<<

UNIVERSITY OF CINCINNATI

Date:______

I, ______, hereby submit this work as part of the requirements for the degree of: in:

It is entitled:

This work and its defense approved by:

Chair: ______

Novel fuel sensing mechanisms in the regulation of food intake

A dissertation submitted to the

Division of Research and Advanced Studies of the University of Cincinnati

in partial fulfillment of the requirements for the degree of

DOCTORATE OF PHILOSOPHY (Ph.D.)

in the Graduate Program in Neuroscience of the College of Medicine

May 22, 2006

by Karine Proulx

B.S., Université Laval, 2000 M.S., McGill University, 2002

Committee Chair: Randy J. Seeley, Ph.D. ABSTRACT

An emerging model is that CNS fuel sensors, such as AMP kinase (AMPK) and

the mammalian target of rapamycin (mTOR), integrate signals from stored and

immediately available fuels, and in turn regulate food intake. The experiments described in this dissertation focus on novel CNS fuel sensing mechanisms by which fatty acid derivatives and compounds that affect fatty acid metabolism modulate food intake.

Oleoylethanolamide (OEA), a derivative of synthesized in the intestine following refeeding, reduces food intake. OEA shares similarities with other nutrient- derived hormones that signal energy status to the CNS, but its mechanisms of action remain unclear. We tested whether OEA-induced anorexia occurs through specific interactions with hormones that modulate food intake through CNS pathways involved in energy homeostasis, or is rather due to unspecific behaviors. Our results indicate that

OEA suppresses feeding without causing visceral illness, and that neither ghrelin, PYY,

GLP-1, apo A-IV nor CCK play a critical role in this effect. OEA is not the only fatty acid metabolism related compound that suppresses food intake. C75 is a fatty acid synthase inhibitor that inhibits food intake via direct actions in the CNS. MTOR, a member of the phosphatidylinositol kinase-related protein kinase family, plays a crucial role in nutrient sensing and the control of protein synthesis. Its inhibition stimulates food intake in rats. We hypothesized that C75-induced anorexia depends on its ability to activate the mTOR pathway in the hypothalamus. Consistent with this hypothesis, C75 increases the phosphorylation of key components of the mTOR pathway and inhibitors of mTOR reverse C75-induced anorexia. Previous work showed that C75 is ineffective when rats are on a ketogenic diet. Consistent with a role for mTOR in mediating the

- I - effects of C75, C75-induced anorexia and activation of the mTOR pathway were abolished in rats maintained on a ketogenic diet. Together, these data argue that neuronal nutrient metabolism is monitored by CNS fuel sensors and contribute to the regulation of food intake.

- II - - III - ACKNOWLEDGEMENTS

Randy: Maybe one of the reasons you always say that you trust my scientific judgment is

that you know you are the one who made me build one over the last four years! As

challenging as those conversations in your office might have been sometimes, I can

honestly say that they are what made me the young scientist I am today. You taught me

how to develop a critical sense, stand up for my ideas and, most importantly, build some

confidence throughout the whole process. I now leave this lab feeling strong and ready to

move to the next step. Thank you also to you and Steve, for showing us what “work hard-

play hard” really means, allowing the passion for science to remain even in tougher

times!

Steve: I remember the very first question Steve asked me during my interview: “Why

would someone coming from such a beautiful city as Montreal (as he said it with his classic Woodsilian French accent!) would want to live in Cincinnati?”. While such a

broad question could have been intimidating coming from someone like Steve Woods, I

exactly knew the answer. My motivation obviously didn’t come from the attractions of

the city (no offense, but that has never been a secret to anyone!). After a few mornings

that I popped my head in his office for questions, he suggested we schedule a weekly

meeting, where we would talk about the philosophy of research. Every time we met from

then until now confirmed that I made the right decision in coming to Cincinnati, and that

I have found exactly the training environment I was looking for. In addition to be a great

mentor, you and Nancy have also been a great second family by making me feel home

during celebrations like Thanksgiving and Easter. Thank you!

- IV - Committee members: Drs. Herman, Jandacek and Williams, I learned so much from our discussions during and outside of our official committee meetings. I truly felt that you were supportive of my research and cared about my career. Our program is blessed to have faculty members like you, who despite their busy schedules maintain an “open- door” policy. You have always made me feel welcome when I needed help for either technical issues or last minute recommendation letters and I want to thank you for that.

Beth: Where should I start, how can I put all those feelings into words? We’ve started together and are finishing it together. We shared so much! You were always there for me, in good as well as bad/ “life threatening” times (I will always remember our infamous

New Orleans episode and how you helped me keep my hot temper low after that guy crashed my car!). I knew I could rely on you whether for intellectual, moral or

“technical” support. I am sure you’ll be a great and successful scientist. I’ll be there to celebrate your tenure-track position on your thirtieth birthday! I wish you the best of luck for your post-doc and I am looking forward to your visit in the UK!

April and Torsten: Thank you for taking me under your wings and getting me started in the lab. You guys were there for my first rat bite, as well as those crazy tail-bleeding sessions at 7h00 AM, after a night of too much celebration at Randall’s house, or at 1h00

AM, because of a light-dark cycle that we chose to accommodate my classes! We can only laugh about it now!

Daniela: You have been not only an awesome colleague, but also a great friend. You were there to celebrate every happy moment, and to support me through all the troubles of this thesis (despite what the ratio between those two might have been, I don’t care anymore!). I want to say how much it meant to me to be able to share ideas and have

- V - scientific conversations that always remained authentic and free of any competition. I say goodbye for now, but I have the feeling it is only temporary. I will probably be meeting

Prof. Cota for a cappuccino in some European café pretty soon!

The Tschoep’s lab: Matthias, Tamara, Paul and Diego: You guys made the OEA

paper happened. I still think that we shouldn’t have had to spend so much time figuring

out how something with a name like “The Clever System” works! Despite how ridiculous

it was to play with the red light until we got the proper “Chinese shadows” to detect the

rat in its cage, I have to admit we had fun! I’ll keep great memories of the time we spent

together in and outside of GRI. Like I said for Daniela, I am sure that our roads will cross

again in the near future my dear European friends.

My non-official committee members: Probably the result of working in such a collaborative environment as the one within the Obesity Research Center and the GRI:

Randall Sakai: Thank you for professional and personal support throughout my time at

UC. Among the many things I will remember from my interactions with you are when

you were there to get me coffee and calm me down the morning of my first big talk at

SFN, your “PR” and networking qualities and, of course, the parties and wine at your

house!

David D’Alessio: Although not an official member of my committee, you surely behaved

like one! You contributed to solving so many issues from the ADX, to OEA to C75

project. I really appreciated you keeping your door opened for my spontaneous questions,

I learned a lot from you.

Patrick Tso: Your enthusiasm for science is contagious and your approach to scientific

questions very unique. I learned a lot from those drawings on the board next to your

- VI - office! Thank you for contributing to my OEA project, and also to regularly check on my

future post-doc career.

Kay Ellis: It was always fun to go drop those plasma samples to you Kay. It was the

occasion to get a little girlie chat, and laugh at how many tubes there were in those boxes!

Matt and Debbie: Although you guys had already left by the time I’ve started working

on C75, you generously offered your help. Sorry for making you go through messy lab

books (mine are not any better!) or trying to find on which paper towel you wrote the

design of the experiment on that day! Thanks for sharing your knowledge of the FAS

system and your insightful discussions.

Past and present members of the “team room” ( Beth, Daniela, Claire, Lynda,

Dong-Hoon, Jason, Darleen, April, Ryuichi, Debbie D., Brad, Debbie C., Stephen,

Matt and Koroh): Despite what Randy says about its lack of light and its small size, this

“team-room” has probably been the most enriching environment I have ever been in. I

grew up so much in that room over the last 4 years, not only as a better scientist, but also

as a better individual. I remember when Koroh and I used to be the only people with an

accent in the first few months I was here, and when all the American fellows could make

fun of how I pronounced “buffet” during my first lab meeting… Not so much anymore!

This room soon became the U.N. as Randy says, bringing people with different cultures

and training backgrounds. I have always been so amazed how such a big group of people, with so many different personalities, can get along and work so well together. It is pretty

rare to find such a big lab free of competition. We always stand up together as a group

and support each other in good and bad times. I couldn’t have wished for better

colleagues and friends, and can only hope to find such an amazing training environment

- VII - during my post-doc. You are all wonderful persons! I’m looking forward to seeing you

again at meetings, and hopefully collaborate when we all become independent researchers.

Neuroscience friends (Beth, Mary, Dennis, Nicole, Susan, Miyuki, Kellie, Alier,

Rick, John D., Derrick, and Brian M.): I couldn’t have made it throughout graduate school without you guys! I felt that we were all on the same boat and there to support each other. Despite what I said earlier about Cincinnati and its attractions, you are the ones who made the difference. You can be in the most entertaining city in the world, without great friends, it doesn’t mean anything. I was glad to be there for those of you who defended already. While I probably won’t be here to celebrate everyone else’s defense, I’ll definitely make sure I’ll have a drink “à votre santé”. Good luck and see you soon!

Deb: You are certainly among the most dedicated and efficient person I met here. From

my first day at UC, when you showed me around Clifton, to the time when you called the

guy who screwed up my application for a US social security number…nothing stops you!

Thanks Deb!

Family and friends: Thank you for supporting my decision in coming to the US. I remember how afraid you were I would not want to come back. You can be reassured, both Randy and Steve would tell you that I have never lost an ounce of my culture

“québécoise” or my French in four years, which by the way, Randy kindly reminded me

when he first read my dissertation! As much as I loved my experience here, I am looking

forward to move back to Canada soon and be closer to you. Merci d’avoir supporté ma

décision de venir étudier aux Etats-Unis. J’ai tellement appris ici en quatre ans mais

- VIII - n’ayez crainte, je n’ai jamais perdu une once de ma culture québécoise! Je vous aime très fort et j’ai hâte de me retrouver parmi vous!

Ian: A ta demande, et pour célébrer le fait que je t’ai officiellement déclaré bilingue, je vais écrire le message en Français mon chéri. Merci de m’avoir compris et encouragé tout au long de ce processus. Merci aussi de m’avoir donné un coup de pied quand je commençais à me laisser aller. Tu avais raison, peu importe ce qui arrive, tout fini

éventuellement par entrer dans l’ordre. Aujourd’hui, je suis fière d’être restée forte malgré les difficultés, et ça c’est grâce à toi. J’ai hâte de débuter notre nouvelle aventure ensemble, enfin fini la distance! Je t’aime.

- IX - TABLE OF CONTENTS

ABSTRACT……………………………………………………………………….………I

ACKNOWLEDGEMENTS……………………………………………………………...IV

TABLE OF CONTENTS…………………………………………………….…………...1

LIST OF TABLES AND FIGURES……….………………..…………………………....2

LIST OF ABBREVIATIONS………………………………………………………...... 4

CHAPTER 1: INTRODUCTORY LITERATURE REVIEW……………………………6

CHAPTER 2: MECHANISMS OF (OEA)-INDUCED CHANGES IN FEEDING BEHAVIOR AND MOTOR ACTIVITY...……36 I. Abstract……………………………………………………...37

II. Introduction……………..…………………………………..38

III. Materials and Methods……………………………………...41

IV. Results……………..………………………………………..49

V. Discussion……….……………………………………….…53

CHAPTER 3: C75 REGULATES FOOD INTAKE VIA ACTIVATION OF HYPOTHALAMIC MTOR SIGNALING………………….………………71 I. Abstract……………………………………………………...72

II. Introduction……………..…………………………………...74

III. Materials and Methods………………………………………78

IV. Results……………..………………………………………...84

V. Discussion……….………………………………………...... 87

CHAPTER 4: GENERAL DISCUSSION……...…………………………...100

REFERENCES……………………………………………………………...112

APPENDICES.……………………………………………………………...126

- 1 - LIST OF TABLES AND FIGURES

Chapter 2:

Figure 1. Mean + SEM food intake at different time points (A), and 24 h (B) after an ip

injection of vehicle (VEH) or three doses of OEA in 24-h fasted rats…………………..61

Figure 2. Mean + SEM preference ratio of flavors previously paired with an ip injection of saline (SAL), lithium chloride (LiCl), VEH or OEA (20 mg/kg)…………………….62

Figure 3. Mean + SEM 2-h sodium intake after an ip injection of SAL, LiCl, VEH or

OEA (20 mg/kg) in sodium-depleted rats………..………………………………… …...63

Figure 4. Mean + SEM time the rats spent in the side previously paired with an ip

injection of SAL, LiCl, VEH or OEA (20 mg/kg)………………………………………64

Figure 5. Mean + SEM heat expenditure at different time points after an ip injection of

either VEH or OEA (20 mg/kg) (A), and mean + SEM heat expenditure per hour during

the 12-h dark phase (B) in fasted rats…………………………………………………...65

Figure 6. Mean + SEM time the rats displayed an extended posture in the 2 h following

an ip injection of either VEH or OEA (20 mg/kg) (A), and either VEH or capsaicin

(CAPS) (1 mg/kg) (B)…………………………………………………………………..66

Figure 7. Mean + SEM time the rats spent in inactivity (A), number of times the rats crossed a square (B), and number of times the rats either reared or groomed (spontaneous activity) (C) after an ip injection of OEA (20 mg/kg), CAPS (1 mg/kg), Wy-14643 (40 mg/kg) or their respective VEH………………………………………………………...67

Figure 8. Mean + SEM plasma concentrations of ghrelin (A), PYY (B), GLP-1 (C), and

apo A-IV (D) at different time points after an ip injection of either VEH or OEA (20

mg/kg)…………………………………………………………………………………..68

- 2 - Figure 9. Mean + SEM food intake at different time points after an ip injection of either

SAL or lorglumide (LORGL) (1mg/kg), followed by an ip injection of either VEH or

OEA (20 mg/kg) (A), and either SAL or CCK-8 (4 μg/kg) (B) in fasted rats…………...69

Chapter 3:

Table 1. Rat Q-PCR Primer Sequences …………………………………………………93

Figure 1. I3vt administration of rapamycin (25 µg in 1 µl DMSO) prevents the effects of

C75 (50 µg in 3 µl RPMI) on food intake (A) and on body weight (B)…………………95

Figure 2. C75 (30 µg in 2 µl RPMI, i3vt) increases hypothalamic mTOR signaling:

Representative Western blots from RPMI- or C75-treated rats and quantification by

image analysis of hypothalamic phosphorylation of mTOR (A), S6K1 (B) and S6 (C)..96

Figure 3. C75 (30 µg in 2 µl RPMI) does not reduce caloric intake in rats given access to

saccharin alongside with the ketogenic diet, but do so with sucrose……………………97

Figure 4. C75 (30 µg in 2 µl RPMI) does not increase hypothalamic S6K1 and S6 phosphorylation in rats given access to saccharin while maintained on a ketogenic diet, but do so in rats receiving sucrose alongside with the diet. Representative Western blots from RPMI- or C75-treated rats from the saccharin and sucrose groups and quantification by image analysis of hypothalamic phosphorylation S6K1 (A) and S6 (B)…………….98

Figure 5. C75 significantly reduced NPY mRNA levels in the hypothalamus as

compared to RPMI controls, as did rapamycin………………………………………….99

- 3 - LIST OF ABBREVIATIONS

α-MSH Alpha-melanocyte stimulating hormone ACC Acetyl-CoA carboxylase AgRP Agouti related protein AICAR Aminoimidazole-4-carboxamide ribonucleoside AMPK AMP-activated protein kinase ANOVA Analysis of variance Apo A-IV Apolipoprotein A-IV ATP BMI Body mass index CAPS Capsaicin CART Cocaine and amphetamine related transcript CB1 and 2 Cannabinoid receptors 1 and 2 CCK Cholecystokinin CEACAM-1 Cell adhesion molecule 1 CNS Central nervous system CPA Conditioned place aversion CPT-1 Carnitine palmitoyl-CoA transferase-1 CTA Conditioned taste aversion DMSO Dimethyl sulfoxide FAAH Fatty acid amide hydrolase FAE Fatty acid ethanolamide FAS Fatty acid synthase FRB FKB12-rapamycin binding GE Glucose-excited GI Glucose-inhibited GLP-1 Glucagon-like peptide 1 i3vt Intra-3rd-cerebroventricular LCFA-CoA Long-chain fatty acid-CoA LiCl Lithium chloride

- 4 - LORGL Lorglumide MC4-R Melanocortin receptor-4 MCD Malonyl-CoA decarboxylase mTOR Mammalian target of rapamycin NPY Neuropeptide Y NPY-Y1 NPY receptor 1 NPY-Y5 NPY receptor 5 OEA Oleoylethanolamide POMC Proopiomelanocortin PPAR-α Peroxisome proliferator-activated receptor-alpha PVN Paraventricular nucleus of the hypothalamus PYY peptide YY Q-PCR Quantitative polymerase chain reaction RAPA Rapamycin S6 S6 ribosomal protein S6K1 S6 kinase 1 SAL Saline SCD-1 Stearoyl-CoA desaturase-1 SEM Standard error of the mean TRPV1 Capsaicin receptor UCP Uncoupling protein VEH Vehicle WAT White adipose tissue

- 5 - CHAPTER 1: INTRODUCTORY LITERATURE REVIEW

- 6 - 1. Obesity: general overview.

Obesity is associated with substantially increased mortality from cardiovascular and cerebrovascular disease, diabetes and certain cancers. It also results in severe morbidity from musculoskeletal, gastrointestinal, psychiatric and reproductive diseases and is associated with lowered quality of life, self-esteem and socio-economic performance (1).

The prevalence of obesity is increasing rapidly throughout the world and the disorder is now recognized as a major global public health threat. While these changes in the prevalence of obesity in the last 10-50 years are clearly driven by secular changes in physical activity and the inexpensive availability of high-caloric foods, it is also clear that inherited factors play a major role in determining adiposity. Family, twin and adoption studies all indicate that adiposity is highly heritable and the estimated genetic contribution to Body Mass Index (BMI) ranges between 50 and 90 percent (2). Thus, the environmental factors that are driving the increase in obesity prevalence are operating against the background of a biological system whose susceptibility or resistance to such environmental stressors has a strong inherited component (2). This might be the result of evolution, which could have selected over the millennia for genes and redundant mechanisms that allowed the body to efficiently store energy in periods of “feast” for survival during times of “famine” (3). Understanding the mechanisms by which these genes regulate body adiposity is fundamental to developing new therapeutic strategies for the treatment of obesity and its related metabolic disorders. The following paragraphs will review the current knowledge of the homeostatic mechanisms that regulate food intake and body weight in mammals.

- 7 - 2. Energy homeostasis.

The interaction of genetic and environmental factors dictates an individualized

level of body adiposity that is defended by a powerful process known as energy

homeostasis. Obesity arises as the result of a sustained mismatch where caloric intake

exceeds caloric expenditure. However, despite what the growing obesity epidemic might

argue, under most circumstances the balance between caloric intake and expenditure is

regulated with tremendous precision since mammals defend a very stable body weight

over time. For example, an average weight male who consumes 900,000 calories in a

year would need to consume 4000 more calories than he burns to gain one pound in that

year, the equivalent of 11 calories (or one potato chip!) a day (4). Statistics indicate that the average increase of weight within the adult U.S. population is actually less than one pound per year (5). Back to our example, this means the organism, and especially the central nervous system (CNS), is accurate at more than 99.5% in matching energy intake to energy expenditure (4).

This example highlights two points with regard to energy balance. First, the

amount of inter-individual variability is relatively large, and becomes obvious when

comparing the average yearly weight gain (5) with the actual number of obese individuals

within the population (6). Second, this would suggest the existence of a regulatory system

where deviations from the defended body adiposity level trigger a signal that can be

monitored by a biochemical sensor. This sensor would then induce adaptive responses in

food intake and metabolic processes that affect fuel oxidation, in order to ensure that body weight remains relatively stable over time (7). Assuming this is true, the question

- 8 - then becomes what is this signal and how is it detected? More importantly, what is the

purpose of this complex regulatory system?

3. Fuel sensing within the central nervous system (CNS).

3. 1. Signals of stored and immediately available fuels.

Not surprisingly, the current data indicate that there is not a single signal. Rather,

growing lists of hormones and nutrients have been proposed to indicate the status of fuels

available to cells for the generation of energy/ATP (8). These fuels can either be stored in

adipose tissue or be derived from recently ingested food, and therefore be immediately available. Among the best studied and characterized signals of stored fuels are leptin and insulin. These hormones, synthesized in adipose tissue and pancreas respectively, circulate in proportion to body fat (9). They inform the CNS that adipose stores are increased and that the organism is therefore in a state of positive energy balance (10, 11).

In addition to negative feedback, the CNS also receives positive feedback when energy stores are low. This role is accomplished by ghrelin, a stomach-derived hormone whose circulating levels are inversely correlated to those of leptin (12). Ghrelin increases food intake and adiposity when directly injected into the brain (13), whereas leptin (14) and insulin (15) do the opposite. In addition to monitoring the status of stored energy, the

CNS also keeps track of immediately available food-derived nutrients such as glucose, fatty acids and amino acids. Infusion of any of the above mentioned substrates in the vicinity of the hypothalamus reduces food consumption and expression of the orexigenic neuropeptide Y (NPY) (16-19), suggesting that they can be sensed by the brain.

- 9 - 3. 2. POMC/CART and NPY/AgRP neurons: a site of integration.

Signals reflecting stored and currently available fuels are detected for the most

part in the arcuate nucleus, where receptors for leptin (20), insulin (21, 22) and ghrelin

(23) are highly expressed, as well as transporters for glucose (24) and amino acids (A.

Sweatt, personal communications). At least two distinct populations of neurons are reciprocally regulated by these signals. One population synthesizes the large precursor peptide proopiomelanocortin (POMC), which can be cleaved into a number of biologically active peptides, including α-melanocyte stimulating hormone (α-MSH).

Central administration of this peptide or its synthetic analogues potently reduces food

consumption and causes body weight loss in rodents (25, 26), presumably through

activation of the melanocortin receptor 4 (MC4-R). It is interesting to note that mutation

in the MC4-R gene is the most common monogenic cause of obesity in humans (27). The

second set of neurons in the arcuate nucleus synthesizes both Agouti-Related Protein

(AgRP) and NPY (28, 29). In contrast to α-MSH, central injection of AgRP or NPY

induces profound hyperphagia and weight gain due to inverse agonism of MC4-R (30,

31) in the case of AgRP, and activation of the NPY-Y1 and NPY-Y5 receptors in the case

of NPY (32).

As will be discussed later in this review, signals of both stored and currently

available fuels appear to be integrated within common intracellular signaling cascades in

NPY/AgRP and POMC/CART neurons (33, 34). These neurons in turn send feedback

signals to modify food intake and metabolic processes so that energy balance is defended.

This concept of common integration is a relatively novel one. For a long time, this area of

- 10 - neuroscience research was dominated by two opposing views, each stating that the

hypothalamus monitors the levels of one signal and adjusts food intake accordingly. For

J. Mayer, the father of the glucostatic theory (35), this signal was the storage and

metabolism of glucose, whereas for G. Kennedy and the proponents of the lipostatic

theory, the signal had to be lipid storage and use (36).

Studies have demonstrated over the last 50 years that each of these hypotheses

have their limitations and caveats (7, 33, 37). Novel concepts have emerged concerning

the neuronal mechanisms that regulate food intake. This led to the working hypothesis behind the experiments of this thesis, which is that signals from both stored and immediately available fuels converge in the same intracellular signaling cascades in the

CNS, where they are monitored in parallel by biochemical fuel sensors that trigger adaptive feeding responses (7). The experiments described in this thesis focus on novel

CNS fuel sensing mechanisms by which fatty acids and fatty acid metabolism regulate food intake.

