91
The Effects of Saturated Fatty Acid Palmitate on Neuropeptide Gene Expression, Signal Transduction, and Insulin Signaling in an Immortalized Hypothalamic Neuronal Cell Model, mHypoA-NPY/GFP By Brian Wong A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Physiology University of Toronto © Copyright by Brian Wong 2015

The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

The Effects of Saturated Fatty Acid Palmitate on Neuropeptide Gene

Expression, Signal Transduction, and Insulin Signaling in an Immortalized

Hypothalamic Neuronal Cell Model, mHypoA-NPY/GFP

By

Brian Wong

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Department of Physiology

University of Toronto

© Copyright by Brian Wong 2015

Page 2: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

ii

The Effects of Saturated Fatty Acid Palmitate on Neuropeptide Gene

Expression, Signal Transduction, and Insulin Signaling in an Immortalized

Hypothalamic Neuronal Cell Model, mHypoA-NPY/GFP

Brian Wong

Master of Science

Department of Physiology

University of Toronto

2015

Abstract

Recent evidence suggests a role for hypothalamic insulin resistance in obesity

pathogenesis, and that obesity-associated hypothalamic inflammation underlies this

resistance. However, few studies have examined the direct effects of saturated fatty acids on

specific hypothalamic neurons. Therefore, an immortalized hypothalamic neuronal cell

model expressing NPY and AgRP was used to determine the effects of palmitate on

neuropeptide gene expression, signal transduction events and insulin signaling. In the

mHypoA-NPY/GFP neuronal cell model, palmitate was found to upregulate the expression

of NF-κB and IκBα within 4 hours of treatment, and upregulate expression of AgRP after 24

hours of treatment. Regulation of AgRP gene expression appeared to be palmitate

metabolism-dependent. Palmitate also induced p38 MAPK phosphorylation, and prolonged

palmitate pre-treatment decreased levels of phosphorylated Akt following insulin re-

challenge. This is the first evidence of palmitate-mediated changes in AgRP gene expression

and its signaling through p38 MAPK in a representative NPY/AgRP neuronal cell model.

Page 3: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

iii

Acknowledgements

First and foremost, I owe my deepest gratitude to Dr. Denise Belsham. I am grateful

to have had you as my mentor and supervisor. The last two years in the laboratory have been

an incredible learning experience. Your continual guidance, support, and encouragement

have enabled me to grow as a person, and have prepared me for the road ahead. Thank you

Denise.

I would also like to thank my committee members, Dr. Michael Wheeler, Dr. Amira

Klip, and Dr. Adria Giacca. Your guidance, mentorship, and valuable insight have been

crucial to the completion of this degree.

I would like to thank my fellow lab mates, who made the laboratory an enjoyable

place to work in. Whether it was generating discussion at lab meetings or simply lending a

helping hand with a new experiment, you have all contributed to this experience.

Finally, I would like to thank my family for their unwavering love and

encouragement. The sacrifices you have made and the many opportunities you have provided

me with do not go unnoticed. I would not be where I am today without you.

Page 4: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

iv

Table of Contents

Acknowledgements .......................................................................................................... iii

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

List of Tables and Figures ............................................................................................. viii

List of Abbreviations ....................................................................................................... ix

Chapter 1 Introduction

1.1 Preface................................................................................................................2

1.2 The Central Melanocortin System and Energy Homeostasis

1.2.1 Adiposity Negative Feedback ..........................................................2

1.2.2 Insulin as an Adiposity Signal .........................................................3

1.2.3 The Hypothalamus ...........................................................................4

1.2.4 The Role of Neuropeptides in the Brain ..........................................6

1.2.5 The Melanocortin System ................................................................6

1.2.6 Neuropeptide Y and Agouti-Related Peptide ..................................7

1.3 Glucose Sensing in Hypothalamic Neurons

1.3.1 Glucose Entry into the Brain and the Discovery

of Glucose Sensing Neurons ............................................................8

1.3.2 Glucose-Excited Neurons ..............................................................10

1.3.3 Glucose-Inhibited Neurons ...........................................................11

1.4 Fatty Acid Sensing in Hypothalamic Neurons

1.4.1 Fatty Acids and Metabolic Physiology ..........................................12

1.4.2 Blood-Brain Barrier Permeability, Fatty Acid Uptake and

Metabolic Fates ..............................................................................13

Page 5: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

v

1.4.3 Fatty Acids as Signaling Molecules in the Hypothalamus ............13

1.5 Obesity and Hypothalamic Inflammation

1.5.1 High-Fat Feeding is Associated with Inflammation ......................15

1.5.2 Evidence that Hypothalamic Inflammation Contributes to

HFD-Induced Obesity ....................................................................16

1.5.3 Palmitate as a Mediator of Hypothalamic Inflammation ...............17

1.6 Obesity and Hypothalamic Insulin Resistance

1.6.1 Obesity, Diabetes and Insulin Resistance ......................................19

1.6.2 Hyperinsulinemia and the Development of Insulin Resistance .....20

1.6.3 High Fat Feeding and Neuronal Insulin Resistance .......................20

1.6.4 FFA Metabolism and Insulin Resistance .......................................21

1.7 Cell Model

1.7.1 The Need for Cell Lines .................................................................24

1.7.2 Adult Hypothalamic Cell Lines (mHypoA-xx) .............................25

1.7.3 mHypoA-NPY/GFP Cell Line .......................................................26

1.8 Hypothesis and Aims .......................................................................................26

Chapter 2 Materials and Methods

2.1 Cell Culture and Reagents ...............................................................................32

2.2 Palmitate Preparation .......................................................................................32

2.3 TNF-α Preparation ...........................................................................................32

2.4 Insulin Preparation ...........................................................................................33

2.5 Quantitative RT-PCR .......................................................................................33

2.6 Western Blot Analysis .....................................................................................34

Page 6: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

vi

2.7 Statistical Analysis ...........................................................................................35

Chapter 3 Results

3.1 Palmitate elicits an inflammatory response and upregulates

Agrp gene expression in mHypoA-NPY/GFP neurons ...................................37

3.2 TNF-α, a pro-inflammatory surrogate of palmitate, also

upregulates AgRP gene expression in mHypoA-NPY/GFP neurons ...............38

3.3 Palmitate-mediated regulation of AgRP gene expression

is metabolism-dependent .................................................................................40

3.4 Palmitate triggers the phosphorylation of p38 MAPK in

mHypoA-NPY/GFP neurons ...........................................................................43

3.5 Palmitate pre-treatment dampens the mHypoA-NPY/GFP

neurons’ response to insulin.............................................................................43

