial

societies in which nighttime light exposure and poor diet often co-occur. Further

understanding of the mechanisms through which LAN contributes to inflammation and

obesity is important for characterizing and treating metabolic disorders.

Page 95: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

79

Figures

Figure 4.1. Light at night exaggerates weight gain on a high fat diet.

Body mass and fat pad mass were elevated by dLAN and HFD. (A) Body mass

throughout the experiment. (B) Final body mass gain expressed as a percentage from

baseline body mass. (C) Epididymal fat pad mass corrected for final body mass. (D)

Blood glucose levels at the conclusion of the study. (* p < 0.05 between lighting

conditions, † p < 0.05 between dietary conditions).

Page 96: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

80

Figure 4.2. High fat diet and light at night alter timing of food intake.

Total daily food intake was comparable between lighting conditions; however, dLAN

mice consumed a higher percentage of food during the light phase. (A) Total daily food

intake. (B) Relative daytime food intake. (* p < 0.05 between lighting conditions, † p <

0.05 between dietary conditions).

Page 97: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

81

Figure 4.3. High fat diet and light at night increase adipose inflammation.

Both dLAN and HFD increased inflammatory gene expression in white adipose tissue.

(A) Relative MAC1 and (B) TNFα gene expression in epididymal fat. (* p < 0.05

between lighting conditions, † p < 0.05 between dietary conditions).

Page 98: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

82

Figure 4.4. Hypothalamic inflammation is elevated by high fat diet but not exposure to

dim light at night.

Hypothalamic inflammation is elevated by HFD but not dLAN. (A) Relative TNFα and

(B) MAC1 gene expression in the hypothalamus. Iba1 staining in the (C) arcuate and (D)

dorsomedial nuclei of the hypothalamus. Representative photomicrographs captured at

20X from the arcuate nucleus of (E) an LD-chow mouse and (F) an LD-HFD mouse.

Page 99: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

83

CHAPTER 5

EXERCISE ATTENUATES THE METABOLIC EFFECTS OF DIM LIGHT AT

NIGHT

Over the course of the 20th

century body mass rapidly increased worldwide. By

the year 2000 the number of adults with excess weight surpassed those who were

underweight for the first time in human history. This excess adiposity is recognized as

one of the world‟s leading health threats because obesity increases the risk of developing

type II diabetes, cardiovascular disease, hypertension, and cancer (Caballero, 2007). The

rapid growth in adiposity during the 20th

century correlates with significant changes in

human environment and lifestyle. In addition to changes in activity levels and dietary

choices, a less appreciated environmental perturbation has been the shift in timing of

daily activities. The invention of electric lighting ~150 years ago has enabled humans to

illuminate their homes, hospitals, factories, and night skies and engage in activities such

as countercyclical shift work (Navara & Nelson, 2007). Widespread adoption of electric

lights occurred well before an understanding of circadian biology, and without any

consideration of the negative biological consequences that artificial light at night (LAN)

may have on physiology and behavior.

Page 100: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

84

Circadian regulation of energy homeostasis is organized by an endogenous

biological clock located in the suprachiasmatic nuclei (SCN) of the hypothalamus. The

circadian clock is entrained by light information that travels directly from light-sensitive

ganglion cells in the retina to the SCN, thereby synchronizing individuals‟ physiology

and behavior to the external day-night cycle (Golombek & Rosenstein, 2010; Reppert &

Weaver, 2002). Because light is the primary signal for the circadian clock, exposure to

light at aberrant times can disrupt clock function (Navara & Nelson, 2007).

Many studies suggest a direct link between the molecular circadian clock and

metabolism (Bray & Young, 2007). Mice harboring a mutation in the core circadian gene

Clock are susceptible to obesity and metabolic syndrome (Turek et al., 2005). Clock

mutants show dramatic changes in circadian rhythmicity, as well as altered timing of food

intake and increased body mass. Serum leptin, glucose, cholesterol, and triglyceride

levels are increased in Clock mutants compared to wild type (WT) mice. Mice with

mutations in other clock related genes including Bmal1, Per1, Per2, Vipr2, and Rev-erbα

display similar metabolic outcomes (Bechtold, Brown, Luckman, & Piggins, 2008;

Carvas et al., 2012; Delezie et al., 2012; Marcheva et al., 2010). Even single tissue clock

gene disruptions can result in metabolic disturbances (Marcheva et al., 2010; Paschos et

al., 2012). Thus, it seems reasonable to propose that disrupted circadian clock function

has the potential to derange normal metabolism.

Mice housed in dim LAN (dLAN) elevate body mass and reduce glucose

tolerance independent of changes in total daily food intake or home cage locomotor

activity (Fonken et al., 2010). dLAN mice increase the percentage of food consumed

Page 101: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

85

during the light phase as compared to mice housed in dark nights; restricting food intake

to the dark phase ameliorates weight gain among dLAN mice (Fonken et al., 2010).

Daytime food intake is associated with weight gain and metabolic disruption in mice in

other contexts (Arble, Bass, Laposky, Vitaterna, & Turek, 2009; Bray et al., 2012).

Furthermore, the relationship between the circadian clock and metabolism appears to be

bi-directional as diet induced obesity can dampen circadian rhythms (Kohsaka et al.,

2007).

As mentioned, light is the dominant entraining factor for the circadian system;

however, non-photic stimuli such as food intake and exercise can alter circadian rhythms

(Fuller, Lu, & Saper, 2008; Mistlberger & Antle, 2011). Activity is both a behavioral

output of the circadian system and an important feedback factor that can modulate

rhythms (Edgar & Dement, 1991; Mistlberger, 1991; Reebs & Stcoeur, 1994). In constant

dark conditions, timed wheel access entrains circadian rhythms in mice (Edgar &

Dement, 1991). Moreover, scheduled access to a running wheel can strengthen circadian

rhythms in mice with disrupted clock function (Power, Hughes, Samuels, & Piggins,

2010). Even under a standard light-dark cycle, ad lib access to wheels can strengthen the

power of circadian rhythms in wild type mice (Schroeder et al., 2012).

In addition to strengthening circadian rhythms, it is well established that exercise

prevents weight gain (Patterson & Levin, 2008). Therefore, I hypothesized providing

mice a running wheel for voluntary exercise would buffer against the effects of LAN on

metabolism. Specifically, I hypothesized that mice exposed to LAN would increase body

mass and alter feeding rhythms, indicating circadian system disruption. I predicted that

Page 102: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

86

providing mice running wheels would strengthen circadian entrainment preventing

altered timing of food intake and LAN-induced weight gain.

Materials and Methods

Animals

Forty male Swiss-Webster mice (~8 weeks of age) were obtained from Charles

River Laboratories. The mice were individually housed in propylene cages (dimensions:

33 x 19 x 14 cm) at an ambient temperature of 22 ± 2° C and provided with Harlan

Teklad 8640 food (Madison, WI) and filtered tap water ad libitum. Upon arrival, mice

were maintained in a standard 14:10 light (150 lux) /dark (0 lux) cycle (LD; lights on at

2:00 EST) for one week in order to habituate to local lighting conditions and recover

from the effects of shipping. After this period mice were randomly assigned a group,

weighed, and transferred to either a cabinet with LD or dim light at night [dLAN; 14:10

light (150 lux) /dim (5 lux) light cycle]. Within each lighting condition mice received

either a locked wheel or a low-profile running wheel (running surface of 15.5 cm

diameter) for voluntary exercise (Med Associates, St. Albans, VT). Wheel running was

constantly monitored using a wireless interface hub system which transmitted the data to

a computer. Locked wheels were provided to control for the presence of a novel object in

the cage. Mice were weighed every week at Zeitgeber Time (ZT) 9.

After 3 weeks in experimental conditions, food was weighed twice daily,

immediately before the onset of the dark phase (ZT 14) and immediately after the onset

of the light phase (ZT 0). Average food intake for the light and dark phases over three

days was used to quantify percentage of daytime food intake. At the conclusion of the

Page 103: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

87

study mice were individually brought into a procedure room, anesthetized with isoflurane

vapors, and rapidly decapitated between ZT 9 and 11; a blood sample was then collected

and epididymal fat pads were removed and weighed.

Statistical analyses

One dLAN mouse with a running wheel was removed from statistical

comparisons because it did not use the wheel and one mouse was removed from the

locked wheel LD group for demonstrating sickness behaviors. Effects of lighting

condition and wheel access on body mass gain, fat pad mass, and percentage of daytime

food intake were analyzed using two-way analysis of variance (ANOVA). A repeated

measures ANOVA was used to assess change in body mass over time. Following a

significant F score, multiple comparisons were conducted with Tukey‟s HSD test. The

above statistical analyses were performed with StatView software (v.5.0.1, Cary, NC).

Running wheel activity was analyzed and actograms were generated using ClockLab

Software (Coulbourn Instruments, Boston, MA). An animal was considered rhythmic

when the highest peak occurred at ~1 cycle/24 h, with an absolute power of at least 0.005

mV/Hz (Kriegsfeld, et al., 2008). In all cases, differences between group means were

considered statistically significant if p ≤ 0.05.

Results

Somatic measures

Over the course of the study, body mass was elevated among all groups (F4,136 =

82.814; p < 0.0001); however, light at night potentiated increases in body mass, whereas

access to a running wheel limited weight gain (Body mass over time: F4,136 = 4.275 and

Page 104: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

88

4.659 respectively, Final body mass gain: F1, 34= 7.711 and 8.203 respectively; p <0.01;

Fig. 1A/B). Final body mass gain did not differ between mice exposed to dLAN with a

running wheel and mice housed in dark nights with either a running or locked wheel (post

hoc; p < 0.05). There were no interactions of the two variables on weight gain. Both light

and access to a running wheel also affected final fat pad mass (F1,33 = 7.505 and 3.791;

Fig. 1C); such that dLAN increased fat pad mass and presence of a running wheel

reduced fat pad mass. In agreement with previous results, mice housed in dLAN

increased percentage of food consumed during the light phase (F1,34 = 14.345; p < 0.001;

Fig. 1D). In contrast to the hypothesis, wheel running also increased the percentage of

food consumed during the light phase (F1,34 = 5.265; p < 0.05; Fig. 1D).

Daily running wheel activity

Total daily wheel running did not differ between mice in the LD and dLAN

conditions (F1,17 = 0.144; p > 0.1; data not shown). All mice in LD showed a dominant

rhythm of 0.042 (or 1 cycle per 24 hours) (Fig. 2A). In contrast, only 5 of the 9 dLAN-

mice demonstrated a dominant 24 hour wheel running rhythm (Fig. 1B).

Discussion

The goal of this study was to assess the effects of voluntary exercise on changes

in body mass and food intake associated with exposure to dLAN in male Swiss Webster

mice. I hypothesized that enhanced activity by means of optional wheel running would

prevent body mass gain in mice exposed to dLAN. Specifically, I predicted that voluntary

exercise would strengthen circadian organization in mice exposed to dLAN, preventing

increased daytime food intake. Here I report that exercise availability limits weight gain

Page 105: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

89

in mice housed under dLAN. In contrast to the hypothesis, reduced body mass occurred

independently of re-establishing nighttime food intake; mice with running wheel access

increased food intake during the light phase. Furthermore, although all mice maintained

under dark nights had a dominant 24 hour activity rhythm, a subset of the dLAN mice

showed disrupted patterns of wheel running.

These results confirm and extend previous findings (Coomans et al., 2013;

Fonken, Kitsmiller, Smale, & Nelson, 2012; Fonken et al., 2010); mice exposed to LAN

without access to wheel running elevated body mass gain over the course of the study.

Consistent with the hypothesis, access to a running wheel reduced weight gain among

mice exposed to dLAN. Final body mass gain was comparable between dLAN mice with

a running wheel and both groups of LD mice. Furthermore, whereas dLAN increased

relative epididymal fat pad mass, an index of overall adiposity, housing mice with a

running wheel reduced fat pad mass. These results indicate that weight gain induced by

dLAN is susceptible to traditional weight loss interventions, i.e. increased exercise.

As in previous studies, dLAN mice ate more food during the light phase

compared to mice exposed to dark nights (Fonken et al., 2010). Eating during the light

part of the day is atypical for nocturnal rodents and is associated with changes in

metabolism (Arble, Bass, Laposky, Vitaterna, & Turek, 2009; Bray et al., 2012).

Moreover, restricting food intake to the dark phase prevents weight gain in models of

obesity (Fonken et al., 2010; Hatori et al., 2012; Mistlberger, Lukman, & Nadeau, 1998;

Sherman et al., 2012). Voluntary wheel running has previously been associated with

increasing the power of ambulatory activity rhythms with greater activity specifically

Page 106: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

90

during the dark phase in mice housed in standard lighting conditions (Schroeder et al.,

2012). Thus, I predicted that access to a running wheel would prevent the shift towards

daytime food intake in dLAN mice. Contrary to my prediction, presence of a running

wheel increased daytime food intake irrespective of lighting condition. These results

differ from rats; rats with either voluntary wheel running or forced exercise consume

more calories during the active phase (Oudot, Larue-Achagiotis, Anton, & Verger, 1996).

To my knowledge the effects of voluntary wheel running on the daily pattern of food

intake in mice has not been investigated. However, mice with access to running wheels

have fewer, but larger, daily meals (Atalayer & Rowland, 2011).

Total daily wheel running did not differ between mice exposed to dark or dimly

illuminated nights. These findings are consistent with previous research demonstrating

that exposure to dLAN does not affect the amount of activity in either an open field or

home cage (Fonken et al., 2009; Fonken et al., 2010). Circadian rhythms of home cage

locomotor activity remain intact in mice exposed to dLAN and comparable to activity

patterns in mice exposed to dark nights (Fonken et al., 2010). In the present study,

however, wheel running activity was disrupted in several dLAN mice. All mice exposed

to dark nights displayed a dominant 24 h rhythm in wheel running compared to 5 of 9

mice exposed to dLAN. Although I have previously asserted that dLAN does not affect

sleep architecture or activity patterns, the existence of running activity during the so-

called inactive periods suggests that dim light may shift circadian activity when paired

with optional wheel running. Future studies should address sleep quantity and quality in

mice exposed to LAN. Disparate results between wheel running and home cage activity

Page 107: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

91

could reflect the different forms of behavior that the two systems monitor (Novak,

Burghardt, & Levine, 2012). Home cage activity occurs for multiple reasons including

grooming, feeding, or locomotor activity, whereas running wheels only monitor

voluntary running. Overall, these results suggest that voluntary exercise prevents weight

gain induced by dLAN without rescuing circadian rhythm disruptions.

Page 108: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

92

Figures

0 1 2 3 430

32

34

36

38

40LD-Sed

dLAN-Sed

LD-Ex

dLAN-Ex*

***

Weeks

Bo

dy m

ass (

g)

LD dLAN0

4

8

12

16 Sed

Ex

*

Bo

dy m

ass g

ain

(% b

aseli

ne)

LD dLAN0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5†

% E

pid

idym

al

fat

pad

mass

LD dLAN0

10

20

30

40

50

60

% D

ayti

me f

oo

d i

nta

ke

A B

C D

Figure 5.1. Voluntary exercise prevents weight gain induced by dim light at night.

(A) Body mass over the course of the study. (B) Body mass gain expressed relative to

baseline body mass. (C) Epididymal fat pad mass expressed relative to final body mass.

(D) Percentage of food consumed during the light phase. All data are presented as mean ±

SEM. *indicates dLAN-Sed differs from all other groups, †indicates main effect of

lighting condition

Page 109: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

93

Figure 5.2. Dim light at night disrupts daily wheel running patterns in a subset of mice.

Representative actograph from a mouse housed in either (A) dark or (B) dim nights.

Page 110: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

94

CHAPTER 6

DIM LIGHT AT NIGHT DISRUPTS MOLECULAR CIRCADIAN RHYTHMS

For >3 billion years, life outside of the highest latitudes has evolved under bright

days and dark nights. Most organisms have developed endogenously driven circadian

rhythms which are synchronized to this daily light/dark cycle. With the widespread

adoption of electric lighting ~150 years ago, humans began brightly illuminating their

nocturnal environments. Exposure to light at night is now pervasive in modern society

and typically considered a mild environmental perturbation. However, the use of light at

night (LAN) began prior to a deep appreciation of the importance of circadian rhythms

for normal biological functions (Fonken & Nelson, 2011; Gerstner, 2012).

Recent evidence suggests that exposure to unnatural light cycles increases the risk for

cancer (Stevens, 2009b), sleep disturbances (Kohyama, 2009), and mood disorders

(Driesen, Jansen, Kant, Mohren, & van Amelsvoort, 2010). Furthermore, exposure to

light at night is increasingly associated with changes in metabolism. Shift workers who

experience sustained nighttime illumination are at increased risk for cardiovascular

disease and elevated body mass index (Ha & Park, 2005; Knutsson, 2003; Parkes, 2002;

van Amelsvoort, Schouten, & Kok, 1999). Increases in nighttime light exposure at home

Page 111: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

95

are associated with increased body mass, waist circumference and triglyceride levels, and

poor cholesterol balance (Obayashi et al., 2013). Even brief exposure to altered light and

food schedules can result in adverse metabolic and cardiovascular consequences (Scheer,

Hilton, Mantzoros, & Shea, 2009). Moreover, I have reported that mice chronically

exposed to dimly illuminated, as opposed to dark, nights elevate body mass

independently of changes in total daily activity or caloric intake (Fonken, Kitsmiller,

Smale, & Nelson, 2012; Fonken et al., 2010). Mice exposed to dim nights shift the timing

of food intake toward the light phase and restricting food access to the dark phase

prevents dLAN-associate body mass gain. The mechanism by which light at night

induces these changes is not fully understood.

Here I propose that light alters metabolic homeostasis in mammals by disrupting

the circadian system. As mentioned, light is the most potent synchronizing factor for the

circadian system. Light information travels directly from intrinsically photosensitive

ganglion cells in the retina to the master circadian clock located in the suprachiasmatic

nuclei (SCN) of the hypothalamus (S. K. Chen, Badea, & Hattar, 2011). Pacemaker

neurons within the SCN thereby drive the circadian clock with an autoregulatory

transcriptional-translational feedback loop of transcription activators and repressors

(Albrecht, 2002). Although the SCN are the primary pacemakers in mammals, most if not

all central and peripheral tissues contain the molecular machinery necessary for self

sustaining circadian oscillation (Mohawk, Green, & Takahashi, 2012). The master clock

converts external light/dark information to neural and endocrine signals that synchronize

Page 112: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

96

peripheral clocks (Guo, Brewer, Champhekar, Harris, & Bittman, 2005; McNamara et al.,

2001).

Exposure to a pulse of light during the night can phase advance or delay the

circadian clock depending on the strength and time of the light signal (Miyake et al.,

2000). Exposure to constant light can alter activity rhythms and ablate circadian rhythms

in glucocorticoids, two principle outputs of the circadian system (Coomans et al., 2013;

Fonken et al., 2010). Moreover, specifically timed nighttime light pulses can be used to

ablate the circadian system (Ruby et al., 2008). Because disruption in circadian clock

genes are associated with significant changes in metabolism (Bechtold, Brown, Luckman,

& Piggins, 2008; Carvas et al., 2012; Delezie et al., 2012; Marcheva et al., 2010; Paschos

et al., 2012), I hypothesized that exposure to light at night alters metabolism through

disrupting the circadian system. To assess the effects of light at night on the circadian

clock I exposed mice to either total darkness or dim light (5 lux) during the night and

then characterized the expression of several circadian clock genes and proteins in the

SCN, hippocampus, liver, and adipose tissue. Five lux of nighttime light exposure was

selected because (1) it is approximately five times bright than maximal moonlight (2) it is

comparable to levels of light pollution found in urban areas (Kloog, Haim, & Portnov,

2009) and sleeping environments (Obayashi et al., 2013), yet (3) it is highly distinct from

daytime light levels.

Methods

Animals

Page 113: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

97

One hundred and twenty male Swiss Webster mice (~8 weeks of age) were

obtained from Charles River for use in this study. Mice were individually housed in

propylene cages (dimensions: 33 x 19 x 14cm) at an ambient temperature of 23 ±2°C and

provided with Harlan Teklad 8640 food (Madison, WI) and filtered tap water ad libitum.

All mice were maintained in a standard light dark cycle (LD; 14:10 light (~150 lux)/dark

(0 lux)) for one week following arrival. After the 1 week acclimation period mice were

randomly assigned a group and transferred to a cabinet with either LD or a light/dim light

cycle (14:10 light (~150 lux)/dim light (5 lux)). Daytime lighting was provided with

white LEDs on the walls of the cabinets and dim light was administered with a flexible

strip of cool white LEDs wrapped around the rack on which the mouse cages were

placed. The lighting intensity was measured inside the home cage and was highly

consistent between cages. Mice were also assigned to 1 of 6 tissue collection time points

(Zeitgeber Time (ZT) 2, 6, 10, 14, 18, 22). Mice were weighed at the start of the

experimental light treatment and weekly throughout the study. After 3 weeks in lighting

conditions food was weighed twice daily for four days to determine the timing of food

intake. Four weeks after placement in light conditions blood and tissue were collected for

either quantitative PCR (qPCR) or immunohistochemical analyses (n=5 per

group/use/time point).

Quantitative PCR

Mice were anesthetized with isofluorane vapors and rapidly decapitated.

Peripheral tissue was dissected out, immediately weighed on a fine balance, and flash

frozen. Weighed tissues include liver, spleen, pancreas, white adipose tissue (epididymal

Page 114: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

98

and inguinal), brown adipose tissue, adrenals, heart, and skeletal muscle. Brains were

collected, placed in RNAlater, and after > 24 h the hypothalamus and hippocampus were

dissected out for PCR. Total RNA was extracted from liver, white adipose, hippocampal,

and hypothalamic tissues using a homogenizer (Ultra-Turrax T8, IKAWorks,

Wilmington, NC) and an RNeasy Mini Kit following the manufacturers protocol (Qiagen,

Austin, TX). For fat extractions an additional chloroform step was added following

homogenization and prior to the use of the Mini Kit. RNA concentration and purity were

measured on an ND-1000 spectrophotometer (Fischer Scientific, PLACE). RNA

concentrations were equalized with sterile water and RNA was reverse transcribed into

cDNA with M-MLV Reverse Transcriptase enzyme (Invitrogen, Carlsbad, CA)

according to the manufacturer‟s protocol. Gene expression for Clock, Bmal1, Per1, Per2,

Cry1, Cry2 and Rev-erbα were determined using inventoried primer and probe assays

(Applied Biosystems, Foster City, CA) on an ABI 7500 Fast Real Time PCR System

using Taqman® Universal PCR Master Mix. The universal two-step RT-PCR cycling

conditions used were: 50 °C for 2 min, 95 °C for 10 min, followed by 40 cycles of 95 °C

for 15 s and 60 °C for 1 min. Relative gene expression of individual samples run in

duplicate was calculated by comparison to a relative standard curve and standardized by

comparison to 18S rRNA signal.

Immonohistochemistry

Mice were given a lethal dose of sodium pentobarbital and perfused transcardially

with ice-cold 0.1M PBS followed by 4% paraformaldehyde. Brains were removed, post-

fixed overnight, cyroprotected in 30% sucrose, and frozen with dry ice. Brains were

Page 115: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

99

serially sectioned at 40 μm into cryoprotectant and stored at -20°C. Sets of tissue

collected at 240 μm intervals were used for immunohistochemical detection of BMAL,

CLOCK, PER1, and PER2 using antibodies generously provided by David Weaver

(LeSauter et al., 2012). Sections were rinsed in PBS, blocked for 1 h in 4% bovine serum

albumin in 0.1 M PBS + 0.3% TX, and then incubated overnight at room temperature

with primary antibody at 1:5000. The following day sections were rinsed and incubated

for 1 h with biotinylated goat anti-rabbit at 1:1000 (Vector Laboratories, Burligame, CA).

Endogenous peroxidase activity was quenched with 3% H2O2 in methanol for 20 min and

then the signal was amplified with avidin-biotin complex (Vectastain Elite ABC kit,

Vector Laboratories) and tissue was developed using DAB. Tissue was then mounted

onto gel coated slides, dehydrated through a series of graded ethanol washes, cleared with

xylene, and coverslipped using Permount. Images of sections containing the SCN were

captured at 20X using a Nikon E800 microscope. The number of immonoreactive cells in

the SCN was counted in ImageJ (NIH) by a condition blind observer and averaged across

section and sides of the bilateral structure to obtain one value per each mouse.

Statistical analyses

Body mass was analyzed using a repeated-measures analysis of variance

(ANOVA) with time as the within subject factor and lighting condition as the between

subject factor. Comparisons between lighting conditions with respect to weight gain,

tissue masses, and percentage of daytime food intake were conducted using a one-way

ANOVA. Blood glucose concentrations, qPCR results, and IHC results were analyzed

using a two-way ANOVA with lighting condition and time as the between subjects

Page 116: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

100

factors. Following a significant F score, multiple comparisons were conducted with

Tukey‟s HSD test. All statistical analyses were performed using StatView software

(v.5.0.1, Cary, NC). In all cases, differences between group means were considered

statistically significant if p ≤ 0.05.

Results

Exposure to dim light at night increases body mass

Body mass increased among both groups over the course of the study (F3,354 =

304.187; p < 0.0001); however, mice exposed to dim light at night significantly elevated

body mass compared to mice housed in dark nights (F3,354 = 15.820; p < 0.0001; Fig 1A).

Overall, mice exposed to dim light at night had a greater body mass gain at the

conclusion of the study compared to mice exposed to dark nights (F1,118 = 15.476; p <

0.001; Fig 1B). There were no differences in spleen, liver, pancreas, BAT, adrenal, or

heart masses between groups (p > 0.05 in all cases). Epididymal fat pad mass was

elevated among mice exposed to dim light at night ( F1,58 = 7.520; p < 0.01; Fig 1D)

suggesting increases in body mass may reflect increases in white adipose tissue.

Light at night attenuates nocturnal feeding behavior

Despite increases in body mass among mice exposed to dLAN, there were no

differences in total daily food intake between groups (p > 0.05; Fig S1). In agreement

with previous results (Fonken, Kitsmiller, Smale, & Nelson, 2012; Fonken et al., 2010),

exposure to light at night increased the percentage of food consumed during the light

phase (F1,118 = 16.595; p < 0.0001; Fig 1D). Mice displayed a diurnal variation in blood

Page 117: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

101

glucose levels (F5,106 = 14.023; p < 0.0001; Fig 1E) with no differences between lighting

conditions (p > 0.05).

Clock gene expression is disrupted by nighttime light exposure

To test the hypothesis that nighttime light exposure affects core clock gene

expression, I analyzed the diurnal expression of transcripts encoding Clock, Bmal1, Per1,

Per2, Cry1, and Cry2 in the hypothalamus, hippocampus, fat, and liver every 4 h after 4

weeks of exposure to dim or dark nights. All of the core clock genes assessed in the

hypothalamus displayed diurnal variation (p < 0.05; Fig. 2A). Expression of Clock, Bmal,

and Cry1 were unaffected by lighting conditions, however, rhythmic expression of Per1,

Per2, and Cry2 were all attenuated by exposure to dim light as compared to dark nights

(p < 0.05; Fig 2A). Specifically, gene expression of Per2 and Cry1 was significantly

reduced at ZT18, 6 hours after lights off, and Per1 expression was reduced at both ZT6

and ZT14.

In order to determine whether the effects of nighttime light exposure in the brain

are specific to the master circadian clock I examined core clock gene expression in the

hippocampus, a brain region known to show robust circadian oscillations. There was

clear cycling of Clock, Bmal1, Per1, Per2, Cry1, and Cry2 in the hippocampus of mice

exposed to both dark and dim nights (p < 0.001; Fig 2B). Overall, lighting condition had

no effect on hippocampal clock gene expression (p > 0.05). This suggests that within the

brain, changes in clock gene expression provoked by exposure to light at night may be

regionally specific.

Page 118: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

102

Recent work has demonstrated the importance of tissue specific clocks in

regulating metabolism (e.g., Marcheva et al., 2010). Thus, I investigated the effects of

nighttime light exposure on core clock gene expression in peripheral white adipose and

liver tissues. All of the clock genes analyzed except for Clock displayed rhythmic

variation in white adipose tissue and there was no effect of nighttime light exposure on

mRNA expression levels (p > 0.05; Fig 2C). In contrast, rhythmic expression of Bmal1,

Per1, Per2, Cry1, and Cry2 were all attenuated in the liver of mice exposed to dim nights

(p < 0.05; Fig 2D). Taken together, these results reveal that exposure to low levels of

light at night produce both tissue- and gene-specific changes in the expression levels of

several core circadian clock genes.

To further explore the effects of light at night on clock transcriptional networks I

studied the 24 h pattern of expression of Rev-Erb mRNA in hypothalamic, hippocampal,

WAT, and liver tissues. Rev-Erb is a nuclear receptor that has prominent functions for

both circadian oscillations and metabolic homeostasis (reviewed in Ribberger and

Albrecht, 2012). Diurnal rhythmicity in Rev-Erb expression was observed in all tissues

evaluated (p < 0.05; Fig 3). However, exposure to dim light at night reduced Rev-Erb

expression levels during the light phase in both the WAT and liver (p < 0.05). Decreased

Rev-Erb expression was specific to the periphery as there were no changes in

hypothalamic or hippocampal Rev-Erb mRNA levels.