4. AMPK and mTOR: CNS fuel sensors.

Recent studies from Kahn’s laboratory and our own have identified two protein

kinases, AMP kinase (AMPK) (34) and the mammalian target of rapamycin (mTOR)

(19), that play a crucial role in integrating both types of signals. These proteins, commonly referred to as “fuel sensors”, were known to monitor the energy status of

peripheral cells, and regulate metabolic pathways in such a way to restore cellular energy

homeostasis (38-40). The observation that the these fuel sensors are phosphorylated at

- 11 - specific residues, an index of their activity, is regulated by the feeding status of the

animal such fuel sensors in hypothalamic neurons that have been linked to the control of

food intake and body weight, together with that their suggest an important function in the

regulation of energy balance at the whole organism level (19, 34, 41).

4. 1. AMPK.

4. 1. 1. AMPK structure and regulation.

AMPK, a cytosolic serine/threonine protein kinase, is comprised of three

subunits: the α-subunit contains the catalytic domain; the β-subunit, a glycogen-binding

domain; and the γ-subunit, the AMP-binding site. Decreases in the energy state of a cell,

as reflected by a decrease in the ratio of ATP/AMP, activate AMPK via several

mechanisms. This includes phosphorylation of its catalytic subunit on Thr172 by an

AMPK kinase (AMPKK), which is itself activated by AMP. AMP binding also inhibits

AMPK dephosphorylation by protein phosphatases. Importantly, all these effects are

antagonized by high concentrations of ATP and thus allowing the system to precisely

monitor changes in the ratio of ATP:AMP (38, 42).

4. 1. 2. AMPK as a peripheral fuel sensor.

AMPK serves as a cellular fuel sensor, and its activation is crucial to protect the viability of the cell in response to ATP depletion (38). During metabolic stress, such as

exercise or fasting, phosphorylated AMPK (Thr172) inactivates a number of regulatory

enzymes and inhibits the expression of several genes and proteins involved in

biosynthetic (anabolic) pathways (43). Consequently, energy-consuming (anabolic)

- 12 - processes are turned off, while ATP-producing (catabolic) ones are switched on. In peripheral tissues, this translates to an activation of signaling cascades that increase fatty acid and glucose oxidation, in order to restore ATP levels (44). In the brain, recent evidence suggests that activation of AMPK stimulates hypothalamic circuits that promote food intake (34, 41).

4. 1. 3. AMPK as a CNS fuel sensor.

In 2004, two independent research groups published data supporting a role for

AMPK as a CNS fuel sensor. They demonstrated that hypothalamic AMPK is regulated by the feeding status of the animal, as well as several hormonal and nutrient-derived signals (34, 41). For instance, fasting or AgRP, the endogenous inverse agonist of MC3-

R and MC4-R, increases AMPK activity in the same regions where refeeding, glucose and the MC3-R and MC4-R agonist MT-II reduce it (34). Furthermore, the potent orexigens ghrelin and cannabinoids (41, 45), in contrast to leptin and insulin (34), stimulate hypothalamic AMPK levels.

The response of neurons to energy depletion is similar to peripheral cells: switching on ATP-producing pathways. For a hypothalamic neuron whose primary role is to regulate energy balance, activation of AMPK translates into stimulation of food intake.

Consistent with this, genetic (34) or pharmacological (41) activation of AMPK using 5- aminoimidazole-4-carboxamide ribonucleoside (AICAR), in the CNS stimulates the expression of the orexigenic neuropeptides AgRP and NPY, which lead to increased feeding and body weight gain. Moreover, the anorectic effect of leptin is completely

- 13 - abolished, and its ability to reduce weight gain is significantly attenuated under those conditions (34). This indicates that suppression of hypothalamic AMPK is required for the catabolic effects of leptin.

Another action of AMPK relevant to the regulation of energy balance is stimulation of fatty acid oxidation in peripheral tissues. AMPK is an inhibitor of acetyl-CoA carboxylase (ACC), the rate-limiting enzyme for the conversion of acetyl-CoA to malonyl-CoA, and causes a reduction in malonyl-CoA concentration (46). This in turn releases the inhibition exerted by malonyl-CoA on carnitine palmitoyl-CoA transferase-1

(CPT-1), the enzyme responsible for the transport of long chain fatty acyl-CoAs into the mitochondria, and therefore promotes fatty acid oxidation (43). As will be discussed later, these enzymes are also present in hypothalamic neurons (47), where our lab and others have demonstrated their involvement CNS fuel sensing (48-51). Leptin-induced inhibition of AMPK regulates this pathway in the hypothalamus. Of interest to the present discussion is the recent report of Moran and colleagues, who proposed that leptin- induced modulation of intermediates of this pathway, specifically malonyl-CoA, is necessary for its anorectic action (52). In muscle and liver, leptin increases fatty acid oxidation through this pathway as well, via activation of the hypothalamic-sympathetic nervous system axis (53). The case of leptin-AMPK provides a perfect illustration of the role of “CNS fuel sensing”. Hypothalamic AMPK integrates signals of energy status, and regulate a cascade of biochemical events that affect both sides of the energy balance equation, in a way that ensures body adiposity remain within the defended range.

- 14 - 4. 2. mTOR.

4. 2. 1. mTOR structure and regulation.

Cells grow to a certain size before they divide (54). So for a cell and, therefore, a whole organism to grow requires the integration of both genetics and environmental cues including fuel availability. Thus, a complex system ensures that growth is tightly coordinated with the nutritional status of the organism. In addition to AMPK, the target of rapamycin (mTOR in mammals and dTOR in Drosophila) is crucial to this system.

MTOR, a member of the phosphatidylinositol kinase-related protein kinase (PIKK) family, plays a crucial role in nutrient sensing and the control of protein synthesis, angiogenesis, and cell cycle progression (55). It was originally identified and cloned in

1994, shortly after the discovery of the two yeast genes, TOR1 and TOR2 during a screen for resistance to the immunosuppressant drug rapamycin (56-58). MTOR is a 2549 amino acid long protein comprised of several domains, including the FRB (FKB12-rapamycin binding) to which rapamycin binds (59).

MTOR integrates signals from growth factors and nutrients, and in turn regulates the cell’s translational machinery in order to regulate cell growth, proliferation and death.

Insulin and amino acids have been shown to induce mTOR phosphorylation at Ser2448

(60), which correlates with activation of the mTOR pathway signaling, including phosphorylation of its downstream effectors S6 kinase (S6K) at Thr 389 and S6 ribosomal protein (S6) at Ser 240/244 (61).

- 15 - MTOR exists in two distinct multi-protein complexes: one containing a protein called raptor and another containing the protein rictor (62). The former complex is involved in nutrient sensing and therefore, the focus of the following section. During nutrient deprivation, the raptor-mTOR complex is stabilized in a manner that inhibits mTOR kinase activity, whereas raptor binds to the N-terminal domain of mTOR with high affinity and serves as a positive regulator of its activity in the presence of nutrients

(59). Raptor also serves as an adaptor protein that recruits mTOR substrates, such as the

40S ribosomal proteins S6 kinases 1 and 2 (S6K1 and S6K2), and is necessary for their phosphorylation by mTOR (63). The action of mTOR on its multiphosphorylated effectors is two-fold: activation of kinases and inhibition of the phosphatases that dephosphorylate them, phosphatase 2A (PP2A) in the case of S6K (64, 65), therefore ensuring a rapid and coordinated response. The mTOR-raptor complex is the best studied and understood of the two, mainly because it alone is inhibited by rapamycin (62).

Rapamycin, also known as sirolimus or RAPA, is a lipophilic macrolide produced by the soil bacterium Streptomyces hygroscopicus (66). This drug does not prevent the association of raptor to mTOR, but rather strongly destabilizes their interaction and inhibits mTOR kinase activity, ultimately causing dephosphorylation and deactivation of downstream components of the mTOR pathway (67). Rapamycin binds to its intracellular receptor FKBP12, which in turn binds the FRB (FKB12-rapamycin binding) domain of mTOR, and thereby inhibits it (59). In the clinic, rapamycin is prescribed as an anti- cancer drug, since it inhibits the growth of tumors, as well as an immunosuppressant (68).

- 16 - In the laboratory, this drug has been widely used to demonstrate the implication of

mTOR in multiple cellular processes.

4. 2. 2. mTOR as a peripheral fuel sensor.

Like AMPK, mTOR plays the role of fuel sensor. In contrast to AMPK, the

mTOR pathway is activated in energy-rich conditions, when the ratio ATP:AMP

increases (69). Consistent with its role of nutrient sensor, it therefore makes intuitive

sense for this kinase to regulate metabolism in a direction opposite to that of AMPK:

inhibiting energy-producing pathway and stimulating energy-consuming ones, such as

protein synthesis. This way, mTOR participates in a nutrient “checkpoint”, coupling

nutrient availability to downstream translation effectors, including S6K and S6 (59). This

way, amino acid levels can act as a feed-forward activator of proteins that activate protein synthesis and cellular growth.

Complete ablation of TOR is lethal (70). However, mice and Drosophila lacking

S6K1 survive. They have reduced cell size and are smaller than wild-type (71, 72). This

shows that TOR signaling, by controlling cell size, also controls the overall body size.

This effect applies to all cell types, so it is also reflected in pancreatic beta cell mass,

which could explain why S6K1-/- mice have reduced circulating insulin levels and are

glucose intolerant (73).

An interesting finding is that S6K1-/- mice are also protected against diet- and age-

induced insulin resistance and obesity, presumably due to increased fatty acid oxidation

- 17 - and mitochondrial density in muscle and adipose tissue (74). This phenotype supports the

fuel sensor role of mTOR signaling in peripheral tissue. It implies that under conditions

of fuel surplus, S6K1 signaling inhibits ATP-producing pathways like fatty acid

oxidation. From an energy homeostasis perspective, this would be the appropriate cellular

response under normal conditions. In conditions of chronic nutrient excess like under

high fat diet, however, the mTOR-S6K pathway may mediate maladaptive responses and

promote obesity (74). This is an important point that will be further emphasized in

chapter 4.

4.2.3. mTOR as a CNS fuel sensor.

The mTOR pathway is present in virtually all cell types. The case of peripheral

cells has been discussed above, and it appears that activation of mTOR-S6K1 signaling in

conditions of nutrient abundance favors energy storage. The pathway is also activated in

the CNS under such conditions. We have recently demonstrated in rats that mTOR plays

a similar role in the hypothalamus, where its phosphorylated form is found both in the

paraventricular (PVN) and the arcuate nucleus of the hypothalamus (19). In the arcuate,

which appears to be the only region where pmTOR levels are controlled by feeding

status, 90% of NPY neurons and 45% of POMC neurons express pS6K1, a downstream

target activated by pmTOR (reference). Interestingly, compounds that activate

hypothalamic S6K1, such as leptin and leucine, have also been reported to reduce NPY expression in this region (19, 75). Intra-third ventricular (i3vt) injection of rapamycin mimics the effects of fasting by inhibiting pS6K1 and causes short-term stimulation of food intake. Moreover, when administered at a sub-threshold dose that does not affect

- 18 - food intake on its own, it prevents the anorexigenic effects of centrally administered

leptin and leucine (19). These data make a case for hypothalamic mTOR signaling to play

a key role in the regulation of food intake. This novel hypothesis is also supported by

studies in Drosophila. Indeed, Drosophila expressing a dominant negative form of S6K

in the CNS eat more food than controls under ad libitum conditions, whereas the normal

hyperphagic response to fasting is reduced in mutants that express a constitutively active

form of the kinase (76).

5. A role for C75 in the regulation of energy balance.

Signals of stored fuels such as leptin, insulin and ghrelin regulate food intake

through modulation of CNS fuel sensors (19, 34, 41, 45). There is little evidence that

physiological fluctuation in the circulating levels of nutrients, including glucose and fatty

acid, controls meal initiation or termination (33, 77). Nonetheless, CNS fuel sensors

integrate these signals among the same intracellular cascade as those of stored fuels (19,

34). Moreover, recent studies indicate that C75, a fatty acid synthase (FAS) inhibitor that

alters neuronal glucose and fatty acid metabolism (78), inhibits hypothalamic AMPK

(79). Of note, this effect is necessary for C75 to reduce food intake (79). Together, these

data argue that neuronal nutrient metabolism is monitored by CNS fuel sensors and

contribute to the regulation of food intake.

5. 1. C75: a FAS inhibitor.

Originally used in cancer biology for its anti-tumor properties, cerulenin (2,3- epoxy-4-oxo-6-dodecadienoylamide), a natural antibiotic from Cephalosporium

- 19 - ceruleans and a FAS inhibitor, was found to cause profound weight loss in vivo (80).

Thus, it was a serendipitous discovery that modulation of the FAS system, a pathway involved in de novo fatty acid synthesis, can regulate energy balance. Interested in further

studying the mechanisms underlying this effect, but concerned by the toxicity and low

solubility of cerulenin, a group of researchers at the John Hopkins University developed

the synthetic analogue C75 (81). C75 is a potent selective inhibitor of FAS that

inactivates its β-ketoacyl-acyl synthase, enoyl reductase and thioesterase activities (82,

83). In vivo, this translates into a reduction of long chain fatty acids synthesis in the liver,

together with an increase in malonyl-CoA, a substrate of FAS. Intraperitoneal

administration of C75 causes profound, dose-dependent, anorexia and weight loss in rats

and mice (48, 49, 84). These effects are also observed with intraventricular (icv)

administration of much lower doses, suggesting a CNS action (48).

5. 2. Fatty acid metabolism in the CNS.

It has long been established that the FAS system is regulated by the nutritional

status of animals in lipogenic tissues such as liver and adipose (85). However, the CNS is

not lipogenic and derives most of its ATP from the oxidation of glucose rather than lipids

(86). Consequently, there was little reason to think that FAS played an important role in

the CNS. So, the discovery that FAS and the other enzymes involved in de novo fatty

acid synthesis are present in areas critical for the regulation of energy balance was quite

surprising (47, 87). Studies using C75, and other pharmacological as well as genetic tools

that modulate the activity of different enzymes of the FAS pathway in the CNS, suggest

that this system senses and integrates signals about the whole body nutrient status (48, 49,

- 20 - 84, 88, 89). This triggers adaptive responses in food intake and energy expenditure that ensures adiposity remains within the defended range over time.

5. 3. Potential mechanisms for the effects of C75 on food intake and energy expenditure.

The effects of C75 on the FAS system are thought to mimic a state of nutrient surplus, like if the animal would be refed (84). Modulation of fatty acid and glucose metabolism in the CNS play a crucial role in the regulation of energy homeostasis (33,

77). Evidence suggests that C75 reduces food intake by modulating these processes, and changing the expression of orexigenic and anorexigenic neuropeptides in the arcuate nucleus (51). There are a number of intracellular signals that have been proposed to mediate the anorexigenic effect of C75 in the CNS. These include increase in the levels of malonyl-CoA (84), long-chain fatty acid-CoA (LCFA-CoA) (88) and ATP (79), and the ratio of (glucose/fatty acid) utilization (49). In addition, some of these mechanisms might also be involved in the effects of C75 on energy expenditure (90, 91), although the latter is a controversial issue with reports of actual decrease after ip C75 (48). Each of these is discussed in the following paragraphs.

5. 3. 1. NPY/AgRP and POMC/CART neurons.

A striking aspect of C75 is its ability to block the normal hypothalamic response to fasting. A single injection of C75 (30 mg/kg, ip), despite suppressing 96% of food intake and causing 45% more weight loss than fasted controls, prevents the stimulation of orexigenic NPY/AgRP neurons (84) and the inhibition of POMC/CART neurons (92).

- 21 - C75-treated rats display hyperphagia in response to NPY injection, suggesting that the

NPY signaling pathway is intact in these animals (84). In addition to regulating CNS

pathways, C75 has also been reported to inhibit the secretion of ghrelin from the stomach

(93). Together, these data suggest that C75 stimulates a signal within the FAS pathway

that mimics the fed state. The organism is fooled into believing that it is in conditions of

energy surplus, and initiates the appropriate responses to restore energy balance: reducing

caloric intake (84) and possibly increasing caloric expenditure (90).

5. 3. 2. C75 and lipid metabolism.

5. 3. 2. 1. Malonyl-CoA.

Since the discovery of leptin, it became increasingly clear that NPY/AgRP- and

POMC/CART-secreting neurons integrate signals about the status of stored and readily

available energy (94). What remain poorly understood are the intracellular signals that inform these neurons about the energy status and influence their activity. Loftus et al. proposed that increased malonyl-CoA and reduced fatty acid oxidation, two features of a positive energy balance, mimic the fed state and underlie the changes observed in NPY and POMC mRNA expression (84, 92).

In most cells, the levels of malonyl-CoA depend on the relative activities of the

enzymes that regulate its synthesis and degradation: acetyl-CoA carboxylase (ACC) and malonyl-CoA decarboxylase (MCD), respectively. Increased fuels elevate cytosolic

malonyl-CoA levels, by increasing acetyl-CoA as well as citrate, which is an activator of

ACC. Thus, in peripheral cells malonyl-CoA is an indicator of cellular energy status (95).

- 22 - A rise in malonyl-CoA results in the inhibition of carnitine palmitoyl CoA transferase 1

(CPT-1), the rate-limiting enzyme for the translocation of LCFA-CoA into the

mitochondria where they undergo β-oxidation favoring lipid storage into triglycerides.

The hypothalamus expresses both isoforms of ACC (ACC-1 and ACC-2) and CPT-1

(CPT-1A and CPT-1B) (96). This is important since ACC-2 is thought to be the isoform responsible for generating the malonyl-CoA that in turn inhibits the CPT-1B isoform.

This mechanism controls the switch from fatty acid to glucose oxidation, and therefore determines the cytoplasmic levels of LCFA-CoA (97).

Loftus et al. reasoned that a signal produced by C75 must mimic the fed state (84).

It was hypothesized that increased hypothalamic malonyl-CoA should be a feature

common to both conditions and so they tested whether it could be a candidate for

mediating the catabolic actions of C75. Indeed, C75 is no longer effective at reducing

food intake and body weight when malonyl-CoA accumulation is prevented. This has

now been demonstrated using both direct (84) and indirect inhibition of ACC (79), as

well as constitutive overexpression of malonyl-CoA decarboxylase (89). To date, there is

still no consensus as to whether or not malonyl-CoA is the signal that makes C75-treated

animals act as if they are in a state of positive energy balance.

Nonetheless, growing evidence supports a role for hypothalamic malonyl-CoA in

the regulation of food intake. Several studies have demonstrated that manipulations that

modulate the activity of ACC, MCD or CPT-1 specifically in the hypothalamus, in a

direction that elevates malonyl-CoA levels, potently inhibit feeding and weight gain, and

- 23 - vice versa (89, 96, 98). Furthermore, hypothalamic malonyl-CoA appears to be a downstream target of leptin signaling, as blocking its elevation with ACC inhibitor also prevents leptin-induced anorexia (52). Recently, Rossetti and colleagues found that rats given ad libitum access to a high fat diet, for a period as short as 3 days, showed reduced hypothalamic levels of malonyl-CoA under basal conditions or following an intravenous infusion of lipids, as compared to rats maintained on standard chow diet (96). While malonyl-CoA is undeniably a nutritionally regulated parameter in the CNS, it is still unclear whether or not it constitutes the metabolic signal that is monitored by neurons and leads to the regulation food intake. An alternative possibility could be that it is regulated in parallel with another factor which actually plays that role.

5. 3. 2. 2. CPT-1 and LCFA-CoA.

Evidence also suggests that C75’s regulation of CPT-1 activity could be involved in causing hypophagia. There are at least two schools of thought regarding the mechanisms that could underlie this effect. One of them refers to the initial inhibitory action of C75 on

CPT-1 (78). While the contribution of this effect to C75-induced anorexia has not been specifically tested, the “LCFA-CoA fuel sensing” model argues that it is a crucial component. According to this model, circulating lipids such as LCFA-CoA regulate feeding by generating an increase in the cellular LCFA-CoA pool in the hypothalamus.

LCFA-CoA, in turn are believed to signal nutrient surplus within the hypothalamus, which activates neural pathways designed to restrict further nutrient intake (88). This hypothesis would predict that the initial inhibitory effect of C75 on CPT-1 may account for the anorexia of C75 by causing LCFA-CoA to accumulate in the cytosol (99). In fact,

- 24 - raising the levels of hypothalamic LCFA through pharmacological (100) or genetic (96)

inhibition of hypothalamic CPT-1A has consequences similar to C75 administration in

terms of food intake and hypothalamic neuropeptide profile (84), and so does icv

administration of oleic acid (18). Furthermore, increased plasma lipid concentration is

indeed accompanied by increased hypothalamic LCFA-CoA levels (96). Intravenous (iv)

infusion of a lipid emulsion (96), or central administration of the LCFA oleic acid (18)

both inhibit food intake supporting the hypothesis that circulating LCFA can signal the

nutritional status of the body to the CNS.

5. 3. 2. 3. Biphasic regulation of CPT-1.

Another school of thought concerns the possible stimulatory effect of C75 on

CPT-1, and its correlation with increased neuronal ATP levels (78, 79). C75 and cerulenin can increase CPT-1 activity in muscle and liver, as well as in cultured adipocytes, hepatocytes and neurons (78, 90, 101). However, these findings have to be interpreted with caution since the stimulatory action of FAS inhibitors is in fact preceded by an initial inhibition of CPT-1 activity, and is therefore characterized as a “biphasic” modulation (101). Nonetheless, according to Ronnett and colleagues, who found this delayed effect in primary cortical neurons (78), C75 could impact both sides of the energy balance equation through this mechanism.

- 25 - 5. 3. 2. 4. CPT-1 and energy expenditure.

There is evidence that systemic administration of C75 (102) or cerulenin (103)

stimulates energy expenditure, a conclusion supported by pair-feeding experiments (104).

This is accompanied by an increase in lipid oxidation, which is unexpected from a compound that elevates cytosolic malonyl-CoA and would be predicted to actually

reduce lipid oxidation through inhibition of CPT-1 (105). This is where the proposed

direct action of C75, i.e. independent of its inhibitory effect on FAS, comes into play

(78). It is difficult to isolate the relative contribution of the inhibitory versus stimulatory

effects of C75 on CPT-1 in regards to caloric expenditure and intake. Nonetheless, the

CPT-1 inhibitor Etomoxir abolishes the thermogenic effect of C75, supporting a role for

CPT-1 activation in C75’s effects (90).

5. 3. 2. 5. CPT-1, ATP and AMPK.

C75 has been reported to modulate neuronal ATP levels in a pattern that correlates

with its biphasic action on CPT-1 activity. In primary cortical neurons, C75 initially

lowers ATP levels, but baseline levels are recovered within an hour after which, ATP

levels rise for at least 15 hours (78). Ronnett and colleagues proposed that a similar

phenomenon could be going on in hypothalamic neurons in vivo, where changes in cellular energy balance may underlie the catabolic actions of C75. This increase in ATP concentration could be a direct effect of CPT-1 activation, or an indirect consequence of the initial decrease of the ATP/AMP ratio (78). This signal might then be integrated by fuel sensors, including AMPK. Such a model has been proposed based on the observation

- 26 - that C75 dephosphorylates and inactivates hypothalamic AMPK, an effect that is required

for its anorectic action (79).

5. 3. 3. C75 and glucose metabolism.

The mechanisms discussed so far are related to the effects of C75 on fatty acid oxidation, and they suggest that the signal sensed by neurons as a state of positive energy

balance lies within fatty acid metabolism. However, C75 also affects neuronal glucose

metabolism. Indeed, both fatty acid and glucose oxidation are significantly increased by

C75 in neurons in vitro (78). Because both pathways are expected to generate ATP, which seems to be part of the signal monitored by fuel sensors (38, 69), the relative contribution of fatty acid versus glucose metabolism to C75-induced anorexia is a matter of debate.

5. 3. 3. 1. Ratio of glucose/fatty acid utilization.

Wortman and colleagues proposed that it is actually the relative ratio of glucose

utilization to fatty acid utilization that is monitored by neurons and signals the overall

energy balance of the body. Indeed, findings from our laboratory demonstrated that C75-

induced anorexia depends on its ability to increase glucose utilization, as a result of

reduced lipid use (49).