Chapter 4 Discussion

4.1 General Discussion ..........................................................................................48

4.2 Plasma Non-Esterified Fatty Acids and the Determination

of Palmitic Acid Concentrations in the Brain ..................................................49

4.3 Transcriptional Effects of Palmitate on mHypoA-NPY/GFP Neurons ...........52

4.4 Transcriptional Effects of TNF-α on mHypoA-NPY/GFP Neurons ...............55

4.5 Palmitate Metabolism and Signaling Dynamics in

mHypoA-NPY/GFP neurons ...........................................................................56

4.6 Palmitate Impairs Insulin Signaling in mHypoA-NPY/GFP Neurons ............58

4.7 Limitations .......................................................................................................60

4.8 Future Directions .............................................................................................63

Page 7: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

vii

4.9 Conclusion .......................................................................................................64

References .........................................................................................................................66

Page 8: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

viii

List of Tables and Figures

Table 1.1 Characterization of the mHypoA-NPY/GFP cell line .......................................30

Fig. 1.1 Schematic illustrating the PI3K-Akt pathway ........................................................5

Fig. 1.2 NPY/AgRP and POMC neurons are directly regulated by insulin .........................9

Fig. 1.3 Metabolic fates of palmitate upon entering the cell .............................................14

Fig. 1.4 Activation of the IKKβ/NF-κB pathway leads to inflammation

and impaired insulin signaling .............................................................................18

Fig. 1.5 Mechanisms of palmitate-mediated inhibition of insulin signaling .....................22

Fig. 1.6 Generation of the mHypoA-NPY/GFP cell line ...................................................27

Fig. 3.1 Saturated fatty acid palmitate upregulates pro-inflammatory and Agrp

gene expression ......................................................................................................39

Fig. 3.2 TNF-α upregulates pro-inflammatory and Agrp gene expression ........................41

Fig. 3.3 Methyl palmitate does not regulate Agrp gene expression ...................................42

Fig. 3.4 Palmitate induces phosphorylation of p38 MAPK ...............................................44

Fig.3.5 Prolonged palmitate or insulin exposure dampens the

insulin-mediated increase in phospho-Akt .............................................................46

Page 9: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

ix

Abbreviations

AgRP agouti-related peptide

ARC arcuate nucleus

β-oxidation beta oxidation

BBB blood-brain barrier

cDNA complementary deoxyribonucleic acid

CNS central nervous system

CNTF ciliary neurotrophic factor

CPT-1 carnitine palmitoyltransferase-1

CREB cAMP response element binding protein

DAG diacylglycerol

DIO diet-induced obesity

DMEM Dulbecco’s modified eagle medium

DMN dorsomedial nucleus

DNA deoxyribonucleic acid

eIF2 eukaryotic initiation factor 2

ELK-1 ETS domain-containing protein-1

ER endoplasmic reticulum

ERK extracellular-related kinase

FABP fatty acid binding protein

FATP fatty acid transport protein

FBS fetal bovine serum

FFA free fatty acid

Page 10: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

x

FOXO1 forkhead box protein 01

GLUT4 glucose transporter type 4

GPAT glycerol-3 phosphate acyltransferase

HFD high-fat diet

ICC immunocytochemistry

ICV intracerebroventricular

IκBα inhibitor of nuclear factor kappa B alpha

IKK-β inhibitor of IkappaB kinase beta

IL-1β interleukin-1 beta

IL-6 interleukin-6

IR insulin receptor

IRS insulin receptor substrate

JNK c-Jun N-terminal kinase

LHA lateral hypothalamic area

LPL lipoprotein lipase

MAPK mitogen-activated protein kinase

MC3/4R melanocortin 3/4 receptor

MPO medial preoptic area

mRNA messenger ribonucleic acid

miRNA microRNA

α-MSH alpha-melanocyte stimulating hormone

NEFA non-esterified fatty acid

NF-κB nuclear factor kappa B

Page 11: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

xi

NPY neuropeptide Y

NTS nucleus of the solitary tract

PBS phosphate buffer saline

PCR polymerase chain reaction

PFA perifornical area

PI3K phosphatidylinositol 3-kinase

PKB protein kinase B

PKC protein kinase C

POMC proopiomelanocortin

PTP1B protein tyrosine phosphatase 1 B

PVN paraventricular nucleus

qRT-PCR quantitative reverse transcriptase polymerase chain reaction

RNA ribonucleic acid

siRNA small interfering RNA

SPT serine palmitoyltransferase

STAT signal transducer and activator of transcription

SV40 simian virus 40

T-Ag T-antigen

TG triglyceride

TAG triacylglycerol

TLR toll-like receptor

TNF-α tumor necrosis factor-alpha

T2DM type 2 diabetes mellitus

Page 12: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

1

Chapter 1

Introduction

Page 13: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

2

Introduction

1.1 Preface

Most overweight or obese individuals develop hyperlipidemia, low-grade

inflammation, and insulin resistance often leading to type 2 diabetes mellitus (T2DM). While

obesity and T2DM may both originate from a primary hypothalamic disease, little is known

about how specific neurons within the hypothalamus sense and respond to nutrient

(particularly fat) excess. The Belsham laboratory has generated several immortalized,

hypothalamic neuronal cell lines from primary fetal and adult hypothalamic neuronal cell

cultures, which have already provided insight into the direct control of neuropeptide

synthesis by nutrients at a mechanistic level not practical in the whole brain. The purpose of

this thesis was to evaluate the effects of palmitate (the most abundant non-esterified saturated

fatty acid) on neuropeptide gene expression, signal transduction events and insulin signaling

in an immortalized, hypothalamic neuronal cell model representative of the NPY/AgRP

neuron. Using the mHypoA-NPY/GFP cell line, I provide evidence of palmitate-mediated

changes in AgRP gene expression and the dependency of such changes on palmitate

metabolism. In addition, these studies begin to elucidate palmitate-mediated signal

transduction events and provide further evidence of palmitate’s ability to impair insulin

signaling in distinct hypothalamic neurons. Taken together, these studies have direct

relevance to the molecular mechanisms involved in the overall development of complex

metabolic disorders, such as obesity.

1.2 The Central Melanocortin System and Energy Homeostasis

1.2.1 Adiposity Negative Feedback

Page 14: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

3

Despite daily variations in energy intake, the body fuel stored in adipose tissue

remains relatively constant over time (1). This observation suggests that short-term

differences in energy balance (the difference between energy consumed and energy

expended) may be offset in the long term by a mechanism that maintains overall energy

homeostasis. Indeed, changes in body fat content through dieting (2), behavior modification

(3) or experimental over-feeding (4) have been shown to induce compensatory responses that

restore adiposity to homeostatic levels.