Core clock protein expression in the suprachiasmatic nucleus

In order to fully characterize changes in circadian clock function within the SCN

after nighttime light exposure I evaluated clock protein expression every 4 h in mice

Page 119: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

103

housed under dark or dimly lit nights for 4 weeks. Rhythmic CLOCK and BMAL protein

expression was observed within the SCN with no effect of nighttime light exposure (Fig

4A/B). Expression levels of both PER1 and PER2 were altered by exposure to dim light

as compared to dark nights (p < 0.05; Fig 4C/D). Whereas mice exposed to dark nights

had rhythmic expression of PER1, mice exposed to dim light at night showed no diurnal

variation in PER1. Specifically, the peak in PER1 expression at ZT10 was abolished by

exposure to dim light at night. PER2 expression was also suppressed in mice exposed to

dim light at night at ZT14. These results confirm gene expression findings demonstrating

dim light at night specifically targets hypothalamic Per1 and Per2.

Discussion

Exposure to electric light at night can lead to disruptions in metabolic energy

homeostasis in rodents and humans (Ha & Park, 2005; Knutsson, 2003; Obayashi et al.,

2013; Parkes, 2002; van Amelsvoort, Schouten, & Kok, 1999). However, it remains

unclear how environmental lighting affects metabolism. Thus, I exposed mice to dim

light at night and investigated changes in body mass and the circadian system. Here I

establish that exposure to ecologically relevant levels of dim light during the night

attenuate circadian clock gene and protein rhythms, change feeding behavior, and lead to

weight gain. These observations indicate that exposure to dim light at night, a

commonplace and innocuous seeming environmental manipulation, can influence the

circadian system and metabolism.

Mice exposed to dim light at night showed rapid and sustained body mass

elevations. Increases in body mass may reflect increases in white adipose tissue as

Page 120: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

104

epididymal fat pad mass was elevated among mice exposed to dim nights. Although total

daily caloric intake was comparable between groups, mice exposed to light at night

consumed more food during the light period and less during the dark period than mice

housed in dark nights. Disorganization in the feeding rhythm may contribute to increased

body weight. Indeed, altered timing of food intake can cause weight gain and restricting

feeding to specific hours prevents development of obesity (Arble, Bass, Laposky,

Vitaterna, & Turek, 2009; Bray et al., 2010; Hatori et al., 2012; Sherman et al., 2012).

Furthermore, mice fed a high fat diet disrupt daily feeding patterns and attenuate

circadian clock gene rhythms in peripheral tissue (Kohsaka et al., 2007).

Genetic models indicate a close association between the molecular events

underlying metabolism and those involved in the generation of circadian rhythms. For

example, Clock mutant mice become overweight on a high fat diet and develop

symptoms characteristic of metabolic syndrome (Turek et al., 2005). Mice with mutations

in other clock related genes including Bmal1, Per1, Per2, Vipr2, and Rev-erbα display

similar metabolic outcomes (Bechtold, Brown, Luckman, & Piggins, 2008; Carvas et al.,

2012; Delezie et al., 2012; Marcheva et al., 2010). These studies indicate alterations in

core clock transcription factors within both the central clock and peripheral tissues alter

metabolism. Importantly, these results suggest that changes in the circadian clock genes

can be induced with exposure to ecologically relevant levels of dim light at night. Mice

exposed to dim light at night suppress Per1 and Per2 expression at both the gene and

protein level in the SCN. Importantly, there were no differences in clock gene expression

Page 121: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

105

in the hippocampus, suggesting changes in central clock gene expression provoked by

exposure to light at night are regionally specific.

In addition to altering clock gene expression in the hypothalamus, exposure to

dim as opposed to dark nights attenuated the rhythm in all but one of the core circadian

clock genes assessed in the liver. Peripheral clocks are entrained by neural and endocrine

signaling from the SCN (Guo, Brewer, Champhekar, Harris, & Bittman, 2005;

McNamara et al., 2001), as well as local factors such as nutritional signals (Vollmers et

al., 2009). Recent work highlights the importance of peripheral clocks in regulating

metabolism as single tissue clock gene deletions in the liver or fat can result in metabolic

disturbances (Lamia, Storch, & Weitz, 2008; Marcheva et al., 2010; Paschos et al., 2012).

In addition to disruption in core clock mechanisms, mice exposed to dim light at

night attenuated Rev-Erb expression in the liver and adipose tissue. Although previously

considered an accessory feedback loop, REV-ERBs are increasingly demonstrated to be

essential for circadian clock function and regulation of rhythmic metabolism. Mice

deficient in both isoforms of REV-ERB show circadian rhythm adjustments and

pronounced changes in metabolically related functions (Bugge et al., 2012; H. Cho et al.,

2012).

Here I provide an extensive characterization of expression of circadian clock

genes and proteins in both the master circadian oscillator in the brain and tissue specific

clocks in mice exposed to light at night. Overall, these findings indicate that exposure to

light at night attenuates core circadian clock mechanisms in the SCN at both the gene and

protein level. Moreover, circadian clock function is disrupted in metabolically relevant

Page 122: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

106

peripheral tissue (i.e., white adipose tissue and liver) by nighttime light exposure. These

changes in circadian clock function are associated with alterations in feeding behavior

and increased weight gain. These finding are significant because they provide evidence

for how mild changes in environmental lighting can alter circadian and metabolic

function. Exposure to light at night is pervasive in modern society and typically

considered a harmless environmental perturbation; however, these results demonstrate

nighttime light exposure alters homeostatic functions. Detailed analysis of temporal

changes induced by nighttime light exposure may provide insight into the onset and

progression of obesity and metabolic syndrome and other disorders involving sleep and

circadian disruption.

Page 123: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

107

Figures

1 2 3 432

34

36

38

LD

dLAN

**

*B

od

y m

ass

LD dLAN0

5

10

15 *

Bo

dy m

ass g

ain

(% b

aseli

ne)

LD dLAN0

1

2

*

Ep

idid

ym

al

fat

pad

mass (

g)

LD dLAN0

10

20

30

40

50

*

Perc

en

t d

ayti

me

foo

d i

nta

ke

0 12 24

100

150

200

250LD

dLAN

Blo

od

glu

co

se (

mg

/dl)

A

E

C

B

D

Figure 6.1. Mice exposed to dim light at night alter somatic measures.

(A) Overall body mass (B) body mass gain and (C) epididymal fat pad mass were

increased in mice exposed to dimly lit as compared to dark nights. (D) Mice exposed to

dim light at night altered timing of food intake consuming more during the light phase

than mice exposed to dark nights. (E) Blood glucose was rhythmic and did not differ

between groups. All data are presented as (mean ± SEM). *indicates dLAN differs from

LD.

Page 124: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

108

0.4

1.2

0

2

0.0

1.5

**

0.00

1.25

*

0.0

1.5

0

2 *

0.0

3.5

0

3

0.0

3.5

0.0

3.5

0

3

0

1

0.0

1.5

0

2

0

3

0.0

0.5

0

3

*

0.0

2.5

0 12 24

0.0

1.5

0 12 24

0.0

3.5

*

0 12 24

0.0

1.5

*

*

0 12 24

0

2

**

0 12 24

0

3

*

0 12 24

0

1

2

Clock Bmal Per1 Per2 Cry1 Cry2

Liv

er

WA

TH

ipp

oca

mp

us

Hyp

oth

ala

mu

s

Re

lati

ve

mR

NA

ex

pre

ss

ion

A

B

C

D

Figure 6.2. Dim light at night attenuates clock gene expression.

(A) Hypothalamic, (B) hippocampal, (C) white adipose, and (D) liver tissues were collected at 4 h intervals from mice

exposed to dark (black lines) or dimly lit (dotted lines) nights and Clock, Bmal1, Per1, Per2, Cry1, and Cry2 mRNA

expression were quantified. Values are relative to a standard curve and normalized to 18S. * dLAN differs from LD

108

Page 125: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

109

0

1

0.0

0.5

0 12 24

0.0

3.5

*

0 12 24

0.0

1.2

*

*

Re

lativ

e R

ev-

Erb

mR

NA

exp

ress

ion

WAT Liver

Hypothalamus Hippocampus

Figure 6.3. Dim light at night suppresses Rev-Erb expression in peripheral tissue.

Rev-Erb gene expression was quantified in the (A) hypothalamus (B) hippocampus, (C)

white adipose tissue, and (D) liver. Values are expressed as relative abundance (mean ±

SEM) after normalization to 18S *indicates dLAN differs from LD.

Page 126: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

110

Figure 6.4. Clock protein expression is reduced in the SCN of mice exposed to dimly lit

nights.

Page 127: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

111

(A) PER1, (B) PER2, (C) CLOCK, and (D) BMAL immunoreactivity were analyzed in

hypothalamic tissue collected every 4 h from mice exposed to either dark (black lines) or

dimly lit (dotted lines) nights for 4 weeks. Images of sections containing the SCN were

captured at 20X and the number of immonoreactive cells was counted and averaged

across section and sides of the bilateral structure. Data are presented as (mean ± SEM).

*indicates dLAN differs from LD.

Page 128: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

112

CHAPTER 7

LIGHT AT NIGHT AFFECTS CARDIAC ARREST OUTCOME

Global ischemia produces high levels of central nervous system (CNS) damage

that profoundly affect patient survival and long-term cognitive and psychological

recovery. Minimizing this CNS injury to improve patient outcome is an important goal of

current research. Importantly, the majority of damage to the CNS produced by cerebral

ischemia is mediated by endogenous secondary processes (Krause, Kumar, White, Aust,

& Wiegenstein, 1986; Saito, Suyama, Nishida, Sei, & Basile, 1996). Excitotoxicity,

inflammation, and apoptosis develop in the days following injury and extensively

contributing to outcome (Eltzschig & Eckle, 2011; Kirino, 1982; Nitatori et al., 1995;

Weil, Norman, DeVries, & Nelson, 2008). The delay in CNS damage following injury

provides a potential therapeutic window for influencing recovery. This suggests changes

in environment may affect neural damage that develops post-ischemia.

One inconsistent environmental factor in hospitals is nighttime light exposure.

Hospital intensive care units have variable levels of lighting during both day and night

(Fig. 1; (Dunn, Anderson, & Hill, 2010)). These disruptive lighting conditions may

influence patient recovery because light is the most potent entraining signal for the

Page 129: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

113

mammalian circadian clock (suprachiasmatic nuclei; SCN). Extrinsic light information

travels directly from the intrinsically photosensitive retina ganglion cells (ipRGCs) to the

SCN via the retinohypothalamic tract (Hattar, Liao, Takao, Berson, & Yau, 2002)

synchronizing daily physiological rhythms to the external light-dark cycle (Reppert &

Weaver, 2002). Aberrant light exposure can disrupt the circadian system creating

desynchrony between internal rhythms and the external environment.

Circadian rhythms are important for many homeostatic functions including those

associated with the immune system (Lange, Dimitrov, & Born, 2010). There are circadian

components to many immunological processes including antigen presentation, toll-like

receptor function, cytokine production, and lymphocyte proliferation (Arjona & Sarkar,

2006; A. C. Silver, Arjona, Walker, & Fikrig, 2012) and many immune cells such as

natural killer cells, macrophages, dendritic cells, and B cells possess molecular clock

mechanisms necessary for self-sustaining oscillations (Arjona & Sarkar, 2005; Keller et

al., 2009; A. C. Silver, Arjona, Hughes, Nitabach, & Fikrig, 2012). This reciprocal

relationship between the circadian system and immune function has led to multiple

studies evaluating the effects of chronic circadian disruption on human physiology. Shift-

workers, who are chronically exposed to LAN, are at increased risk for several

inflammatory disorders including heart disease (Ha & Park, 2005), cancer (Davis &

Mirick, 2006; Schernhammer et al., 2001), disrupted rhythmicity of neuroendocrine

function (such as corticotrophin releasing hormone, glucocorticoids, and prolactin)

(Claustrat, Valatx, Harthe, & Brun, 2008; Persengiev, Kanchev, & Vezenkova, 1991),

metabolic disorders and diabetes, as well as mood disorders (Dumont & Beaulieu, 2007).

Page 130: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

114

Indeed, shift workers display several altered immune parameters, including elevated C-

reactive protein and increased leukocyte count (Puttonen, Viitasalo, & Harma, 2011).

Importantly, circadian disruption dysregulates inflammatory responses independently of

sleep loss or stress (Castanon-Cervantes et al., 2010). In healthy individuals, circadian

misalignment can be rapidly induced with aberrant lighting schedules, resulting in

adverse metabolic and cardiovascular consequences (Scheer, Hilton, Mantzoros, & Shea,

2009).

Cardiac arrest has both seasonal and circadian (i.e., time of day) patterns of

incidence and recovery (M. C. Cohen, Rohtla, Lavery, Muller, & Mittleman, 1997;

Spencer, Goldberg, Becker, & Gore, 1998; Weil et al., 2009), implicating light in altering

the physiological response to cerebral ischemia. Moreover, disruption of the core clock

gene Per2, impairs recovery following myocardial ischemia (Eckle et al., 2012). Light

exposure may be particularly salient to CA-induced neuroinflammation. Although several

mechanisms contribute to damage following ischemic injury, including energetic failure,

excitotoxicity, and oxidative damage (reviewed in (Eltzschig & Eckle, 2011; Weil,

Norman, DeVries, & Nelson, 2008), manipulation of inflammatory responses are

considered a prime target for improving recovery. Following CA an inflammatory

response is triggered by activation of microglia and astrocytes with a corresponding

upregulation of pro-inflammatory cytokines (Stoll, Jander, & Schroeter, 1998). IL-1β and

TNFα are pro-inflammatory cytokines that are upregulated within hours following global

ischemia (Saito, Suyama, Nishida, Sei, & Basile, 1996) and exacerbate injury. Therefore,

if dim light at night promotes inflammation, then it could be a critical factor for

Page 131: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

115

consideration in cardiac arrest outcome. I hypothesized that light exposure at night would

potentiate injury following cardiac arrest. Consistent with my hypotheses, light at night

following CA enhanced acute cytokine responses, increased neuronal death, and resulted

in higher short-term mortality. Moreover, the effects of light at night were ameliorated

through inhibition of cytokines and manipulation of the light source.

Methods

Animals

8-week old male Swiss Webster mice (~30g; Charles River, Kingston, NY) were

housed in a temperature- and humidity-controlled vivarium and provided ad libitum

access to food and water. Mice were left unmanipulated for 1 week to recover from the

effects of shipping and adjust to a 14:10 light/dark (LD) cycle prior to experimental

manipulations.

Cardiac arrest and cardiopulmonary resuscitation procedure

Mice were anesthetized with 3% isoflurane in air, intubated, and maintained

thereafter on 1.5% isoflurane. Mice were ventilated a tidal volume of 150 µL at a

respiratory rate of 160 breaths/min. A temperature probe was inserted in the temporalis

muscle on the left side of the head as an indicator of brain temperature (correlation

between brain and temporalis muscle temp r2 = 0.942) (Neigh, Kofler et al., 2004). A

second probe monitored rectal temperature. A PE10 catheter was placed into the right

jugular vein for epinephrine (EPI) and potassium chloride (KCl) administration. Blood

pressure was monitored through a cannula inserted into the right femoral artery and

connected to a blood pressure transducer (Columbus, Instruments). Mice were stabilized

Page 132: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

116

for 10 min and blood pressure and temperature recorded at 1 min intervals (Fig. 2).

Following the 10 min acclimation, body and tail (but not head) temperature were lowered

by circulating cold water through a coil system beneath the mouse to induce peripheral

hypothermia restricting damage to the CNS during the CA/CPR procedure. CA was

induced with an injection of KCL (50 μl, 0.5 M, 4°C) into the jugular catheter and the

mouse was disconnected from the ventilator. Once a body temperature of 27°C was

reached after approximately 4 min of arrest slow re-warming via a heat lamp and thermal

blanket began. After 7 min 45 sec of arrest mice were reattached to the ventilator and 100

% oxygen at a tidal volume of 150 µL and a respiratory rate of 160 breaths/min was

ventilated. After 8 min of arrest CPR was initiated with an injection of EPI (16 µg in 0.6

cc saline, 37°C) into the jugular catheter and chest compressions (300/min); 0.5 µg

injections of EPI were administered until the mouse resuscitated (with a maximal dose of

32 µg). Mice were maintained on 100% oxygen for 25 min after return of spontaneous

circulation and catheters were removed and incisions sutured.

Tissue collection and staining

One week following CA mice were individually brought into a procedure room,

anesthetized with isoflurane vapors and a blood sample was collected via the retro-orbital

sinus. Mice then received a lethal injection of sodium pentobarbital and were perfused

transcardially with ice-cold 0.1M PBS followed by 50 µL of 4% paraformaldehyde.

Brains were post-fixed overnight, cryoprotected in 30% sucrose, frozen on crushed dry

ice, and stored at -80ºC. Fourteen µm brain sections were sliced at -22ºC using a cryostat

and thaw mounted onto Super Frost Plus slides (Fisher, Hampton, NH). Sections were

Page 133: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

117

stored at -20ºC until stained with Fluoro-Jade C (FJ-C) or Iba1 using procedures already

established in our lab (Weil et al., 2009). Fluoro-Jade C. Cell death was quantified by

labeling degenerating neurons with the fluorescein derivative FJ-C (Millipore, Temecula,

CA). Mounted sections were thoroughly dried on a slide warmer, immersed in a basic

ethanol solution (80% EtOH with 1% NaOH) and rinsed with 70% ethanol followed by

water. Slides were placed in a 0.06% potassium permanganate solution for 10 min and

rinsed twice. Sections were simultaneously incubated in FJ-C (0.0001% in a 1% acetic

acid solution) and counterstained with DAPI (Sigma, St. Louis). Slides were rinsed 3X,

aspirated, completely dried, cleared in xylene for 1 min, and coverslipped with DPX

(Sigma). FJ positive cells were counted in the CA1, CA3, and DG of both hippocampal

hemispheres by a condition blind experimenter using a Nikon E800 microscope at 200X

magnification. Microglia. Microglia were visualized using a Iba1 directed antibody.

Slides were dried, rinsed in phosphate buffered saline (PBS), and blocked with bovine

serum albumin (BSA). Slides were incubated at room temperature for 24 h with rabbit

anti-Iba1 antibody (Wako, Richmond, VA) diluted 1:1000 in PBS containing 0.1%

Triton-X and BSA. Slides were rinsed and incubated with biotinylated goat anti-rabbit

secondary antibody (1:1000; Vector Labs, Burlingame, CA) for 1 h. Sections were

quenched in H2O2 in methanol, rinsed, and treated with Elite ABC reagent for 60 min

(Vector). Sections were rinsed, developed with DAB (Vector), rinsed, dehydrated,

cleared, and coverslipped. Photomicrographs of the hippocampus were taken with a

Nikon E800 microscope at 20X and images were assessed using Image J software (NIH)

to determine immunoreactive regions.

Page 134: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

118

Cytokine expression

A separate cohort of mice that underwent CA or SHAM was used for detection of

hippocampal cytokine gene expression using RT-PCR. Mice were brought individually

into a procedure room, anesthetized with isoflurane vapors, blood was drawn via the

retro-orbital sinus, and mice were rapidly decapitated. Brains were removed, divided

sagittally along the midline, and hippocampi extracted and immersed in RNALater

stabilizing solution RT-PCR. From hippocampi stored in RNALater, total RNA was

extracted using a homogenizer (Ultra-Turrax T8, IKAWorks, Wilmington, NC) and an

RNeasy Mini Kit (Qiagen). RNA was reverse transcribed into cDNA with M-MLV

Reverse Transcriptase enzyme (Invitrogen) according to the manufacturer‟s protocol.

Pro-inflammatory cytokine expression for IL-1β, IL-6, and TNFα was determined using

inventoried primer and probe assays (Applied Biosystems, Foster City, CA) on an ABI

7500 Fast Real Time PCR System using Taqman® Universal PCR Master Mix. The

universal two-step RT-PCR cycling conditions used were: 50 °C for 2 min, 95 °C for 10

min, followed by 40 cycles of 95 °C for 15 s and 60 °C for 1 min. Relative gene

expression of individual samples run in duplicate was calculated by comparison to a

relative standard curve and standardized by comparison to 18S rRNA signal.

Microglial isolation and RNA extraction

A separate cohort of mice that underwent CA or SHAM procedures was used for

microglial cytokine analysis. Microglia were extracted following a previously reported

protocol (Wynne, Henry, Huang, Cleland, & Godbout, 2010). Briefly, 24 h after surgery

and one night of either dLAN or LD mice were anesthetized with isoflurane vapors,

Page 135: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

119

blood was drawn via the retro-orbital sinus and mice were rapidly decapitated. Brains

were removed and placed in ice-cold Hank‟s Balanced Salt Solution (HBSS). Brains

were transferred to a sterile flow hood and crushed through a 70 µm nylon cell strainer.

The resulting homogenate was transferred to a 15 mL tube, topped off with additional

HBSS and centrifuged for 7 min at 600g at 15°C. Supernatant was discarded and cell

pellets re-suspended in 70% isotonic Percoll (GE Healthcare, Uppsala, Sweden). A

discontinuous Percoll gradient was created by applying layers of 70%, 50%, 35%, and

0% isotonic Percoll (bottom to top). Percoll gradient was centrifuged for 30m at 2000g at

15°C and microglia were extracted from the interphase layer between the 70% and 50%

Percoll. Microglia were then washed and RNA immediately extracted using a Sonicator

(Microson XL2000, Misonix Inc., Farmingdale, NY) and an RNAeasy Micro Kit

(Qiagen, Valencia CA). RNA was reverse transcribed into cDNA and processed as

described above (in Real-time (RT) PCR).

Administration of cytokine inhibitors

An indwelling cannula was inserted into the left lateral ventricle (cannula

position: +0.02 posterior and -0.95 lateral to bregma, extending 2.75 mm below the skull;

Plastics One, Roanoke, VA) of anesthetized mice using a stereotaxic apparatus three days

prior to CA or SHAM surgeries (Neigh et al., 2009). One hour following CA mice

received a 2 μL injection of either a vehicle solution (artificial cerebral spinal fluid,

aCSF), a monoclonal TNF antibody (Infliximab. IFX), an IL1 receptor antagonist (IL1-

ra), or an IL6 neutralizing antibody (IL6na) based on pre-assigned group and were then

Page 136: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

120

placed back in their respective lighting conditions. Cannula placement was verified on

Iba1 stained tissue.

Corticosterone radioimmunoassay

Within 30 min of collection, blood samples were centrifuged at 3000g for 30 min

at 4°C. Plasma was collected and stored at -80°C until assayed. The samples were

assayed using and I125

corticosterone kit (MP Biomedicals, Solon, OH). The standard

curve was run in triplicate and samples in duplicate. All samples within an experiment

were run in a single assay.

Statistical Analyses

Statistical comparisons among groups were conducted using ANOVA. In the case

of significant differences (p<0.05), a post-hoc Tukey‟s test was conducted. When

conditions of normality or equal variance were not met, the data were log transformed.

Above statistical tests will be conducted using StatView Software v. 5.0.1. Survival plots

were analyzed using Kaplan-Meier survival analysis. Differences were considered

statistically significant when p < 0.05.

Results

Hospital lighting levels

In order to assess typical lighting environments for patients recovering in hospital

settings, HOBO light loggers (Onset, Bourne, MA) were placed in patient rooms at The

Ohio State University Wexner Medical Center Hospital. Significant nighttime light

intrusions were observed in all units monitored, including the intensive care, post-

surgical, and cardiac intensive care units (Fig. 1; representative patient lighting). Patients

Page 137: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

121

experienced light levels as high as 100 lux several times each night between the hours of

11PM and 6AM. After confirming nighttime light disruptions in patient rooms, I used a

mouse model of CA to determine whether exposure to dim light at night influences

recovery following global cerebral ischemia.

Dim light at night increases mortality and hippocampal damage following CA

Eight week old Swiss Webster mice were acclimated to a standard light dark

cycle [14h light (150 lux): 10h dark (0 lux); LD] and then underwent a CA or SHAM

procedure (Fig. 2; surgical measures). I used nocturnal mice to avoid the confound of

sleep disruption. Following the procedure mice either remained in LD cycle or were

transferred to a bright-dim light cycle [14h light (150 lux): 10h dim light (5 lux); dLAN].

As anticipated, there was 100% survival in both SHAM groups. In contrast, CA

significantly reduced survival compared to the SHAM procedure (p < 0.05; Fig 3a).

Among the CA mice, mortality in the dLAN group was four-fold higher than in the LD

group suggesting that modest changes in the recovery environment markedly affect

cardiac arrest survival.

The reduced survival rate of mice exposed to dLAN may reflect increased

neuroinflammation and hippocampal cell death. One week following CA or SHAM

procedures mice were anesthetized and perfused transcardially with ice cold saline

followed by 4% paraformaldehyde. Brains were cryoprotected, frozen, sectioned, and

processed for Fluoro-JadeC, a marker for degenerating neurons. The Fluoro-JadeC

labeled tissue was used to evaluate cell death in the hippocampus, a brain region

particularly vulnerable to ischemic damage (Nikonenko, Radenovic, Andjus, & Skibo,

Page 138: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

122

2009). As expected, cell death was uniformly low among SHAM mice (Fig. 3b-d), and

significantly elevated among mice subjected to CA (p < 0.05; Fig. 3b,e,f). Moreover,

mice exposed to dLAN had significantly more cell death in the hippocampus one week

after CA compared with mice exposed to dark nights (p < 0.05; Fig. 3b-f). Hippocampal

cell death is a reliable proxy for overall recovery after global ischemia as increased

hippocampal damage is associated with elevated mortality and memory deficits, as well

as impaired affective responses (M. Fujioka et al., 2000; Langdon, Granter-Button, &

Corbett, 2008; Neigh, Glasper et al., 2004; Neigh, Kofler et al., 2004; Zola-Morgan,

Squire, & Amaral, 1986). Investigation of corticosterone concentration at 6 h intervals

following CA did not reveal any significant differences between the CA-dLAN and CA-

LD groups during the first 24 h of recovery (p > 0.05; Fig. 4).

Dim light at night alters acute inflammatory status

The inflammatory response following global ischemia is an important factor in

recovery. Thus, I investigated whether dLAN alters the expression of pro-inflammatory

cytokines following CA. Brain tissue was collected 24 h after CA or SHAM procedures,

i.e., after only a single night of post-ischemic dLAN or LD. The brains were rapidly

removed and placed in RNA later. The following day, hippocampi were dissected out and

used for quantitative real time PCR (q-PCR) analyses of pro-inflammatory cytokines. As

expected, TNFα, IL1β, and IL6 gene expression were elevated among CA compared to

SHAM mice (p < 0.05; Fig 5a-c). Moreover, exposure to a single night of dLAN after CA

was sufficient to upregulate expression of TNFα and IL1β, compared to mice exposed to

a dark night (p < 0.05; Fig. 5a,b). The increase in pro-inflammatory cytokine expression

Page 139: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

123

may occur very early after placement back in lighting conditions. Indeed, a difference in

post-CA TNFα expression was apparent after as little as 4 h of total darkness versus dim

light (p < 0.05; Fig. 5d). There were no differences in hippocampal cytokine gene

expression between dLAN and LD mice that underwent the SHAM procedure (p > 0.05).

In sum, acute upregulation of inflammatory markers among CA mice housed in dLAN

may contribute to the increased hippocampal neuronal death and mortality observed in

this group.

Microglia are the resident immune cells in CNS and perturbations of the

microenvironment can induce microglial activation, resulting in altered morphology and

secretion of pro-inflammatory mediators (Graeber, 2010; Nimmerjahn, Kirchhoff, &

Helmchen, 2005). Therefore, I hypothesized that microglia increase cytokine expression

24 h after CA in dLAN mice, contributing to the overall elevation in inflammatory status

in the dLAN-CA group. Microglia were extracted from whole brain tissue using a Percoll

gradient 24 h after CA or SHAM procedures (a single night of dLAN or LD lighting

conditions). mRNA was extracted immediately following microglial isolation and pro-

inflammatory cytokine expression was evaluated. IL-1β and TNF-α mRNA expression

were significantly higher in microglia isolated from CA mice as compared to SHAM

mice (p < 0.05; Fig. 5e,f). Furthermore, exposure to a single night of dLAN after CA

elevated microglial IL-1β and IL-6 mRNA relative to LD (p < 0.05; Fig 5f,g). These

results suggest that microglia may be partially responsible for the pro-inflammatory bias

observed among mice that were housed in dLAN after CA.