5. 3. 3. 2. Glucose: the primary fuel of the brain.

The brain is different from most other tissues in having specific requirement for

glucose as its primary fuel, and oxidizing few fatty acids (86). Blood glucose

- 27 - concentration is a well regulated parameter in mammals, and it remains within a

relatively narrow range under most circumstances. Clearly, large reductions or elevations

in blood glucose levels that occurs during hypoglycemia or hyperglycemia can drive

rapid and profound changes in food intake (106, 107). However, there is very little

evidence that fluctuations within physiological limits play any role in meal initiation or

termination (108-110), in contrast to what the glucostatic hypothesis proposes (35). One

hypothesis is that basal fluctuation in blood glucose concentration participates in a “state-

dependent” regulation of food intake, as one of the several signals that appear to be

integrated by fuel sensors in metabolic neurons such as NPY/AgRP and POMC/CART

neurons of the hypothalamus (33).

While for most neurons glucose serves as a fuel, specific populations of neurons,

referred to as “glucosensing” neurons, also use glucose as a signal to regulate their

activity (111). In contrast to most neurons, glucosensing neurons are localized next to

areas with fenestrated capillaries and ependymal and tanycyte lining cells, including the arcuate and the ventromedial nucleus of the hypothalamus. Glucosensing neurons are therefore likely to be exposed to fluctuations in glucose concentration that are reflective of those occurring in the brain, CSF and blood. They are believed to monitor and integrate the quantitative and temporal changes in glucose concentration, and in turn regulate their activity and neurotransmitter release accordingly (33). They are subdivided in two categories that refer to their response in the presence of increased glucose concentration: glucose-excited (GE) and glucose-inhibited (GI) neurons. Relevant to the present discussion is that GE neurons in the arcuate secrete POMC (112) and GI neurons

- 28 - secrete NPY (113) , and despite their names, glucosensing neurons actually integrate a

variety of metabolic signals (33). This includes signals of stored fuels such as leptin and

insulin, as well as signals of currently available fuels such as free fatty acids (114).

Moreover, selective ablation of glucosensing neurons causes an obese phenotype (115).

Together, these data strongly argue for glucose metabolism as being an important factor

in CNS fuel sensing.

5. 3. 3. 3. Glucose and alternate sources of fuels.

The brain has glucose transporters that maintain a relatively high glucose concentration in the extracellular space, even when circulating glucose levels are at their

lowest levels (116). This system ensures that glucose availability is sufficient for energy

production under most circumstances. Astrocytes also play a crucial role in maintaining

the brain in a “protected” environment, by providing neurons with fuels, such as lactate, glutamine and ketones (117, 118), which can be used instead of glucose to generate ATP.

This relationship ensures that the brain always has proper energy supplies, including

during fasting or intense neuronal activity. Hence, elevation of any of these factors in the

CNS is presumed to decrease glycolysis and oxidative glucose metabolism.

5. 3. 3. 4. Effects of C75 in conditions of reduced neuronal glucose utilization.

As mentioned earlier, the levels of malonyl-CoA are determined by the levels of

acetyl-CoA, and the relative activity of ACC and MCD. There is no evidence that any of

these factors are affected by the presence of lactate, glutamine or elevated ketones, and

therefore increasing the levels of any of these alternate fuels is not expected to prevent

- 29 - malonyl-CoA accumulation in the cytosol. Thus, CPT-1 should still be inhibited and lipid oxidation should still be reduced. According to the “malonyl-CoA hypothesis”, C75 would be predicted to reduce food intake under these conditions, regardless of reduced glucose utilization (84). This was the rationale behind the experiments of Wortman et al., who demonstrated that providing rats with any of these alternative fuels abolished the anorectic action of C75 (49). These data imply that the anorectic signal of C75 depends on increased glucose utilization which occur secondary to malonyl-CoA-induced inhibition of fatty acid oxidation. An additional implication is that the signal perceived by neurons as a state of energy surplus must be generated during glycolysis since glutamine and lactate, two post-glycolytic intermediates, also attenuate the anorexigenic effect of

C75 (49). In summary, increased malonyl-CoA and reduced lipid use would appear to be insufficient per se to elicit the anorexigenic action of C75.

5. 4. Rationale for the research.

C75 alters neuronal glucose and fatty acid metabolism (78) and inhibits

hypothalamic AMPK, and this effect is necessary for C75 to reduce food intake (79). In

contrast to AMPK, hypothalamic mTOR signaling is activated in conditions of positive

energy balance, and its inhibition stimulates food intake in rats (19). These data, together

with the observation that AMPK inhibits mTOR in vitro (119), lead to the hypothesis that

C75-induced anorexia depends on its ability to activate hypothalamic mTOR signaling.

Inhibitors of glycolysis reduce the phosphorylation and activation of the mTOR pathway

in vitro (39). The anorexigenic action of C75 is abolished in conditions of decreased neuronal glucose utilization. Therefore, it is likely that hypothalamic mTOR signaling is

- 30 - reduced under conditions of reduced neuronal glucose metabolism, such as during

ketosis. Chapter 3 describes experiments testing this hypothesis.

6. A role for oleoylethanolamide (OEA) in the regulation of energy balance.

It has been discussed earlier that the long-term maintenance of energy balance is dependent on the coordination and interpretation of signals indicating sufficient long- term energy stores, such as those of leptin and insulin, as well as short-term meal-related

signals. This second category includes hormones secreted from the gastrointestinal tract in response to the mechanical and chemical stimulation of nutrients, a well-characterized example being cholecystokinin (CCK) (120). They mediate their action neurally through vagal afferents, and/or humorally as circulating ligands for receptors located in the

periphery and the CNS (121). Until recently, most of the meal-induced gastrointestinal

signals discovered have been peptides. It was quite unexpected to find that nutrient- derived fatty acids can also be involved in the “meal to meal” regulation of feeding behavior.

6. 1. OEA synthesis and regulation.

OEA is member of the fatty acid ethanolamide (FAE) family, a group of lipid-

derived signaling factors found in animals and plants (122). FAE are characterized by an

ethanolamine residue linked to a long chain fatty acid through an amide bound. The

physiological significance of FAE was first recognized about 50 years ago, with the

demonstration that palmitoylethanolamine is an anti-inflammatory factor (123). However,

these lipids received little attention until the early 90’s, when the endocannabinoid

- 31 - anandamide and its receptors, cannabinoid receptors 1 and 2 (CB1 and 2), were

discovered (124-126). Five years ago, a physiological role has been described for OEA.

Based on the findings that OEA synthesis and secretion is regulated by nutrient ingestion,

and that it reduces food intake and body weight gain when injected to rodents, the

research group of Piomelli proposed the involvement of OEA in the regulation of energy

balance (127).

OEA is synthesized from a phospholipid precursor, which is found in the lipid

bilayer of the cell membrane, through the concerted action of two enzymes:

phospholipase D (PLD) and N-acyltransferase (NAT) (128). Rather than being stored in

vesicles, like classical neurotransmitters and hormones, OEA is produced and released in

a stimulus-dependent manner (129). After release, it is transported back into cells and

rapidly degraded by fatty acid amide hydrolase (FAAH) (130) or -

preferring acid amidase (PAA) (131), giving rise to oleic acid and ethanolamine. FAAH

is present in all mammalian tissues but highly expressed in liver and brain (132), while

PAA is found predominantly in lung, spleen and small intestine (131).

6. 2. OEA: a nutrient-derived lipid mediator.

While a variety of pharmacological agents activate the synthesis of OEA in

neurons in vitro (133), the physiological stimuli in the brain remain unknown. In the intestine (duodenum and jejunum) however, OEA synthesis and release is regulated in a nutrient-dependent pattern: reduced during fasting and increased upon refeeding. The same is true for NAT, the enzyme that synthesizes it (134). This pattern of secretion is

- 32 - consistent with the anorectic action of OEA (134). In addition to inhibition of food

intake, OEA also stimulates lipolysis and fatty acid oxidation, and therefore reduces

adiposity via actions on both sides of the energy balance equation (135).

6. 3. OEA and its receptors.

Although OEA is a structural analogue of the endocannabinoid anandamide, it does

not bind cannabinoid receptors (136). Rather, the activation of peroxisome proliferator-

activated receptor alpha (PPAR-α), a member of the nuclear hormone receptor family of

transcription factors, accounts for its effect on food intake and lipid metabolism (127,

135). In addition, OEA binds with high affinity to the capsaicin receptor (TRPV1), an ion

channel (137) known for its role in pain (138) and locomotion (139). Recent work also

indicates that it is an agonist at GPR119 (140), which is an orphan G protein coupled

receptor (GPCR) expressed predominantly in the brain, pancreas and gastrointestinal tract

(141). Synthetic agonists of this receptor also reduce food intake and body weight (140),

suggesting that it could also be involved in the ability of OEA to regulate energy balance.

6. 4. Hypothetical model for OEA-induced anorexia.

The role of PPAR-α in mitochondrial and peroxisomal fatty acid oxidation has been well characterized, but has never been linked to the control of food intake before

studies involving OEA (127, 142). Because both OEA (143) and its receptors are found

in the brain (144, 145), including in the hypothalamus, it is quite intriguing that OEA

does not affect food intake when injected into either the lateral (134) or third ventricle (K.

Proulx, unpublished data). Nonetheless, systemic administration of OEA stimulates c-

- 33 - FOS in neurons of the PVN, supraoptic nucleus (SON) and nucleus tractus solitarius

(NTS) (134). Very little is known about the relative contribution of these neuronal populations to OEA-induced anorexia. A model has been proposed where OEA activates

PPAR-α located in peripheral tissues, and regulates brain circuits involved in the control of food intake through the vagus nerve (133). This is based on a report that the anorexigenic effect of OEA is abolished in rats in which the subdiaphragmatic vagus nerve has been severed (134). The latter point is a controversial one, since another group found that OEA remains effective at reducing food intake in vagotomized rats (146).

Since it is not clear whether or not OEA mediates its action neurally through vagal afferents, the possibility that OEA alters a peripheral factor, which itself regulates hypothalamic neurons, cannot be ruled out. In fact, an activation of PVN neurons secreting the anorexigenic neuropeptide corticotropin releasing hormone (CRH), or an inhibition of arcuate NPY neurons (such effect would not be expected to be detected by c-FOS, which is a marker of neuronal activation) would be consistent with the CNS c-

FOS pattern induced by OEA (134).

6. 5. Rationale for the research.

It is important to note that OEA reduces food intake, primarily by delaying meal onset and the size of the first meal only (147). This is a crucial observation given that it also reduces motor behavior (134). Although OEA did not cause a conditioned taste aversion per se in the paradigm used in the original report of Piomelli, the drug abolished the preference rats normally display for saccharin over water (134). Therefore, reduced motor activity or illness could each be responsible for the delayed meal initiation and

- 34 - reduced first meal size caused by OEA. Chapter 2 describes experiments that assess

whether the effects of OEA on food intake are due to unspecific mechanisms or whether

they occur through specific actions on other nutrients-derived fuel signals, including

CCK and ghrelin.

- 35 -

CHAPTER 2: MECHANISMS OF OLEOYLETHANOLAMIDE (OEA)-INDUCED

CHANGES IN FEEDING BEHAVIOR AND MOTOR ACTIVITY

Karine Proulx , Daniela Cota , Tamara R. Castañeda , Matthias H. Tschöp, David A.

D’Alessio, Patrick Tso, Stephen C. Woods and Randy J. Seeley.

36

ABSTRACT

Oleoylethanolamide (OEA), a lipid synthesized in the intestine, reduces food intake and stimulates lipolysis through peroxisome proliferator-activated receptor-alpha

(PPAR-α). OEA also activates transient receptor potential vanilloid type 1 (TRPV1) in vitro. Because the anorexigenic effect of OEA is associated with delayed feeding onset and reduced locomotion, we examined whether ip administration of OEA results in non- specific behavioral effects that contribute to the anorexia in rats. Moreover, we determined whether circulating levels of other gut hormones are modulated by OEA, and whether cholecystokinin (CCK) is involved in OEA-induced anorexia. Our results indicate that OEA reduces food intake without causing a conditioned taste aversion or reducing sodium appetite. It also failed to induce a conditioned place aversion.

However, OEA induced changes in posture and reduced spontaneous activity in the open- field. This likely underlies the reduced heat expenditure and sodium consumption observed after OEA injection, which disappeared within 1 hour. The effects of OEA on motor activity were similar to those of the TRPV1 agonist capsaicin, and were also observed with the PPAR-α agonist Wy-14643. Plasma levels of ghrelin, peptide YY

(PYY), glucagon-like peptide 1 (GLP-1) and apolipoprotein A-IV (apo A-IV) were not

changed by OEA. Finally, antagonism of CCK-1 receptors did not affect OEA-induced anorexia. These results suggest that OEA suppresses feeding without causing visceral

illness, and that neither ghrelin, PYY, GLP-1, apo A-IV nor CCK play a critical role in this effect. Despite that OEA-induced anorexia is unlikely to be due to impaired motor activity, our data raise a cautionary note in how specific behavioral and metabolic effects

of OEA should be interpreted.

37

INTRODUCTION

Energy homeostasis in mammals is tightly regulated by complex neuroendocrine

systems matching caloric intake to energy expenditure. In addition to hormones, fatty

acids (18, 28) and fatty acid derivatives (148-150) also signal to central nervous system

(CNS) pathways that control body weight regulation. Recent evidence suggests that

oleoylethanolamide (OEA), the amide of oleic acid and ethanolamine, could play a

significant role in the regulation of energy balance (134). This lipid is synthesized in

astrocytes (151) and neurons (152), as well as in cells of the small intestine where its

levels are reduced by fasting and increased upon refeeding (134).

OEA is a structural analogue of the endocannabinoid anandamide but does not

activate cannabinoid receptors (CB) (136). It is rather an endogenous ligand for the

peroxisome proliferator-activated receptor-alpha (PPAR-α), through which it has been

reported to stimulate lipolysis (135) and inhibit feeding (127). Indeed, OEA reduces food intake when administered peripherally, but appears to be ineffective at doing so when administered centrally (127). In ad libitum fed rats the suppression of food intake is associated with increased latency to initiate a meal without change in meal size or postmeal interval, whereas in fasted rats OEA increases latency to initiate a meal and reduces size of the first meal (147). Furthermore, chronic administration of OEA can reduce the rate of body weight gain in both lean (134) and obese animals (135). All of

these effects are absent in mice that lack PPAR-α, supporting a critical role for these

receptors in the metabolic effects of OEA (127).

38

Rodriguez de Fonseca et al. have reported that although intraperitoneal (ip)

administration of OEA reduces food intake without causing a conditioned taste aversion,

it potently reduces locomotor activity in rats (134). While the effects of OEA on feeding

appear to be mediated via PPAR-α activation (127), the mechanisms by which it reduces

locomotion are unknown. It is also unknown whether other PPAR-α agonists have

independent effects on locomotor activity as well. On the other hand, OEA activates the

transient receptor potential vanilloid type 1 (TRPV1) in vitro (137). This receptor is

widely distributed in the nervous system, including on dopaminergic neurons of the substantia nigra (145), a region well known to be involved in the regulation of motor activity. Interestingly, capsaicin-induced activation of the TRPV1 causes a reduction in ambulation as well as other motor behaviors such as rearing and grooming in the open- field test (139).

Although the effects of OEA on feeding behavior are intriguing, consideration of

OEA as a target for pharmacological treatment of obesity requires that several basic

questions be addressed. Thus, the fact that OEA reduces locomotor behavior (134) is

noteworthy given that OEA reduces food intake by delaying meal initiation and reducing

first meal size (147). Given these behavioral effects, we reassessed the possibility that ip

administration of OEA results in non-specific behavioral effects, visceral illness and/or

aversion, which contribute to the reduced food intake. To examine this possibility, we

used three very different paradigms: conditioned taste aversion (CTA), need-induced

sodium appetite and conditioned place aversion (CPA). Furthermore, because of the

potential involvement of reduced motor activity in the anorexigenic effect of OEA, we

39

characterized the effects of OEA on motor behavior using computer-based video analysis

and an open-field test. We explored whether TRPV1 receptor activation can alter motor

behavior, and we predicted that a TRPV1 agonist would recapitulate the hypolocomotor effects of OEA while a PPAR-α agonist would not. Assessing the metabolic consequences of OEA administration is also important in studying its role in energy homeostasis. Thus, because OEA up-regulates uncoupling protein-2 (UCP-2) mRNA in both white adipose tissue (WAT) and skeletal muscle (135), we tested whether OEA increases energy expenditure using indirect calorimetry.

OEA shares some similarities with several gut hormones involved in the

regulation of food intake. Like OEA, ghrelin, peptide YY (PYY), glucagon-like peptide

1 (GLP-1), apolipoprotein A-IV (apo A-IV) and cholecystokinin (CCK) are influenced by

the presence of nutrients in the gut (121). Thus, in addition to an interest in the

behavioral mechanisms by which OEA might suppress food intake, we were also

interested in how other gut hormones that have been linked to the control of food intake

might be influenced by OEA. To this end, we measured the plasma levels of ghrelin,

PYY, GLP-1 and apo A-IV at several time points after OEA administration. Given that

CCK (153), like OEA (134), is synthesized in the intestine and mediates its anorexigenic

signal via the vagus nerve, we tested a specific role for endogenous CCK in the effects of

OEA with the use of a pharmacological antagonist of the CCK-1 receptor.

40

MATERIALS AND METHODS

Animals: Male Long-Evans rats, weighing 250-300 g at the beginning of the

experiments, were individually housed and maintained on a 12:12-h light-dark cycle. All animal protocols were approved by the University of Cincinnati Institutional Animal

Care and Use Committee.

Drugs: OEA was purchased from Tocris Cookson Inc. (Ellisville, MO). Lorglumide,

CCK-8, Wy-14643 and capsaicin were purchased from Sigma (St. Louis, MO). OEA

was dissolved in 5% Tween-80, 5% propylene glycol and 90% physiological saline.

Capsaicin was first dissolved in 100% ethanol. The stock solution was then dried under

nitrogen after addition of Tween-80, and reconstituted in physiological saline (Tween-

80/saline; 1:16 v/v). The PPAR-α agonist Wy-14643 was dissolved in 70% DMSO and

30% physiological saline. Lithium chloride (LiCl) was dissolved in sterile distilled water

(except in Experiment 2) while lorglumide, CCK-8, and furosemide were dissolved in

physiological saline. For each experiment, vehicle served as the control solution, except

for LiCl where saline was used as a control. All drugs were injected ip in a volume of 1

ml/kg, unless otherwise specified. OEA was administered at 5, 10 and 20 mg/kg (134) in

Experiment 1. Given that 20 mg/kg was the only dose that remained effective at reducing

food intake 1 h after the injection, this dose was used in all subsequent experiments.

Experimental design:

Experiment 1: Effect of OEA on food intake. Rats were assigned to one of four treatment groups (n = 8-9/group). Thirty minutes prior to the onset of the dark phase, 24-h fasted

rats received an injection of either vehicle or OEA (5, 10 or 20 mg/kg, ip) (134) and then

41

received access to food. The food hoppers were weighed at 30 min, 1, 2, 4 and 24 h after

the injection.

Experiment 2: Effect of OEA on CTA. If rats become ill after consumption of a novel

flavor, they will avoid consumption of this flavor at future opportunities (154). An

example of this is reduced consumption of a flavor that has been previously paired with

toxins such as LiCl (155). In order to test whether the anorexigenic effect of OEA is

secondary to illness, rats were assigned to one of two groups (saline-LiCl or vehicle-

OEA, n = 10/group) and preference ratios for novel flavors were determined after pairing

with the drugs. The preference ratio for a drug is calculated as the intake of the drug-

paired flavor over the total intake of both flavors and the critical comparison is whether the ratio for either flavor deviates from 0.5. All animals were trained for 10 days on a water deprivation schedule during which they had access to two water bottles for 1 h/day.

On conditioning day 1, all rats had access to two bottles of saccharin-sweetened Kool-

Aid (5.25 g of saccharin, 3500 ml of water and one packet of either cherry or grape Kool-

Aid; both bottles contained the same flavor, in a counterbalanced manner across rats and

groups) for 1 h. After access to flavor 1, half of the rats from each group received either

saline or vehicle and the other half received either LiCl (0.15 M in a volume equivalent to

2% of the rat’s body weight) or OEA (20 mg/kg, ip). The following day, rats had access

to two water bottles for 1 h, as a rest day. On conditioning day 2, rats had access to two

bottles of flavor 2 for 1 h. Rats that were injected with saline or vehicle on conditioning

day 1 were then injected with LiCl or OEA, and vice versa. Rats then had 2 rest days in

which they had access to two bottles of water for 1 h. On the test day, rats received one

42

bottle of each flavor, with the side of flavor presentation being switched at 30 min to avoid any side preference. Fluid intake was measured at 1 h, and water was returned at

the end of the experiment.

Experiment 3: Effect of OEA on need-induced sodium appetite. Another reliable index of

visceral illness is failure to consume hypertonic saline solution in response to sodium

depletion. For instance, when rats rendered sodium-depleted are injected with emetic

and/or toxic agents such as LiCl, they reduce their “need-induced” consumption of

hypertonic saline (156). In order to assess whether OEA is aversive by this definition,

rats were assigned to one of two treatment groups (vehicle or OEA, n = 7/group) and

need-induced sodium appetite was assessed. For 7 days, rats had access to two bottles,

one that contained water and another one that contained a 0.5 M NaCl solution. Twenty-

four hours prior to the experiment, the saline bottle was removed and the food hopper

with regular diet was replaced with one that contained sodium-free rat diet (ICN

Biochemicals, Cleveland, OH). Water remained available at all time. Rats were then

weighed and received two subcutaneous injections of the diuretic furosemide (5mg/kg,

ip), 1 h apart (157). Rats were weighed again 3 h after the first injection, and diuresis

(and presumed sodium depletion) was confirmed by observing at least 18 g of body

weight loss. Twenty-four hours after the first furosemide injection, rats received an ip

injection of isotonic LiCl (0.15 M in a volume equivalent to 2% of the rat’s body weight),

an equal volume of physiological saline, OEA (20 mg/kg) or vehicle. Fifteen minutes

later, rats received two bottles; one that contained distilled water and another one that

43

contained a 0.5 M NaCl solution. Intake of both fluids was measured every 30 min for 2

h, and 24 h after the injection.

Experiment 4: Effect of OEA on conditioned place aversion (CPA). The conditioned

place aversion test is a behavioral procedure in which an animal forms an association

between a stimulus of negative value and the location in which it experienced the

stimulus during training. CPA training and testing took place in a 78 x 21-cm box (Med

Associates, Inc.). The box was divided into a black side and a white side (each 25.5 x 21 cm) separated by middle chamber (12 x 21 cm) with two doors. The floor of each side was made of two distinct textures. The training procedure consisted of 6 contiguous days in which rats were placed in alternating sides of the CPA box for 30 min. On one set of alternate days (Days 1, 3, and 5 or Days 2, 4, and 6), rats were injected with either LiCl

(0.15 M in a volume equivalent to 2% of the rat’s body weight) or OEA (20 mg/kg, ip)

and were immediately placed on one side of the CPA box with the door closed. On the

other set of alternate days, rats received either saline or vehicle and were immediately

placed in the alternate side of the box from that paired with the drug on the previous day,

and the door was closed. Conditions were counterbalanced across rats and groups. On

the 7th day (test), rats were placed into the middle chamber and were allowed to explore

the apparatus with both guillotine doors opened for 15 min. Rats were videotaped and

time spent in each chamber was scored by an investigator who was blind to the drug

treatment. An animal was considered to be in a chamber when its head and forepaws were inside it. Results are expressed as the time spent in the drug-paired side.