To explain this phenomenon, Kennedy proposed that inhibitory signals were

generated in proportion to body fat stores and acted in the brain to reduce food intake (5).

Weight loss reduced the plasma levels of these inhibitory signals, causing food intake to

increase until body fat stores returned to normal levels (6).

1.2.2 Insulin as an Adiposity Signal

Insulin, a peptide hormone produced by the pancreatic β-cells, was the first hormonal

signal implicated in the central nervous system control of energy homeostasis (7). It provides

information regarding the amount of body fat stored and causes a long-term catabolic

response, decreasing food intake and increasing energy expenditure (8). Insulin is secreted

acutely in response to increases in blood glucose (i.e. after consumption of a meal) and its

levels are directly correlated to the extent of body adiposity (9). As in peripheral tissues,

insulin binds to its cognate receptor in the CNS. The receptor belongs to the family of

tyrosine kinase receptors, and binding of insulin to its receptor triggers an intracellular

signaling cascade (10).

Page 15: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

4

Binding of insulin leads to rapid autophosphorylation of its receptor, followed by

tyrosine phosphorylation and recruitment of insulin receptor substrate (IRS) proteins. This

leads to activation of downstream pathways such as the phosphatidylinositol 3 kinase (PI3K)

and the mitogen-activated protein kinase (MAPK) cascades (11). Activation of PI3K results

in activation of protein kinase B/Akt and subsequent phosphorylation of the transcription

factor FOXO, which is a critical downstream regulator of energy homeostasis in the CNS

(Figure 1.1) (12).

The hypothalamus contains the highest concentration of insulin receptors (IR) in the

central nervous system. However, IRs are also expressed in the olfactory bulb, cerebral

cortex, cerebellum and hippocampus (13, 14). Neurons within the hypothalamus are capable

of sensing circulating insulin because of their location near the third ventricle, where insulin

can enter via a saturable transporter across the blood-brain barrier (15).

1.2.3 The Hypothalamus

The hypothalamus is a key brain region controlling energy homeostasis. Histological

techniques reveal nuclei as clusters of neurons within the hypothalamus that have distinct

neuronal phenotypes. These neurons express a specific complement of neuropeptides,

neurotransmitters and receptors. Classical lesion studies have shown that some of these

hypothalamic nuclei act as discrete “feeding” and “satiety” centres (16). Lesions of the

ventromedial, paraventricular or dorsal medial hypothalamus lead to hyperphagia, while

lesions of the lateral hypothalamus lead to hypophagia (17).

Besides regulating energy homeostasis, the hypothalamus is also the control centre

for many other endocrine processes. Physiological processes that are under hypothalamic

control include: stress, growth, temperature regulation, water balance, sexual behavior and

Page 16: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

5

Insulin

Cell Membrane

IRS-1

PI3K

PIP3

PDK1

AKT FoxO1

FoxO1

Nucleus

Insulin Receptor

P

P

P

POMC AgRP

Fig. 1.1 Schematic illustrating the PI3K-Akt pathway.

Insulin binds to the insulin receptor, which is a receptor tyrosine kinase that autophosphorylates itself.

This allows IRS proteins to dock. IRS proteins are then activated, and can recruit PI3K which

phosphorylates PIP2 to PIP3. PIP3 acts as a docking site for PDK1 and AKT, allowing for the

phosphorylation of Akt by PDK1. Phosphorylation of Akt leads to its nuclear translocation where it

phosphorylates FoxO1 transcription factor. Phosphorylated FoxO1 can no longer repress POMC

expression and stimulate AgRP gene expression, which results in decreased feeding.

Page 17: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

6

reproduction, and circadian rhythms. Situated below the thalamus, posterior to the optic

chiasm and surrounding the third ventricle, the hypothalamus has access to circulating factors

that cross the blood-brain barrier (BBB) via diffusion or saturable transport mechanisms.

1.2.4 The Role of Neuropeptides in the Brain

Over 70 genes in the mammalian genome encode for neuropeptides (16).

Neuropeptides are peptide molecules synthesized by neurons, are released in a regulated

manner and act on receptors present on other neurons. Compared to some classical

neurotransmitters, such as epinephrine, neuropeptides are large with nanomolar affinities for

their receptors. Neuropeptides can also diffuse over larger distances within the CNS than

some classical neurotransmitters. Indeed, neurotransmitters like glutamate have been shown

to have extrasynaptic effects. However, they are more likely to travel only as far as their

nearest neighbouring synapse. Glutamate, in particular, has been found to travel distances of

less than half a micrometer. In contrast, oxytocin released from neurons in the supraoptic

nucleus of the hypothalamus results in biologically relevant concentrations throughout the

anterior hypothalamus. In having high receptor binding affinity and the ability to affect

distant populations of neurons, neuropeptide release can mediate changes in neuronal activity

across multiple brain regions (17).

A growing number of neuropeptides and neurotransmitters have been implicated in

the regulation of feeding behavior in vivo. These neuropeptides are expressed in distinct

neuronal populations located in specific regions of the hypothalamus, including the arcuate,

paraventricular, and ventromedial nuclei (18).

Page 18: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

7

1.2.5 The Melanocortin System

The melanocortin system is central to the neuronal control of energy homeostasis.

Here, the arcuate nucleus (ARC) of the hypothalamus is particularly important (19). Neurons

within the ARC are strategically located close to fenestrated capillaries at the base of the

hypothalamus such that they have access to circulating humoral signals (20). These neurons

are controlled by neurotransmitters that are released from neighbouring axons, express

receptors for metabolic hormones (20) and respond rapidly to nutritional cues (21).

At present, the mammalian central melanocortin system is defined as a collection of

CNS circuits that include: ARC neurons expressing hypothalamic neuropeptide Y (NPY) and

agouti-related peptide (AgRP) or proopiomelanocortin (POMC), brainstem POMC neurons

within the nucleus of the solitary tract (NTS) and downstream targets of these POMC and

AgRP neurons which express melanocortin 3 (MC3R) and melanocortin 4 (MC4R) receptors

(22).

1.2.6 Neuropeptide Y and Agouti-Related Peptide

Neuropeptides involved in food intake can be grouped into one of two categories:

orexigenic (appetite-stimulating) or anorexigenic (appetite-suppressing). The main

orexigenic neuron in the ARC is the NPY/AgRP neuron. Neuropeptide Y is a 36 amino acid

peptide that is expressed throughout the central nervous system (23), and has notably high

expression in the ARC (24). Agouti-related peptide is a 132 amino acid peptide that, unlike

NPY, is only found in the ARC. ARC NPY/AgRP neurons project to nearby hypothalamic

areas such as the paraventricular nucleus (PVN), dorsomedial nucleus (DMN), perifornical

Page 19: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

8

area (PFA), lateral hypothalamic area (LHA) and the medial preoptic area (MPO), which are

integrative centers for the regulation of both feeding and energy expenditure (25).