Page 140: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

124

We also examined whether altered circulating glucocorticoid concentrations could

be contributing to the impaired recovery after CA because elevated corticosterone has

previously been associated with increased CA-induced neuroinflammation and neuronal

death (Neigh et al., 2009). However, altered corticosteroid responses do not appear to

underlie the differences in ischemic outcome between the CA-LD and CA-dLAN groups;

there were no differences in corticosterone concentrations between CA or SHAM groups

at 24h (p > 0.05; Fig. 5h).

Inhibition of selective cytokines ameliorates light induced damage

Although several mechanisms contribute to damage following ischemic injury,

including energetic failure, excitotoxicity, and oxidative stress (reviewed in (Weil,

Norman, DeVries, & Nelson, 2008), manipulation of inflammatory responses are

considered a prime target for prevention of damage. Following ischemic brain damage

both selective targeting of specific cytokines and non-selective (e.g., minocycline)

inhibition of pro-inflammatory cytokines ameliorate damage improving recovery and

behavioral outcomes (Craft & DeVries, 2006; Karelina et al., 2009; Mizushima et al.,

2002; Neigh et al., 2009). Because the results indicate that IL-1β, TNF-α, and IL-6

mRNA expression are greater among CA-dLAN mice compared to CA-LD mice, I

hypothesized that selective inhibition of these pro-inflammatory cytokines would

improve outcome following CA-dLAN. Three days prior to the CA procedure, mice were

implanted with a cannula directed at the lateral ventricle. Two hours following CA, mice

were administered a single 2μL ICV injection of either vehicle (artificial cerebrospinal

fluid; aCSF), mouse IL6 neutralizing antibody (IL6-na), TNF monoclonal antibody

Page 141: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

125

(infliximab; IFX), or recombinant mouse IL-1 receptor antagonist (IL1-ra). Hippocampal

cell death was evaluated using Fluoro-JadeC as described above. IL1-ra and IFX

decreased hippocampal cell death compared to aCSF treatment among CA-dLAN mice (p

< 0.05; Fig. 6b), producing levels of neuronal death that were similar to CA-LD mice

treated with the vehicle (p>0.05). In contrast, treatment with IL6-na did not ameliorate

hippocampal neuronal damage associated with CA-dLAN.

A similar pattern was apparent for microglial activation, which is often used as an

index of neuroinflammation (Amantea, Nappi, Bernardi, Bagetta, & Corasaniti, 2009).

Brain tissue was labeled with Iba-1, an antibody directed against microglia; increased

Iba-1 surface area suggests microglial activation (Donnelly, Gensel, Ankeny, van

Rooijen, & Popovich, 2009). Significantly greater microglial activation in the CA1, CA2,

and CA3 subfields of the hippocampus was apparent among the CA-dLAN mice treated

with the vehicle (aCSF) relative to the CA-LD mice treated with the vehicle (p < 0.05;

Fig. 6c-i). Furthermore, treatment of CA-dLAN mice with IL1-ra or IFX reduced

microglia activation in the CA1, CA2, and CA3 (p < 0.05; Fig. 6c-i) relative to the CA-

dLAN mice treated with vehicle. Iba1 expression in CA-dLAN mice treated with IL1-ra

or IFX did not differ from CA-LD mice treated with the vehicle in any of the

hippocampal subfields quantified. In contrast, CA-dLAN mice treated with IL6-na had

levels of microglial activation in the CA1 that were comparable to CA-dLAN mice

treated with the vehicle, while levels of microglial activation in the CA2 and CA3 regions

were intermediate between vehicle treated CA-dLAN and CA-LD mice. Thus, inhibiting

IL-1 and TNF-α signaling in CA-dLAN mice normalized the microglial and

Page 142: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

126

neurodegenerative responses. Furthermore, the results suggest IL1 and TNFα cytokine

pathways may be more involved than IL6 in inducing hippocampal damage.

Alternative spectra of lighting minimize light induced damage

The circadian system is not equally responsive to all wavelengths of lighting. The

intrinsically photosensitive retinal ganglion cells (ipRGCs) that project to the master

circadian pacemaker in the SCN contain melanopsin and are most responsive to the blue

region of the visible light spectrum ranging from 450 to 485 nm. These wavelengths are

present in broad spectrum white light such as natural sunlight and the majority of indoor

lighting. Longer wavelengths of lighting, such as red light, do not activate ipRGCs and

therefore minimally influence the circadian system (Brainard et al., 2008; Figueiro &

Rea, 2010). Thus, I hypothesized that the circadian system is involved in dLAN induced

damage following CA with ipRGCs communicating the light information. To test this

hypothesis I examined whether mice exposed to red light at night would more closely

resemble the LD phenotype than the dLAN phenotype. Following CA, mice were placed

in LD, dLAN [14 h light (150 lux): 10 h dim light (5 lux; 6500K cool white light-

containing blue wavelengths)] or a bright-dim red light cycle [rLAN; 14 h light (150 lux):

10 h dim red (5 lux; 636 nm)]. Tissue was collected either seven days later for analysis of

neuronal damage (assessed by Fluoro-JadeC) and microglial activation (via Iba-1) or

after 24 h for evaluation of pro-inflammatory cytokine expression.

Unlike full spectrum light at night, dim red light at night did not increase

mortality following CA. There were no differences in mortality between CA mice

exposed to rLAN versus LD (p > 0.05; Fig. 7a). As in the first experiment, CA-dLAN

Page 143: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

127

increased hippocampal cell death compared to CA-LD (p < 0.05; Fig. 7b,d-f). In contrast,

rLAN did not exacerbate ischemic cell death. Hippocampal neuronal damage among CA-

rLAN mice resembled that of CA-LD mice (p > 0.05) and there was significantly less

damage among rLAN mice compared to dLAN mice following CA (p < 0.05; Fig. 7b,d-

f). I similarly replicated the findings that CA-dLAN mice exhibited increased microglia

activation in multiple hippocampal subfields compared to CA-LD conspecifics (p < 0.05;

Fig. 7c,g-i), whereas rLAN did not increase post-ischemic microglia activation relative to

CA-LD (p > 0.05; Fig. 7c,g-i).

Because the previous results indicate that light at night affects recovery by

altering acute changes in the inflammatory response I evaluated hippocampal pro-

inflammatory cytokine expression 24 h after CA and a single night of dark, dim white

light or dim red light. Again, dLAN mice elevated hippocampal TNFα and IL6

expression compared to LD mice 24 h post-ischemia (p < 0.05; Fig. 7j,l). Red light did

not elevate pro-inflammatory cytokine expression compared to LD controls (p > 0.10);

dLAN mice had significantly elevated TNFα, IL6 and IL1β expression compared to

rLAN mice (p < 0.05; Fig 7j-l). These results indicate that standard indoor lighting could

potentiate neuroinflammation and neuronal damage following CA, whereas alternative

lighting using wavelengths greater than ~600 nm are not likely to produce the same

detrimental biological responses.

Discussion

Cardiovascular disease is the leading causes of death in the US (CDC, 2009). The

survival rate for cardiac arrest is very low, and the majority of patients who survive live

Page 144: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

128

with extensive physical, cognitive, and affective disabilities (Elliott, Rodgers, & Brett,

2011; Keuper, Dieker, Brouwer, & Verheugt, 2007; Lim, Alexander, LaFleche, Schnyer,

& Verfaellie, 2004). However, the results presented in this dissertation indicate that

adjusting environmental lighting could prove to be an inexpensive and effective way to

improve patient outcome in cardiac intensive care units. Because of patient safety

concerns and the need for monitoring, hospital ICU rooms are rarely completely dark and

it is not uncommon for patients to be exposed to bright lights (100 lux) several times per

night (Figure 1; (Dunn, Anderson, & Hill, 2010)). Indeed, even patients whose eyelids

are closed may be affected by the light intrusion (Robinson, Bayliss, & Fielder, 1991).

Here I show that exposing mice to as little as 5 lux of dim light at night after resuscitation

from CA exacerbates neuroinflammation and neuronal damage, and increases short-term

mortality four-fold relative to mice that are maintained in a consistent light-dark cycle.

The effects on neuronal damage and mortality appear to be mediated by increased

neuroinflammation among CA mice exposed to dLAN. Indeed, TNF-α mRNA expression

in the hippocampus is elevated as early as 4 h after exposure to dLAN, and by 24 h the

CA-dLAN group has significantly greater TNF-α, IL-1β, and IL-6 mRNA expression

compared to the CA-LD group (Fig. 2). These pro-inflammatory cytokines are known to

contribute to damage after cerebral ischemia (Betz, Schielke, & Yang, 1996; Hurn et al.,

2007). Thus, early changes in the inflammatory response caused by light at night may

alter the trajectory of recovery resulting in higher mortality and increased neuronal

damage characterized after one week.

Page 145: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

129

Inhibition of TNF-α, IL-1, or IL-6 signaling among CA-dLAN mice produced 7-

day survival rates that approximated or exceeded the survival rate for the CA-LD group

(Fig. 3a), although only treatment with IL-1ra or IFX significantly reduced microglial

activation and neuronal damage relative to the CA-dLAN mice. Thus, two bodies of

evidence point to a role of increased inflammatory responses in mediating elevated

neuronal damage among the CA-dLAN mice: (1) both proinflammatory cytokine gene

expression and neuronal damage were elevated after exposure to dLAN relative to LD

and (2) treatment with IL1ra or IFX prevented the exacerbation of neuronal damage and

microglial activation observed among vehicle treated CA-dLAN mice.

Although pharmacological intervention clearly reduced the detrimental effects of

dLAN on CA-induced microglial activation, neuronal damage, and short-term mortality,

a far simpler approach to improving CA outcome is to modify the physical qualities of

the nighttime light to prevent increased neuroinflammation. For example, night time red

light of the same illuminance as the dim white light did not exacerbate CA-induced

neuronal damage or microglial activation in mice. Indeed, the CA-rLAN mice did not

differ significantly from CA-LD mice in either of these measures (Fig. 3). These data are

consistent with studies reporting that red light at night does not affect other aspects of

physiology and behavior in humans or other animals to the same extent as broad

spectrum white light that contains blue wavelengths (Figueiro, Wood, Plitnick, & Rea,

2011). The effects of night time light are likely mediated by the suprachiasmatic nucleus

or “master circadian clock”, which receives input from melanopsin containing ipRGCs in

the retina. The ipRCGs are activated by blue light (~480nm, found in outdoor and most

Page 146: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

130

indoor lighting, especially fluorescent lights), but are unaffected by long wavelength

light, such as red light. The minimal influence of red light compared to white light on CA

recovery suggests that light recognition by the circadian system is involved in dim white

light induced exacerbation of CA damage. Importantly, exposure to dim light at night is

an equally potent facilitator of inflammation in diurnal rodents (Fonken, Haim, & Nelson,

2011).

In sum, the mouse data presented here suggest that exposure to light at night, a

common occurrence in hospital rooms, increases short-term mortality and compromises

recovery from cerebral ischemia by exacerbating neuroinflammation. Using red lights at

night in hospital rooms or having patients wear goggles that filter lower wave length light

could be inexpensive solutions that allow visibility without priming the immune system

of the patients. If the effects of white light at night are replicated in cardiovascular

patients, then these results could have important implications for the design of lighting in

clinical settings and could apply to a broad number of conditions and medical procedures

that involve ischemia and inflammation, such as stroke, cardiovascular artery bypass

graft, sickle cell disease, sleep apnea, and organ transplant.

Page 147: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

131

Figures

Figure 7.1. Ambient lighting in cardiac intensive care unit patient rooms.

Page 148: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

132

0

10

20

30

40

50

60

70

80

90

100

110

120

130

140

CASHAM

Me

an

Art

eri

al B

loo

dP

res

su

re (

mm

Hg

)

20.0

22.5

25.0

27.5

30.0

32.5

35.0

37.5

40.0

42.5

Te

mp

ora

lis

te

mp

era

ture

C

-3 -2 -1 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 1620.0

22.5

25.0

27.5

30.0

32.5

35.0

37.5

40.0

42.5

Time (min)

Co

re t

em

pe

ratu

re C

Figure 7.2. Cardiac arrest and cardiopulmonary resuscitation surgical parameters.

Mean arterial blood pressure, temporalis temperature, and core body temperature across

the CA/CPR procedure.

Page 149: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

133

Figure 7.3. Dim light at night impairs cardiac arrest recovery.

(A) dLAN increases short-term mortality following CA in a rodent model. (B-F) dLAN

exacerbates CA induced hippocampal cell death as indicated by Fluoro-JadeC staining.

Representative Fluoro-JadeC stained sections from the CA1 of (C) LD-SH (D) dLAN-SH

(E) LD-CA and (F) dLAN-CA mice one week following the CA procedure.

Page 150: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

134

0 6 12 18 240

100

200

300

400

500

600

700

800 LD

dLAN

Time after surgery

Co

rtic

oste

ron

e

co

ncen

trati

on

s (

ng

/mL

)

Figure 7.4. Corticosterone concentrations in the 24 h following cardiac arrest.

Page 151: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

135

Figure 7.5. Cytokine expression is elevated in the hippocampus and microglia of mice

exposed to dim light at night following cardiac arrest.

Page 152: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

136

Hippocampal (A) TNFα, (B) IL1β, and (C) IL6 gene expression are upregulated 24 h

following CA and placement in dLAN. (D) TNF-α expression is elevated as early as 6

hours post-CA and only 4 hours of dLAN. Microglial (E) TNFα, (F) IL1β, and (G) IL6

gene expression are also altered 24 h following CA and placement in dLAN. (h) Serum

corticosterone concentrations are unaffected by light at night.

Page 153: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

137

Figure 7.6. Selective inhibition of specific cytokines attenuates inflammation and

neuronal cell death following cardiac arrest and exposure to dim light at night.

(A) Percent survival following CA and treatment with different cytokine inhibitors. (B)

Neuronal damage in the hippocampus as indicated by Fluoro-JadeC staining. (C)

Proportional area of Iba1 staining in the CA1. Representative photomicrographs from the

CA1 of mice treated with (D) LD-Veh (E) dLAN-Veh (F) dLAN-IL1ra (G) dLAN-IL6ab

(H) dLAN-IFX. (I) Proportional area of Iba1 staining throughout the hippocampus.

Page 154: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

138

Figure 7.7. Manipulation of lighting wavelength minimizes light at night induced

damage following CA.

(A) Percent survival following CA and placement in LD, dLAN, or rLAN. (B) Neuronal

damage in the hippocampus as indicated by Fluoro-JadeC staining. (C) Proportional area

Page 155: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

139

of Iba1 staining in the CA1. Representative photomicrographs of Flourojade staining

from the CA1 of (D) LD (E) dLAN and (F) rLAN mice. Representative

photomicrographs of Iba1 staining from the CA1 of (G) LD (H) dLAN and (I) rLAN

mice. (J) Proportional area of Iba1 staining throughout the hippocampus. Compared to

LD controls (K) TNFα, (L) IL1β, and (M) IL6 gene expression are elevated 24 h

following CA and placement in dLAN but not rLAN (*dLAN significantly differs from

both rLAN and LD, #dLAN

Page 156: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

140

CHAPTER 8

DIM LIGHT AT NIGHT ELEVATES INFLAMMATORY RESPONSES IN NILE

GRASS RATS, A DIURNAL RODENT

Most organisms possess an endogenous biological clock that is synchronized by a

very reliable exogenous cue: the daily cycle of light and dark produced by the rotation of

the Earth about its axis. This biological clock is adaptive as it helps to maintain both daily

and seasonal rhythms that allow animals to anticipate changes in the external

environment (Hut & Beersma, 2011). The natural light-dark cycle, however, is now

disrupted for many humans and nonhuman animals. With the advent of electric lights,

light exposure is no longer limited to the natural pattern. Instead of aligning the circadian

system with a stable cyclical factor, individuals currently experience a variety of lighting

schedules. This divergence from the natural environment is not without repercussions.

Disruptive lighting affects many physiological and behavioral functions (Fonken &

Nelson, 2011; Navara & Nelson, 2007). For example, individuals exposed to altered light

cycles are at increased risk for heart disease (Ha & Park, 2005), cancer (Davis & Mirick,

2006; Kloog, Portnov, Rennert, & Haim, 2011; Schernhammer et al., 2001), sleep

disturbances (Deboer, Detari, & Meijer, 2007; Kohyama, 2009), circadian rhythm

Page 157: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

141

dysfunctions (Borugian, Gallagher, Friesen, Switzer, & Aronson, 2005), disrupted

rhythmicity of neuroendocrine function (Claustrat, Valatx, Harthe, & Brun, 2008;

Persengiev, Kanchev, & Vezenkova, 1991), mood disorders (Dumont & Beaulieu, 2007;

Fonken et al., 2009), metabolic dysfunction (Fonken et al., 2010; Reiter, Tan, Korkmaz,

& Ma, 2011), and reproductive dysfunction (Fiske, 1941; Thomas, Oommen, &

Ashadevi, 2001). One common factor for many of these pathologies is altered immune

function.

Circadian timing in mammals is organized by a hierarchy of oscillating tissues, at

the top of which are the suprachiasmatic nuclei (SCN) of the hypothalamus (Reppert &

Weaver, 2002). Light information is the primary entraining cue for this master circadian

clock. Light travels from the external environment through the intrinsically

photosensitive retinal ganglion cells (ipRGCs) to the SCN. The SCN then influences

downstream “slave” oscillators via the autonomic nervous system and control of the

sleep-wake cycle. SCN driven sympathetic innervation of the pineal gland regulates the

release of melatonin; nighttime sympathetic neural stimulation leads to the production of

melatonin from its precursor serotonin in the pineal gland. This nocturnal melatonin

signal provides time of day information to cells throughout the body and is the most

reliable peripheral marker of central clock activity (Blask, 2009). Nocturnal lighting, if

sufficiently bright, disrupts the synthesis of melatonin (Brainard et al., 1985; Brainard,

Richardson, Petterborg, & Reiter, 1982; Dauchy et al., 2010). Importantly, modulation of

both the circadian system and melatonin alters immunological measures.

Page 158: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

142

Multiple immune markers such as interleukin 2 (IL2), IL10, IL6, IL1β, tumor

necrosis factor alpha (TNFα), interferon gamma (IFNγ), and chemokine receptor 2

(CCR2) are expressed in a circadian pattern (Lundkvist, Robertson, Mhlanga, Rottenberg,

& Kristensson, 1998; Young et al., 1995). Disruption of the circadian system through jet-

lag, genetic mutations, or light exposure, changes the normal pattern of immune

parameters. For example, mice with a loss of function mutation in the clock gene Period

2 (Per2) have irregular production of IL10 and IFNγ in response to lipopolysaccharide

(LPS) injection (Arjona & Sarkar, 2006). Mice deficient in Bmal1, another critical clock

component, show early signs of aging such as sarcopenia, cataracts, organ shrinkage and

elevated reactive oxygen species (ROS) in the kidney and spleen. The lifespans of Bmal1

deficient mice are also reduced (Kondratov, Kondratova, Gorbacheva, Vykhovanets, &

Antoch, 2006). Cry 1 and 2 knockout mice have exacerbated cytokine and joint swelling

after arthritic induction (Hashiramoto et al., 2010). Furthermore, using a phase advancing

chronic jet-lag (CJL) protocol causes persistent hypothermia and reduced survival

following LPS administration in mice, and macrophages extracted from these mice have

increased cytokine response to LPS (Castanon-Cervantes et al., 2010).

The nighttime increase in pineal melatonin production and secretion correlates

with reduced innate immune responses (Markus, Ferreira, Fernandes, & Cecon, 2007).

NF-κB, a pleiotropic transcription factor involved in the regulation of genes encoding for

immune related enzymes, displays daily variation (Cecon, Fernandes, Pinato, Ferreira, &

Markus, 2010; Z. Chen, Gardi, Kushikata, Fang, & Krueger, 1999). NF-κB induces

multiple pro-inflammatory cytokines, inducible nitric oxide synthase (iNOS), and

Page 159: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

143

cyclooxygenase-2 (COX-2). Melatonin blocks NF-κB nuclear translocation in leukocytes

and endothelial cells, suppressing immune gene transcription activity (Gilad et al., 1998;

Tamura, Cecon, Monteiro, Silva, & Markus, 2009). Darkness induced suppression in NF-

κB transcription is blocked by propranolol (a drug that inhibits melatonin production in

addition to altering catecholaminergic function). In vitro work has further supported the

role of melatonin in NF-κB signaling as melatonin reduces NF-κB in cultured pineal

glands (Cecon, Fernandes, Pinato, Ferreira, & Markus, 2010). In a rat model of diabetes

increased expression of NF-κB and pro-inflammatory cytokines including TNFα and IL6

were reduced with melatonin treatment (Negi, Kumar, & Sharma, 2011). Melatonin also

impairs capacity for rolling and adhesion among leukocytes (Lotufo, Lopes, Dubocovich,

Farsky, & Markus, 2001). Furthermore, communication between the circadian and

immune system is bidirectional with multiple studies characterizing an immune-pineal

axis (Couto-Moraes, Palermo-Neto, & Markus, 2009; Lopes, Mariano, & Markus, 2001;

Markus, Ferreira, Fernandes, & Cecon, 2007). For example, administration of LPS

inhibits norephinephrine (NE) induced N-acetylserotonin (NAS) production resulting in

decreased nocturnal melatonin production (da Silveira Cruz-Machado et al., 2010).

Taken together, these studies suggest that nighttime lighting may affect immune

function through disruption of the circadian system and suppression of melatonin.

Recently our lab demonstrated that Siberian hamsters (Phodopus sungorus) exposed to

dim nighttime lighting (dLAN) reduce immunological capabilities (Bedrosian, Fonken,

Walton, & Nelson, 2011a). Housing female Siberian hamsters under dLAN for 4 weeks

reduces delayed-type hypersensitivity responses, decreases bactericidal capacity of blood,

Page 160: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

144

and prevents fever-associated reductions in locomotor activity. Because Siberian

hamsters are nocturnal, and in the laboratory are typically exposed to light at night during

their active period, I asked whether light at night would evoke similar responses in a

diurnal species. Thus, I investigated the effects of nighttime light exposure on immune

parameters in Nile grass rats (Arvicanthis niloticus), a diurnal rodent species. Male grass

rats were exposed to either a standard light- dark cycle or dim light at night for 3 weeks

and then tested for delayed type hypersensitivity, bacteria killing capacity, and antibody

production.

Methods

Animals

Male grass rats (Arvicanthis niloticus) used in this study were bred in the Nelson

lab colony at the Ohio State University from a wild stock obtained by Dr. Laura Smale,

Michigan State University, from the Masai Mara reserve in Kenya. Grass rats were bred

under a light-dark (LD) cycle (14:10 light (~150 lux) /dark (0 lux); lights illuminated at

7:00 Eastern Standard Time [EST]) and all animals were provided food (ProLab RMH

2000, LabDiet) and water ad libitum. Experimental grass rats were weaned between 21

and 24 days of age and housed with same sex siblings in polypropylene cages (40 cm x

20 cm x 20 cm) with straw bedding. Colony rooms were maintained at a temperature of

20 ± 4º C and a relative humidity of 50% ± 10%. At approximately 3 months of age grass

rats were singly housed, randomly assigned a number, and were either maintained in LD

(n=9) or placed in dLAN (n=9; 14:10 light (~150 lux)/ dim (~5 lux); lights illuminated at

Page 161: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

145

7:00 EST) for the remainder of the study. Immunological testing began after 3 weeks in

the lighting conditions.

Delayed-type hypersensitivity (DTH)

DTH is a cell mediated response that provides information about the primary

immune reaction to invading pathogens. After 3 weeks in lighting conditions grass rats

were assessed for DTH response to the chemical antigen 2,4-dinitro-1-fluorobenzene

(DNFB; Sigma, St. Louis, MO). Grass rats were individually brought into a procedure

room between 14:00 and 15:00 EST, lightly anesthetized with isoflurane vapors,

weighed, and a blood sample was collected from the retro-orbital sinus for use in the

bacteria killing and corticosterone assays (see below). Following blood collection a 1 x 2

cm patch of fur was shaved on the dorsum and 25 μl of DNFB in a 0.5% solution (wt/vol)

of 4:1 acetone to olive oil (prepared fresh daily) was applied to the dorsal skin in the

same location on two consecutive days. To obtain a baseline measurement, both right and

left pinna were measured during sensitization with a constant loading dial micrometer

(Mitutoyo, America Corp., Aurora, IL, USA). Grass rats were then left undisturbed for 1

week, after which they were again anesthetized, pinna thickness measured, and

challenged on the surface of the right pinna with 20 µl of 0.2% (wt/vol) DNFB in 4:1

acetone to olive oil. Left pinna was treated with the vehicle solution and both pinnae were

measured every 24 h for 7 days. Pinna swelling values obtained on each day were

expressed as a percentage of baseline thickness. All measurements occurred between

14:00 and 15:00 EST. DTH is an in vivo measure of cell mediated immune responses that

is characterized by swelling at the site of DNFB challenge. Swelling of the right pinna is

Page 162: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

146

due to infiltration of leukocytes into the epidermis and dermis (Vadas, Miller, Gamble, &

Whitelaw, 1975). This immune measure was previously validated; pinna swelling is

positively correlated to the intensity of the immune reaction (Phanuphak, Moorhead, &

Claman, 1974).

Keyhole limpet hemocyanin (KLH)

Two weeks following the conclusion of DTH measures humoral immune function

was assessed by injecting grass rats with 140 µg KLH suspended in 0.2 mL sterile saline.

KLH is a respiratory protein from the giant keyhole limpet (Megathura crenulata) that

produces a robust antigenic response without inducing fever or a long-term inflammatory

response. KLH production has not previously been assessed in grass rats, therefore, this

dose was determined based on other arvicoline rodents and resulted in similar patterns of

plasma immunoglobulin G (IgG) production (Klein & Nelson, 1999; Weil, Martin, &

Nelson, 2006). Blood was drawn from the retro-orbital sinus at the time of injection and

5, 10, and 15 days post injection in order to capture peak immunoglobulin production

(Demas, Chefer, Talan, & Nelson, 1997). All blood sampling occurred between 14:00

and 15:00 EST. Blood samples were centrifuged at 4° C for 30 min at 3.3 g and plasma

was pulled off and stored in microcentrifuge tubes at -80° C.

KLH ELISA

Plasma concentrations of anti-KLH IgG were determined using an enzyme-linked

immunosorbent assay (ELISA) as previously described (Demas, Chefer, Talan, &

Nelson, 1997). Plates were coated overnight with dialyzed KLH antigen, washed, and

blocked the subsequent night with a milk blocking buffer. Plates were then washed and

Page 163: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

147

150 µL of plasma diluted 1:80 in PBS+Tween was added in duplicated to the wells.

Following a 3 h incubation plates were again washed and incubated for 1.5 h with a

secondary antibody (alkaline phosphatase-conjugated anti-mouse IgG). Plates were then

treated with the enzyme substrate (p-nitrophenyl phosphate) before determining the

optical density of each well with a plate reader (Benchmark Microplate Reader, Biorad,

Hercules, CA) and 405 nm wavelength filter. To minimize intra-assay variability optical

density was averaged over duplicate wells and expressed as a percentage of the plate-

positive control value for statistical analyses.

Bactericidal capacity of blood plasma

Blood samples collected directly prior to DTH sensitization were used in this

assay. Blood was centrifuged at 3300 g for 30 min at 4° C and plasma was pulled off and

stored at -80° C. Plasma samples were diluted 1:20 in L-glutamine CO2-independent

media (Gibco, Carlsbad, CA, USA). A standard number of colony-forming units (CFUs)

of Escherichia coli (Epower 0483E7, Fisher Scientific) were added to each sample and

samples were incubated for 30min at 37° C. Using sterile techniques 75 μL of each

sample was plated in duplicate on tryptic soy agar plate. Two positive controls of diluted

bacteria alone and two negative controls of CO2-independent media were also plated.

Plates were inverted and incubated overnight and total CFUs were counted and expressed

as a percent of the positive control.

Radioimmunoassay Procedure (RIA)

Blood samples were collected for RIA of corticosterone from the retro-orbital

sinus of grass rats on the first day of DTH sensitization. Blood samples were centrifuged

Page 164: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

148

at 4° C for 30 min at 3.3g and plasma was pulled off and stored in sealable polypropylene

microcentrifuge tubes at -80° C until assayed. Total plasma corticosterone concentrations

were determined in duplicate using an ICN Diagnostics 125

I double antibody kit (Costa

Mesa, CA, USA). The high and low limits of detectability of the assay were 1200 and 3

ng/ml, respectively. The intra-assay coefficient of variation was 11%. All procedures

followed the manufacturer guidelines.

Activity analyses

A separate cohort of grass rats were implanted intraperitoneally (i.p.; under sterile

conditions) with telemeters (PDT-4000; Minimitter, Bend, OR) while under isoflurane

anesthesia. Surgical wounds were treated topically with Betadine (Sigma Chemical, St.