44

Experiment 5: Effect of OEA on heat expenditure. Rats were assigned to one of two

treatment groups (vehicle or OEA, n = 8/group). Four hours before testing, rats were placed in indirect calorimeter chambers for habituation. They were then injected with either vehicle or OEA (20 mg/kg, ip) 30 min before the onset of the dark and returned to the indirect calorimeter chamber immediately after the injection. Food was removed but water remained available throughout the experiment. Heat production was recorded every 24 min for 24 h.

Experiment 6: Comparison of the effects of OEA and capsaicin on behaviors in home

cage. This study examined the effects of OEA on behaviors displayed by rats in their

home cage. Fifteen minutes before the onset of the dark, rats (n = 5-7/group) received an

injection of OEA (20 mg/kg, ip), capsaicin (1 mg/kg, ip) (12) or their respective vehicles,

and were immediately returned to their home cage. Rats were videotaped for 2 h and

behavioral analysis was conducted using a computerized system (HomeCageScan

software, Clever Sys. Inc.), that recognizes, records and quantifies upwards of 22 distinct

rodent behaviors in a home cage environment.

Experiment 7: Comparison of the effects of OEA, capsaicin and Wy-14643 in the open-

field test. This experiment compared the effects of OEA, capsaicin and Wy-14643 on

motor behaviors. Rats (n = 5-7/group) were injected with OEA (20 mg/kg, ip), capsaicin

(1mg/kg, ip) (12), Wy-14643 (40 mg/kg, ip) (13) or their respective vehicles 10 min prior

to testing. The dimensions of the open-field were 36 x 45 cm and the height of the walls

was 30 cm. The floor was divided into 20 squares (9 x 9 cm). Rats were placed in the

45

middle of the open-field, and they were videotaped for 10 min. Only the last 5 min of the tape were analyzed, the first 5 min serving as a habituation period. Time spent in inactivity, ambulation (number of squares crossed) and spontaneous activity (number of rearing or grooming events) were measured (139). Rats were considered to have crossed a square when they placed their four paws into an adjacent square. Tapes were scored by an investigator who was blind to the drug treatment.

Experiment 8: Effects of OEA on plasma levels of ghrelin, PYY, GLP-1 and apo A-IV.

Rats were injected with either OEA (20 mg/kg, ip) or vehicle 30 min prior to onset of the

dark and were sacrificed at 15, 30, 90 and 120 min after the injection. An additional

group of rats (n = 8) remained uninjected and served as baseline controls. Trunk blood

samples were collected into tubes that contained an anti-proteolytic cocktail (25 mM

EDTA plus 500 kallikrein inhibitory units aprotinin and 80 units Heparin per ml of

blood). Blood was then centrifuged at 4° C for 10 min at 4000 rpm followed by 2 min at

12,000 rpm. Separated plasma was stored at –20°C until assay. Plasma ghrelin (158)

and PYY (159) concentrations were determined using commercial RIA kits according to

manufacturer’s directions (Phoenix Pharmaceuticals, Inc., Belmont, CA.) as previously

described. The ghrelin assay measures both acylated and des-acetylated ghrelin, and the

PPY assay measures both PYY (1-36) and PYY (3-36). Total plasma GLP-1

concentration was measured by RIA using antiserum 89390 (kindly provided by Dr. Jens

Holst, Paanum Institute, Copenhagen, Denmark) from ethanol extracts of plasma as

previously described (160). This antibody recognizes both the intact hormone GLP-1 (7-

36)-NH2 and the metabolite GLP-1 (9-36)-NH2. The intra- and interassay coefficients

46

of variation for these assays were < 5% and < 13%, respectively for ghrelin, < 7 % and

<11% for GLP-1, and < 8.42% and < 14.52% for PYY. Plasma apo A- IV concentration

was measured by an ELISA, as previously described (161).

Experiment 9: Effect of the CCK-1 antagonist lorglumide on OEA-induced reduction of food intake. To test the involvement of CCK-1 receptors in the anorexigenic effect of

OEA, rats were assigned to one of four treatment groups (saline + vehicle, saline + OEA,

lorglumide + vehicle or lorglumide + OEA, n = 6-7/group). Thirty minutes prior to the onset of the dark, 24-h fasted rats received an injection of either saline or lorglumide (1 mg/kg) (162). Fifteen minutes later, rats received a second injection of either vehicle or

OEA (20 mg/kg, ip) and then received access to food. The food hoppers were weighed at

30 min, 1, 2, 4 and 24 h after the last injection. As a positive control, we tested the ability of the same dose of lorglumide to block the anorexigenic effect of CCK-8. Rats were assigned to one of four treatment groups (saline + saline, saline + CCK-8,

lorglumide + saline or lorglumide + CCK-8, n = 8-9/group). Two hours prior to the onset

of the dark, 22-h fasted rats received an injection of either saline or lorglumide (1 mg/kg,

ip) (162). Fifteen minutes later, rats received a second injection of either saline or CCK-

8 (4 μg/kg, ip) (163) and then received access to food. Food hoppers were weighed at 15,

30, 45 min and 1 h after the last injection.

Statistical analysis: Changes in food intake, sodium intake and heat expenditure across time were analyzed by one-way or two-way repeated measures ANOVA, unless otherwise specified in the results. Significant ANOVAs were followed by Least

47

Significant Difference post hoc tests. Preference ratios, time spent in the drug-paired

side, hormones levels, and behaviors in the home cage and in the open-field were

analyzed by two-tailed Student’s t-tests. The level of significance was set at P < 0.05.

All values are expressed as the mean ± SEM.

48

RESULTS

Experiment 1: Effect of OEA on food intake. OEA significantly reduced 30-min food intake as compared to saline at 10 mg/kg (P < 0.05; see Figure 1A) and 20 mg/kg (P <

0.001; Figure 1A), with the 20 mg/kg suppressing 99.48% of food intake. One hour

following the injection, only the 20 mg/kg dose caused a significant reduction in food

intake (P < 0.01; Figure 1A) and this effect remained significant 24 h after the injection.

(P < 0.05; Figure 1B).

Experiment 2: Effect of OEA on CTA. The positive control LiCl caused a CTA as indicated by the significantly lower preference ratio for the LiCl-paired flavor compared to the saline-paired flavor (P < 0.001; Figure 2). In contrast, there was no significant difference between the preference ratio of the flavor previously paired with OEA as compared to the vehicle-paired flavor (P = 0.746; Figure 2) indicating that rats did not develop a CTA to OEA.

Experiment 3: Effect of OEA on need-induced sodium appetite. LiCl significantly

reduced need-induced sodium appetite as compared to saline at all time points (P <

0.001; Figure 3 for 2 h, data at other time points not shown) for at least 24 h (P < 0.05).

Two-tailed t-test comparisons indicated that sodium intake was significantly suppressed

by OEA relative to vehicle at 30-min (OEA: 4.37 + 1.03 g vs. vehicle: 10.03 + 0.91 g; P

< 0.01), although to a lesser extent than by LiCl (LiCl: 1.67 + 0.12 g; P < 0.05). In

contrast to rats injected with LiCl, OEA-injected rats rapidly compensated for this acute reduction as this effect had disappeared at 1-h (OEA: 8.43 + 1.06 g vs. vehicle: 11.12 +

49

2.66 g; P = 0.090). Indeed, one-way repeated measures ANOVA revealed that OEA did not have a significant effect relative to vehicle over 24 h (P = 0.482; Figure 3 for 2 h, data at other time point not shown).

Experiment 4: Effect of OEA on CPA. Rats in the LiCl-saline group spent significantly

less time in the LiCl-paired side as compared to the saline-paired side (P < 0.05; Figure

4), indicating the formation of a CPA to LiCl. In contrast, OEA did not cause a CPA

since rats in the OEA-vehicle group spent a similar amount in both the OEA-paired side

and the vehicle-paired side (P = 0.849). This result together with the lack of a sustained

reduction of sodium appetite and induction of a CTA suggest that acute administration of

OEA does not have a toxic or illness-inducing effect in rats.

Experiment 5: Effect of OEA on heat expenditure. Two-tailed t-test comparison revealed

that OEA significantly reduced heat expenditure relative to vehicle when measured for

the first 48 min following the injection (P < 0.001; Figure 5A). However, this effect

waned by 72 min (P = 0.341; Figure 5A), and one-way repeated measures ANOVA

indicates that there was no significant effect of OEA treatment over the course of either

the 12-h dark (P = 0.367; Figure 5B) or the 12-h light phase (P = 0.403; data not shown).

Experiment 6: Comparison of the effects of OEA and capsaicin on behaviors in home

cage. OEA significantly increased the time rats displayed an extended posture, that is

pushing their abdomen against the floor of the cage with splayed hind limbs, as compared

with vehicle (P < 0.01; Figure 6A), while capsaicin did not cause a significant effect on

50

this behavior (P = 0.334; Figure 6B). There was no significant change in the other behaviors analyzed after either OEA or capsaicin administration as compared with their respective vehicles (P > 0.05).

Experiment 7: Comparison of the effects of OEA, capsaicin and Wy-14643 in the open- field test. Rats treated with OEA were significantly more inactive (P < 0.001; Figure 7A) and had significant decreases in ambulation (P < 0.01; Figure 7B) and spontaneous activity (P < 0.001; Figure 7C) as compared to vehicle-treated rats. The effect of OEA on inactivity was recapitulated by Wy-14643 (P < 0.05; Figure 7A), but not by capsaicin

(P = 0.164; Figure 7A). However, neither Wy-14643 (P = 0.420; Figure 7B) nor capsaicin (P = 0.208; Figure 7B) had a significant effect on ambulation. Similar to OEA, both Wy-14643 (P < 0.01; Figure 7C) and capsaicin (P < 0.05; Figure 7C) significantly inhibited spontaneous activity as compared to their respective vehicles.

Experiment 8: Effects of OEA on plasma levels of ghrelin, PYY, GLP-1, and apo A-IV.

OEA did not cause any significant change in plasma levels of ghrelin, PYY, GLP-1 and apo A-IV relative to vehicle, at any of the time points that were examined (P > 0.05;

Figure 8).

Experiment 9: Effect of lorglumide on OEA-induced reduction of food intake. As reported in Experiment 1, OEA significantly reduced food intake relative to vehicle (1-h intake: P < 0.01 and 2-h intake: P < 0.001; Figure 9A). At none of the time points examined was this effect significantly modified by pre-treatment with lorglumide relative

51

to saline (P > 0.05; Figure 9A), whereas the same dose of lorglumide was effective at blocking the anorexigenic effect of CCK-8 relative to saline (15- and 30-min intakes: P <

0.05; Figure 9B).

52

DISCUSSION

As has been previously reported (127, 134), ip administration of OEA reduces food intake and, at 20 mg/kg, food intake is almost completely suppressed for the first 30 minutes. The anorexigenic effect of OEA is accompanied by a suppression of locomotor activity. In particular, the injection of OEA is followed by a unique behavior in which the rat takes on an extended posture (pushing its abdomen against the floor of the cage) with splayed hind limbs and little ambulatory activity. This behavior resembles the

“lying on belly” that is caused by toxins that produce visceral illness (155). However, on two sensitive assessments of visceral illness OEA had minimal effect, consistent with previous findings (134). Moreover, OEA did not induce the formation of a conditioned place aversion. Based on these results, it is difficult to ascribe the anorectic effects of

OEA to illness or aversion. Therefore, it seems likely that suppression of food intake by

OEA involves specific actions on pathways regulating energy homeostasis.

Illness in rats manifests as a pattern of well-established stereotypic behaviors.

CTA is one of the hallmark behaviors of visceral illness, and CTA tests rely on the ability to learn the association between a flavor and an aversive stimulus (154). In these studies

OEA did not support a CTA. However, other members of the fatty acid ethanolamide family, such as anandamide, have been reported to cause anterograde amnesia (164).

Thus, it is possible that OEA could induce visceral illness, but not produce a CTA because of a separate effect to impair memory. To avoid being misled by this potential confound, we used the additional measure of need-induced sodium appetite (156). While

OEA caused some reduction in sodium consumption as compared to vehicle within the

53

first hour following the injection, this effect dissipated more rapidly than the effect on

food intake and was much shorter than the effect of LiCl. Taken together, these data are

consistent with the hypothesis that the effect of OEA to suppress food intake is not

secondary to visceral illness. Moreover, since the ingestion of the NaCl solution involves much of the same motor behavior as ingestion of food, it is unlikely that the anorexia is

simply a result of the reduced motor activity, at least at time points beyond 1 hour.

Given the negative results of the visceral illness and conditioned aversion tests,

the reduced locomotor activity and unusual posturing we observed after OEA administration requires an alternative explanation. We therefore further characterized the

behavior caused by OEA administration using computer-based video analysis. This assay

confirmed that rats display an extended posture with splayed hindlimbs early after an ip

injection of OEA. This behavior is also followed by signs that resemble those of

catalepsy. While the pharmacological effects of OEA are not due to activation of any of

the known cannabinoid receptors (136), it is noteworthy that we have observed

comparable behavior in rats given ip anandamide (unpublished data). Reduced

locomotor activity, ataxia and catalepsy are indeed commonly observed after

administration of cannabinoid agonists (165). Interestingly, stearoylethanolamide is

another member of the fatty acid ethanolamide family that does not activate cannabinoid

receptors but that causes cannabimimetic effects, including catalepsy (166).

In the open field test, OEA not only reduced locomotion but also grooming and

rearing, while increasing time spent in inactivity. We compared these behavioral effects

54

of OEA to agonists of either PPAR-α or TRPV1. Agonists of TRPV1 such as capsaicin

have previously been reported to reduce motor activity (139) and our data indicate that

this effect is similar to what occurs with OEA. Like OEA, capsaicin significantly

reduced spontaneous activity. Capsaicin also showed a trend to reduce ambulation, and

increase inactivity and extended posture. Because of this trend, we cannot rule out the

possibility that this is a real effect of capsaicin that would be revealed with higher

numbers of animals.

Given that there have been no previous descriptions of the effects of PPAR-α to

reduce motor behavior, we were surprised to see a potent effect of the PPAR- α agonist

Wy-14643 to reduce motor activity. While these observations are consistent with the distribution of TRPV1 and PPAR-α in areas associated with motor activity such as the substantia nigra compacta and the striatum (144, 145), both receptors are widely distributed and so a firm conclusion about the mechanism for this effect is difficult. Thus it remains unclear how OEA induces postural changes and suppresses motor activity.

Since activation of the PPAR-α is necessary for OEA’s effect on food intake

(127), the current data raise the possibility that the locomotor effects are also the result of

agonism of PPAR-α. Interestingly, it has been reported that capsaicin-induced activation

of TRPV1 leads to OEA production in vitro (139). This raises the hypothesis that PPAR-

α activation, by ligands of either endogenous (capsaicin-induced production of OEA) or

exogenous (administration of either Wy-14643 or OEA) sources could mediate the motor

effects of OEA, capsaicin and Wy-14643. Nonetheless, further experiments making use

55

of either highly specific receptor antagonists or targeted genetic disruption of these

receptors will be necessary to conclude whether PPAR-α and/or TRPV1 are involved in

the behavioral effects of OEA on motor activity.

Activation of PPAR-α has been associated with reduced body weight gain due to

effects on lipid catabolism and energy expenditure. For instance, the PPAR-α ligands

Wy-14643 and the drugs , and are all

hypolipemic agents that reduce weight gain in rodent models (167). Moreover, fibrate

treatment has been associated with elevation in UCP-1 mRNA in white adipose tissue

(WAT) and in UCP-3 in WAT and skeletal muscle in rodents (168). The effects of OEA

are consistent with those of , since OEA up-regulates UCP-2 mRNA in both WAT

and skeletal muscle (135). However, contrary to what we have predicted, OEA did not

increase energy expenditure in rats. We observed a significant reduction in heat during

the first 24 min that followed the OEA administration as compared to vehicle. This effect

is likely the result of the reduced motor activity caused by OEA.

Even though it is unlikely that the effect of OEA on food intake is merely by-

product of its action on other motor behaviors, our results raise a cautionary note as to

how specific behavioral and metabolic data should be interpreted in the light of these potent actions. Recent data continue to point to a critical role in the control of food

intake for a number of gut hormones whose secretion is either increased or decreased by

nutrients being absorbed from the gastrointestinal tract (121). Thus, the possibility that

OEA could influence food intake by altering the secretion of one or more of these

56

hormones was necessary to consider. We measured circulating plasma levels of ghrelin,

PYY, GLP-1 and apo A-IV at several time points after the administration of OEA at a dose that reduces food intake. OEA did not have a significant impact on plasma levels of any of these hormones at any time point compared to vehicle-treated rats. These data are consistent with a recent report indicating that OEA failed to modulate either GLP-1 or ghrelin under free-feeding conditions (168). However, in this study OEA did reduce plasma levels of acylated ghrelin in fasted animals. Conclusions from the current data are limited by the fact that rats did not have exposure to nutrients after the administration of OEA. Such a design was used to avoid the possibility that drug-induced change in feeding could influence hormone levels. Consequently, we cannot completely rule out the possibility that OEA modulates the ability of nutrients to alter gut hormone secretion.

Another gut hormone that has been linked to the control of food intake is CCK

(25). However, the critical form (CCK-8) is difficult to measure in plasma and in the rodent several lines of evidence point to paracrine rather than endocrine action of CCK to reduce feeding (169). Thus, to examine whether there may be a role for increased CCK release in the anorexigenic effect of OEA, we took advantage of a well-validated CCK-1 receptor antagonist (162). While pharmacological antagonism of CCK-1 receptors with lorglumide can block the anorexia induced by exogenous CCK-8, it cannot block the anorexia induced by OEA. Thus, it seems unlikely that CCK mediates the effects of

OEA on food intake.

57

Perspectives. Taken together, these data argue against the hypothesis that ghrelin,

PYY, GLP-1, apo A-IV or CCK play a critical role in the effect of OEA to suppress food intake, at least under free-feeding conditions. Previous data indicated that neither corticosterone, leptin nor insulin play a role in OEA-induced anorexia (134). While the current experiments were not designed to test this specific hypothesis, it is tempting to speculate that OEA reduces food intake by regulating the expression of genes in the periphery that are involved in lipid metabolism. Ip administration of the fatty acid ethanolamide stearoylethanolamide has been shown to cause anorexia via such mechanism (170). Furthermore, OEA increases the expression of PPAR-α (127), which represses the transcription of lipogenic genes (171) such as hepatic fatty acid synthase

(172, 173). Growing evidence points to a role for lipid metabolism in the regulation of feeding (18, 77). These findings raise the possibility that ongoing modulation of lipid metabolism in the periphery is involved in the anorexigenic effect of OEA.

58

FIGURE LEGENDS

Figure 1. Mean + SEM food intake at different time points (A), and 24 h (B) after an ip

injection of vehicle (VEH) or three doses of OEA in 24-h fasted rats.

* = P < 0.05, ** = P < 0.01 and *** = P < 0.001.

Figure 2. Mean + SEM preference ratio of flavors previously paired with an ip injection of saline (SAL), lithium chloride (LiCl), VEH or OEA (20 mg/kg).

*** = P < 0.001.

Figure 3. Mean + SEM 2-h sodium intake after an ip injection of SAL, LiCl, VEH or

OEA (20 mg/kg) in sodium-depleted rats. *** = P < 0.001.

Figure 4. Mean + SEM time the rats spent in the side previously paired with an ip

injection of SAL, LiCl, VEH or OEA (20 mg/kg). * = P < 0.05.

Figure 5. Mean + SEM heat expenditure at different time points after an ip injection of

either VEH or OEA (20 mg/kg) (A), and mean + SEM heat expenditure per hour during

the 12-h dark phase (B) in fasted rats. *** = P < 0.001.

Figure 6. Mean + SEM time the rats displayed an extended posture in the 2 h following

an ip injection of either VEH or OEA (20 mg/kg) (A), and either VEH or capsaicin

(CAPS) (1 mg/kg) (B). ** = P < 0.01.

Figure 7. Mean + SEM time the rats spent in inactivity (A), number of times the rats crossed a square (B), and number of times the rats either reared or groomed (spontaneous activity) (C) after an ip injection of OEA (20 mg/kg), CAPS (1 mg/kg), Wy-14643 (40 mg/kg) or their respective VEH. * = P < 0.05, ** = P < 0.01 and *** = P < 0.001.

59

Figure 8. Mean + SEM plasma concentrations of ghrelin (A), PYY (B), GLP-1 (C), and apo A-IV (D) at different time points after an ip injection of either VEH or OEA (20 mg/kg).

Figure 9. Mean + SEM food intake at different time points after an ip injection of either

SAL or lorglumide (LORGL) (1mg/kg), followed by an ip injection of either VEH or

OEA (20 mg/kg) (A), and either SAL or CCK-8 (4 μg/kg) (B) in fasted rats.

* = P < 0.05 and ** = P < 0.01.

60

FIGURES

A VEH 14 OEA (5 mg/kg)

12 OEA (10 mg/kg) OEA (20 mg/kg) 10

8 6 **

Food intake (g) intake Food 4 ** 2 * 0 *** ** 0 1 2 3 4 5 Time (h) B

35

30 * 25

20

15

Food intake (g) 10

5

0 VEH 5 10 20 Dose (mg/kg)

Figure 1

61

1.00

0.75

0.50

*** 0.25 Preference ratio Preference

0.00 SAL LiCl VEH OEA

Figure 2

62

18

16

14

12

10

8

6

4 *** 0.5 intake M NaCl (g) 2

0 SAL LiCl VEH OEA

Figure 3

63

500

400 * 300

200

100

0 SAL LiCl VEH OEA Time spent in the drug-paired side (sec.) side drug-paired the in spent Time

Figure 4

64

A

3.0 VEH OEA

2.5

P = 0.05

Heat (kCal/h) Heat 2.0

1.5 *** 0 24 48 72 96 120 144 168 192 216 240 Time (min)

B 3.0

2.5

2.0

1.5

1.0 12-h Heat (kCal/h) Heat 12-h 0.5

0.0 VEH OEA

Figure 5

65

A

10

8 **

6

4

2 Time in extension in Time (min)

0 VEH OEA

B

10

8

6

4

2 Timeextension in (min)

0 VEH CAPS

Figure 6

66

A

280 *** 280 280

240 240 240 *

200 200 200

160 160 160

120 120 120

80 80 80

40 40 40 Time in inactivityTime in (sec.) (sec.) inactivity in Time Time ininactivity (sec.) 0 0 0 VEH OEA VEH CAPS VEH Wy-14643

B

80 80 80

60 60 60

40 40 40 Ambulation Ambulation Ambulation 20 ** 20 20 (# of squares crossed) (# of squarescrossed) (#of squares crossed) 0 0 0 VEH OEA VEH CAPS VEH Wy-14643

C

50 50 50

40 40 40

30 30 * 30

20 20 20 ** (# of events) (# of events) (# of events) of (# 10 10 10 Spontaneous activity Spontaneous activity *** Spontaneous activity

0 0 0 VEH OEA VEH CAPS VEH Wy-14643

Figure 7

67

A B

VEH 1.50 10.0 OEA

1.25 7.5

1.00 5.0

0.75 2.5 Plasma GLP-1(pg/ml) Plasma ghrelin (ng/ml) Plasma ghrelin

0.50 0.0 0 30 60 90 120 150 0 30 60 90 120 150 Time (min) Time (min)

C D

100 26

24

75 22

20

50 18

Plasma PYY (pg/ml) PYY Plasma 16 Plasma apo A-IV (mg/dl)

25 14 0 30 60 90 120 150 0 30 60 90 120 150 Time (min) Time (min)

Figure 8

68

A SAL/VEH 14 LORGL/VEH

12 SAL/OEA LORGL/OEA 10

8

6

Food intake (g) intake Food 4 ** * 2

0 0 1 2 3 4 5 Time (h)

B SAL/SAL 7 LORGL/SAL

6 SAL/CCK-8 LORGL/CCK-8 5

4

3 * 2 Food intake (g) intake Food * 1

0 0 15 30 45 60 75 Time (min)

Figure 9

69

The OEA-induced aversion hypothesis, revisited:

Our attempts at assessing the contribution of TRPV1, a receptor linked with pain

and motor activity, in the effects of OEA were problematic. We did not replicate the

effects of the TRPV1 inhibitor capsazepine at blocking capsaicin-induced

hypolocomotion. Without this positive control, it was difficult to interpret our data. In the

meanwhile, another research group answered the question we aimed to address. In a

series of very elegant studies, Wang et al. have demonstrated that short-term food intake is reduced in response to OEA in wild-type but not in TRPV1 knockout mice (174). They proposed that TRPV1 mediates the initial hypophagic action of OEA, which they

attributed to pain. Our data, together with those of Wang et al., strongly indicate that in

order to realize the therapeutic potential of OEA, it will be important to develop

analogues with high affinity for PPAR-alpha, while limiting activation of TRPV1.