NPY acts at multiple sites to increase food intake. Locally, NPY released from the

ARC acts to inhibit neighbouring POMC neurons by activation of Y1 and Y2 receptors (26).

NPY also acts on neurons in the PVN to stimulate food intake, and this effect appears to be

mediated by both Y1 and Y5 receptors (27, 28). However, unlike NPY, AgRP acts to increase

food intake by acting as an endogenous antagonist to the melanocortin 3 and 4 receptors (31).

This prevents the constitutive activity of these receptors (32), resulting in an inhibition of the

anorexigenic melanocortin pathway and an increase in food intake (Figure 1.2).

Insulin, among other hormones, regulates feeding and energy balance by modulating

the expression of these hypothalamic neuropeptides. Insulin may have anorexigenic effects

by increasing Pomc and decreasing Agrp gene expression (33), and this effect is mediated by

the phosphorylation of forkhead transcription factor 1 (FOXO1). FOXO1 is a transcription

factor that represses POMC gene expression and stimulates Agrp gene expression. Thus,

insulin-mediated phosphorylation of FOXO1 leads to its export from the nucleus which

relieves the repression on the POMC promoter (34). Concomitantly, FOXO1-induced

expression of Agrp in NPY/AgRP neurons is inhibited (35).

The importance of NPY and AgRP in the regulation of food intake and energy

homeostasis has been well documented. Central administration of either NPY (36) or AgRP

(29) increases food intake and body weight, and chronic administration results in obesity. A

single dose of AgRP results in an increase in food intake that is sustained for 7 days,

indicating its potency as an orexigenic neuropeptide (37). Inhibiting AgRP with arcuate-

Page 20: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

9

Figure 1.2 NPY/AgRP and POMC neurons are directly regulated by insulin.

A representative diagram of the NPY/AgRP and POMC neurons, and how insulin regulates these neurons.

Insulin exerts its anorexigenic effects by increasing Pomc and decreasing AgRP gene expression. The

overall effect is a reduction in food intake and an increase in energy expenditure.

AgRP = agouti-related peptide

MC3R = melanocortin 3 receptor

MC4R = melanocortin 4 receptor

α-MSH = alpha-melanocyte stimulating

hormone

NPY = neuropeptide Y

Y1 Receptor = neuropeptide Y Y1 receptor

Page 21: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

10

specific siRNA leads to decreases in both food intake and body weight (38). Furthermore,

ablation of these neurons in adult mice leads to extreme starvation (39, 40).

1.3 Glucose Sensing in Hypothalamic Neurons

1.3.1 Glucose Entry into the Brain and the Discovery of Glucose Sensing Neurons

Glucose is the primary energy substrate of the brain, and glucose metabolism accounts for

the majority of brain oxygen consumption. Stereospecific, but insulin-independent, GLUT-1

glucose transporters are highly expressed in brain capillary endothelial cells of the blood

brain barrier. GLUT-1 mediates the facilitated diffusion of glucose through the blood-brain

barrier, and can transport two to three times more glucose than is actually metabolized in the

brain (139). The stereospecificity of the GLUT-1 transporter allows D-glucose, but not L-

glucose, to pass into the brain.

Brain glucose varies depending on blood glucose, and declines to approximately 0.7

mM after an overnight fast. During peripheral hypoglycemia, hypothalamic glucose

concentrations have been shown to fall to as low as 0.3 mM (140). These and other studies

have indicated that hypothalamic glucose levels may range anywhere from 0.2 to 4.5 mM as

blood glucose levels vary from pathological hypoglycemia to hyperglycemia.

In 1964, two independent groups suggested the existence of glucose sensing neurons

(141, 142). In these studies, reciprocal changes in activity were measured in the ventromedial

hypothalamus (VMH) and lateral hypothalamus (regions referred to as the “satiety” and

“feeding” centers of the brain, respectively) following intravenous glucose or insulin

injections. In the VMH, glucose increased neuronal activity. In the lateral hypothalamus,

however, the opposite occurred. Later, Oomura et al. demonstrated that hypothalamic

Page 22: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

11

neurons were directly regulated by glucose in vitro. This finding led to the terms “glucose

responsive” for neurons that increased their activity in response to increased glucose, and

“glucose sensitive” for neurons that decreased their activity in response to increased glucose.

Today, glucose sensing neurons are more commonly referred to as either glucose-exicted

(GE) or glucose-inhibited (GI) based on their physiological response to changes in

extracellular glucose (143).

1.3.2 Glucose-Excited Neurons

The expression of glucokinase (GK) (144) and ATP-sensitive potassium (KATP)

channels composed of Kir6.2 and SUR1 subunits (145) has led to the idea that GE neurons

sense changes in extracellular glucose concentrations via a mechanism that is similar to that

which operates in pancreatic β-cells. In this proposed model, increased glucose

concentrations are detected primarily through increased oxidation of glucose and generation

of ATP. The subsequent changes in electrical activity are mediated by closure of KATP

channels. Studies using transgenic POMC-green fluorescent protein (GFP) mice have shown

that ARC POMC neurons exhibit typical GE responses and express the KATP channel (146).

However, recent studies have suggested an additional population of GE neurons that

sense glucose independently of changes in KATP channel activity. A KATP channel-

independent glucose-sensing mechanism has been identified in a population of ARC GE

neurons, which is believed to involve cellular depolarization from the opening of a non-

specific cation channel in response to elevated glucose concentrations (147).

Page 23: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

12

1.3.3 Glucose-Inhibited Neurons

In contrast to GE neurons, changes in AMP-activated protein kinase (AMPK) activity

are likely to mediate the inhibitory effects of glucose on ARC hypothalamic GI neurons

(148). AMPK is an evolutionarily conserved enzyme that acts as an intracellular energy

sensor to regulate fuel availability within a cell. AMPK is a heterotrimeric protein that

becomes activated allosterically by an increase in the intracellular AMP/ATP ratio. It has

been proposed that at low glucose concentrations, the rate of glucose uptake through GLUT3

and metabolism through GK and the glycolytic pathway are low. The resulting increase in

the AMP:ATP ratio would lead to activation of AMPK, which may then act to directly

phosphorylate and inactivate different ion channels leading to cellular depolarization (143).