Louis, MO) to discourage infection, and grass rats were injected (i.p.) with buprenorphine

(0.1mg/kg; Sigma Chemical) in sterile saline to alleviate pain during recovery. Following

surgery, grass rats were placed in a clean cage, which was placed on a receiver

(Minimitter) connected to a computer. Receivers collated emitted body temperature and

movement activity frequencies continuously over 30 min intervals and converted them to

raw data based on pre-programmed calibration curves for each transmitter.

Data analyses

Tissue weight, body mass, and total surviving CFU‟s were compared between

lighting conditions using a one-way analysis of variance (ANOVA). Delayed-type

hypersensitivity swelling and anti-KLH were analyzed with a repeated measures

ANOVA with lighting condition as the between-subject factor and time as the within

subject variable. Following a significant result on repeated measures ANOVA, single

Page 165: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

149

time point comparisons were made. Plasma corticosterone concentrations were analyzed

using a one tailed t- test based on a priori hypotheses (Keppel & Wickens, 2004). Fourier

analysis was used to determine whether locomotor activity was rhythmic and followed 24

h periodicity using Clocklab software from Actimetrics (Wilmette, IL). Grass rats were

considered rhythmic when the highest peak occurred at ~1 cycle per day with an absolute

power of at least 0.005 mV/Hz as previously described (Kriegsfeld et al., 2008). FFT

power values for 0.083 cycles per day were compared between lighting conditions by one

way ANOVA. Percentage of daytime activity and total daily activity were also analyzed

by one way ANOVA. Nonlinear regression analysis was used in GraphPad Prism

software (v. 4 La Jolla, CA). In all cases, differences between group means and

correlation coefficients were considered statistically significant if p ≤ 0.05.

Results

Reproductive and somatic measures

There were no differences in body, reproductive tissue, adrenal, spleen, or thymus

mass between groups (p > 0.05; Table 1).

Circadian activity pattern

There were no differences between grass rats housed with dLAN and those in LD

with respect to circadian pattern in activity, total daily activity, or percentage of activity

occurring during the light phase (p>0.05; Fig. 1).

Plasma corticosterone concentrations

Samples for corticosterone analysis were collected directly prior to DTH

sensitization. Plasma corticosterone concentrations were elevated among grass grass rats

Page 166: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

150

exposed to dLAN as compared to those exposed to dark nights (t16 = 1.815; p ≤ 0.05; Fig.

2a).

DTH swelling responses

One dLAN grass rat did not develop a swelling response and was excluded from

comparisons. The remaining grass rats all exhibited robust swelling of the right pinna

over the measurement period (F5,75 = 19.58, p ≤ 0.05; Fig. 2b). There was a main effect of

lighting condition, such that grass rats exposed to dLAN had significantly elevated

swelling of the right pinna as compared to conspecifics housed with dark nights (F5,75 =

4.30, p ≤ 0.05). dLAN grass rats had greater swelling on days 2 and 3 post challenge than

grass rats exposed to dark nights (F1,15 = 5.62 and 5.19, respectively, p ≤ 0.05).

Furthermore, there was a positive association between plasma corticosterone

concentrations and pinna swelling on days 2, 3, and 5 post challenge (r2 = 0.327, 0.219,

0.391, p ≤ 0.05; day 2 shown; Fig. 2c).

KLH antibody production

Two grass rats, one per group, produced no antibody in response to the KLH

injection and were excluded from the analyses. Grass rats significantly elevated antibody

production following KLH injection (F3,42 = 105.60, p ≤ 0.05). There was an interaction

between light condition and time (F3,42 = 5.21, p ≤ 0.05), such that grass rats exposed to

dLAN increased anti-KLH IgG production 10 and 15 days following injection (F1,14 =

13.11, 7.67, p ≤ 0.05; Fig. 3a).

Bacteria colony killing

Page 167: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

151

Grass rats housed with dLAN decreased the percentage of surviving CFUs

compared to grass rats housed in standard LD conditions (F1,16 = 8.155, p ≤ 0.05; Fig.

3b).

Discussion

Light at night influenced immune function in male Nile grass rats. Rats exposed

to dLAN elevated bacteriacidal capacity, and humoral and cell-mediated immune

responses. Increased immune activity occurred independently of overt changes in

circadian locomotor activity. dLAN grass rats increased plasma corticosterone

concentrations during the active phase after three weeks in lighting conditions which may

have affected immunological measures. These results contrast with previously reported

results in nocturnal rodents undergoing similar experimental nighttime light exposure.

The results suggest that male and female, as well as diurnal and nocturnal, rodents may

respond differently to the effects of nighttime light exposure.

Rats exposed to dLAN increased pinna swelling compared to LD rats in response

to the antigen DNFB. DTH is a measure of cell-mediated immunity that demonstrates

primary somatic immune response to an invading pathogen. Pinna swelling is caused by

increased infiltration of macrophages and lymphocytes into the epidermis and dermis

(Vadas, Miller, Gamble, & Whitelaw, 1975) and has previously been positively

correlated to the intensity of the immune reaction (Phanuphak, Moorhead, & Claman,

1974). Functionally, elevated swelling in DTH testing is indicative of increased

resistance to viruses, bacteria, and fungi (Bilbo et al., 2002).

Page 168: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

152

Increased pinna swelling in dLAN grass rats contrasts with previously reported

results in which swelling was suppressed in Siberian hamsters exposed to light at night

(Bedrosian, Fonken, Walton, & Nelson, 2011a). DTH responses may vary over the

course of the day. In the previous study, Siberian hamsters underwent DTH testing during

the light phase when they are generally inactive. In the present study DTH testing also

occurred during the light phase, however, grass rats are diurnal and active at this time.

Other factors that vary in a circadian pattern such as immune cells and hormones may

contribute to the equivocal t-cell mediated results (Bollinger, Bollinger, Naujoks, Lange,

& Solbach, 2010). For example, exposure to dLAN elevates glucocorticoid

concentrations in grass rats but not Siberian hamsters (Bedrosian, Fonken, Walton, Haim,

& Nelson, 2011a). Furthermore, glucocorticoid concentration can alter diurnal rhythms in

T-cell mediated inflammatory responses, an effect which may be partially mediated by

melatonin. Adrenalectomy abolishes the diurnal rhythm in BCG inflammation; however,

the rhythm can be recovered with exogenous administration of melatonin (Lopes,

Mariano, & Markus, 2001). In the previous study in Siberian hamsters (6) DTH was also

assessed in female Siberian hamsters as compared to male grass rats in the current

experiments. Although female Siberian hamsters were ovariectomized, varying levels of

sex steroids may have contributed to divergent results (Kanda & Watanabe, 2005).

Overall, assessing DTH responses during the active phase in this study is more

ecologically relevant because it is more likely that grass rats would encounter a pathogen

while awake and interacting with the external environment.

Page 169: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

153

Exposure to light at night suppresses melatonin production in both rodents and

humans (Brainard et al., 1985; Brainard, Richardson, Petterborg, & Reiter, 1982; Dauchy

et al., 2010). Even very low levels of light exposure can alter melatonin concentrations in

rodents (Evans, Elliott, & Gorman, 2007). Although I did not measure melatonin

concentrations in this study, it is likely that they were decreased with exposure to light at

night. It is unlikely, however, that changes in DTH response reflect suppression of

melatonin among dLAN rats. Melatonin is positively associated with DTH responses in

diurnal and nocturnal rodent species (Drazen & Nelson, 2001; Haldar & Singh, 2001).

Glucocorticoids both increase and suppress DTH responses depending on the type

and duration of the stressor (Dhabhar, 2002; Dhabhar & McEwen, 1999). Typically,

acute stress increases DTH, whereas chronic stress suppresses DTH reactions, indicating

that changes in DTH are in some cases related to altered glucocorticoid concentrations

(Dhabhar & McEwen, 1999). During acute stress, blood leukocytes redistribute to the

skin, mucosal linings, lung, liver, and lymph nodes, key areas in preventing breaching of

immune defenses. In this study the positive association between corticosterone

concentrations and pinna swelling suggests that the two may be related. It is possible that

a long-term stressor such as light at night induces a state of functional glucocorticoid

resistance. Previous work has demonstrated that psychosocial stressors can cause splenic

macrophages to become resistant to the suppressive effects of glucocorticoid hormones

(Avitsur, Stark, Dhabhar, Padgett, & Sheridan, 2002; Avitsur, Stark, & Sheridan, 2001;

M. T. Bailey, Avitsur, Engler, Padgett, & Sheridan, 2004). Alternatively, the elevation in

glucocorticoids among dLAN rats may be sufficiently low to exert an atypical effect on

Page 170: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

154

DTH swelling. Glucocorticoids exert a U-shaped influence on multiple factors; for

example, basal or low stress levels of corticosterone enhance glucose utilization,

hippocampal synaptic excitability, hippocampal-dependent learning, and cerebral

perfusion rate whereas higher physiological levels of corticosterone exert opposite effects

(Sapolsky, 2004). Moreover, melatonin and glucocorticoids interact in modulating

immunological processes. Acute and chronic stress increase plasma melatonin

concentrations in rodents (Couto-Moraes, Palermo-Neto, & Markus, 2009; Dagnino-

Subiabre et al., 2006). This may be a compensatory mechanism as melatonin protects

against some effects of chronic stress (Brotto, Gorzalka, & LaMarre, 2001). This

interaction has important implications for this study because it suggests that

glucocorticoids may increase melatonin concentrations partially compensating for the

light induced suppression in melatonin.

Rats exposed to dLAN enhanced antibody production following injection with

KLH. Anti-KLH production is a general indicator of B cell activity. Previous studies

have reported no differences in anti-KLH production or enhanced production in

melatonin treated rodents (Demas, Chefer, Talan, & Nelson, 1997; Drazen & Nelson,

2001). Furthermore, primary and secondary antibody production is decreased in mice

treated with propranolol (Maestroni, Conti, & Pierpaoli, 1986). Thus, differences in

antibody production between dLAN and LD rats may be independent of putative changes

in melatonin. Elevated concentrations of glucocorticoids following social defeat are

associated with enhanced lymphocyte release of IFNγ and IL6. However, changes in anti-

KLH production are not apparent (Merlot, Moze, Dantzer, & Neveu, 2004). Furthermore,

Page 171: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

155

in another model of social defeat elevated glucocorticoid concentrations were associated

with impairment in antiviral immunological memory (de Groot, Boersma, Scholten, &

Koolhaas, 2002). Again this indicates that changes in anti-KLH production may occur

independently of changes in corticosteroids. Alternatively, light at night may be an

atypical stressor affecting the glucocorticoid system in a different manner than other

chronic stressors.

Bactericidal capacity was enhanced in dLAN rats as compared to those housed in

standard lighting conditions. Bacteria killing is a low-cost nonspecific immune response

predominately mediated by plasma proteins (L. B. Martin, 2nd, Weil, & Nelson, 2007).

Plasma bactericidal capacity increases with immune challenge and represents an

enhanced ability to clear a bacterial infection (Weinrauch, Abad, Liang, Lowry, & Weiss,

1998). Elevated glucocorticoids concentrations have been associated with enhanced

bactericidal capacity in spleen cells from mice that underwent social disruption stress (M.

T. Bailey, Engler, Powell, Padgett, & Sheridan, 2007). Melatonin, however, generally

enhances bactericidal capacity (Terron et al., 2009).

Overall, nighttime light exposure increased immunocompetence in grass rats

exposed to light at night. Light at night may exert its effects through changes in the

hypothalamic-pituitary adrenal axis as corticosteroid concentrations were elevated among

dLAN rats. It is widely accepted that stress affects immune responses, with chronic

stress generally exerting an immunosuppressive effect. The role of glucocorticoids in

immunological processes are complex however (Sorrells, Caso, Munhoz, & Sapolsky,

2009); depending on the type of stress, glucocorticoids can have opposite effects. Light at

Page 172: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

156

night may also disrupt circadian processes leading to changes in immune function.

Although gross changes in locomotor activity were not apparent, disruption of the

circadian system at the molecular level may have occurred. Previous work has indicated

disruption of clock function can lead to exacerbated immune activity (Arjona & Sarkar,

2006; Castanon-Cervantes et al., 2010; Hashiramoto et al., 2010). Melatonin can also be

directly produced by immune cells (Pontes, Cardoso, Carneiro-Sampaio, & Markus,

2006). Importantly, this study did not evaluate all aspects of immune function; it is

possible that other arms of the immune system could be differentially affected by

nighttime light exposure (L. B. Martin, 2nd, Weil, & Nelson, 2007).

Enhanced immune responses are not always favorable. Immunological processes

are energetically costly and a delegation of energy to immune responses in unnecessary

situations can reduce fitness (L. B. Martin, Weil, & Nelson, 2008). Moreover,

enhancement of the immune response in cases such as allergic asthma is detrimental

(Wills-Karp, 1999). Because of the contrasting results obtained in this study and another

study conducted in a nocturnal rodent species, further research on the effects of light at

night on immune function are warranted. Nighttime light exposure is currently

experienced by over 99% of the population of the US and Europe. Light pollution has

significant ecological consequences for animals living in urban and suburban areas and

may contribute to species loss (Navara & Nelson, 2007). It is important to understand the

physiological implications of this exposure in order to work toward preventing

ecologically related complications.

Page 173: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

157

Tables

LD dLAN

Body mass (g) 93.02 +/- 6.47 93.55 +/- 6.66

Epididymides (mg) 447 +/- 19 474 +/- 29

Epididymal fat (mg) 2108 +/- 315 2230 +/- 370

Testes (mg) 1779 +/- 67 1713 +/- 48

Adrenal (mg) 66 +/- 8 62 +/- 6

Thymus (mg) 112 +/- 20 110 +/- 21

Spleen (mg) 142 +/- 9 148 +/- 12

Table 8.1. Tissue masses in Nile grass rats exposed to dimly lit or dark nights

Page 174: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

158

Figures

LD dLAN0

100

200

300

400

500

600

700

*C

ort

ico

ste

ron

e

co

ncen

trati

on

(n

g/m

L)

0 1 2 3 4 5

0

20

40

60

80

100

120 LD

dLAN* *

Days after challenge

% S

we

llin

g

0 200 400 600 800 10000

50

100

150

200

r2=0.327p=0.013

Corticosterone (ng/mL)

Pin

na s

well

ing

A B

C

Figure 8.1. Nile grass rats exposed to dimly nights elevate corticosterone concentrations

and delayed-type hypersensitivity swelling responses.

Rats exposed to dim light at night (A) elevated plasma corticosterone concentrations

compared to rats with dark night. (B) Dim light rats exhibited increased pinna swelling 2

and 3 days following challenge with the antigen 2,4-dinitro-1-fluorobenzene. (C) There

was an association between peak pinna swelling and corticosterone concentrations 2 days

after challenge. Data are presented as mean ± SEM. *p ≤ 0.05.

Page 175: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

159

0 5 10 15

0

25

50

75

100

125

LDdLAN

**

Days Post Innoculation

An

ti-K

LH

Ig

G

LD dLAN0

5

10

15

20

25

30

35

40

45

*

Bacte

rial

co

lon

ies

(% o

f p

osit

ive c

on

tro

l)

A B

Figure 8.2. Rats exposed to light at night enhance humoral immune function and plasma

bactericidal capacity.

(A) Anti-KLH IgG responses were elevated in dLAN rats 10 and 15 days following

innoculation. (B) Plasma obtained from dLAN grass rats caused fewer surviving bacterial

colonies. Data are presented as mean ± SEM. *p ≤ 0.05.

Page 176: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

160

Figure 8.3. Grass rats exposed to dark and dimly lit nights have comparable activity.

Representative actograph of a grass rat housed in (A) dark or (B) dimly lit nights.

Page 177: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

161

CHAPTER 9

DIM NIGHTTIME LIGHT IMPAIRS COGNITION AND PROVOKES

DEPRESSIVE-LIKE RESPONSES IN A DIURNAL RODENT

Biological rhythms are highly adaptive, aligning individuals to daily fluctuations

in the external environment, as well as synchronizing internal homeostatic processes. The

master mammalian circadian clock is located in the suprachiasmatic nuclei (SCN) and

regulates timing of subordinate oscillators throughout the central nervous system and

periphery. External lighting is important in synchronizing the circadian system and

maintaining daily temporal organization. Prior to the widespread adoption of electric

lighting, individuals‟ biological clocks were entrained to a consistent pattern of light and

dark; in contrast, modern light exists in several temporal patterns. Moreover, shift-work,

trans-meridian travel, and inconsistent sleep schedules have rapidly increased during the

past century. Because the change in nighttime lighting has occurred so rapidly in terms of

evolutionary history, it is likely that significant physiological and ecological

perturbations have resulted. For example, disruption of the circadian system results in

adverse health conditions such as heart disease (Ha & Park, 2005), cancer (Davis &

Page 178: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

162

Mirick, 2006; Schernhammer et al., 2001), and metabolic dysfunction (Reiter, Tan,

Korkmaz, & Ma, 2011).

Circadian disruption and light at night are implicated in impaired cognition. Rats

housed in constant illumination perform poorly in the Morris water maze (A. Fujioka et

al., 2011; Ling et al., 2009; Ma et al., 2007). Constant light causes tau

hyperphosphorylation, increased expression of endoplasmic reticulum (ER) stress-related

proteins, thinner synapses, and increased superoxide dismutase and monoamine oxidase

(Ling et al., 2009). Similarly, rats housed in constant light display impaired spatial

learning in the Morris water maze with accompanying changes in long-term depression in

the CA1 area of the hippocampus (Ma et al., 2007). Constant light also impairs learning

and memory in mice, which may be related to decreased neurogenesis (A. Fujioka et al.,

2011). Furthermore, mice undergoing experimental jet lag decrease neurogenesis and

have prolonged deficits in learning and memory as evaluated in a conditioned place

preference task (Gibson, Wang, Tjho, Khattar, & Kriegsfeld, 2010).

In addition to influencing learning and memory, circadian disruption changes

mood (Monteleone, Martiadis, & Maj, 2010). Seasonal lighting, abnormalities in the

circadian clock (Benedetti et al., 2008), and sleep disorders are associated with

depression (Bunney & Bunney, 2000). Constant light alters anxiety and depressive-like

behaviors in mice (Fonken et al., 2009; Martynhak et al., 2011). Furthermore, Siberian

hamsters exposed to dim light during the dark phase increase depressive-like responses

and have reduced spine density in the CA1 area of the hippocampus (Bedrosian, Fonken,

Walton, Haim, & Nelson, 2011a).

Page 179: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

163

In all of these studies, only nocturnal rodents were used. Both circadian influences

on behavior and masking effects of light are very different in nocturnal and diurnal

species. Thus, in the present experiment, I examined behavioral and brain responses of

diurnal male Nile grass rats to dim light at night (dLAN).

Methods

Animals

Male grass rats (Arvicanthis niloticus) used in this study were bred at The Ohio

State University from a wild stock obtained from LS. Grass rats were bred under a

standard light-dark (LD) cycle (14:10 light (~150 lux) /dark (0 lux)). All animals were

provided food (ProLab RMH 2000, LabDiet) and filtered tap water ad libitum.

Experimental grass rats were weaned between 21 and 24 days of age and housed with

same sex siblings in polypropylene cages (40 cm x 20 cm x 20 cm) with straw bedding.

Colony rooms were maintained at a temperature of 20 ± 4º C and a relative humidity of

50% ± 10%.

At 10 weeks of age grass rats were singly housed, randomly assigned a number,

and either maintained in LD or placed in dLAN (14:10 light (~150 lux)/ dim (~5 lux)).

Blood samples were collected at Zeitgeber Time (ZT) 6 via retro-orbital bleed after two

weeks for corticosterone analysis and one week later grass rats underwent behavioral

testing to assess cognitive and affective behaviors. Testing occurred in the following

order: Barnes maze, sucrose anhedonia, and forced swim test. The sucrose anhedonia test

occurred between ZT 8-13; all other tests were conducted between ZT 1-ZT 6.

Page 180: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

164

Retro orbital blood samples (~0.20 ml) were collected from grass rats

anesthetized with isoflurane vapors for RIA of corticosterone concentrations, prior to the

onset of behavioral testing. Blood samples were centrifuged at 4°C for 30 min at 3.3 g

and plasma aliquots were aspirated and stored in sealable polypropylene microcentrifuge

tubes at -80°C until assayed for corticosterone concentrations using a radioimmunoassay

(RIA). Total plasma corticosterone concentrations for grass rats were determined in

duplicate in an assay using an ICN Diagnostics 125

I double antibody kit (Costa Mesa, CA,

USA). The high and low limits of detectability of the assay were ~1000 and 5 ng/ml,

respectively. All procedures followed those described by the manufacturer guidelines.

Behavioral testing

Barnes maze. The Barnes maze is a brightly lit arena with 18 evenly spaced

holes, one leading to dark box and the others blocked off with black inserts (Sunyer,

2007). On the first day animals were acclimated to the maze; a bright light and loud fan

were turned on as they were guided from the center of the maze to the target hole. After

entering, the bright light and fan were turned off and the grass rat were left undisturbed

for 30 sec. Animals then underwent four days of training consisting of 3, 90 sec trials

separated by 10 min intervals in the home cage. One day after the last training trial

animals were given a 60 sec probe trial in which the escape box was blocked off. Latency

to find the target hole and number of errors were scored during all trials. Sucrose

Anhedonia. Consumption of a 2% sucrose solution between ZT 8 and ZT 13, was

recorded in all grass rats to measure sucrose anhedonia (Willner, Muscat, & Papp, 1992).

Before presentation of the sucrose solution, grass rats were administered water in

Page 181: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

165

modified water bottles for three consecutive days, to control for novelty of the bottles.

The bottles were weighed before and after the 5 h sample time; the next day animals were

provided a choice between a 2% sucrose solution and water. Sucrose consumption was

normalized to water consumption. Forced Swim Test. To assess depressive-like

responses, grass rats were placed in ~17 cm water (22 ±1°C), within an opaque,

cylindrical tank (diameter = 24 cm, height = 53 cm). Swimming behavior was videotaped

for 5 min and scored by a condition-blind observer with the Observer software (Noldus

Corp, Leesburg, VA, USA). Latency to float and time spent floating served as dependent

measures; both are used in rodents, including grass rats, to assess depressive-like

response (Ashkenazy-Frolinger, Kronfeld-Schor, Juetten, & Einat, 2010; Porsolt, Bertin,

& Jalfre, 1977).

Hippocampal Morphology

Grass rats were killed between ZT 3 and ZT 5. and brains were removed, and

processed for Golgi impregnation using the FD Rapid GolgiStain™ Kit (FD

NeuroTechnologies Inc., Ellicott City, MD) according to the manufacturer„s instructions.

Brains were sliced at 100μm, thaw mounted onto gelatin coated slides counterstained

with cresyl violet (Sigma), dehydrated, and coverslipped. Brains were assessed for

hippocampal cell morphology and spine density in the dentate gyrus (DG), CA1, and

CA3 using a Nikon E800 brightfield microscope. Tracings were done with Neurolucida

software (MicroBrightField, Burlington, VT, USA) at a magnification of 200× for

neuronal morphology and 1000× for spine density. Six representative neurons were

selected per area, per animal. Whole cell traces were analyzed using NeuroExplorer

Page 182: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

166

software, for cell body size and perimeter, and dendritic length (MicroBrightField,

Burlington, VT). Sholl analysis defines dendritic complexity by the number of dendritic

branch points at fixed intervals from the cell bodies (Sholl, 1956) and was conducted on

apical and basilar dendrites. From each neuron >20 µm were selected in the apical and

basilar areas respectively (except in the DG where granule cells lack bidirectional

projections). All spine segments selected were at least 50 µm distal to the cell body.

Spine density (spines per 1 μm) was calculated for each trace and averaged per cell, per

area, and per animal.

Statistical Analyses

Comparisons for behavior analyses and hormone concentrations were conducted

using a one-way ANOVA. Neuronal characteristics and spine densities were averaged

per animal and then analyzed using a one-way ANOVA. Sholl analyses were also

averaged per animal and then analyzed using a repeated measures ANOVA with lighting

condition as the between subject factor and distance from the cell body as the with-in

subject factor. For each Barnes maze session (1–4) latencies and error rates, respectively,

were averaged per session for each grass rat; data were subject to repeated-measures

ANOVA (lighting condition as the between subject factor and session as the with-in

subject factor). Fourier analysis was used to determine whether locomotor activity was

rhythmic and followed 24 h periodicity using Clocklab software from Actimetrics

(Wilmette, IL). Grass rats were considered rhythmic when the highest peak occurred at

~1 cycle per day with an absolute power of at least 0.005 mV/Hz as previously described

(Kriegsfeld et al., 2008). FFT power values for 0.083 cycles per day were compared

Page 183: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

167

between lighting conditions by one way ANOVA. Percentage of daytime activity and

total daily activity were also analyzed by one way ANOVA. The above statistical

analyses were conducted with StatView software (v. 5.0.1, Cary, NC). Nonlinear

regression analysis was used in GraphPad Prism software (v. 4 La Jolla, CA). In all

cases, differences between group means were considered statistically significant if p ≤

0.05.

Results

Somatic Measures

There were no differences in body or reproductive tissue mass (p > 0.05; data not

shown).

Learning and Memory

Grass rats exposed to dLAN decreased learning abilities in the Barnes maze.

dLAN grass rats had an increased latency to reach the target hole and a higher error rate

as compared to grass rats housed under dark nights. There was a main effect of lighting

condition, such that dLAN increased latency to reach the target hole over the course of

training trials (F1,42 = 6.064, p < 0.05; Fig. 1A). Post-hoc analysis revealed dLAN

increased latency to reach the target hole on days 2 and 4 of the training trials (p < 0.05).

Furthermore, there was a main effect of lighting condition with respect to error rate; grass

rats housed under dLAN increased errors over the course of training trials (F1,42 = 8.184,

p < 0.05; Fig. 1B). On day two of the training trials dLAN grass rats showed increased

error rate compared to grass rats housed under standard LD conditions (p < 0.05).

Page 184: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

168

Twenty-four hours after the final training session, memory retention was assessed

with a probe trial. During the probe trial the target box was removed and behavior on the

maze was recorded for 60 sec. dLAN grass rats spent a lower percentage of time

investigating the target hole compared to LD grass rats; these data indicate decreased

memory retention (F1,14 = 7.084, p < 0.05; Fig. 1C). Our lab has previously found no

differences in spontaneous locomotor activity between grass rats housed under dLAN and

LD in a 5 min brightly lit open field task (unpublished observations). This indicates

Barnes maze differences are due to changes in learning and memory and not differences

in motivation when exposed to a brightly lit open space.

Affective Responses

Depressive-like responses were evaluated using a sucrose anhedonia and forced

swim task. Grass rats exposed to dLAN increased depressive-like responses in the

sucrose anhedonia test. One grass rat was excluded from analyses because of a leaky

water bottle. dLAN grass rats reduced consumption of a sucrose solution demonstrating

an anhedonic-like response (F1,15 = 4.711, p < 0.05; Fig. 2A).

Grass rats exposed to dLAN also increased behavioral despair in the forced swim

task. dLAN grass rats reduced latency to first float indicating they more rapidly reach a

state of behavioral despair (F1,16 = 4.774; p<0.05; Fig. 2B). No differences, however,

were observed between groups with respect to float duration which is the primary

depressive-like response evaluated in the forced swim test (p > 0.10).

Corticosterone Concentrations

Page 185: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

169

Grass rats displayed elevated corticosterone concentrations 2 weeks after

placement in dLAN compared to LD (F1,16 = 4.521, p < 0.05; Fig. 2D).

Hippocampal Neuronal Morphology

Exposure to dLAN is associated with changes in neuronal morphology in the CA1

and DG regions of the hippocampus. Rats housed under dLAN reduced dendritic length

in the DG and CA1 basilar dendrites (F1,11 = 4.875, 7.357 respectively, p < 0.05; Fig 3).

Furthermore, there was a positive association between dendritic length in the dentate

gyrus and sucrose consumption in the sucrose anhedonia test (r = 0.455, p = .016; Fig,

3C). Groups did not differ with respect to spine density, cell body area, or cell body

perimeter in any area (Table 1).

Discussion

This study investigated the effect of dim nighttime light exposure on depressive-

like responses and learning and memory in Nile grass rats, a diurnal rodent. Here I show

that exposing grass rats to light at night impaired their spatial learning and memory as

evaluated by the Barnes maze (Sunyer, 2007). Animals were trained to find a target hole

on the maze over the course of 4 days and then evaluated in a probe trial. During the

training trials, dLAN impaired performance as compared to LD. dLAN increased the

latency for grass rats to reach the target hole and increased the number of visits to

incorrect holes. Spatial memory was similarly impaired by dLAN. Twenty-four hours

after the final training session, memory retention was assessed with a probe trial. dLAN

decreased the percentage of time spent investigating the target hole, indicating decreased

memory retention.