70

CHAPTER 3: C75 REGULATES FOOD INTAKE VIA ACTIVATION OF

HYPOTHALAMIC MTOR SIGNALING.

Karine Proulx, Daniela Cota, Stephen C. Woods and Randy J. Seeley

71

ABSTRACT

C75 is a fatty acid synthase inhibitor that reduces food intake and body weight in rodents when administered either in the central nervous system (CNS) or the periphery.

Such effects are believed to occur via inhibition of the hypothalamic fuel sensor AMP- activated protein kinase (AMPK), as they are prevented by central administration of the

AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR). Recent evidence from our laboratory strongly indicates that the mammalian target of rapamycin

(mTOR), a protein kinase that regulates protein synthesis and cell growth, also serves as an energy sensor in the CNS. In contrast to AMPK, hypothalamic mTOR signaling is activated in conditions of positive energy balance and its inhibition with rapamycin stimulates food intake in rats. These data, together with the ability of AMPK to inhibit mTOR signaling in vitro, led us to hypothesize that C75 reduces food intake through activation of the mTOR pathway in the hypothalamus. Using Western blotting, we found that intra-third ventricular (i3vt) administration of C75 (30 μg in 2 μl of RPMI) activates the mTOR pathway as indicated by a significant increase in the phosphorylation of mTOR (P < 0.05) and its downstream effector S6 ribosomal protein (S6) (P < 0.001) in the hypothalamus. As reported before, i3vt administration of C75 (50 μg in 3 μl of

RPMI) caused anorexia and body weight loss (P < 0.05), effects that were absent in rats receiving an i3vt injection of the mTOR inhibitor rapamycin prior to C75 (P < 0.05). We also replicated our previous finding that C75 is ineffective at reducing caloric intake in rats maintained on a ketogenic diet. Interestingly, C75 is also unable to activate the hypothalamic mTOR signaling under these conditions. In conclusion, our results indicate

72

that the effects of C75 on energy balance depend on its ability to activate mTOR signaling in the brain.

73

INTRODUCTION

Energy balance is achieved when caloric intake is accurately matched to caloric

expenditure. A complex neuroendocrine system underlies this process and thus regulates

energy homeostasis in mammals. In addition to sensing hormonal signals of stored fuels,

such as leptin and insulin (9), specific populations of neurons in the central nervous

system (CNS) have the ability to sense the level of available nutrients. In turn, they

regulate their activity to adjust caloric intake and expenditure accordingly. Glucose (33)

and oleic acid (99) inhibit NPY neurons and reduce food intake when injected centrally.

This phenomenon is also observed with intracerebroventricular (icv) administration of

compounds that modify either glucose (175) or fatty acid metabolism (88). One such

compound is the fatty acid synthase (FAS) inhibitor C75.

C75 reduces food intake and body weight in rodents when administered in the

periphery or directly into the CNS (48-50, 84). The C75-induced anorexia is

accompanied by reduced NPY/AgRP and increased POMC/CART mRNA expression

(92). Hypothalamic malonyl-CoA, which accumulates as a result of FAS inhibition, has

been proposed to underlie these changes and thus, to play a key role in mediating the

anorexia of C75 (176). In fact, manipulations that reduce the rate of synthesis of malonyl-

CoA (84) or increase its degradation (89) prevent C75 from reducing food intake.

The serine/threonine protein kinase AMP-activated protein kinase (AMPK) is a

cellular fuel sensor that is activated by signals of energy depletion, specifically a reduction of ATP:AMP, and that exerts rapid regulation of energy-producing and -

74

consuming metabolic pathways (177). In the hypothalamus, AMPK is inhibited by

signals of positive energy status such as leptin, insulin and glucose (34, 41). Genetic or

pharmacological manipulations that increase AMPK activity in the CNS stimulate food

consumption (34, 41) and prevent the anorexigenic effect of leptin (34). C75, which

mimics a state of positive energy balance (84), also reduces hypothalamic AMPK activity

(78, 79). This latter effect has been proposed to mediate C75 anorexigenic action, since

C75-induced anorexia does not occur following central administration of the AMPK

activator AICAR (79). Given that AMPK is an inhibitor of acetyl-CoA carboxylase

(ACC), Hu et al. have proposed a model where the C75-induced disinhibition of ACC

elevates malonyl-CoA levels, which in turn acts as a signal to reduce food intake (89).

Like AMPK, the mammalian target of rapamycin (mTOR) is a serine/threonine

protein kinase that resides at the interface between nutrient sensing and regulation of

energy balance (39). MTOR integrates signals of nutrient availability, including amino

acids and growth factors such as leucine and insulin, and in turn regulates protein

synthesis and cell growth (178). For instance, mice and Drosophila lacking S6 kinase 1

(S6K1), a downstream component of TOR, have smaller cells and consequently smaller bodies than controls (72). In contrast to AMPK, the mTOR pathway is activated in conditions of energy surplus, and is therefore activated when ATP:AMP increases (39).

This occurs in peripheral tissues, and possibly in the brain as well. Our laboratory has recently demonstrated that mTOR signaling is controlled by energy status and co- localizes with neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons in the arcuate nucleus (19). Leptin and the branched-chain amino acid leucine activate the

75

pathway when administered into the third ventricle (i3vt). This is necessary for leptin and

leucine to reduce food intake, since their anorectic action is abolished by pre-treatment

with the mTOR inhibitor rapamycin (19).

Evidence supports the existence of a regulatory relationship between AMPK and mTOR. Activated AMPK indirectly inhibits mTOR through activation of the TSC2 tumor

suppressor gene (119). These data, together with the inhibitory effect of C75 on AMPK

(79) and the role of hypothalamic mTOR in feeding (19), led us to suggest that C75

reduces food intake through activation of mTOR in the hypothalamus. To test this

hypothesis, we first assessed by Western blots whether C75 increases the phosphorylation

of mTOR and its downstream effectors S6K1 and S6 ribosomal protein (S6). We then

determined whether inhibition of mTOR signaling prevents the inhibitory effects of C75

on food consumption and modulates the mRNA expression of NPY, AgRP and POMC.

Our laboratory has previously reported that the anorectic signal of C75 is derived from its ability to increase CNS glucose metabolism (49). Neurons minimally oxidize

fatty acids, and instead, rely on glucose as their primary fuel under most conditions.

Therefore, we reasoned that the increased glucose utilization relative to the reduced lipid

use would be more likely to serve as the primary biochemical sensor for nutrient excess

in neurons (49); as opposed to the reduction in lipid use per se, caused by the malonyl-

CoA-induced inhibition of carnitine palmitoyl-CoA transferase-1 (CPT-1) (84).

Consistent with this view, we have demonstrated that C75 does not reduce food intake in

various conditions in which its effect on neuronal glucose oxidation is prevented,

76

including when ketone bodies are available as an alternative, and preferred, source of fuel

(49). Here, we explored the possibility that C75 fails to reduce caloric intake under the ketogenic diet because of its inability to activate mTOR signaling in the hypothalamus.

77

MATERIALS AND METHODS

Animals: Adult male Long-Evans rats (250–300 g at the beginning of the study) were housed individually and maintained on a 12:12-h light-dark cycle. Animals had ad libitum access to standard lab chow (15% lipid, 23% protein, and 62% carbohydrate calories)

(Harlan-Teklad, Indianapolis, IN) and water unless specified. All animal procedures were approved by the Institutional Animal Care and Use Committee of the University of

Cincinnati.

I3vt cannulation: Rats were implanted with third-ventricular (i3vt) cannulas as previously described (179). Briefly, rats were anaesthetized with ketamine/xylazine

(10: 6.5 solution; 1.0 ml/kg) and placed into a stereotaxic frame. A 22-gauge cannula

(Plastics One, Roanoke, VA, USA) was lowered along the midsagittal plane into the third ventricle (-2.2 mm anteroposterior and -7.5 mm dorsoventral with respect to bregma) and then secured to the skull with screws and dental acrylic. Animals were allowed to recover for a minimum of 7 days. Verification of cannula placement was confirmed by i3vt injection of 10 ng of angiotensin II in 1 µl 0.9% saline. Only animals that consumed at least 5 ml of water within 60 min of injection were included in the studies.

Drugs: All drugs were administered through an i3vt cannula. C75 (Calbiochem, EMD

Bioscience Inc., La Jolla, CA) was dissolved in RPMI (Gibco, Carlsbad, CA) and administered in a 2-µl volume, unless specified otherwise. Rapamycin (RAPA)

(Calbiochem, EMD Bioscience Inc., La Jolla, CA) was dissolved in dimethylsulfoxide

(DMSO) and administered in a 1-µl volume. All vehicle solutions served as controls.

78

Ketogenic diet: Rats were placed on a ketogenic diet (80.7% lipid, 14.3% protein, and

5% carbohydrate calories) (Dyet # 100959 Dyets Inc., Bethlehem, Pennsylvania) for 4

weeks before the experiment. Rats had ad libitum access to the ketogenic diet and water,

and to a second solution containing either 10% sucrose or 0.1 % saccharin, unless stated

otherwise (49).

β-hydroxybutyrate measurement: Fifty μl of blood was taken from the tip of the tail

after 24 h of fasting. Blood samples were collected into heparinised Natelson tubes and

immediately placed on ice in 1.5 ml heparinised Eppendorf tubes. β-hydroxybutyrate

concentration was measured using the KetoSite® test (Stanbio laboratory, Boerne,

Texas). Ketosis was confirmed by a significant elevation of blood β-hydroxybutyrate in

rats that had access to saccharin as compared to those given glucose while maintained on

the ketogenic diet.

Westerns: A wedge of the mediobasal hypothalamus was dissected using as landmarks the mammillary bodies caudally, the optic chiasm rostrally, the optic tract laterally and the apex of the third ventricle. Tissues were homogenized in RIPA lysis buffer (Santa

Cruz Biotechnology, Santa Cruz, CA) with addition of phosphatase and protease

inhibitors (Santa Cruz Biotechnology). Protein concentration was determined by BCA kit

(Pierce, Rockford, IL), following manufacturer’s instructions. Seventy µg of samples

were suspended in Laemmli sample buffer (BioRad, Hercules, CA), with 5% 2-

mercaptoethanol (Fisher Scientific, Hampton, NH) and lysates were denatured at 95°C

for 5 min. Samples were loaded on a 10% SDS-polyacrylamide gel (BioRad). Proteins

were separated by electrophoresis and transferred to nitrocellulose membranes.

Membranes were then incubated 1 h at room temperature in 1x TBS (20 mM Tris base

79

and 136 mM NaCl, pH 7.6) containing 0.1% Tween 20 (TBS-T) and 5% skim milk

powder and washed in TBS-T at room temperature before overnight incubation at 4°C

with one of the following primary antibody: pmTOR at Ser 2448 (Cell Signaling

Technology, Beverly, MA) (1:1000), pS6K1 at Thr 389 (Cell Signaling Technology,

1:500), pS6 at Ser 240/244 (Cell Signaling Technology, 1:500). After washing in TBS-T,

the membranes were incubated for 1 h at room temperature with secondary antibody

conjugated with horseradish peroxidase (goat anti-rabbit, 1:2000, Cell Signaling

Technology). After washes in TBS-T, immunopositive bands were visualized by

chemiluminescence (Lumiglo reagent and peroxide kit, Cell Signaling Technology) using exposure to radiographic films (Denville Scientific, South Plainfield, NJ). After protein detection, membranes were stripped for 15 min at 55 °C with a solution containing 62.5 mM Tris-HCl, 100 mM mercaptoethanol, and 2% SDS before 2 h blocking in TBS-T with

5% skim milk powder at room temperature, and re-blotted with either rabbit anti mTOR

(Cell Signaling Technology, 1:1000), rabbit anti p70 S6K1 (Cell Signaling Technology,

1:250) and rabbit anti S6 (Cell Signaling Technology, 1:500). The optical densitometry

of immunoreactive bands was measured using the NIH program Scion image.

q-PCR: Hypothalami were dissected using as landmarks the mammillary bodies

caudally, the optic chiasm rostrally and the optic tract laterally. Tissues were

homogenized in Tri-Reagent (Molecular Research Center, Cincinnati, OH), and total

RNA was isolated. cDNA was synthesized using SuperScript III kits (Invitrogen Corp.) and verified by L32 amplification products in agarose gel. PCR was performed using

Failsafe PCR kits (EPICENTRE Biotechnologies). For amplification of the reference gene L32, reaction mixture included Buffer I and forward primer 5’-

80

CCTCTGGTGAAGCCCAAGAT-3’ and reverse 5’-CTAGGCAGCATGTGCTTGGT-

3’. Thermocycler conditions were: 3 minutes at 95°C; 30 seconds at 95°C, 30 seconds at

55°C, and 45 seconds at 72°C for 25 cycles; and 5 minutes at 72°C. Q-PCR primer

sequences are shown in TABLE 1. Each primer set was optimized such that the

correlation coefficient was 0.99–1.0 and the PCR efficiency was 90–100%. Q-PCR was

performed in triplicate using an iCycler and the iQ SYBR Green Supermix (Bio-Rad

Laboratories) with 2-step amplification (95°C for 10 seconds, annealing temperature for

30 seconds) for 40 cycles. L32 was amplified from every sample for use as an

endogenous control. For the data analysis, the average threshold cycle (CT) of each set of

triplicates was calculated. To normalize the data, the CT was calculated for each sample

by subtracting the average CT of L32 from the average CT of the gene of interest. For

relative quantitation, the CT was averaged for the defined control group and was then

subtracted from the CT of each experimental sample to generate the CT. The CT was then used to calculate the approximate fold difference, 2 CT.

Experimental design:

Experiment 1: Effect of C75 on pmTOR, pS6K1 and pS6 levels. Rats were divided in 2

treatment groups based on their baseline food intake and body weight. Food was

removed and rats were weighed 2 h before the experiment. One hour before dark, rats

were injected with RPMI or C75 (30 μg in 2 μl, i3vt) and sacrificed 30 min and 1 h later.

Brains were removed, immediately frozen in isopentane and stored at -80 °C until

performing Western blot analysis.

81

Experiment 2: Effect of rapamycin (RAPA) on C75-induced anorexia. Rats were assigned

to one of four treatment groups (DMSO-RPMI, RAPA-RPMI, DMSO-C75, RAPA-C75) based on their baseline food intake and body weight. On the test day, rats were weighed and food was removed 2 h before the experiment. Three hours before the onset of the dark phase, rats received an injection of either DMSO or RAPA (25 μg in 1 μl, i3vt) followed by an injection of either RPMI or C75 (50 μg in 3 μl, i3vt) 2 h later. Food was immediately returned and weighed 1, 2, 4, 8 and 24 h after the last injection and body

weight was measured at 24 h.

Experiment 3: Effect of the ketogenic diet on C75-induced anorexia. Rats were divided

into 2 groups (saccharin or sucrose) based on their baseline body weight. All rats were

given ad libitum access to a ketogenic diet and to a sucrose or similarly sweet saccharin

solution. Water was available at all time. Four weeks later, 100 μl of blood was collected

into heparinised Natelson from the tip of the rat’s tail and blood β-hydroxybutyrate

concentration was measured using the KetoSite® test according to manufacturer’s

instruction (Stanbio Laboratory, Boerne, Texas). Three days later, the food and the solution of sucrose or saccharin were removed from the cages at the onset of the dark phase. The next day, each group (sucrose or saccharin) was subdivided into 2 treatment groups (RPMI or C75), based on their baseline intake of the ketogenic diet and their body weight, and rats were injected with either RPMI or C75 (30 μg in 2 μl, i3vt) 1 h prior to lights off. Food and the bottles of sucrose or saccharin were immediately returned and

weighed 4 and 24 h later.

82

Experiment 4: Effect of the ketogenic diet on C75-induced changes in pS6K1 and pS6 levels. The protocol was the same as described for Experiment 3, except that food, sucrose and saccharin were not returned after the injection in order to avoid any confounding effect of caloric intake. Rats were sacrificed 1 h later, and brains were taken for Western blot analysis of pS6K1 and pS6.

Experiment 5: Effect of C75 on NPY, AgRP and POMC mRNA expression, and consequence of rapamycin pre-treatment. Twenty-four hour fasted rats were injected with C75 (30 μg in 2 μl, i3vt) or RPMI 1 h before the onset of the dark phase, and sacrificed 2 h and 30 min later (92). Brains were rapidly removed and stored in RNA later (Ambion, Austin, Texas) at 4 °C until processed for RNA extraction and q-PCR.

Statistical analysis: For multiple group designs, the data were analyzed by two-way

ANOVA and Tukey’s post hoc tests. For designs with only two groups, statistical validity was assessed with unpaired t tests. Experiment-wise significance was set at P < 0.05, two- tailed. All values are expressed as the mean ± SEM.

83

RESULTS

Experiment 1: Effect of rapamycin on C75-induced anorexia. C75 significantly reduced

food intake compared with RPMI starting 4 h after the injection (DMSO-C75 = 1.86 +

0.68 vs. DMSO-RPMI = 6.51 + 0.52 g; P < 0.001; data not shown). This effect was still

present at 24 h (DMSO-C75 vs. DMSO-RPMI, P < 0.001) but was prevented by pre- treatment with the mTOR inhibitor RAPA (DMSO-C75 vs. RAPA-C75; P < 0.05, Fig.

1A) at a dose that did not affect food intake by itself (RAPA-RPMI vs. DMSO-RPMI; P

= 0.999, Fig. 1A). RAPA was also effective at preventing the body weight loss induced

by C75 over 24 h (DMSO-RPMI vs. DMSO-C75; P < 0.01 and DMSO-C75 vs. RAPA-

C75; P < 0.05, Fig. 1B).

Experiment 2: Effect of C75 on pmTOR, pS6K1 and pS6 levels. There was no significant

effect of the drug on the phosphorylation of S6K1 (pS6K1/S6K1: RPMI = 100.00 +

8.36% vs. C75 = 86.49 + 11.68 % of RPMI; P = 0.365) or S6 (pS6/S6: RPMI = 100 +

18.36% vs. C75 = 105.59 + 14.45 % of RPMI; P = 0.816) at 30 min post-injection.

However, C75 administration activates mTOR signaling at 1 h, as revealed by a

significant increase in the level of pmTOR (C75 vs. RPMI: P < 0.05; Fig. 2A) and pS6

(C75 vs. RPMI: P < 0.05; Fig. 2C) relative to vehicle. There was also a trend for C75 to

increase the phosphorylation of S6K1 at this time point (P = 0.05, 1-tailed t-test; Fig.

2B).

Experiment 3: Effect of the ketogenic diet on C75-induced anorexia. After 4 weeks on the

ketogenic diet, blood β-hydroxybutyrate concentration was significantly elevated in rats

84

given access to the saccharin solution as compared to sucrose (saccharin = 0.5 + 0.04 mM vs. sucrose = 0.17 + 0.03; P < 0.001). Consistent with its effect on the standard chow diet

(Fig. 1A), C75 significantly reduced caloric intake in rats given access to sucrose while maintained on the ketogenic diet (RPMI vs. C75: P < 0.05, Fig. 3). However, the anorexigenic effect of C75 was absent in rats receiving the saccharin solution (RPMI vs.

C75: P = 0.585; Fig. 3).

Experiment 4: Effect of the ketogenic diet on C75-induced changes in pS6K1 and pS6

levels. The ketogenic diet did not have any significant effect on the basal activation of the

mTOR pathway (levels of pS6K1/S6K1: saccharin-RPMI = 101.92 + 5.17 % of sucrose-

RPMI levels = 100.00 + 4.40 %; P = 0.783; data not shown), although there was a trend

for pS6 to be increased in rats exposed to saccharin as compared with those exposed to sucrose (pS6/S6: saccharin-RPMI = 152.27 + 17.93 % of sucrose-RPMI levels = 100.00

+ 15.17 %; P = 0.09). C75 administration stimulated mTOR signaling, as indicated by a

significant increase in pS6K1 and pS6, only in rats that consumed sucrose along with the

ketogenic diet [saccharin: (RPMI vs. C75; P = 0.729) vs. sucrose: (RPMI vs. C75; P <

0.05), Fig. 4].

Experiment 5: Effect of rapamycin on NPY, AgRP and POMC mRNA expression. The

anorexigenic effect of C75 was also accompanied by a significant reduction in

hypothalamic NPY mRNA levels (DMSO-RPMI vs. DMSO-C75, P < 0.05, Fig.5). There

was a trend for rapamycin to reverse the effect of C75 (DMSO-C75 vs. RAPA-C75, P =

0.07, Fig.5), although significantly reducing NPY levels as compared to DMSO (RAPA-

85

RPMI vs. DMSO-RPMI, P < 0.05, Fig.5). No changes were reported in the levels of

expression of AgRP (DMSO-RPMI = 100.00 + 5.58 % vs. DMSO-C75 = 83.88 + 11.09

%; P = 0.834) and POMC (DMSO-RPMI = 100.00 + 9.97 % vs. DMSO-C75 = 90.69 +

18.92 %; P = 0.856).

86

DISCUSSION

In this series of experiments, we present evidence that the FAS inhibitor C75

activates hypothalamic mTOR signaling in rats. This effect is necessary for the anorectic action of this compound, since pre-treatment with rapamycin, a well-known mTOR inhibitor (180), blocks C75-induced anorexia. We observed a potent effect of C75 at phosphorylating S6 1h after the injection, and a trend for pS6K1 to be increased by C75 as well. Since the phosphorylation of S6K is an event preceding that of S6, perhaps this time point was too late for observing a significant phosphorylation of the kinase. We have also evaluated possible changes in phosphorylation 30 min after the injection, but failed to detect any modification in either protein at this time point. Others have reported examples of conditions under which S6 is phosphorylated without parallel changes in

S6K1. In some cases, S6 activation was attributed to a second S6 kinase, termed S6K2, and the effect was mTOR-dependent (181). Therefore, one interpretation would be that

C75 stimulates S6 phosphorylation mainly via S6K2, which has not been measured in the present study. Another possibility is that C75 directly inhibits the phosphatase that dephosphorylates S6 (182, 183). Thus, the degree of S6 phosphorylation might depend on the balance between S6K and specific phosphatase activities at a specific time point.

In the past, we have proposed that C75’s ability to reduce caloric consumption

depends on its ability to increase glucose use in the brain, rather than reducing fatty acid

use per se (49). Consistent with this view, neuronal exposure to alternative fuels, and

consequent reduction in neuronal demand for glucose, attenuate the anorexigenic effect

of C75 (49). Central administration of glutamine or lactate, which is produced by

87

astrocytes and can be used by neurons as an alternative to glucose for ATP production

(117), significantly reduces the anorectic effect of C75. A similar phenomenon occurs

when rats are maintained on a very low-carbohydrate (ketogenic) diet for a prolonged

period of time (118). The latter condition mimics what happens during starvation, when

blood glucose levels fall and gluconeogenesis is not sufficient to respond to the brain’s

needs for glucose. Metabolic adaptations insure that the brain preferentially uses ketone

bodies over glucose for energy production (184).