While the mechanism of glucose-induced inhibition remains unclear, much more is

known about the physiological identities of these GI neurons. In the ARC, GI neurons were

found to co-express NPY and AgRP. Similarly, 94% of rat ARC neurons that were

stimulated by lowering extracellular glucose concentrations contained NPY

immunoreactivity (149). By switching extracellular glucose between 0.5 and 5 mM, 40% of

ARC NPY neurons were reversibly hyperpolarized and inhibited. Since NPY and AgRP are

orexigenic in nature, their co-localization in GI neurons implicates these neurons in the

mechanisms which lead to a stimulation of feeding.

1.4 Fatty Acid Sensing in Hypothalamic Neurons

1.4.1 Fatty Acids and Metabolic Physiology

The function of non-esterified fatty acids (NEFAs) was elucidated in the 1950’s

through the work of Vincent Dole (41) and Robert Gordon (42). Gordon demonstrated that

Page 24: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

13

plasma NEFAs originate from adipose tissues, and elucidated their use by tissues such as the

liver and myocardium. We now understand that NEFAs are the primary fuel for most tissues

under fasting conditions (43). The release of NEFAs into the circulation results partly from

the hydrolysis of triacylglycerol-rich lipids via the action of lipoprotein lipase (LPL) (43). In

addition to being an important source of energy, NEFAs are also necessary for membrane

lipid synthesis and lipid signaling (44). Although mostly bound to albumin, NEFA turnover

is fast. The circulating half-life of NEFAs is only 3-4 minutes (43). In the fasting state,

plasma NEFAs arise almost entirely from the hydrolysis of trigylcerides (TG) in adipocytes

(45). However, after a meal, there is an additional source of plasma NEFA. LPL in the

capillaries of adipose tissue hydrolyzes circulating TG, which constitutes much of the dietary

fat carried in chylomicrons. Though fatty acids thereafter become taken up by adipocytes for

storage, there is always a proportion that escapes and joins the plasma NEFA pool (46).

Therefore, the plasma NEFA pool composition changes in accordance with the composition

of meal fat (47).

Fat mobilization is rapidly suppressed by insulin. Therefore, plasma NEFA

concentrations fall after any meal containing carbohydrates. Typical plasma NEFA

concentrations range from 300-600 µmol/L in an overnight fasting state to approximately

1,300 µmol/L after a 72 hour fast (48).

1.4.2 Blood-Brain Barrier Permeability, Fatty Acid Uptake and Metabolic Fates

Once in the plasma, free fatty acids are bound by the carrier protein albumin.

Albumin increases the solubility of these FFAs and facilitates their transport across

membranes (44). To cross the blood-brain barrier, FFAs readily desorb from albumin and are

Page 25: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

14

rapidly taken up by a “flip-flop” diffusion process (49) and/or transport proteins. Transport

proteins include CD36, fatty acid transport protein (FATP) and plasma membrane fatty acid

binding protein (FABP) (50). Upon entry into the cell, FFAs become coupled to FABPs,

which carry FFAs from the plasma membrane to their target organelles.

After cellular uptake, fatty acids become rapidly esterified to a fatty acyl-coenzyme A

(fatty acyl-CoA). This reaction is catalyzed by the enzyme acyl-CoA synthetase (51). In this

activated aycl-CoA form, fatty acids can be (i) degraded by mitochondrial β-oxidation to

provide cellular energy, (ii) esterified to membrane lipids or (iii) enter the sphingolipid

pathway and contribute to the generation of ceramide metabolites (Figure 1.3).

1.4.3 Fatty Acids as Signaling Molecules in the Hypothalamus

Fatty acyl-CoA’s and the pathways regulating fatty acyl-CoA metabolism have been

implicated in the hypothalamic control of feeding behavior and energy homeostasis. One

hypothesis is that circulating lipids regulate feeding behavior by generating an increase in the

hypothalamic fatty acyl-CoA pool. In turn, these fatty acyl-CoA’s signal an energy surplus

within the hypothalamus, which activates neuronal circuits to decrease both food intake and

liver glucose production (52). Indeed, intracerebroventricular (icv) administration of the

monounsaturated fatty acid oleic acid is sufficient to inhibit food intake and liver glucose

production. Furthermore, icv oleic acid inhibits the expression of orexigenic NPY and AgRP

in the hypothalamus (53).

Given these findings, it was hypothesized that similar metabolic and behavioral

effects would be seen by increasing fatty acyl-CoA availability. Under genetic or

pharmacological inhibition of hypothalamic CPT1 (an enzyme which facilitates the transport

Page 26: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

15

Fig. 1.3 Metabolic fates of palmitate upon entering the cell.

Once palmitate enters the cell, it becomes “primed” in order to cross the mitochondrial membrane.

This occurs in the peroxisome, where peroxisomal acyl-CoA synthetase catalyzes the reaction

between the fatty acid and CoA. The resultant palmitoyl-CoA can then: 1) enter the mitochondria

where it undergoes β-oxidation, 2) participate in protein palmitoylation via the actions of protein

acyltransferase or 3) contribute towards de novo ceramide synthesis.

Page 27: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

16

of fatty acyl-CoA molecules into the mitochondria for β-oxidation), the concentration of

hypothalamic fatty acyl-CoA’s increased, whereas the expression of orexigenic NPY and

AgRP decreased (54). Therefore, these data lend support to the idea that fatty acids and the

availability of fatty acyl-CoAs are important components of hypothalamic lipid sensing.

1.5 Obesity and Hypothalamic Inflammation

1.5.1 High-Fat Feeding is Associated with Inflammation

Excessive caloric intake is a primary risk factor for the development of obesity.

Epidemiological studies have shown that individuals consuming high fat diets are

particularly prone to gaining body mass (55). In peripheral tissues, the deleterious metabolic

consequences of obesity arise, in part, from cellular inflammation triggered by this nutrient

excess. Excess visceral adiposity is accompanied by chronic low grade inflammation in the

liver, adipose tissue, skeletal muscle and vasculature. This inflammation is associated with

increased circulating levels of pro-inflammatory cytokines (56). Circulating saturated fatty

acids are also capable of triggering Toll-like receptor (TLR) signaling, which results in

subsequent activation of intracellular inflammatory signals such as inhibitor of kB-kinase-β

(IKKβ)/nuclear factor-kB (NF-κB) and c-Jun N-terminal kinase (JNK) (57). The end result is

a vicious cycle of inflammation that produces progressive, systemic metabolic impairment.

In 2005, evidence emerged that inflammatory changes could be detected in the brains

of high fat diet-fed animals. A 20-week HFD-feeding study found increased NF-κB signaling

in the rat cerebral cortex (58). Honing in on the hypothalamus, De Souza et al. (59) tested the

hypothesis that high fat consumption could modulate gene expression in the hypothalamus.