Page 186: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

170

These results confirm and extend previous findings (A. Fujioka et al., 2011; Ling

et al., 2009; Ma et al., 2007) indicating that housing nocturnal rodents under light at night

impairs spatial learning and memory. The results demonstrate that nighttime light

exposure also impairs spatial learning and memory in diurnal rodents. Furthermore,

previous studies have used 24 h lighting of the same intensity while in this study grass

rats were exposed to a distinctly darker phase. Grass rats exsposed to dark or dimly lit

nights display maintain a diurnal activity pattern and have equivalent levels of total daily

locomotor activity and similar locomotor activity rhythms (Fonken, Haim, & Nelson,

2011; McElhinny, Smale, & Holekamp, 1997). Constant lighting conditions used in

previous studies can result in an arrhythmic activity rhythm (Cambras, Castejon, & Diez-

Noguera, 2011). These results demonstrate that cognitive impairments occur in animals

with light at night in the absence of disruption in locomotor activity rhythm.

The effects of light at night on other cognitive functions remain unspecified

(Castro et al., 2005). It is possible that nighttime light exposure specifically targets

hippocampal dependent learning and memory through disruption of circadian processes

(Ruby et al., 2008). Alternatively, dLAN may represent a mild chronic stressor producing

deficits in learning and memory via reduced neurogenesis, changes in hippocampal

architecture, or both processes (A. Fujioka et al., 2011; Gould & Gross, 2002; McEwen

& Sapolsky, 1995). Studies in nocturnal rodents have reported glucocorticoid

concentrations to be both elevated or unaffected by nighttime light exposure (Abilio,

Freitas, Dolnikoff, Castrucci, & Frussa-Filho, 1999; Fonken et al., 2009; Fonken et al.,

2010; Van der Meer, Van Loo, & Baumans, 2004). Grass rats housed under dLAN

Page 187: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

171

elevated plasma corticosterone concentrations. dLAN may be a stronger stressor to

diurnal as compared to nocturnal rodents. Circulating glucocorticoid concentrations are

predictive of hippocampal atrophy and memory deficits in both humans and rodents

(Bodnoff et al., 1995; Lupien et al., 1998). Because corticosterone concentrations were

only measured at a single time point however, conclusions drawn from the results must

be constrained.

Grass rats exposed to dLAN increased depressive-like responses in a sucrose

preference test. dLAN reduced consumption of a sucrose solution in the sucrose

anhedonia test. LD rats consumed a higher percentage of sucrose than water, whereas

dLAN grass rats showed no preference for the sucrose solution. This implies that the

sucrose solution had diminished hedonic valence for grass rats exposed to dLAN which

models a key feature of human depression (Willner, Muscat, & Papp, 1992). Results in

the forced swim test were equivocal. Increased floating time in the forced swim test is

considered “behavioral despair” because rodents putatively stop searching for an escape

mechanism (Porsolt, Bertin, & Jalfre, 1977). There were no differences in total floating

time in the forced swim test between LD and dLAN grass rats, but grass rat housed under

dLAN reduced latency to first float. The forced swim test has not been extensively used

in grass rats, although one study reported increases in floating duration in grass rats

housed in short photoperiods (Ashkenazy-Frolinger, Kronfeld-Schor, Juetten, & Einat,

2010). Grass rats are poor swimmers, which suggests the forced swim test may not be a

reliable behavioral measure (Duplantier & Ba, 2001).

Page 188: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

172

The depressive-like phenotype of the dLAN grass rats is consistent with my

predictions based on depressive disorders related to both stress (Willner, 1997) and

circadian dysfunction (Turek, 2007). Furthermore, nighttime light exposure increases

depressive-like responses in nocturnal rodents (Bedrosian, Fonken, Walton, Haim, &

Nelson, 2011b). Light at night may increase depressive-like responses through changes in

hippocampal circuitry. dLAN decreased dendritic length in DG and CA1 basilar

dendrites. Furthermore, there was a positive association between dendritic length in the

dentate gyrus and sucrose consumption in the sucrose anhedonia test. The hippocampus

is a critical structure in the pathophysiology of depressive disorders. Depression is

associated with changes in glucocorticoids and hippocampal atrophy (Sapolsky, 2000;

Sheline, Wang, Gado, Csernansky, & Vannier, 1996). Moreover, changes in hippocampal

morphology are associated with chronic stress and depressive-like responses in rodents

(Hajszan et al., 2009; Magarinos et al., 2011; Magarinos, McEwen, Flugge, & Fuchs,

1996).

In summary, these results suggest that exposure to dim nighttime lighting can

alter hippocampal neuronal morphology, impair learning and memory, and increase

depressive-like responses in a diurnal rodent. Opportunities for exposure to light at night

have rapidly increased during the past century. The present results suggest that this

exposure may have accompanying maladaptive effects. Finding an appropriate model to

test whether changes in environmental lighting are related to mood disorders is critical.

Many animal models are potentially confounded by the use of nocturnal rodents that may

not experience the same form of disruption as diurnal animals when exposed to light at

Page 189: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

173

night. The responses to light at night seen in the present study are comparable to those

described in previous reports using nocturnal rodents (e.g., Bedrosian et al., 2011a),

which increases confidence that nocturnal rodents are appropriate subjects of study of this

issue. In addition, these results raise questions about the use of rodent vivaria that have

windows in the doors of animal rooms and continuous lighting in the halls.

Page 190: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

174

Tables

Table 9.1. Characteristics of hippocampal neurons in grass rats exposed to dark or dimly

lit nights.

Characteristics of neurons in the CA1, CA3, and DG of the hippocampus of grass rats

exposed to dark or dimly lit nights. Represented as mean ± SEM micrometers; AD =

apical dendrite; BD = basilar dendrite.

Page 191: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

175

Figures

1 2 3 40

25

50

75

100LD

dLAN

*

*

Days

La

ten

cy

(s

ec)

1 2 3 40

5

10

15

20

25

*

Days

# E

rro

rs

LD dLAN0

5

10

15

20

25

30

35

*

% v

isit

s t

o c

orr

ec

t h

ole

A B

C

Figure 9.1. Grass rats exposed to dim light at night show impairments in learning and

memory.

Dim light at night impaired spatial learning and memory in the Barnes maze. (A) Latency

to reach the target hole in the Barnes maze by day (average of 3 trials). (B) Number of

visits to false holes by session. (C) Relative visits to the target hole versus false holes

during the probe trial.

Page 192: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

176

500 650 800 950 1100 1250 14000

20

40

60

80

100

R=0.45p=0.02

Dendritic length

% S

ucro

se c

on

su

med

LD dLAN0

25

50

75

100

*%

Su

cro

se c

on

su

med

LD dLAN0

10

20

30

40

50

60

*

Late

ncy t

o f

loat

(sec)

A B

C

LD dLAN0

100

200

300

400

500

600*

Co

rtic

oste

ron

e

co

ncen

trati

on

(n

g/m

L)

D

Figure 9.2. Exposure to dim light at night increases depressive-like responses in grass

rats.

(A) Amount of sucrose solution versus total liquid consumed in a sucrose anhedonia task.

(B) Latency to first float the forced swim test. (C) Correlation between percentage of

sucrose consumed and dendritic length in the dentate gyrus of the hippocampus. (D)

Corticosterone concentrations after two weeks in lighting conditions. Data are expressed

as mean ± standard error of the mean (SEM). *p<0.05 between groups.

Page 193: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

177

Figure 9.3. Exposure to dim light at night alters hippocampal morphology in grass rats.

(A) Total dendritic length was reduced in CA1 basilar dendrites and (B) in the dentate

gyrus (DG). (C) Representative tracing from the CA1 of an LD grass rat (black)

compared to a dLAN grass rat (red). (D) Representative tracing from the DG of an LD

grass rat (black) and dLAN grass rat (red). Data are expressed as mean ± SEM. *p<0.05

between groups.

Page 194: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

178

CONCLUSIONS

Exposure to light at night is generally considered an innocuous environmental

manipulation. The perceived harmlessness of nocturnal illumination likely persists

because the invention and widespread adoption of the electric lighting occurred prior to

an understanding of circadian biology. The goal of this dissertation was to determine the

physiological consequence of exposing nocturnal (Swiss Webster mice) and diurnal (Nile

grass rats) rodents to ecologically relevant levels of dim (~5 lux) light at night. In this

dissertation, I demonstrate that exposure to light at night has significant metabolic,

immunological, and behavioral repercussions.

Summary

The global increase in the prevalence of obesity and metabolic disorders coincides

with the increase of exposure to light at night and shift work. Circadian regulation of

energy homeostasis is controlled by an endogenous biological clock that is synchronized

by light information (Reppert & Weaver, 2002). To promote optimal adaptive

functioning, the circadian clock prepares individuals for predictable events such as food

availability and sleep, and disruption of clock function causes circadian and metabolic

disturbances (Green, Takahashi, & Bass, 2008). To determine whether a causal

relationship exists between nighttime light exposure and obesity I examined the effects of

exposure to light at night on body mass in male mice (Chapter 2). Mice exposed to dim

Page 195: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

179

light at night or continuous lighting increase body mass and impair glucose processing as

compared to mice exposed to dark nights. Changes in body mass occur independently of

changes in total daily food intake or activity. However, mice attenuate the daily pattern in

food intake, eating more during the rest phase than mice exposed to dark nights. Blocking

daytime food intake in mice with light at night prevents weight gain. Furthermore,

placing mice back in dark nights following exposure to 4 weeks of dim light at night

prevents changes in glucose tolerance and partially restores body mass (Chapter 3).

In industrialized societies, exposure to light at night and more typical obesogenic

factors such as a high fat diet and sedentary lifestyle, often occur in tandem and may

contribute to the increasing obesity epidemic. Thus, I examined the effects of a high fat

diet and exercise availability on dim light at night associated weight gain in Chapters 4

and 5, respectively. Dim light at night exaggerates weight gain in mice fed a high fat diet

(Chapter 4). Moreover, both high fat feeding and dim light at night increase daytime

food intake and elevate peripheral inflammation. Exposure to a high fat diet but not dim

light at night elevates hypothalamic inflammation suggesting that these two factors may

work through different physiological mechanisms to affect weight regulation. As

anticipated, access to a functional running wheel prevents body mass gain in mice

exposed to dim light at night (Chapter 5). Voluntary exercise suppresses weight gain in

mice exposed to dimly lit nights without rescuing changes to the circadian system;

increases in daytime food intake induced by exposure to dim light at night are not

diminished by exercise availability. Furthermore, exposure to light at night disrupts

wheel running behavior in a subset of mice exposed to dim light at night.

Page 196: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

180

Because changes in circadian clock mechanisms affect metabolism (Kornmann,

Schaad, Bujard, Takahashi, & Schibler, 2007; Lamia, Storch, & Weitz, 2008; Marcheva

et al., 2010; Oishi et al., 2006; Paschos et al., 2012; Turek et al., 2005) and exposure to

light at night affects circadian rhythms (Albrecht, Sun, Eichele, & Lee, 1997; Schwartz,

Tavakoli-Nezhad, Lambert, Weaver, & de la Iglesia, 2012; Shigeyoshi et al., 1997), in

Chapter 6, I investigated the effects of exposure to light at night on both central and

peripheral core circadian clock mechanisms. Mice exposed to dim light at night attenuate

core circadian clock rhythms in the SCN at both the gene and protein levels. Circadian

clock rhythms are also perturbed in the liver of mice exposed to dimly lit as compared to

dark nights.

In addition to affecting metabolism, circadian system disruptions are linked to

alterations in immune function. For example, many of the pathologies associated with

exposure to light at night such as cancer (Stevens, 2009b), obesity (X. S. Wang,

Armstrong, Cairns, Key, & Travis, 2011), and mood disorders (Driesen, Jansen, Kant,

Mohren, & van Amelsvoort, 2010) involve changes in inflammation. Moreover, multiple

immune related parameters show circadian oscillations (Lange, Dimitrov, & Born, 2010)

(Arjona & Sarkar, 2006; Narasimamurthy et al., 2012; A. C. Silver, Arjona, Walker, &

Fikrig, 2012) (Arjona & Sarkar, 2005; Keller et al., 2009; A. C. Silver, Arjona, Hughes,

Nitabach, & Fikrig, 2012). Therefore, Chapter 7 of this dissertation addressed the effects

of exposure to light at night on recovery from cardiac arrest in mice. Following either a

cardiac arrest or a sham procedure, mice were exposed to dark or dimly lit nights. Mice

exposed to dimly illuminated as compared to dark nights elevate mortality in the week

Page 197: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

181

following cardiac arrest. Furthermore, mice exposed to dim light at night post-cardiac

arrest increase neuroinflammation and hippocampal cell death. Dim light at night likely

affects cardiac arrest recovery by elevating inflammation; selective inhibition of IL-1β or

TNFα ameliorates the effects of dim light at night on cardiac arrest recovery.

Additionally, restricting the wavelength of the nighttime light exposure to ~640 nm

eliminates the detrimental effects of nighttime light exposure on cardiac arrest outcome.

The experiments described in the first 7 chapters of this dissertation were

conducted using nocturnal rodents in order to examine the effects of nighttime light

exposure without disrupting sleep. However, in diurnal species many hormones and

immune parameters vary with secretion patterns 180º out of phase to those of nocturnal

rodents. Furthermore, light can have very different effects on activity and masking in

diurnal versus nocturnal rodents. Thus, in the final two chapters of this dissertation

(Chapters 8 and 9) I investigated the effects of nighttime light on behavior, hippocampal

connectivity, and immune related parameters, in grass rats. Rats exposed to dim light at

night increase delayed-type hypersensitivity pinna swelling which is consistent with

enhanced cell-mediated immune function. Similarly, rats exposed to dimly lit as

compared to dark nights increase antibody production following inoculation with keyhole

lymphocyte hemocyanin (KLH) and increase bactericidal capacity. In contrast to

nocturnal rodents, daytime corticosterone concentrations are elevated in grass rats

exposed to nighttime lighting.

In addition to influencing immune function, three behavioral effects are apparent

in grass rats exposed to dim light at night: (1) decreased preference for a sucrose solution,

Page 198: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

182

(2) increased latency to float in a forced swim test, and (3) impaired learning and memory

in the Barnes maze. Light at night also reduces dendritic length in dentate gyrus and

basilar CA1 dendrites. In agreement with findings in nocturnal rodents (Fonken et al.,

2010), nighttime light exposure does not disrupt the pattern of circadian locomotor

activity in grass rats.

Mechanisms

There are several mechanisms by which nighttime light exposure can affect

physiology. In this dissertation, I specifically focused on how light at night may influence

metabolism through disruption of the circadian system. Exposure to unnatural light at

night can also affect physiological processes through melatonin suppression, alterations

in glucocorticoids, and changes in sleep architecture.

Melatonin is an endogenously synthesized molecule that is secreted by the pineal

gland during the night in both nocturnal and diurnal mammals (Reiter, 1991). Melatonin

secretion is potently inhibited by exposure to sufficient levels and durations of nighttime

lighting in both rodents and humans (Brainard, Rollag, & Hanifin, 1997). For example,

exposing humans to one hour of 45 lux of nighttime light exposure decreases plasma

melatonin by ~60% (Brainard, Richardson, Petterborg, & Reiter, 1982). Similar to

circadian clock gene disruption, suppressing melatonin secretion is associated with

increased risk for developing cancer (Blask, 2009; Blask et al., 2005), obesity (Mantele et

al., 2012; Tan, Manchester, Fuentes-Broto, Paredes, & Reiter, 2011), and mood disorders

(Srinivasan, De Berardis, Shillcutt, & Brzezinski, 2012). Although there is compelling

evidence that suppression of melatonin secretion can contribute to weight gain, I

Page 199: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

183

specifically did not focus on melatonin for several reasons: (1) pineal melatonin

suppression requires high and sustained levels of nighttime light exposure (Brainard,

Rollag, & Hanifin, 1997), (2) nighttime light exposure below the threshold for melatonin

suppression is associated with changes in metabolism (Obayashi et al., 2013), and (3)

multiple strains of laboratory mice that lack pineal melatonin demonstrate changes in

metabolism with nighttime light exposure (Coomans et al., 2013; Fonken et al., 2010).

Diurnal variations in other hormones may be disrupted by exposure to light at

night. Glucocorticoids are of particular interest in the context of nighttime light exposure

because (1) light at night may be interpreted as a stressor (Ma et al., 2007) and (2)

glucocorticoids are a primary output of, and feedback signal for the circadian system

(Kiessling, Eichele, & Oster, 2010; Sage et al., 2004). Importantly, I did not focus on

glucocorticoids as a primary mechanism for physiological changes associated with

nighttime light exposure because exposure to light at night appears to affect

glucocorticoid secretion in only a subset of mammals. For example, Nile grass rats show

elevations in serum corticosterone concentrations after chronic exposure to dim light at

night, but mice do not (Fonken, Haim, & Nelson, 2011; Fonken et al., 2010). Despite

disparate effects of light at night on glucocorticoids, Swiss Webster mice and Nile grass

rats show similar changes in physiology and behavior following exposure to light at night

(Fonken et al., 2009; Fonken, Kitsmiller, Smale, & Nelson, 2012; Fonken & Nelson,

2013). This suggests that alterations in glucocorticoids are not critical for dim light at

night-associated changes.

Page 200: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

184

Finally, exposure to light at night may affect metabolism through disturbing sleep

architecture. Sleep disruptions can profoundly affect physiology and contribute to

multiple pathological conditions including metabolic syndrome (Mullington, Haack,

Toth, Serrador, & Meier-Ewert, 2009; Spiegel, Tasali, Leproult, & Van Cauter, 2009). In

order to dissociated the effects of nighttime light exposure from sleep disruptions I

specifically worked with nocturnal Swiss Webster mice in the majority of the studies in

this dissertation. These studies indicate that light at night causes changes in metabolism

independently of sleep disruptions. However, due to the synergistic effects of light at

night and disrupted sleep on metabolism, future research should address whether these

variables act through similar mechanisms to affect metabolism.

Overall, it is difficult to tease apart which mechanism is the greatest contributing

force to the negative effects of nighttime light exposure. Melatonin suppression, circadian

disruption, changes in the HPA axis, and sleep disturbances likely all contribute to the

deleterious outcomes associated with exposure to light at night.

Implications

Over 99% of the population in the US and Europe is exposed to light at night

(Cinzano, Falchi, & Elvidge, 2001). In addition to experiencing urban light pollution,

many people bring light into their homes by turning on electric lights after sunset,

watching TV late into the night, or using computers directly prior to bed. It is estimated

that two-thirds of the population experience this form of “social jet lag” (Roenneberg,

Allebrandt, Merrow, & Vetter, 2012). Moreover, shift workers make up approximately

20% of the populations and are exposed to high and prolonged levels of light at night.

Page 201: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

185

It is also important to note that exposure to light at night is not just a human issue.

Many plant and animal species are affected by nighttime light exposure, as lighting from

infrastructure strays into the atmosphere creating a general nighttime glow termed “light

pollution” (Navara & Nelson, 2007). The results presented in this dissertation suggest

that unnatural exposure to light at night may have significant ecological implications.

Indeed, exposure to light at night is known to affect mating (Dominoni, Quetting, &

Partecke, 2013; Kempenaers, Borgstrom, Loes, Schlicht, & Valcu, 2010), foraging and

predation (Davies, Bennie, & Gaston, 2012; Dwyer, Bearhop, Campbell, & Bryant, 2012;

Rydell, 1992; Stone, Jones, & Harris, 2009), and migration in multiple species (Z. Wang

et al., 2011).

Prevention and Interventions

Preventing the general population from excessive exposure to light at night can be

achieved with relatively low-cost manipulations, such as using curtains to block out street

lights, turning off hallway lights, and removing all light sources, including televisions

and computers, from bedrooms. Furthermore, adhering to a consistent schedule and

avoiding rapid phase shifts can minimizing “social jet lag” (Roenneberg, Allebrandt,

Merrow, & Vetter, 2012).

In shift-working populations, avoiding phase shifts and nighttime light exposure

is often unavoidable. However, not all nocturnal illumination equally affects the circadian

system. The intrinsically photosensitive retinal ganglion cells that project to the SCN are

most responsive to the blue region of the visible spectrum (ranging from 450 to 485 nm)

with longer wavelengths of lighting minimally influencing the circadian system (Brainard

Page 202: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

186

et al., 1985; Brainard, Richardson, Petterborg, & Reiter, 1982). Manipulation of lighting

wavelength may prove effective in blocking out light-induced physiological changes. To

that end, ongoing research is investigating the effectiveness of preventing exposure to

blue wavelength light with specially designed goggles and light fixtures.

Future Directions

Future studies should confirm the importance of the circadian system in mediating

light at night-associated physiological changes. This could be accomplished by several

means including (1) examining the effects of exposure to red light at night, (2) using a

mouse strain lacking ipRGCs such as Opn4aDTA/aDTA

mice (LeGates et al., 2012), or (3) by

ablating ipRGCs with saporin conjugated to a melanopsin polyclonal antibody (Ingham,

Gunhan, Fuller, & Fuller, 2009). Additionally, these studies should particularly focus on

potential interventions to limit deleterious changes associated with exposure to light at

night.

Because the research presented in this dissertation may have important

implications for human health, future studies should directly investigate the effects of

exposure to light at night on humans. First, conducting comparative work in the Amish,

who are minimally exposed to light at night, may provide insight into some of the

unintended consequences of exposure to light at night. For example, Amish have reduced

risk compared to the general population for developing breast cancer and metabolic

syndrome, two conditions associated with exposure to light at night (Fonken & Nelson,

2011). Second, the effects of nighttime light exposure on patient recovery should be

evaluated. This could be achieved by introducing altered light fixtures into hospital and

Page 203: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

187

retroactively monitoring variables such as length of patients stay. Finally, the effects of

dim light at night on human physiology should be investigated in a controlled laboratory

setting. Exposing people to a single night, or week, of dim light at night and monitoring

various circadian and metabolic outputs may highlight the mechanism by which exposure

to electril light at night influences human health

Page 204: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

188

REFERENCES

Abilio, V. C., Freitas, F. M., Dolnikoff, M. S., Castrucci, A. M., & Frussa-Filho, R.

(1999). Effects of continuous exposure to light on behavioral dopaminergic

supersensitivity. Biol Psychiatry, 45(12), 1622-1629.

Akhtar, R. A., Reddy, A. B., Maywood, E. S., Clayton, J. D., King, V. M., Smith, A. G.,

et al. (2002). Circadian cycling of the mouse liver transcriptome, as revealed by

cDNA microarray, is driven by the suprachiasmatic nucleus. Curr Biol, 12(7),

540-550.

Albrecht, U. (2002). Invited review: regulation of mammalian circadian clock genes. J

Appl Physiol, 92(3), 1348-1355.

Albrecht, U., Sun, Z. S., Eichele, G., & Lee, C. C. (1997). A differential response of two

putative mammalian circadian regulators, mper1 and mper2, to light. Cell, 91(7),

1055-1064.

Allison, K. C., Lundgren, J. D., O'Reardon, J. P., Geliebter, A., Gluck, M. E., Vinai, P., et

al. (2010). Proposed diagnostic criteria for night eating syndrome. Int J Eat

Disord, 43(3), 241-247.

Amantea, D., Nappi, G., Bernardi, G., Bagetta, G., & Corasaniti, M. T. (2009). Post-

ischemic brain damage: pathophysiology and role of inflammatory mediators.

Febs J, 276(1), 13-26.

Page 205: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

189

Ando, H., Kumazaki, M., Motosugi, Y., Ushijima, K., Maekawa, T., Ishikawa, E., et al.

(2011). Impairment of peripheral circadian clocks precedes metabolic

abnormalities in ob/ob mice. Endocrinology, 152(4), 1347-1354.

Ando, H., Yanagihara, H., Hayashi, Y., Obi, Y., Tsuruoka, S., Takamura, T., et al.

(2005). Rhythmic messenger ribonucleic acid expression of clock genes and

adipocytokines in mouse visceral adipose tissue. Endocrinology, 146(12), 5631-

5636.

Antipatis, V. J., & Gill, T. P. (2001). Obesity as a Global Problem: John Wiley & Sons

Ltd.

Arble, D. M., Bass, J., Laposky, A. D., Vitaterna, M. H., & Turek, F. W. (2009).

Circadian timing of food intake contributes to weight gain. Obesity (Silver

Spring), 17(11), 2100-2102.

Arble, D. M., Vitaterna, M. H., & Turek, F. W. (2011). Rhythmic leptin is required for

weight gain from circadian desynchronized feeding in the mouse. PLoS One, 6(9),

e25079.

Arjona, A., & Sarkar, D. K. (2005). Circadian oscillations of clock genes, cytolytic

factors, and cytokines in rat NK cells. J Immunol, 174(12), 7618-7624.

Arjona, A., & Sarkar, D. K. (2006). The circadian gene mPer2 regulates the daily rhythm

of IFN-gamma. J Interferon Cytokine Res, 26(9), 645-649.

Ashkenazy-Frolinger, T., Kronfeld-Schor, N., Juetten, J., & Einat, H. (2010). It is

darkness and not light: Depression-like behaviors of diurnal unstriped Nile grass

Page 206: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

190

rats maintained under a short photoperiod schedule. J Neurosci Methods, 186(2),

165-170.

Atalayer, D., & Rowland, N. E. (2011). Comparison of voluntary and foraging running

wheel activity on food demand in mice. Physiol Behav, 102(1), 22-29.

Avitsur, R., Stark, J. L., Dhabhar, F. S., Padgett, D. A., & Sheridan, J. F. (2002). Social

disruption-induced glucocorticoid resistance: kinetics and site specificity. J

Neuroimmunol, 124(1-2), 54-61.

Avitsur, R., Stark, J. L., & Sheridan, J. F. (2001). Social stress induces glucocorticoid

resistance in subordinate animals. Horm Behav, 39(4), 247-257.

Bae, K., Jin, X., Maywood, E. S., Hastings, M. H., Reppert, S. M., & Weaver, D. R.

(2001). Differential functions of mPer1, mPer2, and mPer3 in the SCN circadian

clock. Neuron, 30(2), 525-536.

Baggs, J. E., & Green, C. B. (2003). Nocturnin, a deadenylase in Xenopus laevis retina: a

mechanism for posttranscriptional control of circadian-related mRNA. Curr Biol,

13(3), 189-198.

Bailey, M. T., Avitsur, R., Engler, H., Padgett, D. A., & Sheridan, J. F. (2004). Physical

defeat reduces the sensitivity of murine splenocytes to the suppressive effects of

corticosterone. Brain Behav Immun, 18(5), 416-424.

Bailey, M. T., Engler, H., Powell, N. D., Padgett, D. A., & Sheridan, J. F. (2007).

Repeated social defeat increases the bactericidal activity of splenic macrophages

through a Toll-like receptor-dependent pathway. Am J Physiol Regul Integr Comp

Physiol, 293(3), R1180-1190.

Page 207: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

191

Bailey, S. L., & Heitkemper, M. M. (2001). Circadian rhythmicity of cortisol and body

temperature: morningness-eveningness effects. Chronobiol Int, 18(2), 249-261.

Balagura, S., & Devenport, L. D. (1970). Feeding patterns of normal and ventromedial

hypothalamic lesioned male and female rats. J Comp Physiol Psychol, 71(3), 357-

364.

Balsalobre, A., Damiola, F., & Schibler, U. (1998). A serum shock induces circadian

gene expression in mammalian tissue culture cells. Cell, 93(6), 929-937.

Barclay, J. L., Husse, J., Bode, B., Naujokat, N., Meyer-Kovac, J., Schmid, S. M., et al.

(2012). Circadian desynchrony promotes metabolic disruption in a mouse model

of shiftwork. PLoS One, 7(5), e37150.

Barnea, M., Madar, Z., & Froy, O. (2009). High-Fat Diet Delays and Fasting Advances

the Circadian Expression of Adiponectin Signaling Components in Mouse Liver.

Endocrinology, 150(1), 161-168.

Baron, K. G., Reid, K. J., Horn, L. V., & Zee, P. C. (2013). Contribution of evening

macronutrient intake to total caloric intake and body mass index. Appetite, 60(1),

246-251.

Baron, K. G., Reid, K. J., Kern, A. S., & Zee, P. C. (2011). Role of sleep timing in caloric

intake and BMI. Obesity (Silver Spring), 19(7), 1374-1381.

Bechtold, D. A., Brown, T. M., Luckman, S. M., & Piggins, H. D. (2008). Metabolic

rhythm abnormalities in mice lacking VIP-VPAC2 signaling. Am J Physiol Regul

Integr Comp Physiol, 294(2), R344-351.

Page 208: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

192

Becker, E. E., & Grinker, J. A. (1977). Meal patterns in the genetically obese Zucker rat.

Physiol Behav, 18(4), 685-692.

Bedrosian, T. A., Fonken, L. K., Walton, J. C., Haim, A., & Nelson, R. J. (2011a). Dim

light at night provokes depression-like behaviors and reduces CA1 dendritic spine

density in female hamsters. Psychoneuroendocrinology, In press.

Bedrosian, T. A., Fonken, L. K., Walton, J. C., Haim, A., & Nelson, R. J. (2011b). Dim

light at night provokes depression-like behaviors and reduces CA1 dendritic spine

density in female hamsters. Psychoneuroendocrinology, 36(7), 1062-1069.