In the current study, not only did we replicate our previous findings describing the

inability of C75 to reduce caloric intake in rats maintained on a ketogenic diet (49), but

also we found that C75 is ineffective at activating the mTOR pathway under this

condition. In contrast, when sucrose is provided alongside with the ketogenic diet, rats

remain sensitive to these C75-mediated effects, in a similar fashion to rats exposed to

normal chow. The implication of these findings is that the ability of C75 to activate

mTOR signaling requires increased CNS glucose utilization.

We observed a non-significant increase in the level of hypothalamic pS6 in the vehicle-injected rats exposed to ketogenic-saccharin diet. Interestingly, ketone bodies induce phosphorylation of S6K1 in primary cultured rat hepatocytes (185). Given that leucine is known to activate hypothalamic mTOR signaling (19), and that the levels of

leucine in the brain are increased following exposure to a ketogenic diet (186), these

events could have contributed to the non-significant elevation of pS6 in the vehicle-

88

treated rats in our study. Furthermore, this higher baseline could also have contributed to the lack of effect of C75 in the saccharin group.

The implication of our findings is two-fold. First, they demonstrate that activation of mTOR signaling is necessary for the anorexigenic effect of C75. Second, these results suggest that neuronal glucose metabolism is crucial for the effect of C75 on hypothalamic mTOR activation. CNS glucose metabolism is a key biochemical signal of nutrient status monitored by AMPK. Indeed, intracerebroventricular (icv) administration of glucose decreases AMPK activity and food intake, whereas 2-deoxyglucose, a well-known inhibitor of intracellular glucose utilization, does the opposite (34, 187). We have previously demonstrated that hypothalamic mTOR integrates nutrient signals from hormones and amino acids, such as leptin and leucine (19). Although our experiments did not specifically test this possibility, it is tempting to speculate that hypothalamic mTOR, like AMPK, also monitors glucose utilization. In the periphery, glucose-induced activation of S6K requires glucose metabolism, and the signals have therefore been proposed to directly emanate from glycolysis and/or glucose mitochondrial oxidation

(188). Given the established role of mTOR as an ATP sensor (39), an obvious candidate would be ATP.

C75 increases glucose oxidation as well as ATP levels in several cell types, including neurons (78, 90). Specifically, a study found that neuronal ATP levels are affected in a biphasic manner by C75: an initial decrease is then followed by a prolonged increase relative to control (78). Thus, the ability of C75 to stimulate mTOR might

89

involve its regulatory action on neuronal ATP levels. Future studies are necessary to

determine whether manipulations that inhibit ATP synthesis impair the ability of C75 to

activate hypothalamic mTOR signaling and to reduce food intake.

An interesting observation in the original report of Loftus et al. was that despite eating very little food, animals treated with C75 had a hypothalamic neuropeptide profile comparable to that of the fed state. Indeed, C75 reduced NPY levels relative to fasting, while suppressing 96% of food intake (84). We have recently reported that the phosphorylated form of mTOR and S6K1 is highly expressed in NPY/AgRP neurons, and to a lesser extent, in POMC/CART neurons. In addition, we found that the mTOR pathway is activated in the arcuate nucleus of the hypothalamus following refeeding

(189). The present data reveals another resemblance between C75 treatment and refeeding (189), since both conditions are able to drive hypothalamic mTOR signaling.

Furthermore, there is a trend for rapamycin to reverse the effect of C75 on NPY.

However, C75’s effect is rather small in comparison to previous reports (47, 92), therefore limiting the interpretation of the effects of rapamycin. Another caveat of our

study is that rapamycin reduced NPY relative to its vehicle DMSO. However, the

direction of this effect would suggest that it is unlikely that it contributed to block the

C75-induced reduction of NPY. Hence, it is still tempting to speculate the possible involvement of mTOR signaling in the effect of C75 at regulating NPY expression. The inhibition of NPY/AgRP neurons is believed to be an important component of the anorexigenic effect of C75 (47, 79, 84), although little is known about the intracellular events leading to this effect. The role of mTOR in regulating protein synthesis is well

90

established (55). Thus, an attractive hypothesis is that C75-induced activation of S6, a downstream target of S6K, might be linked to reduced NPY levels. Additional experiments are required to confirm the consequences of rapamycin on the ability of C75 to reduce NPY, and therefore to address the exact role of mTOR in this context.

Our experiments have not addressed whether the effects of C75 on the mTOR pathway are directly mediated, or rather occur indirectly through inhibition of hypothalamic AMPK. mTOR is recognized as a downstream target of AMPK, which inhibits its phosphorylation through activation of the TSC2 (119). Kim et al. have demonstrated the ability of C75 to dephosphorylate and inactivate AMPK. They proposed that this mechanism contributes to the ability of C75 to reduce food intake, since the

AMPK activator AICAR inhibits C75-induced anorexia (79). We cannot rule out the possibility that our observations result from a disinhibition of mTOR, through the inhibitory action of C75 on AMPK, rather than a direct stimulatory effect of the compound on the mTOR pathway. However, the time-course reported for AICAR (79) is very different from the one we observed with rapamycin. AICAR blocks C75’s anorexia exclusively in the first hour following the injection (79), while the effect of rapamycin occurs after 8 h and is still significant at 24 h. While this could be due to differences in experimental design and drug kinetics, it is still a difference that is worth considering.

In conclusion, these data provide evidence that link the CNS FAS system to the hypothalamic mTOR signaling cascade. This connects two separate metabolic pathways that have independently been linked to the regulation of energy balance. Indeed, the

91

anorectic action of leptin and the amino acid leucine also depends on its ability to activate hypothalamic mTOR signaling (19). Thus, a hypothetical model would be that C75 increases neuronal glucose metabolism, triggering increase ATP that would be sensed

and integrated by mTOR. The subsequent phosphorylation of this kinase would therefore

activate a cascade of intracellular events aimed at preventing further nutrient intake and

maintaining energy balance.

92

TABLE 1

Rat Q-PCR Primer Sequences

Gene Rat Q-PCR primer sequences T (°C)

L32 Forward: 5'-GCC AGG AGA CGA CAA AAA T 61.2

Reverse: 5'-AAT CCT CTT GCC CTG ATC C

AGRP Forward: 5'-TGT GTA AGG CTG CAC GAG TC 61.2

Reverse: 5'-GGC AGT AGC AAA AGG CAT TG

NPY Forward: 5'-AGG CTT GAA GAC CCT TCC AT 61.2

Reverse: 5'-ACA GGC AGA CTG GTT TCA GG

POMC Forward: 5'-CGC CCG TGT TTC CA 58.0

Reverse: 5'-TGA CCC ATG ACG TAC TTC C

93

FIGURE LEGEND

Figure 1. I3vt administration of rapamycin (25 µg in 1 µl DMSO) prevents the effects of

C75 (50 µg in 3 µl RPMI) on food intake (A) and on body weight (B). Mean ± SEM of 9

to 15 rats in each treatment group. ** P < 0.01; *** P < 0.001 vs. DMSO/RPMI-treated rats; 1 vs. DMSO/RPMI-treated rats; # P < 0.05 vs. DMSO/C75 rats.

Figure 2. C75 (30 µg in 2 µl RPMI, i3vt) increases hypothalamic mTOR signaling:

Representative Western blots from RPMI- or C75-treated rats and quantification by

image analysis of hypothalamic phosphorylation of mTOR (A), S6K1 (B) and S6 (C).**

P < 0.01; *** P < 0.001 vs. RPMI-treated rats. 7-8 brains examined in each condition.

Figure 3. C75 (30 µg in 2 µl RPMI) does not reduce caloric intake in rats given access to

saccharin alongside with the ketogenic diet, but do so with sucrose. * P < 0.05 vs. RPMI-

treated rats from the same group. Seven to eight rats in each treatment group.

Figure 4. C75 (30 µg in 2 µl RPMI) does not increase hypothalamic S6K1 and S6 phosphorylation in rats given access to saccharin while maintained on a ketogenic diet, but do so in rats receiving sucrose alongside with the diet. Representative Western blots from RPMI- or C75-treated rats from the saccharin and sucrose groups and quantification by image analysis of hypothalamic phosphorylation S6K1 (A) and S6 (B). * P < 0.05; P

< 0.01 vs. RPMI-treated rats from the same group. Five to eight brains were examined in each condition.

Figure 5. C75 significantly reduced NPY mRNA levels in the hypothalamus as

compared to RPMI controls, as did rapamycin. There was a trend for rapamycin to prevent the effect of C75 on NPY expression. * P < 0.05 vs. DMSO/RPMI-treated rats.

Seven to twelve brains were examined in each condition.

94

FIGURES

A

30 DMSO/RPMI 25 RAPA/RPMI 20 DMSO/C75 (50 μg) # RAPA/C75 (50 μg) 15 *** 10

5 24-h food intake (g)

0

B

-5

-15

-25 ** # -35 24-h weight body (g) change

Figure 1 Figure 1

95

A 150 RPMI ** 125 C75 (30 μg)

100

75

50 (% of RPMI) of (% pmTOR/mTOR 25

0

RPMI C75

pmTOR B mTOR 175 150 pS6K1 125 S6K1 100 75 (% of RPMI) of (% pS6 pS6K1/S6K1 50 25 S6 0

C 1200 *** 1000

800

600

pS6/S6 400 (% of RPMI) of (% 200 0 Figure 2

96

RPMI 140 C75 (30 μg) 120 100 80 * 60 40 20

24-h energy intake (kcal) intake energy 24-h 0 sucrose saccharin

Figure 3

97

RPMI C75 RPMI C75

pS6K1

S6K1

pS6

S6

sucrose saccharin

A B 200 200 RPMI C75 (30 μg) ** 150 * 150

100 100 pS6/S6 (% of RPMI) of (% (% of RPMI) of (% pS6K1/S6K1 50 50

0 0 sucrose saccharin sucrose saccharin

Figure 4

98

120 DMSO/RPMI 100 # * RAPA/RPMI 80 DMSO/C75 (30 μg) RAPA/C75 (30 μg) 60

40

20

0 NPY mRNA (% control DMSO/RPMI)

Figure 5

99

CHAPTER 4: GENERAL DISCUSSION

100

OEA as a derivative of oleic acid.

Very little is known about the intracellular mechanisms by which OEA-induced stimulation of PPAR-α leads to activation of brain regions involved in the control of energy balance (127). Oleic acid, the precursor of OEA, reduces food intake when directly administered into the CNS (18). As reviewed in the introduction, this causes an increase in the cytoplasmic concentration of long chain fatty acyl-CoAs (LCFA-CoAs), which is believed to mimic conditions of energy surplus and act as a signal to reduce food intake (88). Thus, an attractive hypothesis would be to attribute the anorectic effects of

OEA to the action of its catabolite oleic acid in the CNS. However, OEA does not affect food intake when delivered directly into the brain or when peripheral sensory fibres are removed by treatment with capsaicin (127). These findings suggest that OEA-induced anorexia is vagally mediated rather than a consequence of increased oleic acid levels

(127).

Our laboratory has collected data indicating that the susceptibility to develop obesity on a high fat diet depends on the fatty acid composition of the diet (Seeley R.J. and Woods S.C., unpublished data). Studies suggest that the “satiating” effect of ingested lipids depends not only on their quantity, but also on their properties (chain length, and degree of saturation and conjugation) (77). These properties have been proposed to influence the release of meal-related gastrointestinal signals that inhibit feeding, such as

CCK (190, 191). As such, it is interesting that rats maintained on a high-fat diet rich in monounsaturated fatty acids, such as oleic acid, are protected against diet-induced obesity

(Seeley R.J. and Woods SC., unpublished data). Because oleic acid is a precursor for

101

OEA and because it can reduce food intake (18), it is tempting to speculate that the lipid

composition of the diet modulates the synthesis of OEA, and that OEA consequently

contributes to the reduced weight gain and caloric consumption observed under these conditions.

OEA as an inhibitor of AMPK and FAS?

An interesting parallel can be drawn between OEA and its structural analogue

anandamide. Although they do not share the same receptor target, it is possible that

anandamide and OEA, two nutrient-derived signals whose pattern of secretion and

regulatory action on food intake are inversely related (192), share a common target within

the CNS. This is true for leptin and ghrelin. The secretion of these two adiposity signals

is also inversely related (193), and their opposite actions on hypothalamic AMPK are

required for their respective anorexigenic and orexigenic effects (34, 45).

OEA (127), anandamide (149) and stearoylethanolamide (SEA) (170) have all been

reported to control energy balance. An interesting point is that although these lipids are

all part of the same FAE family, and therefore have some degree of structural homology,

they do not regulate food intake through a common receptor. Anandamide appears to

mediate its effect on CNS feeding circuits via both a peripheral and central actions (149,

192), while OEA signal indirectly through the vagus nerve (127). The anorexigenic

action of SEA is accompanied by a reduction of stearoyl-CoA desaturase-1 (SCD-1)

mRNA expression in the liver, but its effects in the CNS have not been evaluated (170).

Previous work has linked the orexigenic effect of anandamide to activation of AMPK and

102

FAS in the hypothalamus (45, 194). OEA-induced anorexia occurs through stimulation of

the PPAR-alpha (127). Of interest, this nuclear receptor is involved in the regulation of

several aspects of lipid metabolism in the liver, partly through its ability to inhibit the

transcription of FAS (195). One might speculate that nutrient-derived lipids such as OEA

and anandamide have redundant signaling pathways or could share a common, yet

unidentified, target in the CNS. Given that anandamide is inversely related to OEA, and

that it mediates its orexigenic effect through activation of FAS and AMPK in the hypothalamus, an attractive hypothesis is that OEA and anandamide both converge on

FAS to regulate food intake in an opposite direction.

FAS inhibitors and mTOR.

In addition to AMPK, mTOR is an important CNS biochemical fuel sensor that

participates in the recognition of the energy status of the body and the regulation of

feedback mechanisms to restore energy balance. Hypothalamic mTOR integrates signals

from leptin and amino acids, and its phosphorylation is necessary for their anorexigenic

actions (19). The findings described in chapter 3 suggest that mTOR also integrates

metabolic signals derived from changes in neuronal glucose and fatty acid metabolism

induced by the FAS inhibitor C75.

To investigate the possible role of mTOR in the effects of C75, we assessed

whether rapamycin, a well-known inhibitor of the mTOR-raptor complex, would impair

the anorexia and weight loss produced by C75. Our results demonstrate that C75 increases the phosphorylation of mTOR and its downstream effectors S6K1 and S6 on

103

amino acids residues that are specifically related to activation of the mTOR pathway, and

we demonstrate that increased mTOR signaling is necessary for C75-induced anorexia.

These results provide a novel mechanism for the potent action of C75 on the regulation of

energy balance.

Neurons derive their energy primarily from glucose, and oxidize very small

amounts of fatty acids. In the original report of Loftus et al, increased malonyl-CoA and

reduced fatty acid oxidation were believed to mediate C75-induced anorexia (84). M.

Wortman rather proposed that it is the increase in the ratio of glucose to fatty acid

oxidation (glucose: fatty acid oxidation) that is monitored by neurons and interpreted as a

signal of energy surplus (49). Because neurons oxidize relatively more glucose than fatty

acids (86), the former was predicted to contribute to a larger extent to changes in the ratio

of glucose: fatty acid oxidation. Increased glucose utilization was hypothesized to be

necessary for C75 to signal a state of positive energy balance and induce anorexia. CNS

glucose utilization is at its nadir during ketosis, forcing neurons to use ketones as an

alternate source of energy (117). Consistent with this, the anorexigenic effect of C75 was

abolished in conditions that prevent it from increasing glucose use, including when rats

were in ketosis. Providing rats with a sucrose solution alongside with the ketogenic diet, which is presumed to reverse ketosis, restored the anorexigenic effect of C75. Given that malonyl-CoA levels are determined by the presence of acetyl-CoA, as well as the relative activity of ACC and MCD, the effects of C75 at reducing fatty acid oxidation would therefore not expected to be altered under this condition. The implication of these studies

104

is that increased glucose use is the primary signal necessary for the anorexigenic action of

C75 (49).

The lack of anorexigenic effect of C75 during ketosis is associated with changes in

the ability of C75 to increase mTOR signaling. This model is also an indirect way to

assess whether the increased neuronal glucose metabolism caused by C75 could be

sensed and integrated by mTOR, as measured by increased mTOR signaling. Our results

indicate the existence of a relationship between CNS glucose metabolism and the mTOR

pathway. Consistent with this view, the hypothalamic mTOR signaling pathway might not only be a target of the action of C75, but also an integrator of the status of glucose

and fatty acid metabolism in the CNS. One way to further confirm this hypothesis, would

be to examine whether inhibitors of glycolysis such as 2-deoxyglucose, recapitulate the

effects of ketosis on the mTOR pathway.

Relative contribution of AMPK and mTOR to C75-induced anorexia.

Activating AMPK or inhibiting mTOR signaling in the CNS both stimulate feeding

(19, 34, 41). Either one of these manipulations also reverses the phenotype induced by

C75 in terms of food intake, and possibly neuropeptide expression, making it difficult to

delineate the relative contribution of AMPK and mTOR to the metabolic effects of C75.

Redundancy is a critical feature of the neurocircuitry regulating food intake. While it is unlikely that a single pathway accounts for the effects of any one compound, an alternative possibility is that different pathways are more or less important to sense and integrate the signal of different fuels. Interestingly, the inhibitory effect of AMPK

105

activation on C75-induced anorexia occurs in the first hour following treatment and has

dissipated 3 hours later (79), whereas the effect of mTOR inhibition is revealed after 8

hours and is still significant 24 hours after injection of the drug. These observations

would suggest that inhibition of AMPK is predominantly involved in the acute anorectic

effect of C75, whereas activation of mTOR participates in the delayed and long-lasting

effects of C75.

Hypothetical model of C75-induced anorexia.

Chapter 3 describes experiments where rapamycin tends to prevent the reduction in

NPY levels that normally accompany C75. The fact that the effect of C75 on its own was relatively modest as compared to previous reports (47, 84) limited our interpretation of the data. Nonetheless, a point worth considering here is the similarities that C75 shares with leptin and leucine. All three factors activate the mTOR pathway, suppress food intake in an mTOR-dependent fashion and reduce NPY expression (19, 75). Furthermore, previous studies have demonstrated that this inhibitory action of leptin (196) and C75

(84) on NPY neurons is necessary for their anorectic effect, while this remains unknown in the case of leucine. This is consistent with a model in which mTOR acts as a convergence point within metabolic sensing neurons, through which signals reflecting the levels of stored and currently available fuels affect the synthesis of proteins involved in triggering the appropriate changes in terms of caloric intake and expenditure, so that energy homeostasis is maintained (see hypothetical model 1, Appendix 1). Further experiments are warranted to confirm this model, especially the effect of mTOR inhibition on leptin-, leucine- and C75-induced changes in neuropeptide expression.

106

Nevertheless, the data described in this thesis, together with those linking leptin and leucine to the mTOR pathway (19), support this model.

Based on the observation that reduced hypothalamic AMPK activity is required for

the anorexigenic action of both leptin (34) and C75 (79), Moran and colleagues have

proposed that leptin may affect food intake through interactions with the hypothalamic

fatty acid synthesis pathway. They found that leptin, like C75 (176), elevates malonyl-

CoA and that this is necessary for leptin to reduce food intake (52). Therefore it is

reasonable to propose that leptin may modulate mTOR through interaction with the FAS

pathway through the mechanism illustrated in “hypothetical model 2” (Appendix 2).

What is the purpose of this complex regulatory system?

From the perspective of food intake and energy balance regulation, it makes

intuitive sense for CNS fuel sensors to monitor the circulating levels of adiposity signals.

In contrast, meal-induced increase of circulating nutrients such as glucose and fatty acids

is poorly correlated to meal termination (33). Hence, the reasons for which signals of

immediately available fuels are integrated among the same circuits as those of stored

fuels are less clear. At the cellular level, however, this system might be in place to

prevent disruption of energy homeostasis, since this would impact on the ability of the

cell to grow and to survive. The control of cell size impacts on the size of organs, limbs

and the entire organism itself, which can somehow be compared to a large cell. One

difference, for example, is the extent of energy depletion necessary to induce cell versus

organism death, given that most mammals can survive for a relatively long time without

107

food. From a philosophical point of view, could these be reasons why the CNS monitors

current fuel status, in addition to stored energy? CNS fuel sensing systems might be

designed in a way to ensure that both the cell and the entire organism engage in the

appropriate anabolic and catabolic responses to maintain energy homeostasis, in order to

maintain their functions and preserve their viability.

Clinical perspectives.

The importance of understanding the molecular pathways underlying CNS fuel

sensing is highlighted by recent evidence indicating that these elegant mechanisms can

become disrupted under conditions of nutrient excess. Indeed, reduced sensitivity to the

anorexigenic effects of leptin, insulin and long chain fatty acids occurs relatively rapidly

after exposure to high fat diet (88, 197). Animals are therefore more susceptible to gain

weight and eventually, to become obese. Another point that is worth considering from a

clinical perspective is the rather large contribution these systems have to the lack of

success achieved so far by behavioral and pharmacological approaches for treating obesity. They counteract for decreased adiposity by driving appetite and hyperphagia,

and reducing energy expenditure. While these compensatory responses might have

evolved to favor animals survival in times of famine (3), they render weight loss difficult

to achieve and even more difficult to maintain in an environment of nutrient abundance

(198).

Thus, the redundancy of intracellular mechanisms that are in place to achieve such

energy homeostasis is undeniable. A example of this is that NPY knock-out mice eat and

108

gain weight similarly to wild-type animals, despite its presumed cardinal role as an

orexigenic neuropeptide (199). There is compelling evidence that hypothalamic AMPK

and mTOR are important integrators of fuel signals, but it is likely that other molecules

will emerge as CNS fuel sensors in the future as well. A large portion of the current

knowledge of CNS fuel sensing comes from hypotheses generated from what was already

known about fuel sensing in peripheral cells, and adapted to take into account the unique

environment of the brain. Such approach has been successful in identifying novel

mechanisms by which FAS inhibitors regulate food intake (49) and more recently, in identifying AMPK (34) and mTOR (19) as a CNS fuel sensors.

In that regard, it has recently been reported that carcinoembryonic antigen-related

cell adhesion molecule 1 (CEACAM-1), a glycoprotein downstream of insulin signaling,

is an endogenous inhibitor of FAS in the liver (200). In the fed state, a condition under

which ACC would be expected to be active and its substrate acetyl-CoA elevated, there is

an elevation of hypothalamic malonyl-CoA (176). This suggests that either: 1) FAS

activity is relatively too low to compensate, therefore allowing malonyl-CoA to

accumulate, or 2) there is a simultaneous inhibition of FAS. While Kim et al. have not

observed any change in hypothalamic FAS mRNA, protein or enzymatic activity in free

fed rats relative to animals that been fasted for 24 h (47), we found that refeeding after a

24-h fast acutely reduces FAS activity in the hypothalamus, supporting the latter view (K.

Proulx, unpublished data). Given that refeeding acutely reduces hepatic FAS activity

through activation of CEACAM-1 (200), and that the temporal pattern of this effect is

comparable to the one we observed in the hypothalamus (K. Proulx, unpublished data), it

109

is tempting to hypothesize that CEACAM-1 is an endogenous inhibitor of FAS in the hypothalamus as well. In preliminary experiments, we found that CEACAM-1 mRNA is regulated by feeding and insulin in the hypothalamus (K. Proulx, unpublished data). The potent anorexia observed with pharmacological inhibitors of FAS supports the clinical potential of agents that could increase the action of such putative endogenous FAS inhibitor in the brain.