Using a macroarray, the expression of more than 1,000 hypothalamic genes was

Page 28: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

17

simultaneously measured. Of the 1,000 genes examined, more than 15% were modulated by

diet. Grouping the genes based on function revealed that inflammatory genes were most

affected after 16 weeks of HFD feeding (59).

1.5.2 Evidence that Hypothalamic Inflammation Contributes to HFD-Induced Obesity

Consistent with a role for hypothalamic inflammation in diet-induced obesity,

neuron-specific disruption of TLR4 or IKKβ/NF-κB pathways protected against diet-induced

obesity and its associated metabolic consequences (60). Viral deletion of IKKβ or over-

expression of a dominant-negative IKKβ isoform in the mediobasal hypothalamus also

reduced food intake and weight gain during HFD feeding (60). Moreover, in genetically

normal animals, central infusion of an IKKβ inhibitor or antibodies to TLR4 can reduce food

intake in diet-induced obese (DIO) mice (61). Taken together, these studies demonstrate the

causal role of hypothalamic inflammation in HFD-induced weight gain.

Complementing these findings is the fact that augmented hypothalamic inflammation

is associated with HFD-induced obesity. For example, neuronal expression of a constitutively

active IKKβ isoform increases food intake (60). Furthermore, infusion of the cytokine IL-4

directly into the brain of HFD-fed rats exerts a pro-inflammatory effect on the hypothalamus

that exacerbates weight gain in an IKKβ-dependent manner (62). These data suggest that

hypothalamic inflammation is both necessary and sufficient for initial and sustained weight

gain during HFD feeding.

Yet, if hypothalamic inflammation is to be implicated in obesity pathogenesis, it must

occur prior to obesity onset. Indeed, hypothalamic inflammation is observed weeks before

Page 29: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

18

peripheral cytokines are produced in the liver and adipose tissue, and before alterations in

body weight occur (63).

1.5.3 Palmitate as a Mediator of Hypothalamic Inflammation

Whether hypothalamic inflammation is a consequence of excess caloric intake

irrespective of diet composition has been the subject of considerable debate. One mechanism

that has received attention is the ability of saturated versus unsaturated fatty acids to activate

TLR4/NF-κB signaling. A predominant saturated fatty acid in our diet, palmitic acid (16:0),

is found in high concentrations in all animal products and accounts for approximately 20-

30% of the total FFAs in humans. Palmitic acid enters the brain linearly with time and is

rapidly incorporated into brain lipids (64). Many studies that have investigated the role of

FFAs in HFD-induced hypothalamic inflammation have utilized palmitic acid. Recent work

demonstrates that saturated fatty acid palmitate (16:0) induces NF-κB signaling through a

TLR4-dependent mechanism when administered in neuronal cell culture and after infusion

into the brain (65). Signaling through NF-κB leads to the induction of cytokine gene

expression, which causes local levels of TNF-α, IL-1β and IL-6 to rise and exacerbate the

inflammatory state (Figure 1.4) (66).

1.6 Obesity and Hypothalamic Insulin Resistance

1.6.1 Obesity, Diabetes and Insulin Resistance

Currently, more than one third of U.S. adults are obese (which is defined as having a

BMI >30 kg/m2) and over 11% of individuals aged 20 or over have diabetes (67). Due to the

strong association between T2DM and obesity, Zimmet et al. coined the term “diabesity”

(68). However, only 20% of obese individuals develop T2DM, and this is thought to be due

Page 30: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

19

Fig. 1.4 Activation of the IKKβ/NF-κB pathway leads to inflammation and impaired insulin signaling.

Activation of TLR4 or TNF-α receptor by palmitate and TNF-α, respectively, stimulates downstream NF-

κB and AP-1 transcription factors to upregulate gene expression of pro-inflammatory cytokines. Insulin

signaling is inhibited by chronic receptor stimulation by insulin itself or by stimulation of the TLR4 and/or

TNF-α receptors. IKKβ and JNK, in particular, can inhibit insulin signaling by phosphorylating serine

residues on IRS proteins.

AP-1 = activator protein 1

IKKβ = inhibitor of IkappaB kinase beta

Ins = insulin

IR = insulin receptor

IRS = insulin receptor substrate protein

JNK = c-Jun N-terminal kinase

Pal = palmitate

TNF = tumor necrosis factor

TLR4 = toll-like receptor 4

TNFR = tumor necrosis factor receptor

TAK1 = transforming growth factor-β-activated kinase 1

NF-κB = nuclear factor kappa B

Page 31: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

20

to a compensatory response by pancreatic β-cells to increase insulin secretion. Therefore,

T2DM involves both decreased insulin sensitivity and a loss of compensatory insulin

secretion. Insulin resistance, then, is defined as a diminished cellular response to insulin,

resulting in the inability to increase glucose uptake leading to increased blood glucose (69).

Obesity results from an imbalance between energy intake and energy expenditure.

The result is adipocyte hypertrophy, along with increased lipolysis (70). Increased adiposity

can lead to insulin resistance through increased adipocyte-derived FFAs and increased

adipokines (71). Excess FFAs become stored in non-adipose cells such as muscle, where they

may be catabolized into lipid metabolites such as fatty acyl-CoAs, diacylglycerol (DAG),

triacylglycerol (TAG) and ceramide. These lipid metabolites are capable of inhibiting insulin

signal transduction leading to insulin resistance (71). Moreover, in an obese state, there is an

increase in peripherally-derived pro-inflammatory cytokines such as tumor necrosis factor-

alpha (TNF-α), which have also been shown to induce insulin resistance in mice and humans

(72). Another contributing factor to the development of insulin resistance is

hyperinsulinemia, which will be discussed in the following section. Finally, impaired insulin

signaling has the ability to potentiate obesity pathogenesis due to the importance of central

insulin action in regulating energy balance. Therefore, the maintenance of proper insulin

action is critical for maintaining energy homeostasis.

1.6.2 Hyperinsulinemia and the Development of Insulin Resistance

Insulin resistance is a hallmark feature of obesity. There are numerous etiologies for

insulin resistance, including lipotoxicity, inflammation and hyperinsulinemia (73).

Hyperinsulinemia reflects the compensation by insulin-secreting β-cells to systemic insulin

Page 32: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

21

resistance. In vivo studies indicate that prolonged exposure to high levels of insulin can lead

to insulin resistance (74). Indeed, plasma insulin levels are increased in obese states and this

increase occurs prior to a reduction in insulin sensitivity (75). At the cellular level,

hyperinsulinemia impairs insulin signal transduction through homologous desensitization.