Bedrosian, T. A., Fonken, L. K., Walton, J. C., & Nelson, R. J. (2011a). Chronic

exposure to dim light at night suppresses immune responses in Siberian hamsters.

Biol Lett.

Bedrosian, T. A., Fonken, L. K., Walton, J. C., & Nelson, R. J. (2011b). Chronic

exposure to dim light at night suppresses immune responses in Siberian hamsters.

Biol Lett, 7(3), 468-471.

Bell-Pedersen, D., Cassone, V. M., Earnest, D. J., Golden, S. S., Hardin, P. E., Thomas,

T. L., et al. (2005). Circadian rhythms from multiple oscillators: lessons from

diverse organisms. Nat Rev Genet, 6(7), 544-556.

Benca, R., Duncan, M. J., Frank, E., McClung, C., Nelson, R. J., & Vicentic, A. (2009).

Biological rhythms, higher brain function, and behavior: Gaps, opportunities, and

challenges. Brain Res Rev, 62(1), 57-70.

Page 209: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

193

Benedetti, F., Dallaspezia, S., Colombo, C., Pirovano, A., Marino, E., & Smeraldi, E.

(2008). A length polymorphism in the circadian clock gene Per3 influences age at

onset of bipolar disorder. Neurosci Lett, 445(2), 184-187.

Betz, A. L., Schielke, G. P., & Yang, G. Y. (1996). Interleukin-1 in cerebral ischemia.

Keio J Med, 45(3), 230-237; discussion 238.

Bilbo, S. D., Dhabhar, F. S., Viswanathan, K., Saul, A., Yellon, S. M., & Nelson, R. J.

(2002). Short day lengths augment stress-induced leukocyte trafficking and stress-

induced enhancement of skin immune function. Proc Natl Acad Sci U S A, 99(6),

4067-4072.

Birketvedt, G. S., Florholmen, J., Sundsfjord, J., Osterud, B., Dinges, D., Bilker, W., et

al. (1999). Behavioral and neuroendocrine characteristics of the night-eating

syndrome. Jama, 282(7), 657-663.

Blask, D. E. (2009). Melatonin, sleep disturbance and cancer risk. Sleep Med Rev, 13(4),

257-264.

Blask, D. E., Brainard, G. C., Dauchy, R. T., Hanifin, J. P., Davidson, L. K., Krause, J.

A., et al. (2005). Melatonin-depleted blood from premenopausal women exposed

to light at night stimulates growth of human breast cancer xenografts in nude rats.

Cancer Res, 65(23), 11174-11184.

Bodnoff, S. R., Humphreys, A. G., Lehman, J. C., Diamond, D. M., Rose, G. M., &

Meaney, M. J. (1995). Enduring effects of chronic corticosterone treatment on

spatial learning, synaptic plasticity, and hippocampal neuropathology in young

and mid-aged rats. J Neurosci, 15(1 Pt 1), 61-69.

Page 210: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

194

Bollinger, T., Bollinger, A., Naujoks, J., Lange, T., & Solbach, W. (2010). The influence

of regulatory T cells and diurnal hormone rhythms on T helper cell activity.

Immunology, 131(4), 488-500.

Borugian, M. J., Gallagher, R. P., Friesen, M. C., Switzer, T. F., & Aronson, K. J. (2005).

Twenty-four-hour light exposure and melatonin levels among shift workers. J

Occup Environ Med, 47(12), 1268-1275.

Brainard, G. C., Lewy, A. J., Menaker, M., Fredrickson, R. H., Miller, L. S., Weleber, R.

G., et al. (1985). Effect of Light Wavelength on the Suppression of Nocturnal

Plasma Melatonin in Normal Volunteers. Annals of the New York Academy of

Sciences, 453, 376-378.

Brainard, G. C., Richardson, B. A., Petterborg, L. J., & Reiter, R. J. (1982). The effect of

different light intensities on pineal melatonin content. Brain Res, 233(1), 75-81.

Brainard, G. C., Rollag, M. D., & Hanifin, J. P. (1997). Photic regulation of melatonin in

humans: ocular and neural signal transduction. J Biol Rhythms, 12(6), 537-546.

Brainard, G. C., Sliney, D., Hanifin, J. P., Glickman, G., Byrne, B., Greeson, J. M., et al.

(2008). Sensitivity of the human circadian system to short-wavelength (420-nm)

light. J Biol Rhythms, 23(5), 379-386.

Bray, M. S., Ratcliffe, W. F., Grenett, M. H., Brewer, R. A., Gamble, K. L., & Young, M.

E. (2012). Quantitative analysis of light-phase restricted feeding reveals metabolic

dyssynchrony in mice. Int J Obes (Lond).

Bray, M. S., Tsai, J. Y., Villegas-Montoya, C., Boland, B. B., Blasier, Z., Egbejimi, O., et

al. (2010). Time-of-day-dependent dietary fat consumption influences multiple

Page 211: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

195

cardiometabolic syndrome parameters in mice. Int J Obes (Lond), 34(11), 1589-

1598.

Bray, M. S., & Young, M. E. (2007). Circadian rhythms in the development of obesity:

potential role for the circadian clock within the adipocyte. Obes Rev, 8(2), 169-

181.

Brotto, L. A., Gorzalka, B. B., & LaMarre, A. K. (2001). Melatonin protects against the

effects of chronic stress on sexual behaviour in male rats. Neuroreport, 12(16),

3465-3469.

Bugge, A., Feng, D., Everett, L. J., Briggs, E. R., Mullican, S. E., Wang, F., et al. (2012).

Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal

metabolic function. Genes Dev, 26(7), 657-667.

Bunger, M. K., Wilsbacher, L. D., Moran, S. M., Clendenin, C., Radcliffe, L. A.,

Hogenesch, J. B., et al. (2000). Mop3 is an essential component of the master

circadian pacemaker in mammals. Cell, 103(7), 1009-1017.

Bunney, W. E., & Bunney, B. G. (2000). Molecular clock genes in man and lower

animals: possible implications for circadian abnormalities in depression.

Neuropsychopharmacology, 22(4), 335-345.

Caballero, B. (2007). The global epidemic of obesity: an overview. Epidemiol Rev, 29, 1-

5.

Cailotto, C., Lei, J., van der Vliet, J., van Heijningen, C., van Eden, C. G., Kalsbeek, A.,

et al. (2009). Effects of nocturnal light on (clock) gene expression in peripheral

organs: a role for the autonomic innervation of the liver. PLoS One, 4(5), e5650.

Page 212: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

196

Cambras, T., Castejon, L., & Diez-Noguera, A. (2011). Social interaction and sex

differences influence rat temperature circadian rhythm under LD cycles and

constant light. Physiol Behav, 103(3-4), 365-371.

Carmona-Alcocer, V., Fuentes-Granados, C., Carmona-Castro, A., Aguilar-Gonzalez, I.,

Cardenas-Vazquez, R., & Miranda-Anaya, M. (2012). Obesity alters circadian

behavior and metabolism in sex dependent manner in the volcano mouse

Neotomodon alstoni. Physiol Behav, 105(3), 727-733.

Carvas, J. M., Vukolic, A., Yepuri, G., Xiong, Y., Popp, K., Schmutz, I., et al. (2012).

Period2 gene mutant mice show compromised insulin-mediated endothelial nitric

oxide release and altered glucose homeostasis. Front Physiol, 3, 337.

Castanon-Cervantes, O., Wu, M., Ehlen, J. C., Paul, K., Gamble, K. L., Johnson, R. L., et

al. (2010). Dysregulation of inflammatory responses by chronic circadian

disruption. J Immunol, 185(10), 5796-5805.

Castro, J. P., Frussa-Filho, R., Fukushiro, D. F., Chinen, C. C., Abilio, V. C., & Silva, R.

H. (2005). Effects of long-term continuous exposure to light on memory and

anxiety in mice. Physiol Behav, 86(1-2), 218-223.

CDC. (2009). FASTSTATS: Leading causes of death: Center for Disease Control and

Prevention.

Cecon, E., Fernandes, P. A., Pinato, L., Ferreira, Z. S., & Markus, R. P. (2010). Daily

variation of constitutively activated nuclear factor kappa B (NFKB) in rat pineal

gland. Chronobiol Int, 27(1), 52-67.

Page 213: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

197

Chen, S. K., Badea, T. C., & Hattar, S. (2011). Photoentrainment and pupillary light

reflex are mediated by distinct populations of ipRGCs. Nature, 476(7358), 92-95.

Chen, Z., Gardi, J., Kushikata, T., Fang, J., & Krueger, J. M. (1999). Nuclear factor-

kappaB-like activity increases in murine cerebral cortex after sleep deprivation.

Am J Physiol, 276(6 Pt 2), R1812-1818.

Cho, H., Zhao, X., Hatori, M., Yu, R. T., Barish, G. D., Lam, M. T., et al. (2012).

Regulation of circadian behaviour and metabolism by REV-ERB-alpha and REV-

ERB-beta. Nature, 485(7396), 123-127.

Cho, K. (2001). Chronic 'jet lag' produces temporal lobe atrophy and spatial cognitive

deficits. Nat Neurosci, 4(6), 567-568.

Cinzano, P., Falchi, F., & Elvidge, C. D. (2001). The first World Atlas of the artificial

night sky brightness. Monthly Notices of the Royal Astronomical Society, 328(3),

689-707.

Claustrat, B., Valatx, J. L., Harthe, C., & Brun, J. (2008). Effect of constant light on

prolactin and corticosterone rhythms evaluated using a noninvasive urine

sampling protocol in the rat. Horm Metab Res, 40(6), 398-403.

Cohen, M. C., Rohtla, K. M., Lavery, C. E., Muller, J. E., & Mittleman, M. A. (1997).

Meta-analysis of the morning excess of acute myocardial infarction and sudden

cardiac death. Am J Cardiol, 79(11), 1512-1516.

Cohen, R., Kronfeld-Schor, N., Ramanathan, C., Baumgras, A., & Smale, L. (2010). The

substructure of the suprachiasmatic nucleus: Similarities between nocturnal and

diurnal spiny mice. Brain Behav Evol, 75(1), 9-22.

Page 214: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

198

Coomans, C. P., van den Berg, S. A., Houben, T., van Klinken, J. B., van den Berg, R.,

Pronk, A. C., et al. (2013). Detrimental effects of constant light exposure and

high-fat diet on circadian energy metabolism and insulin sensitivity. Faseb J,

[Epub ahead of print].

Corbalan-Tutau, M. D., Madrid, J. A., Ordovas, J. M., Smith, C. E., Nicolas, F., &

Garaulet, M. (2011). Differences in daily rhythms of wrist temperature between

obese and normal-weight women: associations with metabolic syndrome features.

Chronobiol Int, 28(5), 425-433.

Couto-Moraes, R., Palermo-Neto, J., & Markus, R. P. (2009). The immune-pineal axis:

stress as a modulator of pineal gland function. Ann N Y Acad Sci, 1153, 193-202.

Craft, T. K., & DeVries, A. C. (2006). Role of IL-1 in poststroke depressive-like behavior

in mice. Biol Psychiatry, 60(8), 812-818.

Czeisler, C. A., Kronauer, R. E., Allan, J. S., Duffy, J. F., Jewett, M. E., Brown, E. N., et

al. (1989). Bright light induction of strong (type 0) resetting of the human

circadian pacemaker. Science, 244(4910), 1328-1333.

da Silveira Cruz-Machado, S., Carvalho-Sousa, C. E., Tamura, E. K., Pinato, L., Cecon,

E., Fernandes, P. A., et al. (2010). TLR4 and CD14 receptors expressed in rat

pineal gland trigger NFKB pathway. J Pineal Res, 49(2), 183-192.

Dagnino-Subiabre, A., Orellana, J. A., Carmona-Fontaine, C., Montiel, J., Diaz-Veliz, G.,

Seron-Ferre, M., et al. (2006). Chronic stress decreases the expression of

sympathetic markers in the pineal gland and increases plasma melatonin

concentration in rats. J Neurochem, 97(5), 1279-1287.

Page 215: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

199

Dallman, M. F., la Fleur, S. E., Pecoraro, N. C., Gomez, F., Houshyar, H., & Akana, S. F.

(2004). Minireview: glucocorticoids--food intake, abdominal obesity, and wealthy

nations in 2004. Endocrinology, 145(6), 2633-2638.

Dallmann, R., Viola, A. U., Tarokh, L., Cajochen, C., & Brown, S. A. (2012). The human

circadian metabolome. Proc Natl Acad Sci U S A, 109(7), 2625-2629.

Dallmann, R., & Weaver, D. R. (2010). Altered body mass regulation in male mPeriod

mutant mice on high-fat diet. Chronobiol Int, 27(6), 1317-1328.

Damiola, F., Le Minh, N., Preitner, N., Kornmann, B., Fleury-Olela, F., & Schibler, U.

(2000). Restricted feeding uncouples circadian oscillators in peripheral tissues

from the central pacemaker in the suprachiasmatic nucleus. Genes Dev, 14(23),

2950-2961.

Dauchy, R. T., Dauchy, E. M., Tirrell, R. P., Hill, C. R., Davidson, L. K., Greene, M. W.,

et al. (2010). Dark-phase light contamination disrupts circadian rhythms in plasma

measures of endocrine physiology and metabolism in rats. Comp Med, 60(5), 348-

356.

Dauncey, M. J., & Brown, D. (1987). Role of activity-induced thermogenesis in twenty-

four hour energy expenditure of lean and genetically obese (ob/ob) mice. Q J Exp

Physiol, 72(4), 549-559.

Davies, T. W., Bennie, J., & Gaston, K. J. (2012). Street lighting changes the

composition of invertebrate communities. Biol Lett, 8(5), 764-767.

Page 216: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

200

Davis, S., & Mirick, D. K. (2006). Circadian disruption, shift work and the risk of cancer:

a summary of the evidence and studies in Seattle. Cancer Causes Control, 17(4),

539-545.

De Bacquer, D., Van Risseghem, M., Clays, E., Kittel, F., De Backer, G., & Braeckman,

L. (2009). Rotating shift work and the metabolic syndrome: a prospective study.

Int J Epidemiol, 38(3), 848-854.

de Groot, J., Boersma, W. J., Scholten, J. W., & Koolhaas, J. M. (2002). Social stress in

male mice impairs long-term antiviral immunity selectively in wounded subjects.

Physiol Behav, 75(3), 277-285.

De Souza, C. T., Araujo, E. P., Bordin, S., Ashimine, R., Zollner, R. L., Boschero, A. C.,

et al. (2005). Consumption of a fat-rich diet activates a proinflammatory response

and induces insulin resistance in the hypothalamus. Endocrinology, 146(10),

4192-4199.

Deboer, T., Detari, L., & Meijer, J. H. (2007). Long term effects of sleep deprivation on

the mammalian circadian pacemaker. Sleep, 30(3), 257-262.

Delezie, J., Dumont, S., Dardente, H., Oudart, H., Grechez-Cassiau, A., Klosen, P., et al.

(2012). The nuclear receptor REV-ERBalpha is required for the daily balance of

carbohydrate and lipid metabolism. Faseb J, 26(8), 3321-3335.

Demas, G. E., Chefer, V., Talan, M. I., & Nelson, R. J. (1997). Metabolic costs of

mounting an antigen-stimulated immune response in adult and aged C57BL/6J

mice. Am J Physiol, 273(5 Pt 2), R1631-1637.

Page 217: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

201

Dhabhar, F. S. (2002). Stress-induced augmentation of immune function--the role of

stress hormones, leukocyte trafficking, and cytokines. Brain Behav Immun, 16(6),

785-798.

Dhabhar, F. S., & McEwen, B. S. (1999). Enhancing versus suppressive effects of stress

hormones on skin immune function. Proc Natl Acad Sci U S A, 96(3), 1059-1064.

Dominoni, D., Quetting, M., & Partecke, J. (2013). Artificial light at night advances

avian reproductive physiology. Proc Biol Sci, 280(1756), 20123017.

Donnelly, D. J., Gensel, J. C., Ankeny, D. P., van Rooijen, N., & Popovich, P. G. (2009).

An efficient and reproducible method for quantifying macrophages in different

experimental models of central nervous system pathology. J Neurosci Methods,

181(1), 36-44.

Douris, N., Kojima, S., Pan, X., Lerch-Gaggl, A. F., Duong, S. Q., Hussain, M. M., et al.

(2011). Nocturnin regulates circadian trafficking of dietary lipid in intestinal

enterocytes. Curr Biol, 21(16), 1347-1355.

Drazen, D. L., & Nelson, R. J. (2001). Melatonin receptor subtype MT2 (Mel 1b) and not

mt1 (Mel 1a) is associated with melatonin-induced enhancement of cell-mediated

and humoral immunity. Neuroendocrinology, 74(3), 178-184.

Driesen, K., Jansen, N. W., Kant, I., Mohren, D. C., & van Amelsvoort, L. G. (2010).

Depressed mood in the working population: associations with work schedules and

working hours. Chronobiol Int, 27(5), 1062-1079.

Dumont, M., & Beaulieu, C. (2007). Light exposure in the natural environment:

relevance to mood and sleep disorders. Sleep Med, 8(6), 557-565.

Page 218: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

202

Dunlap, J. C., Loros, J. J., & DeCoursey, P. J. (Eds.). (2004). Chronobiology: Biological

Timekeeping. Sunderland, MA: Sinauer Associates, Inc.

Dunn, H., Anderson, M. A., & Hill, P. D. (2010). Nighttime lighting in intensive care

units. Crit Care Nurse, 30(3), 31-37.

Duplantier, J. M., & Ba, K. (2001). Swimming ability in six West-African rodent species

under laboratory conditions - Evaluation of their potentialities to colonize islands.

African Small Mammals, 331-+.

Dwyer, R. G., Bearhop, S., Campbell, H. A., & Bryant, D. M. (2012). Shedding light on

light: benefits of anthropogenic illumination to a nocturnally foraging shorebird. J

Anim Ecol.

Eckel-Mahan, K., & Sassone-Corsi, P. (2009). Metabolism control by the circadian clock

and vice versa. Nat Struct Mol Biol, 16(5), 462-467.

Eckel-Mahan, K. L., Patel, V. R., Mohney, R. P., Vignola, K. S., Baldi, P., & Sassone-

Corsi, P. (2012). Coordination of the transcriptome and metabolome by the

circadian clock. Proc Natl Acad Sci U S A, 109(14), 5541-5546.

Eckle, T., Hartmann, K., Bonney, S., Reithel, S., Mittelbronn, M., Walker, L. A., et al.

(2012). Adora2b-elicited Per2 stabilization promotes a HIF-dependent metabolic

switch crucial for myocardial adaptation to ischemia. Nat Med, 18(5), 774-782.

Edgar, D. M., & Dement, W. C. (1991). Regularly Scheduled Voluntary Exercise

Synchronizes the Mouse Circadian Clock. American Journal of Physiology,

261(4), R928-R933.

Ekirch, A. R. (2005). At Day's Close:Night in Times Past. New York: W.W. Norton&Co.

Page 219: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

203

Elliott, V. J., Rodgers, D. L., & Brett, S. J. (2011). Systematic review of quality of life

and other patient-centred outcomes after cardiac arrest survival. Resuscitation,

82(3), 247-256.

Eltzschig, H. K., & Eckle, T. (2011). Ischemia and reperfusion--from mechanism to

translation. Nat Med, 17(11), 1391-1401.

Engineering, N. A. o. (2000). Greatest Engineering Achievements of the 20th Century.

Esliger, D. W., Tremblay, M. S., Copeland, J. L., Barnes, J. D., Huntington, G. E., &

Bassett, D. R., Jr. (2010). Physical activity profile of Old Order Amish,

Mennonite, and contemporary children. Med Sci Sports Exerc, 42(2), 296-303.

Esquirol, Y., Bongard, V., Mabile, L., Jonnier, B., Soulat, J. M., & Perret, B. (2009).

Shift work and metabolic syndrome: respective impacts of job strain, physical

activity, and dietary rhythms. Chronobiol Int, 26(3), 544-559.

Evans, J. A., Elliott, J. A., & Gorman, M. R. (2005). Circadian entrainment and phase

resetting differ markedly under dimly illuminated versus completely dark nights.

Behavioural Brain Research, 162(1), 116-126.

Evans, J. A., Elliott, J. A., & Gorman, M. R. (2007). Circadian effects of light no brighter

than moonlight. J Biol Rhythms, 22(4), 356-367.

Feng, D., Liu, T., Sun, Z., Bugge, A., Mullican, S. E., Alenghat, T., et al. (2011). A

circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid

metabolism. Science, 331(6022), 1315-1319.

Ferrari, E., Fraschini, F., & Brambilla, F. (1990). Hormonal circadian rhythms in eating

disorders. Biol Psychiatry, 27(9), 1007-1020.

Page 220: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

204

Figueiro, M. G., & Rea, M. S. (2010). The effects of red and blue lights on circadian

variations in cortisol, alpha amylase, and melatonin. Int J Endocrinol, 2010,

829351.

Figueiro, M. G., Wood, B., Plitnick, B., & Rea, M. S. (2011). The impact of light from

computer monitors on melatonin levels in college students. Neuro Endocrinol

Lett, 32(2), 158-163.

Fiske, V. M. (1941). Effect of light on sexual maturation, estrous cycles, and anterior

pituitary of the rat. Endocrinology, 29, 187-196.

Fonken, L. K., Finy, M. S., Walton, J. C., Weil, Z. M., Workman, J. L., Ross, J., et al.

(2009). Influence of light at night on murine anxiety- and depressive-like

responses. Behav Brain Res, 205(2), 349-354.

Fonken, L. K., Haim, A., & Nelson, R. J. (2011). Dim light at night increases immune

function in nile grass rats, a diurnal rodent. Chronobiol Int, 29(1), 26-34.

Fonken, L. K., Kitsmiller, E., Smale, L., & Nelson, R. J. (2012). Dim nighttime light

impairs cognition and provokes depressive-like responses in a diurnal rodent. J

Biol Rhythms, 27(4), 319-327.

Fonken, L. K., & Nelson, R. J. (2011). Illuminating the deleterious effects of light at

night. F1000 Med Rep, 3, 18.

Fonken, L. K., & Nelson, R. J. (2013). Dim light at night increases depressive-like

responses in male C3H/HeNHsd mice. Behav Brain Res, 243C, 74-78.

Page 221: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

205

Fonken, L. K., Weil, Z. M., & Nelson, R. J. (2013). Dark nights reverse metabolic

disruption caused by dim light at night. Obesity (Silver Spring), [Epub ahead of

print].

Fonken, L. K., Workman, J. L., Walton, J. C., Weil, Z. M., Morris, J. S., Haim, A., et al.

(2010). Light at night increases body mass by shifting the time of food intake.

Proc Natl Acad Sci U S A, 107(43), 18664-18669.

Fujioka, A., Fujioka, T., Tsuruta, R., Izumi, T., Kasaoka, S., & Maekawa, T. (2011).

Effects of a constant light environment on hippocampal neurogenesis and memory

in mice. Neurosci Lett, 488(1), 41-44.

Fujioka, M., Nishio, K., Miyamoto, S., Hiramatsu, K. I., Sakaki, T., Okuchi, K., et al.

(2000). Hippocampal damage in the human brain after cardiac arrest. Cerebrovasc

Dis, 10(1), 2-7.

Fuller, P. M., Lu, J., & Saper, C. B. (2008). Differential rescue of light- and food-

entrainable circadian rhythms. Science, 320(5879), 1074-1077.

Fung, T. T., Hu, F. B., Yu, J., Chu, N. F., Spiegelman, D., Tofler, G. H., et al. (2000).

Leisure-time physical activity, television watching, and plasma biomarkers of

obesity and cardiovascular disease risk. Am J Epidemiol, 152(12), 1171-1178.

Garaulet, M., Gomez-Abellan, P., Alburquerque-Bejar, J. J., Lee, Y. C., Ordovas, J. M.,

& Scheer, F. A. (2013). Timing of food intake predicts weight loss effectiveness.

Int J Obes (Lond).

Page 222: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

206

Gaston, K. J., Davies, T. W., Bennie, J., & Hopkins, J. (2012). Reducing the ecological

consequences of night-time light pollution: options and developments. J Appl

Ecol, 49(6), 1256-1266.

Gekakis, N., Staknis, D., Nguyen, H. B., Davis, F. C., Wilsbacher, L. D., King, D. P., et

al. (1998). Role of the CLOCK protein in the mammalian circadian mechanism.

Science, 280(5369), 1564-1569.

Gerstner, J. R. (2012). On the evolution of memory: a time for clocks. Front Mol

Neurosci, 5, 23.

Gibson, E. M., Wang, C., Tjho, S., Khattar, N., & Kriegsfeld, L. J. (2010). Experimental

'jet lag' inhibits adult neurogenesis and produces long-term cognitive deficits in

female hamsters. PLoS One, 5(12), e15267.

Gilad, E., Wong, H. R., Zingarelli, B., Virag, L., O'Connor, M., Salzman, A. L., et al.

(1998). Melatonin inhibits expression of the inducible isoform of nitric oxide

synthase in murine macrophages: role of inhibition of NFkappaB activation.

Faseb J, 12(9), 685-693.

Goel, N., Stunkard, A. J., Rogers, N. L., Van Dongen, H. P., Allison, K. C., O'Reardon, J.

P., et al. (2009). Circadian rhythm profiles in women with night eating syndrome.

J Biol Rhythms, 24(1), 85-94.

Golombek, D. A., & Rosenstein, R. E. (2010). Physiology of circadian entrainment.

Physiol Rev, 90(3), 1063-1102.

Page 223: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

207

Gomez-Abellan, P., Hernandez-Morante, J. J., Lujan, J. A., Madrid, J. A., & Garaulet, M.

(2008). Clock genes are implicated in the human metabolic syndrome. Int J Obes

(Lond), 32(1), 121-128.

Gooley, J. J., Schomer, A., & Saper, C. B. (2006). The dorsomedial hypothalamic

nucleus is critical for the expression of food-entrainable circadian rhythms. Nat

Neurosci, 9(3), 398-407.

Goto, M., Oshima, I., Tomita, T., & Ebihara, S. (1989). Melatonin Content of the Pineal-

Gland in Different Mouse Strains. Journal of Pineal Research, 7(2), 195-204.

Gould, E., & Gross, C. G. (2002). Neurogenesis in adult mammals: some progress and

problems. J Neurosci, 22(3), 619-623.

Graeber, M. B. (2010). Changing face of microglia. Science, 330(6005), 783-788.

Green, C. B., Douris, N., Kojima, S., Strayer, C. A., Fogerty, J., Lourim, D., et al. (2007).

Loss of Nocturnin, a circadian deadenylase, confers resistance to hepatic steatosis

and diet-induced obesity. Proc Natl Acad Sci U S A, 104(23), 9888-9893.

Green, C. B., Takahashi, J. S., & Bass, J. (2008). The meter of metabolism. Cell, 134(5),

728-742.

Guh, D. P., Zhang, W., Bansback, N., Amarsi, Z., Birmingham, C. L., & Anis, A. H.

(2009). The incidence of co-morbidities related to obesity and overweight: a

systematic review and meta-analysis. BMC Public Health, 9, 88.

Guo, H., Brewer, J. M., Champhekar, A., Harris, R. B., & Bittman, E. L. (2005).

Differential control of peripheral circadian rhythms by suprachiasmatic-dependent

neural signals. Proc Natl Acad Sci U S A, 102(8), 3111-3116.

Page 224: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

208

Ha, M., & Park, J. (2005). Shiftwork and metabolic risk factors of cardiovascular disease.

J Occup Health, 47(2), 89-95.

Hajszan, T., Dow, A., Warner-Schmidt, J. L., Szigeti-Buck, K., Sallam, N. L., Parducz,

A., et al. (2009). Remodeling of Hippocampal Spine Synapses in the Rat Learned

Helplessness Model of Depression. Biological Psychiatry, 65(5), 392-400.

Haldar, C., & Singh, R. (2001). Pineal modulation of thymus and immune function in a

seasonally breeding tropical rodent, Funambulus pennanti. J Exp Zool, 289(2), 90-

98.

Hashinaga, T., Wada, N., Otabe, S., Yuan, X., Kurita, Y., Kakino, S., et al. (2012).

Modulation by adiponectin of circadian clock rhythmicity in model mice for

metabolic syndrome. Endocr J.

Hashiramoto, A., Yamane, T., Tsumiyama, K., Yoshida, K., Komai, K., Yamada, H., et

al. (2010). Mammalian clock gene Cryptochrome regulates arthritis via

proinflammatory cytokine TNF-alpha. J Immunol, 184(3), 1560-1565.

Hatori, M., Vollmers, C., Zarrinpar, A., Ditacchio, L., Bushong, E. A., Gill, S., et al.

(2012). Time-restricted feeding without reducing caloric intake prevents

metabolic diseases in mice fed a high-fat diet. Cell Metab, 15(6), 848-860.

Hattar, S., Liao, H. W., Takao, M., Berson, D. M., & Yau, K. W. (2002). Melanopsin-

containing retinal ganglion cells: architecture, projections, and intrinsic

photosensitivity. Science, 295(5557), 1065-1070.