A drug that would affect both sides of the energy balance equation, in a way to reduce food intake and increase fat oxidation in the periphery, would have therapeutic potential. The problem in considering mTOR and AMPK as targets for drug therapy is that their peripheral and central actions oppose each other in terms of energy balance regulation. For instance, pharmacological inhibition of the mTOR pathway in the CNS

(19), as well as total knockout of the S6K1 gene (74) are associated with increased food intake in rodents, supporting that CNS mTOR promotes a negative energy balance by regulating feeding. Despite this, total S6K1 knockout mice maintained on a high fat diet are leaner than controls due to increased fatty acid oxidation (74). These effects might appear to oppose each other in terms of body weight regulation. However, they are consistent with the concept of a fuel sensor. In fact, endogenous factors such as leptin

(34) and α-lipoic acid (187) inhibit AMPK in the CNS while activating it in the periphery, therefore promoting a negative energy balance. The effect of therapeutic agents is more problematic. Insulin therapy is an example. Type II diabetes patients can gain weight as a result of insulin treatment, probably because the anorexigenic action of insulin in the brain is counteracted by its potent stimulatory effect on fatty acid synthesis

110

in the periphery. So from a CNS perspective, one could predict that the potency of mTOR activator or AMPK inhibitor to induce weight loss would be determined by the relative balance between their inhibitory action on food intake and fatty acid oxidation. This dual action complicates the development of therapeutic strategies. On a more optimistic note, understanding the mechanisms by which endogenous factors modulate the activity of fuel sensors in an opposite direction in the CNS versus the periphery might lead to the development of “intelligent” drugs, which would do the same. Again, answers to this dilemma might come from identifying differences between cells of the CNS and those of the periphery in terms of their metabolism and the environment in which they operate.

Such an approach might lead to the identification of features that are specific to each cell type, and that could be used as targets to increase the specificity of novel drugs. Ideally, such treatments would activate mTOR in the CNS while inhibiting it in the periphery, and vice versa for AMPK.

111

REFERENCES

1. Kopelman, P.G. 2000. Obesity as a medical problem. Nature 404:635-643. 2. Barsh, G.S., Farooqi, I.S., and O'Rahilly, S. 2000. Genetics of body-weight regulation. Nature 404:644-651. 3. Neel, J.V. 1962. Diabetes mellitus: a "thrifty" genotype rendered detrimental by "progress?" American Journal Human Genetics 14:353-362. 4. Weigle, D.S. 1994. Appetite and the regulation of body composition. Faseb J 8:302-310. 5. Fox, F.W. 1973. The enigma of obesity. Lancet 2:1487-1488. 6. Friedrich, M.J. 2002. Epidemic of obesity expands its spread to developing countries. Jama 287:1382-1386. 7. Seeley, R.J., and Woods, S.C. 2003. Monitoring of stored and available fuel by the CNS: implications for obesity. Nat Rev Neurosci 4:901-909. 8. Woods, S.C. 2005. Signals that influence food intake and body weight. Physiol Behav 86:709-716. 9. Benoit, S.C., Clegg, D.J., Seeley, R.J., and Woods, S.C. 2004. Insulin and leptin as adiposity signals. Recent Prog Horm Res 59:267-285. 10. Considine, R.V., Sinha, M.K., Heiman, M.L., Kriaucinas, A., Stephens, T.W., Nyce, M.R., Ohannesian, J.P., Marco, C.C., McKee, L.J., Bauer, T.L., et al. 1996. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. The New England Journal of Medicine 334:292-295. 11. Caro, J.F., Kolaczynski, J.W., Nyce, M.R., Ohannesian, J.P., Opentanova, I., Goldman, W.H., Lynn, R.B., Zhang, P.L., Sinha, M.K., and Considine, R.V. 1996. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet 348:159-161. 12. Wisse, B.E., Campfield, L.A., Marliss, E.B., Morais, J.A., Tenenbaum, R., and Gougeon, R. 1999. Effect of prolonged moderate and severe energy restriction and refeeding on plasma leptin concentrations in obese women. American Journal of Clinical Nutrition 70:321-330. 13. Nakazato, M., Murakami, N., Date, Y., Kojima, M., Matsuo, H., Kangawa, K., and Matsukura, S. 2001. A role for ghrelin in the central regulation of feeding. Nature 409:194-198. 14. Seeley, R.J., van Dijk, G., Campfield, L.A., Smith, F.J., Nelligan, J.A., Bell, S.M., Baskin, D.G., Woods, S.C., and Schwartz, M.W. 1996. The effect of intraventricular administration of leptin on food intake and body weight in the rat. Hormone and Metabolic Research 28:664-668. 15. Woods, S.C., Lotter, E.C., McKay, L.D., and Porte, D., Jr. 1979. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282:503-505. 16. Levin, B.E. 2001. Glucosensing neurons do more than just sense glucose. Int J Obes Relat Metab Disord 25 Suppl 5:S68-72. 17. Kurata, K., Fujimoto, K., Sakata, T., Etou, H., and Fukagawa, K. 1986. D-glucose suppression of eating after intra-third ventricle infusion in rat. Physiol Behav 37:615-620.

112

18. Obici, S., Feng, Z., Morgan, K., Stein, D., Karkanias, G., and Rossetti, L. 2002. Central administration of oleic acid inhibits glucose production and food intake. Diabetes 51:271-275. 19. Cota, D., Proulx, K., Smith, K.A., Kozma, S.C., Thomas, G., Woods, S.C., and Seeley, R.J. 2006. Hypothalamic mTOR signaling regulates food intake. Science. 20. Tartaglia, L.A., Dembski, M., Weng, X., Deng, N., Culpepper, J., Devos, R., Richards, G.J., Campfield, L.A., Clark, F.T., Deeds, J., et al. 1995. Identification and expression cloning of a leptin receptor, OB-R. Cell 83:1263-1271. 21. Van Houten, M., Posner, B.I., Kopriwa, B.M., and Brawer, J.R. 1979. Insulin binding sites in the rat brain: in vivo localization to the circumventricular organs by quantitative radioautoradiography. Endocrinology 105:666-673. 22. Van Houten, M., Posner, B.I., Kopriwa, B.M., and Brawer, J.R. 1980. Insulin binding sites localized to nerve terminals in rat median eminence and arcuate nucleus. Science 207:1081-1083. 23. Guan, X.M., Hess, J.F., Yu, H., Hey, P.J., and van der Ploeg, L.H. 1997. Differential expression of mRNA for leptin receptor isoforms in the rat brain. Mol Cell Endocrinol 133:1-7. 24. Kang, L., Routh, V.H., Kuzhikandathil, E.V., Gaspers, L.D., and Levin, B.E. 2004. Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. Diabetes 53:549-559. 25. Fan, W., Boston, B.A., Kesterson, R.A., Hruby, V.J., and Cone, R.D. 1997. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385:165-168. 26. Tsujii, S., and Bray, G.A. 1989. Acetylation alters the feeding response to MSH and beta-endorphin. Brain Research Bulletin 23:165-169. 27. Farooqi, I.S., and O'Rahilly, S. 2005. Monogenic obesity in humans. Annu Rev Med 56:443-458. 28. Hahn, T.M., Breininger, J.F., Baskin, D.G., and Schwartz, M.W. 1998. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nature Neuroscience 1:271-272. 29. Chen, P., Li, C., Haskell-Luevano, C., Cone, R.D., and Smith, M.S. 1999. Altered expression of agouti-related protein and its colocalization with neuropeptide Y in the arcuate nucleus of the hypothalamus during lactation. Endocrinology 140:2645-2650. 30. Nijenhuis, W.A., Oosterom, J., and Adan, R.A. 2001. AgRP(83-132) acts as an inverse agonist on the human-melanocortin-4 receptor. Mol Endocrinol 15:164- 171. 31. Ollmann, M., Wilson, B., Yang, Y., Kerns, J., Chen, Y., Gantz, I., and Barsh, G. 1997. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278:135-138. 32. Xu, B., Li, B.-H., Rowland, N.E., and Kalra, S.P. 1995. Neuropeptide Y injection into the fourth cerebroventricle stimulates c-Fos expression in the paraventricular nucleus and other nuclei in the forebrain: effect of food consumption. Brain Research 698:227-231.

113

33. Levin, B.E., Routh, V.H., Kang, L., Sanders, N.M., and Dunn-Meynell, A.A. 2004. Neuronal glucosensing: what do we know after 50 years? Diabetes 53:2521-2528. 34. Minokoshi, Y., Alquier, T., Furukawa, N., Kim, Y.B., Lee, A., Xue, B., Mu, J., Foufelle, F., Ferre, P., Birnbaum, M.J., et al. 2004. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428:569-574. 35. Mayer, J. 1953. Glucostatic mechanism of regulation of food intake. New England Journal of Medicine 249:13-16. 36. Kennedy, G.C. 1953. The role of depot fat in the hypothalamic control of food intake in the rat. Proc R Soc Lond (Biol) 140:579-592. 37. Mayer, J. 1955. Regulation of energy intake and the body weight: The glucostatic and lipostatic hypothesis. Annals of the New York Academy of Sciences 63:14-42. 38. Hardie, D.G., and Carling, D. 1997. The AMP-activated protein kinase--fuel gauge of the mammalian cell? Eur J Biochem 246:259-273. 39. Dennis, P.B., Jaeschke, A., Saitoh, M., Fowler, B., Kozma, S.C., and Thomas, G. 2001. Mammalian TOR: a homeostatic ATP sensor. Science 294:1102-1105. 40. Thomas, G. 2002. The S6 kinase signaling pathway in the control of development and growth. Biol Res 35:305-313. 41. Andersson, U., Filipsson, K., Abbott, C.R., Woods, A., Smith, K., Bloom, S.R., Carling, D., and Small, C.J. 2004. AMP-activated protein kinase plays a role in the control of food intake. J Biol Chem 279:12005-12008. 42. Kahn, B.B., Alquier, T., Carling, D., and Hardie, D.G. 2005. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 1:15-25. 43. Ruderman, N.B., Park, H., Kaushik, V.K., Dean, D., Constant, S., Prentki, M., and Saha, A.K. 2003. AMPK as a metabolic switch in rat muscle, liver and adipose tissue after exercise. Acta Physiol Scand 178:435-442. 44. Hardie, D.G., Scott, J.W., Pan, D.A., and Hudson, E.R. 2003. Management of cellular energy by the AMP-activated protein kinase system. FEBS Lett 546:113- 120. 45. Kola, B., Hubina, E., Tucci, S.A., Kirkham, T.C., Garcia, E.A., Mitchell, S.E., Williams, L.M., Hawley, S.A., Hardie, D.G., Grossman, A.B., et al. 2005. Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. J Biol Chem 280:25196-25201. 46. Ruderman, N.B., Saha, A.K., Vavvas, D., and Witters, L.A. 1999. Malonyl-CoA, fuel sensing, and insulin resistance. Am J Physiol 276:E1-E18. 47. Kim, E.K., Miller, I., Landree, L.E., Borisy-Rudin, F.F., Brown, P., Tihan, T., Townsend, C.A., Witters, L.A., Moran, T.H., Kuhajda, F.P., et al. 2002. Expression of FAS within hypothalamic neurons: a model for decreased food intake after C75 treatment. American Journal of Physiology 283:E867-E879. 48. Clegg, D.J., Wortman, M.D., Benoit, S.C., McOsker, C.C., and Seeley, R.J. 2002. Comparison of central and peripheral administration of C75 on food intake, body weight, and conditioned taste aversion. Diabetes 51:3196-3201.

114

49. Wortman, M.D., Clegg, D.J., D'Alessio, D., Woods, S.C., and Seeley, R.J. 2003. C75 inhibits food intake by increasing CNS glucose metabolism. Nature Medicine 9:483-485. 50. Ronnett, G.V., Kim, E.K., Landree, L.E., and Tu, Y. 2005. Fatty acid metabolism as a target for obesity treatment. Physiol Behav 85:25-35. 51. Dowell, P., Hu, Z., and Lane, M.D. 2005. Monitoring energy balance: metabolites of fatty acid synthesis as hypothalamic sensors. Annu Rev Biochem 74:515-534. 52. GAO, S., Kinzig, K., AJA, S., LADENHEIM, E.E., and MORAN, T.H. 2005. Hypothalamic de novo fatty acid synthesis mediates leptin’s anorectic effects. Society for the Study of Ingestive Behavior, Annual Meeting 2005, Pittsburgh, PA. 53. Minokoshi, Y., Kim, Y.B., Peroni, O.D., Fryer, L.G., Muller, C., Carling, D., and Kahn, B.B. 2002. Leptin stimulates fatty-acid oxidation by activating AMP- activated protein kinase. Nature 415:339-343. 54. Nurse, P. 1975. Genetic control of cell size at cell division in yeast. Nature 256:547-551. 55. Jacinto, E., and Hall, M.N. 2003. Tor signalling in bugs, brain and brawn. Nat Rev Mol Cell Biol 4:117-126. 56. Sabatini, D.M., Erdjument-Bromage, H., Lui, M., Tempst, P., and Snyder, S.H. 1994. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin- dependent fashion and is homologous to yeast TORs. Cell 78:35-43. 57. Chiu, M.I., Katz, H., and Berlin, V. 1994. RAPT1, a mammalian homolog of yeast Tor, interacts with the FKBP12/rapamycin complex. Proc Natl Acad Sci U S A 91:12574-12578. 58. Brown, E.J., Albers, M.W., Shin, T.B., Ichikawa, K., Keith, C.T., Lane, W.S., and Schreiber, S.L. 1994. A mammalian protein targeted by G1-arresting rapamycin- receptor complex. Nature 369:756-758. 59. Hay, N., and Sonenberg, N. 2004. Upstream and downstream of mTOR. Genes Dev 18:1926-1945. 60. Reynolds, T.H.t., Bodine, S.C., and Lawrence, J.C., Jr. 2002. Control of Ser2448 phosphorylation in the mammalian target of rapamycin by insulin and skeletal muscle load. J Biol Chem 277:17657-17662. 61. Nave, B.T., Ouwens, M., Withers, D.J., Alessi, D.R., and Shepherd, P.R. 1999. Mammalian target of rapamycin is a direct target for protein kinase B: identification of a convergence point for opposing effects of insulin and amino- acid deficiency on protein translation. Biochem J 344 Pt 2:427-431. 62. Sarbassov, D.D., Ali, S.M., Kim, D.H., Guertin, D.A., Latek, R.R., Erdjument- Bromage, H., Tempst, P., and Sabatini, D.M. 2004. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 14:1296-1302. 63. Kim, D.H., Sarbassov, D.D., Ali, S.M., King, J.E., Latek, R.R., Erdjument- Bromage, H., Tempst, P., and Sabatini, D.M. 2002. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110:163-175. 64. Inui, S., Sanjo, H., Maeda, K., Yamamoto, H., Miyamoto, E., and Sakaguchi, N. 1998. Ig receptor binding protein 1 (alpha4) is associated with a rapamycin-

115

sensitive signal transduction in lymphocytes through direct binding to the catalytic subunit of protein phosphatase 2A. Blood 92:539-546. 65. Nanahoshi, M., Nishiuma, T., Tsujishita, Y., Hara, K., Inui, S., Sakaguchi, N., and Yonezawa, K. 1998. Regulation of protein phosphatase 2A catalytic activity by alpha4 protein and its yeast homolog Tap42. Biochem Biophys Res Commun 251:520-526. 66. Sehgal, S.N. 2003. Sirolimus: its discovery, biological properties, and mechanism of action. Transplant Proc 35:7S-14S. 67. Burnett, P.E., Barrow, R.K., Cohen, N.A., Snyder, S.H., and Sabatini, D.M. 1998. RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E- BP1. Proc Natl Acad Sci U S A 95:1432-1437. 68. Guba, M., von Breitenbuch, P., Steinbauer, M., Koehl, G., Flegel, S., Hornung, M., Bruns, C.J., Zuelke, C., Farkas, S., Anthuber, M., et al. 2002. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 8:128-135. 69. Dennis, P.B., Fumagalli, S., and Thomas, G. 1999. Target of rapamycin (TOR): balancing the opposing forces of protein synthesis and degradation. Curr Opin Genet Dev 9:49-54. 70. Gangloff, Y.G., Mueller, M., Dann, S.G., Svoboda, P., Sticker, M., Spetz, J.F., Um, S.H., Brown, E.J., Cereghini, S., Thomas, G., et al. 2004. Disruption of the mouse mTOR gene leads to early postimplantation lethality and prohibits embryonic stem cell development. Mol Cell Biol 24:9508-9516. 71. Shima, H., Pende, M., Chen, Y., Fumagalli, S., Thomas, G., and Kozma, S.C. 1998. Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse phenotype and a new functional S6 kinase. Embo J 17:6649-6659. 72. Montagne, J., Stewart, M.J., Stocker, H., Hafen, E., Kozma, S.C., and Thomas, G. 1999. Drosophila S6 kinase: a regulator of cell size. Science 285:2126-2129. 73. Pende, M., Kozma, S.C., Jaquet, M., Oorschot, V., Burcelin, R., Le Marchand- Brustel, Y., Klumperman, J., Thorens, B., and Thomas, G. 2000. Hypoinsulinaemia, glucose intolerance and diminished beta-cell size in S6K1- deficient mice. Nature 408:994-997. 74. Um, S.H., Frigerio, F., Watanabe, M., Picard, F., Joaquin, M., Sticker, M., Fumagalli, S., Allegrini, P.R., Kozma, S.C., Auwerx, J., et al. 2004. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature 431:200-205. 75. Schwartz, M.W., Seeley, R.J., Campfield, L.A., Burn, P., and Baskin, D.G. 1996. Identification of hypothalmic targets of leptin action. Journal of Clinical Investigation 98:1101-1106. 76. Wu, Q., Zhang, Y., Xu, J., and Shen, P. 2005. Regulation of hunger-driven behaviors by neural ribosomal S6 kinase in Drosophila. Proc Natl Acad Sci U S A 102:13289-13294. 77. Leonhardt, M., and Langhans, W. 2004. Fatty acid oxidation and control of food intake. Physiol Behav 83:645-651. 78. Landree, L.E., Hanlon, A.L., Strong, D.W., Rumbaugh, G., Miller, I.M., Thupari, J.N., Connolly, E.C., Huganir, R.L., Richardson, C., Witters, L.A., et al. 2004.

116

C75, a fatty acid synthase inhibitor, modulates AMP-activated protein kinase to alter neuronal energy metabolism. J Biol Chem 279:3817-3827. 79. Kim, E.K., Miller, I., Aja, S., Landree, L.E., Pinn, M., McFadden, J., Kuhajda, F.P., Moran, T.H., and Ronnett, G.V. 2004. C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase. J Biol Chem 279:19970-19976. 80. Kuhajda, F.P., Jenner, K., Wood, F.D., Hennigar, R.A., Jacobs, L.B., Dick, J.D., and Pasternack, G.R. 1994. Fatty acid synthesis: a potential selective target for antineoplastic therapy. Proc Natl Acad Sci U S A 91:6379-6383. 81. Kuhajda, F.P., Pizer, E.S., Li, J.N., Mani, N.S., Frehywot, G.L., and Townsend, C.A. 2000. Synthesis and antitumor activity of an inhibitor of fatty acid synthase. Proc Natl Acad Sci U S A 97:3450-3454. 82. Rendina, A.R., and Cheng, D. 2005. Characterization of the inactivation of rat fatty acid synthase by C75: inhibition of partial reactions and protection by substrates. Biochem J 388:895-903. 83. Wang, X., and Tian, W. 2001. Green tea epigallocatechin gallate: a natural inhibitor of fatty-acid synthase. Biochem Biophys Res Commun 288:1200-1206. 84. Loftus, T.M., Jaworsky, D.E., Frehywot, G.L., Townsend, C.A., Ronnett, G.V., Lane, M.D., and Kuhajda, F.P. 2000. Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science 288:2299-2300. 85. Jayakumar, A., Tai, M.H., Huang, W.Y., al-Feel, W., Hsu, M., Abu-Elheiga, L., Chirala, S.S., and Wakil, S.J. 1995. Human fatty acid synthase: properties and molecular cloning. Proc Natl Acad Sci U S A 92:8695-8699. 86. Sokoloff, L., Reivich, M., Kennedy, C., Des Rosiers, M.H., Patlak, C.S., Pettigrew, K.D., Sakurada, O., and Shinohara, M. 1977. The [14C]deoxyglucose method for the measurement of local cerebral glucose utilization: theory, procedure, and normal values in the conscious and anesthetized albino rat. J Neurochem 28:897-916. 87. Kusakabe, T., Maeda, M., Hoshi, N., Sugino, T., Watanabe, K., Fukuda, T., and Suzuki, T. 2000. Fatty acid synthase is expressed mainly in adult hormone- sensitive cells or cells with high lipid metabolism and in proliferating fetal cells. J Histochem Cytochem 48:613-622. 88. Lam, T.K., Schwartz, G.J., and Rossetti, L. 2005. Hypothalamic sensing of fatty acids. Nat Neurosci 8:579-584. 89. Hu, Z., Dai, Y., Prentki, M., Chohnan, S., and Lane, M.D. 2005. A role for hypothalamic malonyl-CoA in the control of food intake. J Biol Chem 280:39681- 39683. 90. Thupari, J.N., Landree, L.E., Ronnett, G.V., and Kuhajda, F.P. 2002. C75 increases peripheral energy utilization and fatty acid oxidation in diet-induced obesity. Proc Natl Acad Sci U S A 11:11. 91. Thupari, J.N., Kim, E.K., Moran, T.H., Ronnett, G.V., and Kuhajda, F.P. 2004. Chronic C75 treatment of diet-induced obese mice increases fat oxidation and reduces food intake to reduce adipose mass. Am J Physiol Endocrinol Metab 287:E97-E104.