The insulin receptor itself is involved in negative feedback involving a reduction in

(i) receptor affinity, (ii) the number of receptors expressed on the cell surface and (iii) the

effectiveness of the receptor as a transmitter of stimulatory signals (76). Continual exposure

to insulin can also lead to serine phosphorylation of downstream IRS proteins, which reduces

its ability to activate downstream elements in the insulin signaling pathway (76).

1.6.3 High Fat Feeding and Neuronal Insulin Resistance

The growing trend towards a more sedentary lifestyle combined with the

consumption of fat-rich foods play an important role in the current obesity epidemic (34).

Consumption of HFD for as little as 72 hours is sufficient to reduce hypothalamic insulin

sensitivity in rats (77). Elevated saturated fatty acids not only increase body weight, but also

chronically reduce hypothalamic insulin sensitivity (77). At a molecular level, saturated fatty

acids such as palmitate cross the blood-brain barrier and accumulate in the hypothalamus.

Here, they activate pro-inflammatory signaling pathways including toll-like receptor 4

(TLR4) signaling, resulting in central insulin resistance (65).

Palmitate-mediated central insulin resistance is due, at least in part, to the activation

of protein kinase Cθ (PKCθ). Subsequent translocation of PKCθ to the cell membrane

prevents insulin-mediated activation of PI3K via direct interaction with insulin receptor and

insulin receptor substrate proteins (78). On the other hand, palmitate can also activate NF-κB,

Page 33: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

22

which subsequently induces suppressor of cytokine signaling (SOCS) 3 expression (60).

SOCS3 is one of the principal negative regulators of insulin signaling, interfering with

insulin-mediated phosphorylation of IR and its downstream molecules. It also targets IRS

proteins for proteasomal degradation (79). Elevated NF-κB signaling also triggers

endoplasmic reticulum stress leading to increased activity of c-Jun N-terminal kinase (JNK).

In turn, JNK mediates inhibitory phosphorylation events on IRS serine residues which

contribute to the development of insulin resistance (80,81).

During obesity progression, elevated levels of pro-inflammatory cytokines exacerbate

central insulin resistance by further activating NF-κB and JNK signaling. TNF-α, like

palmitate, can induce expression of protein tyrosine phosphatase 1B (PTP1B), potentially

through transactivation of NF-κB (82). Elevated levels of PTP1B in the ARC, as seen after

20 weeks of HFD feeding, inhibit insulin signaling by direct dephosphorylation of the IR

(Fig. 1.5) (83).

1.6.4 FFA Metabolism and Insulin Resistance

Dysregulated FFA metabolism is thought to play a causal role in the development of insulin

resistance (84). The large majority of FFAs enter the glycerolipid pathway, where they

become substrates for membrane glycerophospholipids and TAG, the primary form of stored

fat (86). Glycerol-3-phosphate acyltransferase (GPAT) is the enzyme that regulates entrance

of FFAs into this pathway, and it transfers the acyl group from fatty acyl-CoA to glycerol-3-

phosphate (86). The saturated fatty acid palmitate increases intracellular levels of DAG, the

precursor to TAG, and TAG itself in rat islets (87). Interestingly, the GPAT knockout mouse

is protected from insulin resistance on a HF diet (88).

Page 34: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

23

Fig. 1.5 Mechanisms of palmitate-mediated inhibition of insulin signaling.

Palmitate has been shown to inhibit insulin signaling through a variety of mechanisms. 1) Palmitate may

enter the cell and contribute to an increase in de novo ceramide synthesis. Ceramides are known to interact

with and activate the phosphatase enzyme PP2A. PP2A dephosphorylates Akt, leading to impaired insulin

signaling. 2) Palmitate can increase intracellular levels of diacylglycerol, leading to activation of PKCθ and

subsequent serine phosphorylation of IRS-1. 3) Palmitate (and inflammation) induces the expression of

PTP1B, which is a protein tyrosine phosphatase enzyme. PTP1B inhibits insulin signaling by removing

phosphate groups from the tyrosine residues of the activated insulin receptor.

AKT = protein kinase B

DAG = diacylglycerol

ER = endoplasmic reticulum

Pal = palmitate

Pal-CoA = palmitoyl-CoA

PKCθ = protein kinase C θ

PP2A = protein phosphatase 2A

PTP1B = protein tyrosine

phosphatase 1B

Page 35: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

24

Rates of ceramide synthesis depend on the availability of long-chain saturated fatty

acids, which participate in the initial rate-limiting step of de novo ceramide synthesis (86). In

this reaction, serine palmitoyltransferase (SPT) catalyzes the condensation of palmitoyl-CoA

and serine to produce 3-ketosphinganine. Subsequent reactions lead to the eventual synthesis

of ceramide, which serves as a precursor for more complex sphingolipids (86). Ceramides

with different fatty acid and long-chain base compositions can be formed in different

compartments or membranes of the cell, each with potentially distinct functions.

Interestingly, ceramide levels are elevated in rodent and human insulin-resistant tissues (89).

As demonstrated in numerous reports, elevated ceramide levels may inhibit insulin-

stimulated glucose uptake, GLUT4 translocation and/or glycogen synthesis (90). This

dysregulation of insulin signaling has been linked to ceramide-mediated regulation of IRS-1

and Akt/PKB.

Three independent groups found that treating cultured cells with short-chain ceramide

analogs blocked insulin-stimulated tyrosine phosphorylation of IRS-1 and its subsequent

activation of PI3K (91). These groups proposed that ceramide may promote the

phosphorylation of IRS-1 on inhibitory serine/threonine residues. In various cell types,

ceramides have been shown to activate extracellular signal-regulated kinase 2 (ERK2), p38,

JNK and IkB kinases (IKKs) (92), which have been implicated in serine/threonine

phosphorylation of IRS-1 (93).

Many groups have demonstrated that ceramide also inhibits phosphorylation and

activation of Akt/PKB. It is now understood that ceramide inhibits activation of Akt/PKB

through two distinct mechanisms. First, ceramide promotes the dephosphorylation of

Akt/PKB through direct activation of protein phosphatase 2A (PP2A) (94). Indeed, the PP2A

Page 36: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

25

inhibitor okadaic acid was sufficient to prevent the inhibitory ceramide effects on Akt/PKB

in C2C12 myotubes and brown adipocytes (95, 96). Second, ceramide may activate PKCζ,

which inhibits Akt/PKB translocation to the membrane by phosphorylating threonine-34

(97).