Hill, J. O., Wyatt, H. R., Reed, G. W., & Peters, J. C. (2003). Obesity and the

environment: where do we go from here? Science, 299(5608), 853-855.

Page 225: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

209

Hotamisligil, G. S. (2006). Inflammation and metabolic disorders. Nature, 444(7121),

860-867.

Hotamisligil, G. S., Shargill, N. S., & Spiegelman, B. M. (1993). Adipose expression of

tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance.

Science, 259(5091), 87-91.

Hsieh, M. C., Yang, S. C., Tseng, H. L., Hwang, L. L., Chen, C. T., & Shieh, K. R.

(2010). Abnormal expressions of circadian-clock and circadian clock-controlled

genes in the livers and kidneys of long-term, high-fat-diet-treated mice. Int J Obes

(Lond), 34(2), 227-239.

Hurn, P. D., Subramanian, S., Parker, S. M., Afentoulis, M. E., Kaler, L. J., Vandenbark,

A. A., et al. (2007). T- and B-cell-deficient mice with experimental stroke have

reduced lesion size and inflammation. Journal of Cerebral Blood Flow and

Metabolism, 27(11), 1798-1805.

Hut, R. A., & Beersma, D. G. (2011). Evolution of time-keeping mechanisms: early

emergence and adaptation to photoperiod. Philos Trans R Soc Lond B Biol Sci,

366(1574), 2141-2154.

Ingham, E. S., Gunhan, E., Fuller, P. M., & Fuller, C. A. (2009). Immunotoxin-induced

ablation of melanopsin retinal ganglion cells in a non-murine mammalian model.

J Comp Neurol, 516(2), 125-140.

Jasser, S. A., Hanifin, J. P., Rollag, M. D., & Brainard, G. C. (2006). Dim light

adaptation attenuates acute melatonin suppression in humans. J Biol Rhythms,

21(5), 394-404.

Page 226: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

210

Jhanwar-Uniyal, M., Beck, B., Burlet, C., & Leibowitz, S. F. (1990). Diurnal rhythm of

neuropeptide Y-like immunoreactivity in the suprachiasmatic, arcuate and

paraventricular nuclei and other hypothalamic sites. Brain Res, 536(1-2), 331-

334.

Kahn, S. E., Hull, R. L., & Utzschneider, K. M. (2006). Mechanisms linking obesity to

insulin resistance and type 2 diabetes. Nature, 444(7121), 840-846.

Kalra, S. P., Dube, M. G., Pu, S., Xu, B., Horvath, T. L., & Kalra, P. S. (1999).

Interacting appetite-regulating pathways in the hypothalamic regulation of body

weight. Endocr Rev, 20(1), 68-100.

Kalsbeek, A., Fliers, E., Romijn, J. A., La Fleur, S. E., Wortel, J., Bakker, O., et al.

(2001). The suprachiasmatic nucleus generates the diurnal changes in plasma

leptin levels. Endocrinology, 142(6), 2677-2685.

Kanda, N., & Watanabe, S. (2005). Regulatory roles of sex hormones in cutaneous

biology and immunology. J Dermatol Sci, 38(1), 1-7.

Karatsoreos, I. N., Bhagat, S., Bloss, E. B., Morrison, J. H., & McEwen, B. S. (2011).

Disruption of circadian clocks has ramifications for metabolism, brain, and

behavior. Proc Natl Acad Sci U S A, 108(4), 1657-1662.

Karelina, K., Norman, G. J., Zhang, N., Morris, J. S., Peng, H., & DeVries, A. C. (2009).

Social isolation alters neuroinflammatory response to stroke. Proc Natl Acad Sci

U S A, 106(14), 5895-5900.

Page 227: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

211

Karlsson, B., Knutsson, A., & Lindahl, B. (2001). Is there an association between shift

work and having a metabolic syndrome? Results from a population based study of

27,485 people. Occup Environ Med, 58(11), 747-752.

Karlsson, B. H., Knutsson, A. K., Lindahl, B. O., & Alfredsson, L. S. (2003). Metabolic

disturbances in male workers with rotating three-shift work. Results of the WOLF

study. Int Arch Occup Environ Health, 76(6), 424-430.

Keller, M., Mazuch, J., Abraham, U., Eom, G. D., Herzog, E. D., Volk, H. D., et al.

(2009). A circadian clock in macrophages controls inflammatory immune

responses. Proc Natl Acad Sci U S A, 106(50), 21407-21412.

Kempenaers, B., Borgstrom, P., Loes, P., Schlicht, E., & Valcu, M. (2010). Artificial

night lighting affects dawn song, extra-pair siring success, and lay date in

songbirds. Curr Biol, 20(19), 1735-1739.

Kennaway, D. J., Owens, J. A., Voultsios, A., Boden, M. J., & Varcoe, T. J. (2007).

Metabolic homeostasis in mice with disrupted Clock gene expression in

peripheral tissues. Am J Physiol Regul Integr Comp Physiol, 293(4), R1528-1537.

Kennaway, D. J., Voultsios, A., Varcoe, T. J., & Moyer, R. W. (2002). Melatonin in

mice: rhythms, response to light, adrenergic stimulation, and metabolism. Am J

Physiol Regul Integr Comp Physiol, 282(2), R358-365.

Keppel, G., & Wickens, T. (2004). Design and Analysis: A Researcher's Handbook (4th

ed.). Upper Sadle River, NJ: Prentice Hall.

Page 228: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

212

Keuper, W., Dieker, H. J., Brouwer, M. A., & Verheugt, F. W. (2007). Reperfusion

therapy in out-of-hospital cardiac arrest: current insights. Resuscitation, 73(2),

189-201.

Kiessling, S., Eichele, G., & Oster, H. (2010). Adrenal glucocorticoids have a key role in

circadian resynchronization in a mouse model of jet lag. J Clin Invest, 120(7),

2600-2609.

Kim, F., Pham, M., Maloney, E., Rizzo, N. O., Morton, G. J., Wisse, B. E., et al. (2008).

Vascular inflammation, insulin resistance, and reduced nitric oxide production

precede the onset of peripheral insulin resistance. Arterioscler Thromb Vasc Biol,

28(11), 1982-1988.

King, D. P., Zhao, Y., Sangoram, A. M., Wilsbacher, L. D., Tanaka, M., Antoch, M. P.,

et al. (1997). Positional cloning of the mouse circadian clock gene. Cell, 89(4),

641-653.

Kirino, T. (1982). Delayed neuronal death in the gerbil hippocampus following ischemia.

Brain Res, 239(1), 57-69.

Klein, S. L., & Nelson, R. J. (1999). Social interactions unmask sex differences in

humoral immunity in voles. Anim Behav, 57(3), 603-610.

Kloog, I., Haim, A., & Portnov, B. A. (2009). Using kernel density function as an urban

analysis tool: Investigating the association between nightlight exposure and the

incidence of breast cancer in Haifa, Israel. Computers Environment and Urban

Systems, 33(1), 55-63.

Page 229: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

213

Kloog, I., Haim, A., Stevens, R. G., Barachana, M., & Portnov, B. A. (2008). Light at

night co-distributes with incident breast but not lung cancer in the female

population of israel. Chronobiol Int, 25(1), 65-81.

Kloog, I., Haim, A., Stevens, R. G., & Portnov, B. A. (2009). Global co-distribution of

light at night (LAN) and cancers of prostate, colon, and lung in men. Chronobiol

Int, 26(1), 108-125.

Kloog, I., Portnov, B. A., Rennert, H. S., & Haim, A. (2011). Does the modern urbanized

sleeping habitat pose a breast cancer risk? Chronobiol Int, 28(1), 76-80.

Knutsson, A. (2003). Health disorders of shift workers. Occup Med (Lond), 53(2), 103-

108.

Ko, C. H., & Takahashi, J. S. (2006). Molecular components of the mammalian circadian

clock. Hum Mol Genet, 15 Spec No 2, R271-277.

Kohsaka, A., & Bass, J. (2007). A sense of time: how molecular clocks organize

metabolism. Trends Endocrinol Metab, 18(1), 4-11.

Kohsaka, A., Laposky, A. D., Ramsey, K. M., Estrada, C., Joshu, C., Kobayashi, Y., et al.

(2007). High-fat diet disrupts behavioral and molecular circadian rhythms in

mice. Cell Metab, 6(5), 414-421.

Kohyama, J. (2009). A newly proposed disease condition produced by light exposure

during night: asynchronization. Brain Dev, 31(4), 255-273.

Kondratov, R. V., Kondratova, A. A., Gorbacheva, V. Y., Vykhovanets, O. V., &

Antoch, M. P. (2006). Early aging and age-related pathologies in mice deficient in

Page 230: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

214

BMAL1, the core componentof the circadian clock. Genes Dev, 20(14), 1868-

1873.

Korkmaz, A., Topal, T., Tan, D. X., & Reiter, R. J. (2009). Role of melatonin in

metabolic regulation. Rev Endocr Metab Disord, 10(4), 261-270.

Kornmann, B., Schaad, O., Bujard, H., Takahashi, J. S., & Schibler, U. (2007). System-

driven and oscillator-dependent circadian transcription in mice with a

conditionally active liver clock. PLoS Biol, 5(2), e34.

Krause, G. S., Kumar, K., White, B. C., Aust, S. D., & Wiegenstein, J. G. (1986).

Ischemia, resuscitation, and reperfusion: mechanisms of tissue injury and

prospects for protection. Am Heart J, 111(4), 768-780.

Krieger, D. T. (1974). Food and water restriction shifts corticosterone, temperature,

activity and brain amine periodicity. Endocrinology, 95(5), 1195-1201.

Kriegsfeld, L. J., Mei, D. F., Yan, L., Witkovsky, P., Lesauter, J., Hamada, T., et al.

(2008). Targeted mutation of the calbindin D28K gene disrupts circadian

rhythmicity and entrainment. Eur J Neurosci, 27(11), 2907-2921.

la Fleur, S. E., Kalsbeek, A., Wortel, J., Fekkes, M. L., & Buijs, R. M. (2001). A daily

rhythm in glucose tolerance: a role for the suprachiasmatic nucleus. Diabetes,

50(6), 1237-1243.

Lamia, K. A., Storch, K. F., & Weitz, C. J. (2008). Physiological significance of a

peripheral tissue circadian clock. Proc Natl Acad Sci U S A, 105(39), 15172-

15177.

Page 231: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

215

Lamont, E. W., Diaz, L. R., Barry-Shaw, J., Stewart, J., & Amir, S. (2005). Daily

restricted feeding rescues a rhythm of period2 expression in the arrhythmic

suprachiasmatic nucleus. Neuroscience, 132(2), 245-248.

Langdon, K. D., Granter-Button, S., & Corbett, D. (2008). Persistent behavioral

impairments and neuroinflammation following global ischemia in the rat. Eur J

Neurosci, 28(11), 2310-2318.

Lange, T., Dimitrov, S., & Born, J. (2010). Effects of sleep and circadian rhythm on the

human immune system. Ann N Y Acad Sci, 1193, 48-59.

Laposky, A. D., Bass, J., Kohsaka, A., & Turek, F. W. (2008). Sleep and circadian

rhythms: key components in the regulation of energy metabolism. FEBS Lett,

582(1), 142-151.

Lee, A., Ader, M., Bray, G. A., & Bergman, R. N. (1992). Diurnal variation in glucose

tolerance. Cyclic suppression of insulin action and insulin secretion in normal-

weight, but not obese, subjects. Diabetes, 41(6), 750-759.

LeGates, T. A., Altimus, C. M., Wang, H., Lee, H. K., Yang, S., Zhao, H., et al. (2012).

Aberrant light directly impairs mood and learning through melanopsin-expressing

neurons. Nature, 491(7425), 594-598.

LeSauter, J., Lambert, C. M., Robotham, M. R., Model, Z., Silver, R., & Weaver, D. R.

(2012). Antibodies for assessing circadian clock proteins in the rodent

suprachiasmatic nucleus. PLoS One, 7(4), e35938.

Page 232: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

216

Lim, C., Alexander, M. P., LaFleche, G., Schnyer, D. M., & Verfaellie, M. (2004). The

neurological and cognitive sequelae of cardiac arrest. Neurology, 63(10), 1774-

1778.

Lin, Y. C., Hsiao, T. J., & Chen, P. C. (2009a). Persistent rotating shift-work exposure

accelerates development of metabolic syndrome among middle-aged female

employees: a five-year follow-up. Chronobiol Int, 26(4), 740-755.

Lin, Y. C., Hsiao, T. J., & Chen, P. C. (2009b). Shift work aggravates metabolic

syndrome development among early-middle-aged males with elevated ALT.

World J Gastroenterol, 15(45), 5654-5661.

Ling, Z. Q., Tian, Q., Wang, L., Fu, Z. Q., Wang, X. C., Wang, Q., et al. (2009). Constant

illumination induces Alzheimer-like damages with endoplasmic reticulum

involvement and the protection of melatonin. J Alzheimers Dis, 16(2), 287-300.

Liu, A. C., Welsh, D. K., Ko, C. H., Tran, H. G., Zhang, E. E., Priest, A. A., et al. (2007).

Intercellular coupling confers robustness against mutations in the SCN circadian

clock network. Cell, 129(3), 605-616.

Liu, C., Li, S., Liu, T., Borjigin, J., & Lin, J. D. (2007). Transcriptional coactivator PGC-

1alpha integrates the mammalian clock and energy metabolism. Nature,

447(7143), 477-481.

Lopes, C., Mariano, M., & Markus, R. P. (2001). Interaction between the adrenal and the

pineal gland in chronic experimental inflammation induced by BCG in mice.

Inflamm Res, 50(1), 6-11.

Page 233: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

217

Lotufo, C. M., Lopes, C., Dubocovich, M. L., Farsky, S. H., & Markus, R. P. (2001).

Melatonin and N-acetylserotonin inhibit leukocyte rolling and adhesion to rat

microcirculation. Eur J Pharmacol, 430(2-3), 351-357.

Lowrey, P. L., & Takahashi, J. S. (2004). Mammalian circadian biology: Elucidating

genome-wide levels of temporal organization. Annual Review of Genomics and

Human Genetics, 5, 407-441.

Lundkvist, G. B., Robertson, B., Mhlanga, J. D., Rottenberg, M. E., & Kristensson, K.

(1998). Expression of an oscillating interferon-gamma receptor in the

suprachiasmatic nuclei. Neuroreport, 9(6), 1059-1063.

Lupien, S. J., de Leon, M., de Santi, S., Convit, A., Tarshish, C., Nair, N. P., et al. (1998).

Cortisol levels during human aging predict hippocampal atrophy and memory

deficits. Nat Neurosci, 1(1), 69-73.

Ma, W. P., Cao, J., Tian, M., Cui, M. H., Han, H. L., Yang, Y. X., et al. (2007). Exposure

to chronic constant light impairs spatial memory and influences long-term

depression in rats. Neurosci Res, 59(2), 224-230.

Maestroni, G. J., Conti, A., & Pierpaoli, W. (1986). Role of the pineal gland in immunity.

Circadian synthesis and release of melatonin modulates the antibody response and

antagonizes the immunosuppressive effect of corticosterone. J Neuroimmunol,

13(1), 19-30.

Magarinos, A. M., Li, C. J., Toth, J. G., Bath, K. G., Jing, D., Lee, F. S., et al. (2011).

Effect of Brain-Derived Neurotrophic Factor Haploinsufficiency on Stress-

Induced Remodeling of Hippocampal Neurons. Hippocampus, 21(3), 253-264.

Page 234: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

218

Magarinos, A. M., McEwen, B. S., Flugge, G., & Fuchs, E. (1996). Chronic psychosocial

stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in

subordinate tree shrews. J Neurosci, 16(10), 3534-3540.

Mantele, S., Otway, D. T., Middleton, B., Bretschneider, S., Wright, J., Robertson, M. D.,

et al. (2012). Daily rhythms of plasma melatonin, but not plasma leptin or leptin

mRNA, vary between lean, obese and type 2 diabetic men. PLoS One, 7(5),

e37123.

Marceau, P., Biron, S., Hould, F. S., Marceau, S., Simard, S., Thung, S. N., et al. (1999).

Liver pathology and the metabolic syndrome X in severe obesity. J Clin

Endocrinol Metab, 84(5), 1513-1517.

Marcheva, B., Ramsey, K. M., Buhr, E. D., Kobayashi, Y., Su, H., Ko, C. H., et al.

(2010). Disruption of the clock components CLOCK and BMAL1 leads to

hypoinsulinaemia and diabetes. Nature, 466(7306), 627-631.

Markus, R. P., Ferreira, Z. S., Fernandes, P. A., & Cecon, E. (2007). The immune-pineal

axis: a shuttle between endocrine and paracrine melatonin sources.

Neuroimmunomodulation, 14(3-4), 126-133.

Martin, L. B., 2nd, Weil, Z. M., & Nelson, R. J. (2007). Immune defense and

reproductive pace of life in Peromyscus mice. Ecology, 88(10), 2516-2528.

Martin, L. B., Weil, Z. M., & Nelson, R. J. (2008). Seasonal changes in vertebrate

immune activity: mediation by physiological trade-offs. Philos Trans R Soc Lond

B Biol Sci, 363(1490), 321-339.

Page 235: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

219

Martynhak, B. J., Correia, D., Morais, L. H., Araujo, P., Andersen, M. L., Lima, M. M.,

et al. (2011). Neonatal exposure to constant light prevents anhedonia-like

behavior induced by constant light exposure in adulthood. Behav Brain Res,

222(1), 10-14.

Maury, E., Ramsey, K. M., & Bass, J. (2010). Circadian rhythms and metabolic

syndrome: from experimental genetics to human disease. Circ Res, 106(3), 447-

462.

McDearmon, E. L., Patel, K. N., Ko, C. H., Walisser, J. A., Schook, A. C., Chong, J. L.,

et al. (2006). Dissecting the functions of the mammalian clock protein BMAL1 by

tissue-specific rescue in mice. Science, 314(5803), 1304-1308.

McElhinny, T. L., Smale, L., & Holekamp, K. E. (1997). Patterns of body temperature,

activity, and reproductive behavior in a tropical murid rodent, Arvicanthis

niloticus. Physiol Behav, 62(1), 91-96.

McEwen, B. S., & Sapolsky, R. M. (1995). Stress and cognitive function. Curr Opin

Neurobiol, 5(2), 205-216.

McNamara, P., Seo, S. B., Rudic, R. D., Sehgal, A., Chakravarti, D., & FitzGerald, G. A.

(2001). Regulation of CLOCK and MOP4 by nuclear hormone receptors in the

vasculature: a humoral mechanism to reset a peripheral clock. Cell, 105(7), 877-

889.

Mendoza, J., Pevet, P., & Challet, E. (2008). High-fat feeding alters the clock

synchronization to light. J Physiol, 586(Pt 24), 5901-5910.

Page 236: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

220

Merlot, E., Moze, E., Dantzer, R., & Neveu, P. J. (2004). Cytokine production by spleen

cells after social defeat in mice: activation of T cells and reduced inhibition by

glucocorticoids. Stress, 7(1), 55-61.

Michel, S., Marek, R., Vanderleest, H. T., Vansteensel, M. J., Schwartz, W. J., Colwell,

C. S., et al. (2013). Mechanism of bilateral communication in the suprachiasmatic

nucleus. Eur J Neurosci.

Mieda, M., & Sakurai, T. (2011). Bmal1 in the nervous system is essential for normal

adaptation of circadian locomotor activity and food intake to periodic feeding. J

Neurosci, 31(43), 15391-15396.

Milagro, F. I., Gomez-Abellan, P., Campion, J., Martinez, J. A., Ordovas, J. M., &

Garaulet, M. (2012). CLOCK, PER2 and BMAL1 DNA methylation: association

with obesity and metabolic syndrome characteristics and monounsaturated fat

intake. Chronobiol Int.

Mistlberger, R. E. (1991). Scheduled daily exercise or feeding alters the phase of photic

entrainment in Syrian hamsters. Physiol Behav, 50(6), 1257-1260.

Mistlberger, R. E. (2009). Food-anticipatory circadian rhythms: concepts and methods.

Eur J Neurosci, 30(9), 1718-1729.

Mistlberger, R. E., & Antle, M. C. (2011). Entrainment of circadian clocks in mammals

by arousal and food. Essays in Biochemistry: Chronobiology, 49, 119-136.

Mistlberger, R. E., Lukman, H., & Nadeau, B. G. (1998). Circadian rhythms in the

Zucker obese rat: assessment and intervention. Appetite, 30(3), 255-267.

Page 237: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

221

Miyake, S., Sumi, Y., Yan, L., Takekida, S., Fukuyama, T., Ishida, Y., et al. (2000).

Phase-dependent responses of Per1 and Per2 genes to a light-stimulus in the

suprachiasmatic nucleus of the rat. Neurosci Lett, 294(1), 41-44.

Mizushima, H., Zhou, C. J., Dohi, K., Horai, R., Asano, M., Iwakura, Y., et al. (2002).

Reduced postischemic apoptosis in the hippocampus of mice deficient in

interleukin-1. J Comp Neurol, 448(2), 203-216.

Mohawk, J. A., Green, C. B., & Takahashi, J. S. (2012). Central and peripheral circadian

clocks in mammals. Annu Rev Neurosci, 35, 445-462.

Monk, T. H. (2000). Shift work. In M. H. Kryger, T. Roth & W. C. Dement (Eds.),

Principles and Practice of Sleep Medicine. (2nd ed., pp. 471-476). Philadelphia:

W.B. Saunders Company.

Monteleone, P., Martiadis, V., & Maj, M. (2010). Circadian rhythms and treatment

implications in depression. Prog Neuropsychopharmacol Biol Psychiatry.

Morris, C. J., Yang, J. N., & Scheer, F. A. (2012). The impact of the circadian timing

system on cardiovascular and metabolic function. Prog Brain Res, 199, 337-358.

Mullington, J. M., Haack, M., Toth, M., Serrador, J. M., & Meier-Ewert, H. K. (2009).

Cardiovascular, inflammatory, and metabolic consequences of sleep deprivation.

Prog Cardiovasc Dis, 51(4), 294-302.

Murakami, D. M., Horwitz, B. A., & Fuller, C. A. (1995). Circadian rhythms of

temperature and activity in obese and lean Zucker rats. Am J Physiol, 269(5 Pt 2),

R1038-1043.

Page 238: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

222

Myers, M. G., Jr., Leibel, R. L., Seeley, R. J., & Schwartz, M. W. (2010). Obesity and

leptin resistance: distinguishing cause from effect. Trends Endocrinol Metab,

21(11), 643-651.

Narasimamurthy, R., Hatori, M., Nayak, S. K., Liu, F., Panda, S., & Verma, I. M. (2012).

Circadian clock protein cryptochrome regulates the expression of

proinflammatory cytokines. Proc Natl Acad Sci U S A, 109(31), 12662-12667.

Navara, K. J., & Nelson, R. J. (2007). The dark side of light at night: physiological,

epidemiological, and ecological consequences. J Pineal Res, 43(3), 215-224.

Negi, G., Kumar, A., & Sharma, S. S. (2011). Melatonin modulates neuroinflammation

and oxidative stress in experimental diabetic neuropathy: effects on NF-kappaB

and Nrf2 cascades. J Pineal Res, 50(2), 124-131.

Neigh, G. N., Glasper, E. R., Kofler, J., Traystman, R. J., Mervis, R. F., Bachstetter, A.,

et al. (2004). Cardiac arrest with cardiopulmonary resuscitation reduces dendritic

spine density in CA1 pyramidal cells and selectively alters acquisition of spatial

memory. Eur J Neurosci, 20(7), 1865-1872.

Neigh, G. N., Karelina, K., Glasper, E. R., Bowers, S. L., Zhang, N., Popovich, P. G., et

al. (2009). Anxiety after cardiac arrest/cardiopulmonary resuscitation:

exacerbated by stress and prevented by minocycline. Stroke, 40(11), 3601-3607.

Neigh, G. N., Kofler, J., Meyers, J. L., Bergdall, V., La Perle, K. M., Traystman, R. J., et

al. (2004). Cardiac arrest/cardiopulmonary resuscitation increases anxiety-like

behavior and decreases social interaction. J Cereb Blood Flow Metab, 24(4), 372-

382.

Page 239: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

223

Nelson, R. J. (1990). Photoperiodic responsiveness in house mice. Physiol Behav, 48(3),

403-408.

Nikonenko, A. G., Radenovic, L., Andjus, P. R., & Skibo, G. G. (2009). Structural

features of ischemic damage in the hippocampus. Anat Rec (Hoboken), 292(12),

1914-1921.

Nimmerjahn, A., Kirchhoff, F., & Helmchen, F. (2005). Resting microglial cells are

highly dynamic surveillants of brain parenchyma in vivo. Science, 308(5726),

1314-1318.

Nishiura, C., Noguchi, J., & Hashimoto, H. (2010). Dietary patterns only partially explain

the effect of short sleep duration on the incidence of obesity. Sleep, 33(6), 753-

757.

Nitatori, T., Sato, N., Waguri, S., Karasawa, Y., Araki, H., Shibanai, K., et al. (1995).

Delayed neuronal death in the CA1 pyramidal cell layer of the gerbil

hippocampus following transient ischemia is apoptosis. J Neurosci, 15(2), 1001-

1011.

Novak, C. M., Burghardt, P. R., & Levine, J. A. (2012). The use of a running wheel to

measure activity in rodents: relationship to energy balance, general activity, and

reward. Neurosci Biobehav Rev, 36(3), 1001-1014.

Obayashi, K., Saeki, K., Iwamoto, J., Okamoto, N., Tomioka, K., Nezu, S., et al. (2013).

Exposure to light at night, nocturnal urinary melatonin excretion, and

obesity/dyslipidemia in the elderly: a cross-sectional analysis of the HEIJO-KYO

study. J Clin Endocrinol Metab, 98(1), 337-344.

Page 240: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

224

Oishi, K., Atsumi, G., Sugiyama, S., Kodomari, I., Kasamatsu, M., Machida, K., et al.

(2006). Disrupted fat absorption attenuates obesity induced by a high-fat diet in

Clock mutant mice. FEBS Lett, 580(1), 127-130.

Oishi, K., Miyazaki, K., Kadota, K., Kikuno, R., Nagase, T., Atsumi, G., et al. (2003).

Genome-wide expression analysis of mouse liver reveals CLOCK-regulated

circadian output genes. J Biol Chem, 278(42), 41519-41527.

Okano, S., Akashi, M., Hayasaka, K., & Nakajima, O. (2009). Unusual circadian

locomotor activity and pathophysiology in mutant CRY1 transgenic mice.

Neurosci Lett, 451(3), 246-251.

Oudot, F., Larue-Achagiotis, C., Anton, G., & Verger, P. (1996). Modifications in dietary

self-selection specifically attributable to voluntary wheel running and exercise

training in the rat. Physiol Behav, 59(6), 1123-1128.

Panda, S., Antoch, M. P., Miller, B. H., Su, A. I., Schook, A. B., Straume, M., et al.

(2002). Coordinated transcription of key pathways in the mouse by the circadian

clock. Cell, 109(3), 307-320.

Parkes, K. R. (2002). Shift work and age as interactive predictors of body mass index

among offshore workers. Scand J Work Environ Health, 28(1), 64-71.

Paschos, G. K., Ibrahim, S., Song, W. L., Kunieda, T., Grant, G., Reyes, T. M., et al.

(2012). Obesity in mice with adipocyte-specific deletion of clock component

Arntl. Nat Med, 18, 1768-1777.

Page 241: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

225

Patterson, C. M., & Levin, B. E. (2008). Role of exercise in the central regulation of

energy homeostasis and in the prevention of obesity. Neuroendocrinology, 87(2),

65-70.

Pendergast, J. S., Branecky, K. L., Yang, W., Ellacott, K. L., Niswender, K. D., &

Yamazaki, S. (2013). High-fat diet acutely affects circadian organisation and

eating behavior. Eur J Neurosci.

Pendergast, J. S., Niswender, K. D., & Yamazaki, S. (2012). Tissue-specific function of

Period3 in circadian rhythmicity. PLoS One, 7(1), e30254.

Persengiev, S., Kanchev, L., & Vezenkova, G. (1991). Circadian patterns of melatonin,

corticosterone, and progesterone in male rats subjected to chronic stress: effect of

constant illumination. J Pineal Res, 11(2), 57-62.

Phanuphak, P., Moorhead, J. W., & Claman, H. N. (1974). Tolerance and contact

sensitivity to DNFB in mice. I. In vivo detection by ear swelling and correlation

with in vitro cell stimulation. J Immunol, 112(1), 115-123.

Phillips, M. S., Liu, Q., Hammond, H. A., Dugan, V., Hey, P. J., Caskey, C. J., et al.

(1996). Leptin receptor missense mutation in the fatty Zucker rat. Nat Genet,

13(1), 18-19.

Pietroiusti, A., Neri, A., Somma, G., Coppeta, L., Iavicoli, I., Bergamaschi, A., et al.