117

92. Shimokawa, T., Kumar, M.V., and Lane, M.D. 2002. Effect of a fatty acid synthase inhibitor on food intake and expression of hypothalamic neuropeptides. Proc Natl Acad Sci U S A 99:66-71. 93. Hu, Z., Cha, S.H., van Haasteren, G., Wang, J., and Lane, M.D. 2005. Effect of centrally administered C75, a fatty acid synthase inhibitor, on ghrelin secretion and its downstream effects. Proc Natl Acad Sci U S A 102:3972-3977. 94. Cowley, M.A. 2003. Hypothalamic melanocortin neurons integrate signals of energy state. Eur J Pharmacol 480:3-11. 95. Ruderman, N., and Prentki, M. 2004. AMP kinase and malonyl-CoA: targets for therapy of the metabolic syndrome. Nat Rev Drug Discov 3:340-351. 96. Pocai, A., Lam, T.K., Obici, S., Gutierrez-Juarez, R., Muse, E.D., Arduini, A., and Rossetti, L. 2006. Restoration of hypothalamic lipid sensing normalizes energy and glucose homeostasis in overfed rats. J Clin Invest 116:1081-1091. 97. McGarry, J.D., Mannaerts, G.P., and Foster, D.W. 1977. A possible role for malonyl-CoA in the regulation of hepatic fatty acid oxidation and ketogenesis. J Clin Invest 60:265-270. 98. He, W., Lam, T.K., Obici, S., and Rossetti, L. 2006. Molecular disruption of hypothalamic nutrient sensing induces obesity. Nat Neurosci 9:227-233. 99. Obici, S., and Rossetti, L. 2003. Minireview: nutrient sensing and the regulation of insulin action and energy balance. Endocrinology 144:5172-5178. 100. Obici, S., Feng, Z., Arduini, A., Conti, R., and Rossetti, L. 2003. Inhibition of hypothalamic carnitine palmitoyltransferase-1 decreases food intake and glucose production. Nature Medicine 9:756-761. 101. Jin, Y.J., Li, S.Z., Zhao, Z.S., An, J.J., Kim, R.Y., Kim, Y.M., Baik, J.H., and Lim, S.K. 2004. Carnitine palmitoyltransferase-1 (CPT-1) activity stimulation by cerulenin via sympathetic nervous system activation overrides cerulenin's peripheral effect. Endocrinology 145:3197-3204. 102. Tu, Y., Thupari, J.N., Kim, E.K., Pinn, M.L., Moran, T.H., Ronnett, G.V., and Kuhajda, F.P. 2005. C75 alters central and peripheral gene expression to reduce food intake and increase energy expenditure. Endocrinology 146:486-493. 103. Makimura, H., Mizuno, T.M., Yang, X.J., Silverstein, J., Beasley, J., and Mobbs, C.V. 2001. Cerulenin mimics effects of leptin on metabolic rate, food intake, and body weight independent of the melanocortin system, but unlike leptin, cerulenin fails to block neuroendocrine effects of fasting. Diabetes 50:733-739. 104. Aja, S., Bi, S., Knipp, S.B., McFadden, J.M., Ronnett, G.V., Kuhajda, F.P., and Moran, T.H. 2006. Intracerebroventricular C75 decreases meal frequency and reduces AgRP gene expression in rats. Am J Physiol Regul Integr Comp Physiol. 105. Bentebibel, A., Sebastian, D., Herrero, L., Lopez-Vinas, E., Serra, D., Asins, G., Gomez-Puertas, P., and Hegardt, F.G. 2006. Novel effect of C75 on carnitine palmitoyltransferase I activity and palmitate oxidation. Biochemistry 45:4339- 4350. 106. Heller, S.R., and Cryer, P.E. 1991. Reduced neuroendocrine and symptomatic responses to subsequent hypoglycemia after 1 episode of hypoglycemia in nondiabetic humans. Diabetes 40:223-226. 107. Bernardis, L.L., Luboshitzky, R., and Bellinger, L.L. 1981. Long-term effects of insulin in weanling rats with dorsomedial hypothalamic hypophagia: food intake,

118

efficiency of food utilization, body weight and composition. Physiol Behav 27:469-474. 108. Campfield, L.A., Brandon, P., and Smith, F.J. 1985. On-line continuous measurement of blood glucose and meal pattern in free-feeding rats: the role of glucose in meal initiation. Brain Research Bulletin 14:605-616. 109. Melanson, K.J., Westerterp-Plantenga, M.S., Saris, W.H., Smith, F.J., and Campfield, L.A. 1999. Blood glucose patterns and appetite in time-blinded humans: carbohydrate versus fat. Am J Physiol 277:R337-345. 110. Kerr, D., Diamond, M.P., Tamborlane, W.V., Kerr, S., and Sherwin, R.S. 1993. Influence of counterregulatory hormones, independently of hypoglycaemia, on cognitive function, warning symptoms and glucose kinetics. Clin Sci (Lond) 85:197-202. 111. Oomura, Y., Kimura, K., Ooyama, H., Maeno, T., Iki, M., and Kuniyoshi, M. 1964. Reciprocal Activities Of The Ventromedial And Lateral Hypothalamic Areas Of Cats. Science 143:484-485. 112. Ibrahim, N., Bosch, M.A., Smart, J.L., Qiu, J., Rubinstein, M., Ronnekleiv, O.K., Low, M.J., and Kelly, M.J. 2003. Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. Endocrinology 144:1331-1340. 113. Muroya, S., Yada, T., Shioda, S., and Takigawa, M. 1999. Glucose-sensitive neurons in the rat arcuate nucleus contain neuropeptide Y. Neurosci Lett 264:113- 116. 114. Oomura, Y., Nakamura, T., Sugimori, M., and Yamada, Y. 1975. Effect of free fatty acid on the rat lateral hypothalamic neurons. Physiol Behav 14:483-486. 115. Borg, W.P., During, M.J., Sherwin, R.S., Borg, M.A., Brines, M.L., and Shulman, G.I. 1994. Ventromedial hypothalamic lesions in rats suppress counterregulatory responses to hypoglycemia. J Clin Invest 93:1677-1682. 116. Levin, B.E., Dunn-Meynell, A.A., and Routh, V.H. 1999. Brain glucose sensing and body energy homeostasis: role in obesity and diabetes. American Journal of Physiology 276:R1223-R1231. 117. Tsacopoulos, M. 2002. Metabolic signaling between neurons and glial cells: a short review. J Physiol Paris 96:283-288. 118. Guzman, M., and Blazquez, C. 2001. Is there an astrocyte-neuron ketone body shuttle? Trends Endocrinol Metab 12:169-173. 119. Inoki, K., Zhu, T., and Guan, K.L. 2003. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115:577-590. 120. Bi, S., and Moran, T.H. 2002. Actions of CCK in the controls of food intake and body weight: lessons from the CCK-A receptor deficient OLETF rat. Neuropeptides 36:171-181. 121. Woods, S.C. 2004. Gastrointestinal Satiety Signals I. An overview of gastrointestinal signals that influence food intake. American Journal of Physiology 286:G7-G13. 122. Bachur, N.R., Masek, K., Melmon, K.L., and Udenfriend, S. 1965. Fatty Acid Amides Of Ethanolamine In Mammalian Tissues. J Biol Chem 240:1019-1024. 123. Kuehl, F.A., Jacob, T.A., Ganley, O.H., Ormond, R.E., and Meisinger, M.A.P. 1957. The identificationof N-(2-hydroxyethyl)-palmitamide as a naturally occuring anti-inflammatory agent. J. Am. Chem. Soc. 79:5577-5578.

119

124. Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G., Gibson, D., Mandelbaum, A., Etinger, A., and Mechoulam, R. 1992. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258:1946-1949. 125. Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., and Bonner, T.I. 1990. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346:561-564. 126. Munro, S., Thomas, K.L., and Abu-Shaar, M. 1993. Molecular characterization of a peripheral receptor for cannabinoids. Nature 365:61-65. 127. Fu, J., Gaetani, S., Oveisi, F., Lo Verme, J., Serrano, A., Rodriguez De Fonseca, F., Rosengarth, A., Luecke, H., Di Giacomo, B., Tarzia, G., et al. 2003. Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-alpha. Nature 425:90-93. 128. Okamoto, Y., Morishita, J., Tsuboi, K., Tonai, T., and Ueda, N. 2004. Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem 279:5298-5305. 129. Schmid, P.C., Zuzarte-Augustin, M.L., and Schmid, H.H. 1985. Properties of rat liver N-acylethanolamine amidohydrolase. J Biol Chem 260:14145-14149. 130. Cravatt, B.F., Giang, D.K., Mayfield, S.P., Boger, D.L., Lerner, R.A., and Gilula, N.B. 1996. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature 384:83-87. 131. Ueda, N., Yamanaka, K., and Yamamoto, S. 2001. Purification and characterization of an acid amidase selective for N-palmitoylethanolamine, a putative endogenous anti-inflammatory substance. J Biol Chem 276:35552-35557. 132. Cravatt, B.F., and Lichtman, A.H. 2002. The enzymatic inactivation of the fatty acid amide class of signaling lipids. Chem Phys Lipids 121:135-148. 133. Lo Verme, J., Gaetani, S., Fu, J., Oveisi, F., Burton, K., and Piomelli, D. 2005. Regulation of food intake by oleoylethanolamide. Cell Mol Life Sci 62:708-716. 134. Rodriguez de Fonseca, F., Navarro, M., Gomez, R., Escuredo, L., Nava, F., Fu, J., Murillo-Rodriguez, E., Giuffrida, A., LoVerme, J., Gaetani, S., et al. 2001. An anorexic lipid mediator regulated by feeding. Nature 414:209-212. 135. Guzman, M., Lo Verme, J., Fu, J., Oveisi, F., Blazquez, C., and Piomelli, D. 2004. Oleoylethanolamide Stimulates Lipolysis by Activating the Nuclear Receptor Peroxisome Proliferator-activated Receptor {alpha} (PPAR-{alpha}). Journal of Biological Chemistry 279:27849-27854. 136. Piomelli, D., Beltramo, M., Giuffrida, A., and Stella, N. 1998. Endogenous cannabinoid signaling. Neurobiol Dis 5:462-473. 137. Ahern, G.P. 2003. Activation of TRPV1 by the satiety factor oleoylethanolamide. J Biol Chem 278:30429-30434. 138. Numazaki, M., and Tominaga, M. 2004. Nociception and TRP Channels. Curr Drug Targets CNS Neurol Disord 3:479-485. 139. Di Marzo, V., Lastres-Becker, I., Bisogno, T., De Petrocellis, L., Milone, A., Davis, J.B., and Fernandez-Ruiz, J.J. 2001. Hypolocomotor effects in rats of capsaicin and two long chain capsaicin homologues. Eur J Pharmacol 420:123- 131.

120

140. Overton, H.A., Babbs, A.J., Doel, S.M., Fyfe, M.C., Gardner, L.S., Griffin, G., Jackson, H.C., Procter, M.J., Rasamison, C.M., Tang-Christensen, M., et al. 2006. Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab 3:167-175. 141. Fredriksson, R., Hoglund, P.J., Gloriam, D.E., Lagerstrom, M.C., and Schioth, H.B. 2003. Seven evolutionarily conserved human rhodopsin G protein-coupled receptors lacking close relatives. FEBS Lett 554:381-388. 142. Schoonjans, K., Staels, B., and Auwerx, J. 1996. Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression. J Lipid Res 37:907-925. 143. Murillo-Rodriguez, E., Desarnaud, F., and Prospero-Garcia, O. 2006. Diurnal variation of arachidonoylethanolamine, palmitoylethanolamide and oleoylethanolamide in the brain of the rat. Life Sci. 144. Moreno, S., Farioli-Vecchioli, S., and Ceru, M.P. 2004. Immunolocalization of peroxisome proliferator-activated receptors and retinoid X receptors in the adult rat CNS. Neuroscience 123:131-145. 145. Mezey, E., Toth, Z.E., Cortright, D.N., Arzubi, M.K., Krause, J.E., Elde, R., Guo, A., Blumberg, P.M., and Szallasi, A. 2000. Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci U S A 97:3655-3660. 146. McKay, B.M., and Ritter, R.C. 2004. Oleoylethanolamide induces rapid reduction of short-term food intake in intact and vagotomized rats. Program No. 427.12, Society for Neuroscience, San Diego, CA. 147. Gaetani, S., Oveisi, F., and Piomelli, D. 2003. Modulation of meal pattern in the rat by the anorexic lipid mediator oleoylethanolamide. Neuropsychopharmacology 28:1311-1316. 148. Cota, D., Marsicano, G., Tschop, M., Grubler, Y., Flachskamm, C., Schubert, M., Auer, D., Yassouridis, A., Thone-Reineke, C., Ortmann, S., et al. 2003. The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J Clin Invest 112:423-431. 149. Di Marzo, V., Goparaju, S.K., Wang, L., Liu, J., Batkai, S., Jarai, Z., Fezza, F., Miura, G.I., Palmiter, R.D., Sugiura, T., et al. 2001. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 410:822-825. 150. Di Marzo, V., Fontana, A., Cadas, H., Schinelli, S., Cimino, G., Schwartz, J.C., and Piomelli, D. 1994. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 372:686-691. 151. Walter, L., Franklin, A., Witting, A., Moller, T., and Stella, N. 2002. Astrocytes in culture produce anandamide and other acylethanolamides. J Biol Chem 277:20869-20876. 152. Cadas, H., di Tomaso, E., and Piomelli, D. 1997. Occurrence and biosynthesis of endogenous cannabinoid precursor, N-arachidonoyl phosphatidylethanolamine, in rat brain. J Neurosci 17:1226-1242. 153. Moran, T.H., and Kinzig, K.P. 2004. Gastrointestinal satiety signals II. Cholecystokinin. American Journal of Physiology 286:G183-G188. 154. Riley, A.L., and Tuck, D.L. 1985. Conditioned taste aversions: a behavioral index of toxicity. Annals of the New York Academy of Sciences 443:272-292.

121

155. Bernstein, I.L., and Goehler, L.E. 1983. Chronic lithium chloride infusions: conditioned suppression of food intake and preference. Behav Neurosci 97:290- 298. 156. Curtis, K.S., Sved, A.F., Verbalis, J.G., and Stricker, E.M. 1994. Lithium chloride-induced anorexia, but not conditioned taste aversions, in rats with area postrema lesions. Brain Res 663:30-37. 157. Breslin, P.A., Kaplan, J.M., Spector, A.C., Zambito, C.M., and Grill, H.J. 1993. Lick rate analysis of sodium taste-state combinations. Am J Physiol 264:R312- 318. 158. Tschöp, M., Weyer, C., Tataranni, P.A., Devanarayan, V., Ravussin, E., and Heiman, M.L. 2001. Circulating ghrelin levels are decreased in human obesity. Diabetes 50:707-709. 159. Strader AD, V.T., Jandacek RJ, Woods SC, D'Alessio DA, Seeley RJ. 2005. Weight loss through ileal transposition is accompanied by increased ileal hormone secretion and synthesis in the rat. Am J Physiol Endocrinol Metab 288:E447- E453. 160. Holst, J.J., Bersani, M., Johnsen, A.H., Kofod, H., Hartmann, B., and Orskov, C. 1994. Proglucagon processing in porcine and human pancreas. J Biol Chem 269:18827-18833. 161. Liu, M., Doi, T., Shen, L., Woods, S.C., Seeley, R.J., Zheng, S., Jackman, A., and Tso, P. 2001. Intestinal satiety protein apolipoprotein AIV is synthesized and regulated in rat hypothalamus. Am J Physiol 280:R1382-R1387. 162. Hayes, M.R., Savastano, D.M., and Covasa, M. 2004. Cholecystokinin-induced satiety is mediated through interdependent cooperation of CCK-A and 5-HT3 receptors. Physiol Behav 82:663-669. 163. Moran, T.H., Ameglio, P.J., Schwartz, G.J., and McHugh, P.R. 1992. Blockade of type A, not type B, CCK receptors attenuates satiety actions of exogenous and endogenous CCK. Am J Physiol 262:R46-50. 164. Barros, D.M., Carlis, V., Maidana, M., Silva, E.S., Baisch, A.L., Ramirez, M.R., and Izquierdo, I. 2004. Interactions between anandamide-induced anterograde amnesia and post-training memory modulatory systems. Brain Res 1016:66-71. 165. Romero, J., Lastres-Becker, I., de Miguel, R., Berrendero, F., Ramos, J.A., and Fernandez-Ruiz, J. 2002. The endogenous cannabinoid system and the basal ganglia. biochemical, pharmacological, and therapeutic aspects. Pharmacol Ther 95:137-152. 166. Maccarrone, M., Cartoni, A., Parolaro, D., Margonelli, A., Massi, P., Bari, M., Battista, N., and Finazzi-Agro, A. 2002. Cannabimimetic activity, binding, and degradation of stearoylethanolamide within the mouse central nervous system. Mol Cell Neurosci 21:126-140. 167. Willson, T.M., Brown, P.J., Sternbach, D.D., and Henke, B.R. 2000. The PPARs: from orphan receptors to drug discovery. J Med Chem 43:527-550. 168. Cabrero, A., Llaverias, G., Roglans, N., Alegret, M., Sanchez, R., Adzet, T., Laguna, J.C., and Vazquez, M. 1999. Uncoupling protein-3 mRNA levels are increased in white adipose tissue and skeletal muscle of bezafibrate-treated rats. Biochem Biophys Res Commun 260:547-556.

122

169. Cox, J.E., Perdue, G.S., and Tyler, W.J. 1995. Suppression of sucrose intake by continuous near-celiac and intravenous cholecystokinin infusions in rats. American Journal of Physiology 268:R150-R155. 170. Terrazzino, S., Berto, F., Dalle Carbonare, M., Fabris, M., Guiotto, A., Bernardini, D., and Leon, A. 2004. Stearoylethanolamide exerts anorexic effects in mice via down-regulation of liver stearoyl-coenzyme A desaturase-1 mRNA expression. Faseb J 18:1580-1582. 171. Miller, C.W., and Ntambi, J.M. 1996. Peroxisome proliferators induce mouse liver stearoyl-CoA desaturase 1 gene expression. Proc Natl Acad Sci U S A 93:9443-9448. 172. Jump, D.B., Ren, B., Clarke, S., and Thelen, A. 1995. Effects of fatty acids on hepatic gene expression. Prostaglandins Leukot Essent Fatty Acids 52:107-111. 173. Xu, J., Nakamura, M.T., Cho, H.P., and Clarke, S.D. 1999. Sterol regulatory element binding protein-1 expression is suppressed by dietary polyunsaturated fatty acids. A mechanism for the coordinate suppression of lipogenic genes by polyunsaturated fats. J Biol Chem 274:23577-23583. 174. Wang, X., Miyares, R.L., and Ahern, G.P. 2005. Oleoylethanolamide excites vagal sensory neurones, induces visceral pain and reduces short-term food intake in mice via capsaicin receptor TRPV1. J Physiol 564:541-547. 175. Minami, S., Kamegai, J., Sugihara, H., Suzuki, N., Higuchi, H., and Wakabayashi, I. 1995. Central glucoprivation evoked by administration of 2- deoxy-D-glucose induces expression of the c-fos gene in a subpopulation of neuropeptide Y neurons in the rat hypothalamus. Brain Res Mol Brain Res 33:305-310. 176. Hu, Z., Cha, S.H., Chohnan, S., and Lane, M.D. 2003. Hypothalamic malonyl- CoA as a mediator of feeding behavior. Proc Natl Acad Sci U S A 100:12624- 12629. 177. Hardie, D.G., Carling, D., and Carlson, M. 1998. The AMP-activated/SNF1 protein kinase subfamily: metabolic sensors of the eukaryotic cell? Annu Rev Biochem 67:821-855. 178. Kozma, S.C., and Thomas, G. 2002. Regulation of cell size in growth, development and human disease: PI3K, PKB and S6K. Bioessays 24:65-71. 179. Chavez, M., Kaiyala, K., Madden, L.J., Schwartz, M.W., and Woods, S.C. 1995. Intraventricular insulin and the level of maintained body weight in rats. Behavioral Neuroscience 109:528-531. 180. Bjornsti, M.A., and Houghton, P.J. 2004. The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4:335-348. 181. Boylan, J.M., Anand, P., and Gruppuso, P.A. 2001. Ribosomal protein S6 phosphorylation and function during late gestation liver development in the rat. J Biol Chem 276:44457-44463. 182. Ballou, L.M., Jeno, P., and Thomas, G. 1988. Protein phosphatase 2A inactivates the mitogen-stimulated S6 kinase from Swiss mouse 3T3 cells. J Biol Chem 263:1188-1194. 183. Olivier, A.R., Ballou, L.M., and Thomas, G. 1988. Differential regulation of S6 phosphorylation by insulin and epidermal growth factor in Swiss mouse 3T3

123

cells: insulin activation of type 1 phosphatase. Proc Natl Acad Sci U S A 85:4720- 4724. 184. Yudkoff, M., Daikhin, Y., Nissim, I., Lazarow, A., and Nissim, I. 2001. Ketogenic diet, amino acid metabolism, and seizure control. J Neurosci Res 66:931-940. 185. Abdelmegeed, M.A., Carruthers, N.J., Woodcroft, K.J., Kim, S.K., and Novak, R.F. 2005. Acetoacetate induces CYP2E1 protein and suppresses CYP2E1 mRNA in primary cultured rat hepatocytes. J Pharmacol Exp Ther 315:203-213. 186. Melo, T.M., Nehlig, A., and Sonnewald, U. 2006. Neuronal-glial interactions in rats fed a ketogenic diet. Neurochem Int 48:498-507. 187. Kim, M.S., Park, J.Y., Namkoong, C., Jang, P.G., Ryu, J.W., Song, H.S., Yun, J.Y., Namgoong, I.S., Ha, J., Park, I.S., et al. 2004. Anti-obesity effects of alpha- lipoic acid mediated by suppression of hypothalamic AMP-activated protein kinase. Nat Med 10:727-733. 188. Patel, J., Wang, X., and Proud, C.G. 2001. Glucose exerts a permissive effect on the regulation of the initiation factor 4E binding protein 4E-BP1. Biochem J 358:497-503. 189. Cota, D., Marsicano, G., Lutz, B., Vicennati, V., Stalla, G.K., Pasquali, R., and Pagotto, U. 2003. Endogenous cannabinoid system as a modulator of food intake. International Journal of Obesity and Related Metabolic Disorders 27:289-301. 190. Beglinger, C., and Degen, L. 2004. Fat in the intestine as a regulator of appetite-- role of CCK. Physiol Behav 83:617-621. 191. Gibbs, J., Young, R.C., and Smith, G.P. 1973. Cholecystokinin decreases food intake in rats. Journal of Comparative and Physiological Psychology 84:488-495. 192. Gomez, R., Navarro, M., Ferrer, B., Trigo, J.M., Bilbao, A., Del Arco, I., Cippitelli, A., Nava, F., Piomelli, D., and Rodriguez de Fonseca, F. 2002. A peripheral mechanism for CB1 cannabinoid receptor-dependent modulation of feeding. J Neurosci 22:9612-9617. 193. Cummings, D.E., and Foster, K.E. 2003. Ghrelin-leptin tango in body-weight regulation. Gastroenterology 124:1532-1535. 194. Osei-Hyiaman, D., Depetrillo, M., Pacher, P., Liu, J., Radaeva, S., Batkai, S., Harvey-White, J., Mackie, K., Offertaler, L., Wang, L., et al. 2005. Endocannabinoid activation at hepatic CB(1) receptors stimulates fatty acid synthesis and contributes to diet-induced obesity. J Clin Invest 115:1298-1305. 195. Hannah, V.C., Ou, J., Luong, A., Goldstein, J.L., and Brown, M.S. 2001. Unsaturated fatty acids down-regulate srebp isoforms 1a and 1c by two mechanisms in HEK-293 cells. J Biol Chem 276:4365-4372. 196. Pralong, F.P., Gonzales, C., Voirol, M.J., Palmiter, R.D., Brunner, H.R., Gaillard, R.C., Seydoux, J., and Pedrazzini, T. 2002. The neuropeptide Y Y1 receptor regulates leptin-mediated control of energy homeostasis and reproductive functions. Faseb J 16:712-714. 197. Clegg, D.J., Benoit, S.C., Air, E.L., Jackman, A., Tso, P., D'Alessio, D., Woods, S.C., and Seeley, R.J. 2003. Increased dietary fat attenuates the anorexic effects of intracerebroventricular injections of MTII. Endocrinology 144:2941-2946. 198. Leibel, R.L., Rosenbaum, M., and Hirsch, J. 1995. Changes in energy expenditure resulting from altered body weight. N Engl J Med 332:621-628.

124

199. Erickson, J.C., Clegg, K.E., and Palmiter, R.D. 1996. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381:415-421. 200. Najjar, S.M., Yang, Y., Fernstrom, M.A., Lee, S.J., Deangelis, A.M., Rjaily, G.A., Al-Share, Q.Y., Dai, T., Miller, T.A., Ratnam, S., et al. 2005. Insulin acutely decreases hepatic fatty acid synthase activity. Cell Metab 2:43-53.

125

Appendix 1

Hypothalamic neuron

NPY - NPY - pS6- pS6 + Food intake pS6K1+ pS6K1+ pmTOR+ pmTOR

+ +

Leptin Glucose/leucine

Stored fuels (adipose tissue) current fuels (nutrients)

Hypothetical model 1: mTOR may integrate signals from both stored and currently available fuels. MTOR might be a convergence point within metabolic sensing neurons, through which signals reflecting the levels of stored and currently available fuels affect the synthesis of proteins involved in triggering the appropriate changes in terms of caloric intake and expenditure, so that energy homeostasis is maintained.

126

Appendix 2

Hypothalamic neuron

C75 NPY - - pS6 FAS Food intake acetyl-CoA Malonyl-CoA LCFA + ACC pS6K1 CPT-1 + AMPK glucose pmTOR

- +

Leptin

Stored fuels (adipose tissue)

Hypothetical model 2: C75 and leptin may modulate mTOR through interaction with the FAS pathway. C75 and leptin might increase glucose oxidation through interaction with the FAS pathway. This would increase the phosphorylation and activation of mTOR signaling, which might in turn reduce NPY levels and, consequently, food intake.

127

128