(2010). Incidence of metabolic syndrome among night-shift healthcare workers.

Occup Environ Med, 67(1), 54-57.

Plomgaard, P., Bouzakri, K., Krogh-Madsen, R., Mittendorfer, B., Zierath, J. R., &

Pedersen, B. K. (2005). Tumor necrosis factor-alpha induces skeletal muscle

Page 242: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

226

insulin resistance in healthy human subjects via inhibition of Akt substrate 160

phosphorylation. Diabetes, 54(10), 2939-2945.

Pontes, G. N., Cardoso, E. C., Carneiro-Sampaio, M. M., & Markus, R. P. (2006). Injury

switches melatonin production source from endocrine (pineal) to paracrine

(phagocytes) - melatonin in human colostrum and colostrum phagocytes. J Pineal

Res, 41(2), 136-141.

Porsolt, R. D., Bertin, A., & Jalfre, M. (1977). Behavioral despair in mice: a primary

screening test for antidepressants. Arch Int Pharmacodyn Ther, 229(2), 327-336.

Poudyal, H., Panchal, S. K., Ward, L. C., Waanders, J., & Brown, L. (2012). Chronic

high-carbohydrate, high-fat feeding in rats induces reversible metabolic,

cardiovascular, and liver changes. Am J Physiol Endocrinol Metab, 302(12),

E1472-1482.

Power, A., Hughes, A. T. L., Samuels, R. E., & Piggins, H. D. (2010). Rhythm-

Promoting Actions of Exercise in Mice with Deficient Neuropeptide Signaling.

Journal of Biological Rhythms, 25(4), 235-246.

Preitner, N., Damiola, F., Lopez-Molina, L., Zakany, J., Duboule, D., Albrecht, U., et al.

(2002). The orphan nuclear receptor REV-ERBalpha controls circadian

transcription within the positive limb of the mammalian circadian oscillator. Cell,

110(2), 251-260.

Preuss, F., Tang, Y., Laposky, A. D., Arble, D., Keshavarzian, A., & Turek, F. W.

(2008). Adverse effects of chronic circadian desynchronization in animals in a

Page 243: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

227

"challenging" environment. Am J Physiol Regul Integr Comp Physiol, 295(6),

R2034-2040.

Purkayastha, S., Zhang, H., Zhang, G., Ahmed, Z., Wang, Y., & Cai, D. (2010). Neural

dysregulation of peripheral insulin action and blood pressure by brain

endoplasmic reticulum stress. Proc Natl Acad Sci U S A, 108(7), 2939-2944.

Puttonen, S., Viitasalo, K., & Harma, M. (2011). Effect of shiftwork on systemic markers

of inflammation. Chronobiol Int, 28(6), 528-535.

Rajaratnam, S. M., & Arendt, J. (2001). Health in a 24-h society. Lancet, 358(9286), 999-

1005.

Rasmussen, D. D., Boldt, B. M., Wilkinson, C. W., Yellon, S. M., & Matsumoto, A. M.

(1999). Daily melatonin administration at middle age suppresses male rat visceral

fat, plasma leptin, and plasma insulin to youthful levels. Endocrinology, 140(2),

1009-1012.

Reddy, A. B., Maywood, E. S., Karp, N. A., King, V. M., Inoue, Y., Gonzalez, F. J., et al.

(2007). Glucocorticoid signaling synchronizes the liver circadian transcriptome.

Hepatology, 45(6), 1478-1488.

Redman, J., Armstrong, S., & Ng, K. T. (1983). Free-running activity rhythms in the rat:

entrainment by melatonin. Science, 219(4588), 1089-1091.

Reebs, S. G., & Stcoeur, J. (1994). Aftereffects of Scheduled Daily Exercise on Free-

Running Circadian Period in Syrian-Hamsters. Physiology & Behavior, 55(6),

1113-1117.

Page 244: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

228

Reed, A. S., Unger, E. K., Olofsson, L. E., Piper, M. L., Myers, M. G., Jr., & Xu, A. W.

(2010). Functional role of suppressor of cytokine signaling 3 upregulation in

hypothalamic leptin resistance and long-term energy homeostasis. Diabetes,

59(4), 894-906.

Reiter, R. J. (1991). Melatonin: the chemical expression of darkness. Mol Cell

Endocrinol, 79(1-3), C153-158.

Reiter, R. J., Tan, D. X., Korkmaz, A., & Ma, S. (2011). Obesity and metabolic

syndrome: Association with chronodisruption, sleep deprivation, and melatonin

suppression. Ann Med.

Reppert, S. M., & Weaver, D. R. (2001). Molecular analysis of mammalian circadian

rhythms. Annu Rev Physiol, 63, 647-676.

Reppert, S. M., & Weaver, D. R. (2002). Coordination of circadian timing in mammals.

Nature, 418(6901), 935-941.

Robinson, J., Bayliss, S. C., & Fielder, A. R. (1991). Transmission of light across the

adult and neonatal eyelid in vivo. Vision Res, 31(10), 1837-1840.

Roenneberg, T., Allebrandt, K. V., Merrow, M., & Vetter, C. (2012). Social jetlag and

obesity. Curr Biol, 22(10), 939-943.

Roesler, W. J., Helgason, C., Gulka, M., & Khandelwal, R. L. (1985). Aberrations in the

diurnal rhythms of plasma glucose, plasma insulin, liver glycogen, and hepatic

glycogen synthase and phosphorylase activities in genetically diabetic (db/db)

mice. Horm Metab Res, 17(11), 572-575.

Page 245: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

229

Rogers, P., & Webb, G. P. (1980). Estimation of body-fat in normal and obese mice.

British Journal of Nutrition, 43(1), 83-93.

Ruby, N. F., Hwang, C. E., Wessells, C., Fernandez, F., Zhang, P., Sapolsky, R., et al.

(2008). Hippocampal-dependent learning requires a functional circadian system.

Proc Natl Acad Sci U S A, 105(40), 15593-15598.

Rudic, R. D., McNamara, P., Curtis, A. M., Boston, R. C., Panda, S., Hogenesch, J. B., et

al. (2004). BMAL1 and CLOCK, two essential components of the circadian

clock, are involved in glucose homeostasis. PLoS Biol, 2(11), e377.

Ruger, M., St Hilaire, M. A., Brainard, G. C., Khalsa, S. B., Kronauer, R. E., Czeisler, C.

A., et al. (2013). Human phase response curve to a single 6.5 h pulse of short-

wavelength light. J Physiol, 591(Pt 1), 353-363.

Ruiter, M., La Fleur, S. E., van Heijningen, C., van der Vliet, J., Kalsbeek, A., & Buijs,

R. M. (2003). The daily rhythm in plasma glucagon concentrations in the rat is

modulated by the biological clock and by feeding behavior. Diabetes, 52(7),

1709-1715.

Rusak, B., & Groos, G. (1982). Suprachiasmatic stimulation phase shifts rodent circadian

rhythms. Science, 215(4538), 1407-1409.

Rusak, B., Robertson, H. A., Wisden, W., & Hunt, S. P. (1990). Light pulses that shift

rhythms induce gene expression in the suprachiasmatic nucleus. Science,

248(4960), 1237-1240.

Rydell, J. (1992). Exploitation of Insects around Streetlamps by Bats in Sweden.

Functional Ecology, 6(6), 744-750.

Page 246: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

230

Sage, D., Ganem, J., Guillaumond, F., Laforge-Anglade, G., Francois-Bellan, A. M.,

Bosler, O., et al. (2004). Influence of the corticosterone rhythm on photic

entrainment of locomotor activity in rats. J Biol Rhythms, 19(2), 144-156.

Saito, K., Suyama, K., Nishida, K., Sei, Y., & Basile, A. S. (1996). Early increases in

TNF-alpha, IL-6 and IL-1 beta levels following transient cerebral ischemia in

gerbil brain. Neurosci Lett, 206(2-3), 149-152.

Salgado-Delgado, R., Angeles-Castellanos, M., Saderi, N., Buijs, R. M., & Escobar, C.

(2010). Food intake during the normal activity phase prevents obesity and

circadian desynchrony in a rat model of night work. Endocrinology, 151(3), 1019-

1029.

Sapolsky, R. M. (2000). Glucocorticoids and hippocampal atrophy in neuropsychiatric

disorders. Archives of General Psychiatry, 57(10), 925-935.

Sapolsky, R. M. (2004). Stress and Cognition. In M. Gazzaniga (Ed.), The Cognitive

Neurosciences (III ed., pp. 1031-1042). Cambridge, MA: MIT press.

Scheer, F. A., Hilton, M. F., Mantzoros, C. S., & Shea, S. A. (2009). Adverse metabolic

and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci

U S A, 106(11), 4453-4458.

Schernhammer, E. S., Laden, F., Speizer, F. E., Willett, W. C., Hunter, D. J., Kawachi, I.,

et al. (2001). Rotating night shifts and risk of breast cancer in women

participating in the nurses' health study. J Natl Cancer Inst, 93(20), 1563-1568.

Schroeder, A. M., Truong, D., Loh, D. H., Jordan, M. C., Roos, K. P., & Colwell, C. S.

(2012). Voluntary scheduled exercise alters diurnal rhythms of behaviour,

Page 247: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

231

physiology and gene expression in wild-type and vasoactive intestinal peptide-

deficient mice. Journal of Physiology-London, 590(23), 6213-6226.

Schwartz, W. J., Tavakoli-Nezhad, M., Lambert, C. M., Weaver, D. R., & de la Iglesia,

H. O. (2012). Distinct patterns of Period gene expression in the suprachiasmatic

nucleus underlie circadian clock photoentrainment by advances or delays. Proc

Natl Acad Sci U S A, 108(41), 17219-17224.

Scott, E. M., Carter, A. M., & Grant, P. J. (2008). Association between polymorphisms in

the Clock gene, obesity and the metabolic syndrome in man. Int J Obes (Lond),

32(4), 658-662.

Scott, S., & Pellman, K. (1990). Living Without Electricity (People's Place Book No. 9).

Intercourse, PA: Good Books.

Sheline, Y. I., Wang, P. W., Gado, M. H., Csernansky, J. G., & Vannier, M. W. (1996).

Hippocampal atrophy in recurrent major depression. Proceedings of the National

Academy of Sciences of the United States of America, 93(9), 3908-3913.

Sherman, H., Genzer, Y., Cohen, R., Chapnik, N., Madar, Z., & Froy, O. (2012). Timed

high-fat diet resets circadian metabolism and prevents obesity. Faseb J, 26(8),

3493-3502.

Shigeyoshi, Y., Taguchi, K., Yamamoto, S., Takekida, S., Yan, L., Tei, H., et al. (1997).

Light-induced resetting of a mammalian circadian clock is associated with rapid

induction of the mPer1 transcript. Cell, 91(7), 1043-1053.

Page 248: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

232

Shuboni, D. D., Cramm, S., Yan, L., Nunez, A. A., & Smale, L. (2012). Acute behavioral

responses to light and darkness in nocturnal Mus musculus and diurnal

Arvicanthis niloticus. J Biol Rhythms, 27(4), 299-307.

Silver, A. C., Arjona, A., Hughes, M. E., Nitabach, M. N., & Fikrig, E. (2012). Circadian

expression of clock genes in mouse macrophages, dendritic cells, and B cells.

Brain Behav Immun, 26(3), 407-413.

Silver, A. C., Arjona, A., Walker, W. E., & Fikrig, E. (2012). The Circadian Clock

Controls Toll-like Receptor 9-Mediated Innate and Adaptive Immunity. Immunity,

36(2), 251-261.

Silver, R., LeSauter, J., Tresco, P. A., & Lehman, M. N. (1996). A diffusible coupling

signal from the transplanted suprachiasmatic nucleus controlling circadian

locomotor rhythms. Nature, 382(6594), 810-813.

Sinha, M. K., Opentanova, I., Ohannesian, J. P., Kolaczynski, J. W., Heiman, M. L.,

Hale, J., et al. (1996). Evidence of free and bound leptin in human circulation.

Studies in lean and obese subjects and during short-term fasting. J Clin Invest,

98(6), 1277-1282.

Smith, M. R., Revell, V. L., & Eastman, C. I. (2009). Phase advancing the human

circadian clock with blue-enriched polychromatic light. Sleep Med, 10(3), 287-

294.

Solt, L. A., Wang, Y., Banerjee, S., Hughes, T., Kojetin, D. J., Lundasen, T., et al.

(2012). Regulation of circadian behaviour and metabolism by synthetic REV-

ERB agonists. Nature, 485(7396), 62-68.

Page 249: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

233

Sookoian, S., Gemma, C., Fernandez Gianotti, T., Burgueno, A., Alvarez, A., Gonzalez,

C. D., et al. (2007). Effects of rotating shift work on biomarkers of metabolic

syndrome and inflammation. J Intern Med, 261(3), 285-292.

Sorrells, S. F., Caso, J. R., Munhoz, C. D., & Sapolsky, R. M. (2009). The stressed CNS:

when glucocorticoids aggravate inflammation. Neuron, 64(1), 33-39.

Spencer, F. A., Goldberg, R. J., Becker, R. C., & Gore, J. M. (1998). Seasonal

distribution of acute myocardial infarction in the second National Registry of

Myocardial Infarction. J Am Coll Cardiol, 31(6), 1226-1233.

Spiegel, K., Tasali, E., Leproult, R., & Van Cauter, E. (2009). Effects of poor and short

sleep on glucose metabolism and obesity risk. Nat Rev Endocrinol, 5(5), 253-261.

Srinivasan, V., De Berardis, D., Shillcutt, S. D., & Brzezinski, A. (2012). Role of

melatonin in mood disorders and the antidepressant effects of agomelatine. Expert

Opin Investig Drugs, 21(10), 1503-1522.

Stephan, F. K., & Zucker, I. (1972). Circadian rhythms in drinking behavior and

locomotor activity of rats are eliminated by hypothalamic lesions. Proc Natl Acad

Sci U S A, 69(6), 1583-1586.

Stevens, R. G. (2009a). Electric light causes cancer? Surely you're joking, Mr. Stevens.

Mutat Res, 682(1), 1-6.

Stevens, R. G. (2009b). Light-at-night, circadian disruption and breast cancer: assessment

of existing evidence. Int J Epidemiol, 38(4), 963-970.

Stokkan, K. A., Yamazaki, S., Tei, H., Sakaki, Y., & Menaker, M. (2001). Entrainment of

the circadian clock in the liver by feeding. Science, 291(5503), 490-493.

Page 250: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

234

Stoll, G., Jander, S., & Schroeter, M. (1998). Inflammation and glial responses in

ischemic brain lesions. Prog Neurobiol, 56(2), 149-171.

Stone, E. L., Jones, G., & Harris, S. (2009). Street lighting disturbs commuting bats. Curr

Biol, 19(13), 1123-1127.

Storch, K. F., Lipan, O., Leykin, I., Viswanathan, N., Davis, F. C., Wong, W. H., et al.

(2002). Extensive and divergent circadian gene expression in liver and heart.

Nature, 417(6884), 78-83.

Stucchi, P., Gil-Ortega, M., Merino, B., Guzman-Ruiz, R., Cano, V., Valladolid-Acebes,

I., et al. (2012). Circadian feeding drive of metabolic activity in adipose tissue and

not hyperphagia triggers overweight in mice: is there a role of the pentose-

phosphate pathway? Endocrinology, 153(2), 690-699.

Stunkard, A. J., Grace, W. J., & Wolff, H. G. (1955). The night-eating syndrome; a

pattern of food intake among certain obese patients. Am J Med, 19(1), 78-86.

Sturm, R. (2007). Increases in morbid obesity in the USA: 2000-2005. Public Health,

121(7), 492-496.

Su, W., Xie, Z., Guo, Z., Duncan, M. J., Lutshumba, J., & Gong, M. C. (2012). Altered

clock gene expression and vascular smooth muscle diurnal contractile variations

in type 2 diabetic db/db mice. Am J Physiol Heart Circ Physiol, 302(3), H621-

633.

Sunyer, B., Patil, S., Hoger, H., Lubec, G. (2007). Barnes maze, a useful task to assess

spatial reference memory in the mice. Nature Protocols.

Page 251: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

235

Symonds, M. E., Sebert, S., & Budge, H. (2011). The obesity epidemic: from the

environment to epigenetics - not simply a response to dietary manipulation in a

thermoneutral environment. Front Genet, 2, 24.

Takahashi, J. S. (2004). Finding new clock components: past and future. J Biol Rhythms,

19(5), 339-347.

Takahashi, J. S., Hong, H. K., Ko, C. H., & McDearmon, E. L. (2008). The genetics of

mammalian circadian order and disorder: implications for physiology and disease.

Nat Rev Genet, 9(10), 764-775.

Tamura, E. K., Cecon, E., Monteiro, A. W., Silva, C. L., & Markus, R. P. (2009).

Melatonin inhibits LPS-induced NO production in rat endothelial cells. J Pineal

Res, 46(3), 268-274.

Tan, D. X., Manchester, L. C., Fuentes-Broto, L., Paredes, S. D., & Reiter, R. J. (2011).

Significance and application of melatonin in the regulation of brown adipose

tissue metabolism: relation to human obesity. Obes Rev, 12(3), 167-188.

Terron, M. P., Delgado, J., Paredes, S. D., Barriga, C., Reiter, R. J., & Rodriguez, A. B.

(2009). Effect of melatonin and tryptophan on humoral immunity in young and

old ringdoves (Streptopelia risoria). Exp Gerontol, 44(10), 653-658.

Thaler, J. P., Yi, C. X., Schur, E. A., Guyenet, S. J., Hwang, B. H., Dietrich, M. O., et al.

(2012). Obesity is associated with hypothalamic injury in rodents and humans

(vol 122, pg 153, 2012). Journal of Clinical Investigation, 122(2), 778-778.

Thomas, B. B., Oommen, M. M., & Ashadevi, O. (2001). Constant light and blinding

effects on reproduction of male South Indian gerbils. J Exp Zool, 289(1), 59-65.

Page 252: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

236

Tosini, G., & Menaker, M. (1998). The clock in the mouse retina: melatonin synthesis

and photoreceptor degeneration. Brain Res, 789(2), 221-228.

Tremblay, M. S., Esliger, D. W., Copeland, J. L., Barnes, J. D., & Bassett, D. R. (2008).

Moving forward by looking back: lessons learned from long-lost lifestyles. Appl

Physiol Nutr Metab, 33(4), 836-842.

Tsai, J. Y., Villegas-Montoya, C., Boland, B. B., Blasier, Z., Egbejimi, O., Gonzalez, R.,

et al. (2012). Influence of dark phase restricted high fat feeding on myocardial

adaptation in mice. J Mol Cell Cardiol.

Tunez, I., del Carmen Munoz, M., Feijoo, M., Valdelvira, M. E., Rafael Munoz-

Castaneda, J., & Montilla, P. (2003). Melatonin effect on renal oxidative stress

under constant light exposure. Cell Biochem Funct, 21(1), 35-40.

Turek, F. W. (2007). From circadian rhythms to clock genes in depression. Int Clin

Psychopharmacol, 22 Suppl 2, S1-8.

Turek, F. W., Joshu, C., Kohsaka, A., Lin, E., Ivanova, G., McDearmon, E., et al. (2005).

Obesity and metabolic syndrome in circadian Clock mutant mice. Science,

308(5724), 1043-1045.

Vadas, M. A., Miller, J. F., Gamble, J., & Whitelaw, A. (1975). A radioisotopic method

to measure delayed type hypersensitivity in the mouse. I. Studies in sensitized and

normal mice. Int Arch Allergy Appl Immunol, 49(5), 670-692.

van Amelsvoort, L. G., Schouten, E. G., & Kok, F. J. (1999). Duration of shiftwork

related to body mass index and waist to hip ratio. Int J Obes Relat Metab Disord,

23(9), 973-978.

Page 253: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

237

van der Horst, G. T., Muijtjens, M., Kobayashi, K., Takano, R., Kanno, S., Takao, M., et

al. (1999). Mammalian Cry1 and Cry2 are essential for maintenance of circadian

rhythms. Nature, 398(6728), 627-630.

Van der Meer, E., Van Loo, P. L., & Baumans, V. (2004). Short-term effects of a

disturbed light-dark cycle and environmental enrichment on aggression and

stress-related parameters in male mice. Lab Anim, 38(4), 376-383.

Vetter, C., Juda, M., & Roenneberg, T. (2012). The influence of internal time, time

awake, and sleep duration on cognitive performance in shiftworkers. Chronobiol

Int, 29(8), 1127-1138.

Vinogradova, I. A. (2007). [Effect of different light regimens on the development of

metabolic syndrome of aging rats]. Adv Gerontol, 20(2), 70-75.

Vinogradova, I. A., Anisimov, V. N., Bukalev, A. V., Semenchenko, A. V., &

Zabezhinski, M. A. (2009). Circadian disruption induced by light-at-night

accelerates aging and promotes tumorigenesis in rats. Aging-Us, 1(10), 855-865.

Vollmers, C., Gill, S., DiTacchio, L., Pulivarthy, S. R., Le, H. D., & Panda, S. (2009).

Time of feeding and the intrinsic circadian clock drive rhythms in hepatic gene

expression. Proc Natl Acad Sci U S A, 106(50), 21453-21458.

Vyas, M. V., Garg, A. X., Iansavichus, A. V., Costella, J., Donner, A., Laugsand, L. E., et

al. (2012). Shift work and vascular events: systematic review and meta-analysis.

Bmj, 345, e4800.

Page 254: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

238

Wang, X. S., Armstrong, M. E., Cairns, B. J., Key, T. J., & Travis, R. C. (2011). Shift

work and chronic disease: the epidemiological evidence. Occup Med (Lond),

61(2), 78-89.

Wang, Y., Beydoun, M. A., Liang, L., Caballero, B., & Kumanyika, S. K. (2008). Will all

Americans become overweight or obese? estimating the progression and cost of

the US obesity epidemic. Obesity (Silver Spring), 16(10), 2323-2330.

Wang, Z., Lin, A., Yuan, Q., Zhou, W., Zhang, W., & Yang, X. J. (2011). Attraction of

night-migrating birds to light-blue structures causes mass bird deaths. Environ Sci

Technol, 45(24), 10296-10297.

Wangsness, P. J., Dilettuso, B. A., & Martin, R. J. (1978). Dietary effects on body

weight, feed intake and diurnal feeding behavior of genetically obese rats. J Nutr,

108(2), 256-264.

Weil, Z. M., Karelina, K., Su, A. J., Barker, J. M., Norman, G. J., Zhang, N., et al.

(2009). Time-of-day determines neuronal damage and mortality after cardiac

arrest. Neurobiol Dis, 36(2), 352-360.

Weil, Z. M., Martin, L. B., 2nd, & Nelson, R. J. (2006). Photoperiod differentially affects

immune function and reproduction in collared lemmings (Dicrostonyx

groenlandicus). J Biol Rhythms, 21(5), 384-393.

Weil, Z. M., Norman, G. J., DeVries, A. C., & Nelson, R. J. (2008). The injured nervous

system: a Darwinian perspective. Prog Neurobiol, 86(1), 48-59.

Page 255: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

239

Weinrauch, Y., Abad, C., Liang, N. S., Lowry, S. F., & Weiss, J. (1998). Mobilization of

potent plasma bactericidal activity during systemic bacterial challenge. Role of

group IIA phospholipase A2. J Clin Invest, 102(3), 633-638.

Weisberg, S. P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R. L., & Ferrante, A.

W., Jr. (2003). Obesity is associated with macrophage accumulation in adipose

tissue. J Clin Invest, 112(12), 1796-1808.

Weiss, A., Xu, F., Storfer-Isser, A., Thomas, A., Ievers-Landis, C. E., & Redline, S.

(2010). The association of sleep duration with adolescents' fat and carbohydrate

consumption. Sleep, 33(9), 1201-1209.

Welsh, D. K., Logothetis, D. E., Meister, M., & Reppert, S. M. (1995). Individual

neurons dissociated from rat suprachiasmatic nucleus express independently

phased circadian firing rhythms. Neuron, 14(4), 697-706.

Westman, J. A., Ferketich, A. K., Kauffman, R. M., MacEachern, S. N., Wilkins, J. R.,

3rd, Wilcox, P. P., et al. (2010). Low cancer incidence rates in Ohio Amish.

Cancer Causes Control, 21(1), 69-75.

WHO. (2000). Obesity: preventing and managing the global epidemic. Report of a WHO

consultation.

Wideman, C. H., & Murphy, H. M. (2009). Constant light induces alterations in

melatonin levels, food intake, feed efficiency, visceral adiposity, and circadian

rhythms in rats. Nutr Neurosci, 12(5), 233-240.

Willner, P. (1997). Stress and depression: Insights from animal models. Stress Medicine,

13(4), 229-233.

Page 256: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

240

Willner, P., Muscat, R., & Papp, M. (1992). An animal model of anhedonia. Clin

Neuropharmacol, 15 Suppl 1 Pt A, 550A-551A.

Wills-Karp, M. (1999). Immunologic basis of antigen-induced airway

hyperresponsiveness. Annu Rev Immunol, 17, 255-281.

Wynne, A. M., Henry, C. J., Huang, Y., Cleland, A., & Godbout, J. P. (2010). Protracted

downregulation of CX3CR1 on microglia of aged mice after lipopolysaccharide

challenge. Brain Behav Immun, 24(7), 1190-1201.

Wyse, C. A., Selman, C., Page, M. M., Coogan, A. N., & Hazlerigg, D. G. (2011).

Circadian desynchrony and metabolic dysfunction; did light pollution make us

fat? Med Hypotheses, 77(6), 1139-1144.

Xu, B., Kalra, P. S., Farmerie, W. G., & Kalra, S. P. (1999). Daily changes in

hypothalamic gene expression of neuropeptide Y, galanin, proopiomelanocortin,

and adipocyte leptin gene expression and secretion: effects of food restriction.

Endocrinology, 140(6), 2868-2875.

Xu, H., Barnes, G. T., Yang, Q., Tan, G., Yang, D., Chou, C. J., et al. (2003). Chronic

inflammation in fat plays a crucial role in the development of obesity-related

insulin resistance. J Clin Invest, 112(12), 1821-1830.

Yamaguchi, S., Isejima, H., Matsuo, T., Okura, R., Yagita, K., Kobayashi, M., et al.

(2003). Synchronization of cellular clocks in the suprachiasmatic nucleus.

Science, 302(5649), 1408-1412.

Page 257: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

241

Yamazaki, S., Numano, R., Abe, M., Hida, A., Takahashi, R., Ueda, M., et al. (2000).

Resetting central and peripheral circadian oscillators in transgenic rats. Science,

288(5466), 682-685.

Yang, X., Downes, M., Yu, R. T., Bookout, A. L., He, W., Straume, M., et al. (2006).

Nuclear receptor expression links the circadian clock to metabolism. Cell, 126(4),

801-810.

Yin, L., Wu, N., Curtin, J. C., Qatanani, M., Szwergold, N. R., Reid, R. A., et al. (2007).

Rev-erbalpha, a heme sensor that coordinates metabolic and circadian pathways.

Science, 318(5857), 1786-1789.

Young, M. R., Matthews, J. P., Kanabrocki, E. L., Sothern, R. B., Roitman-Johnson, B.,

& Scheving, L. E. (1995). Circadian rhythmometry of serum interleukin-2,

interleukin-10, tumor necrosis factor-alpha, and granulocyte-macrophage colony-

stimulating factor in men. Chronobiol Int, 12(1), 19-27.

Zanquetta, M. M., Correa-Giannella, M. L., Giannella-Neto, D., Alonso, P. A.,

Guimaraes, L. M., Meyer, A., et al. (2012). Expression of clock genes in human

subcutaneous and visceral adipose tissues. Chronobiol Int, 29(3), 252-260.

Zee, P. C., Attarian, H., & Videnovic, A. (2013). Circadian rhythm abnormalities.

Continuum (Minneap Minn), 19(1 Sleep Disorders), 132-147.

Zeman, M., Szantoova, K., Stebelova, K., Mravec, B., & Herichova, I. (2009). Effect of

rhythmic melatonin administration on clock gene expression in the

suprachiasmatic nucleus and the heart of hypertensive TGR(mRen2)27 rats. J

Hypertens, 27 Suppl 6, S21-26.

Page 258: PHYSIOLOGICAL CONSEQUENCES OF CIRCADIAN DISRUPTION BY NIGHTTIME LIGHT

242

Zhang, X., Zhang, G., Zhang, H., Karin, M., Bai, H., & Cai, D. (2008). Hypothalamic

IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and

obesity. Cell, 135(1), 61-73.

Zola-Morgan, S., Squire, L. R., & Amaral, D. G. (1986). Human amnesia and the medial

temporal region: enduring memory impairment following a bilateral lesion limited

to field CA1 of the hippocampus. J Neurosci, 6(10), 2950-2967.

Zucker, I. (1971). Light-dark rhythms in rat eating and drinking behavior. Physiol Behav,

6(2), 115-126.