1.7 Cell Model

1.7.1 The Need for Cell Lines

In vivo experimentation is important for determining the overall function of a

molecule (i.e. hormone, receptor or structural protein). Indeed, the use of animal models has

enhanced our basic understanding of physiological processes, such as energy homeostasis.

These studies have elucidated the role of the brain in overall metabolism, and have triggered

the development of brain-specific and neuron-specific mouse models. Yet, despite new

technologies allowing for closer examination of intracellular workings, animal

experimentation has its limits. This is especially true in the context of the hypothalamus,

where a collection of cell phenotypes exist. Therefore, classical in vivo approaches cannot

establish the molecular mechanisms involved in gene regulation and cellular signaling.

Moreover, it is difficult to determine the direct actions of agents, such as nutrients or

hormones, on specific cell types. Given these limitations, researchers have turned to the use

of cell lines (98).

Cell lines allow for experimentation with homogeneous populations of cells in a

controlled environment. However, one cannot state for certain that cell lines function exactly

as the native cells would. For this reason, caution must be taken before extrapolating findings

from cell lines to an in vivo model. When working with neuronal cell lines, it is particularly

Page 37: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

26

important to remember that these models lack the complexity and integrated network of

neurons found in vivo. Regardless, recent studies looking at molecular events in vitro have

found that the results from cell lines mirror that of in vivo studies (98).

Basic tissue culture techniques were established in 1885 by Wilhelm Roux, and it was

not until 1940 that the first immortal cell line was developed (99). Since that time, cell lines

have been produced from many different tissues although the first attempt at immortalizing

neurons was not performed until 1974 by Shaw et al. (100). Shaw et al. transfected primary

hypothalamic cells from embryonic mice with simian virus 40 T-Ag to create an

immortalized cell population labeled HT9. Years later, Cepko et al. developed retroviral

shuttle vectors which would allow researchers to retrovirally infect primary cells with an

immortalizing oncogene and selectively propagate them (101).

1.7.2 Adult Hypothalamic Cell Lines (mHypoA-xx)

Non-transformed primary hypothalamic cultures are difficult to maintain, have a short

lifespan and represent a heterogeneous population of neurons. Contrarily, immortalized,

clonal cell lines represent an unlimited and homogeneous population of specific neuronal cell

types. Since the hypothalamus contains a wide range of cell types, Belsham et al. recognized

the need for mouse cell lines representative of many unique hypothalamic neurons. Belsham

et al. initially developed 38 embryonic, clonal hypothalamic mouse cell lines (98). However,

to understand key molecular mechanisms involved in adult neuroendocrine cells, the

Belsham group also immortalized adult hypothalamic cell models.

In order to immortalize the adult neurons, cells were treated with ciliary neurotrophic

factor (CNTF) to induce cell proliferation. This would render the cell cultures amenable to

Page 38: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

27

retroviral transfer of the SV40 T-antigen oncogene (Figure 1.6) (102). Over 50 adult mouse

cell lines were eventually established, and labeled in the form mHypoA-‘clone number’. Like

the embryonic cell lines before them, the adult cell lines also express mature neuronal

markers, display typical neuronal morphology and have been characterized for the expression

of necessary neuropeptides and receptors. The hypothalamic cell lines made available from

our lab and others allow for the study of molecular events involved in nutrient sensing in

distinct neuronal populations. These novel, representative cell models put us in an

advantageous position to determine the direct effects of nutrients (i.e. palmitate) on

neuropeptide gene expression and signaling events in neuropeptide-expressing neurons.

1.7.3 mHypoA-NPY/GFP Cell Line

To immortalize NPY/AgRP-expressing neurons from the adult hypothalamus, the

Belsham group dissected hypothalamii from NPY-GFP transgenic mice and immortalized as

described above. NPY/AgRP neurons were then selected using flow cytometry. The NPY-

GFP cell line has been thoroughly characterized using RT-PCR, ICC and NPY secretion

assays, and the phenotypic characterization is described in Table 1.1. In this thesis, the

mHypoA-NPY/GFP cell model is used to describe palmitate-mediated regulation of AgRP

gene expression and to elucidate the effects of palmitate on signaling events in representative

NPY/AgRP-expressing neurons.

1.8 Hypothesis and Aims

Neuronal circuits within the hypothalamus form the homeostatic control mechanism

that controls food intake and energy balance. It has been well established that coordinated

regulation

Page 39: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

28

1) 2)

3) 4) 5)

Fig. 1.6 Generation of the mHypoA-NPY/GFP cell line.

Adult NPY/GFP-expressing transgenic mice were generated (1). Cells were harvested from the

GFP-expressing mouse hypothalamus (2). These cells were then treated with CNTF to induce

neurogenesis, and transfected with SV-40 T antigen for immortalization (3). Cells were FAC sorted

for GFP fluorescence with greater than 95% purity (4). The fluorescent cells represent the cells of

interest, mHypoA-NPY/GFP (5).

Page 40: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

29

of neuropeptide gene expression from the hypothalamus is critical to maintain normal energy

homeostasis. Disturbances at the hypothalamic level may lead to metabolic disease. A

leading contributor to diet-induced obesity and T2DM is hypothalamic inflammation and

insulin resistance in response to saturated fatty acids consumed in the diet. In fact,

hypothalamic inflammation occurs weeks before peripheral cytokines are produced and

before alterations in body weight occur. Several molecules and pathways have been

identified as mediators of hypothalamic inflammation during HFD feeding, including JNK,

IKKβ and TLR4. JNK and IKKβ, in particular, can inhibit insulin signaling through

induction of SOCS3 signaling or serine phosphorylation of insulin receptor substrate. For

these reasons, hypothalamic inflammation is an important new target for obesity therapeutics.

Research has since focused on understanding how HFD induces hypothalamic

inflammation and insulin resistance. Palmitate (16:0), a non-esterified saturated fatty acid

which exists at high levels in the plasma of obese individuals, has received considerable

attention due to its ability to activate TLR4/NF-κB signaling. Indeed, icv injection of

palmitate has been shown to attenuate hypothalamic insulin signaling and increase IKKβ

activity.

Though animal models have proven invaluable in establishing our basic understanding

of energy homeostasis, the specific molecular events involved in nutrient (particularly fat)

sensing in distinct neuronal populations remain largely unknown. However, the Belsham

laboratory has generated several immortalized, hypothalamic neuronal cell lines from

primary fetal and adult hypothalamic neuronal cell culture. These representative cell models

enable us to determine the direct effects of nutrients (i.e. palmitate) on neuropeptide gene

expression and signaling events in neuropeptide-expressing neurons. This thesis involved the

Page 41: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

30

use of the mHypoA-NPY/GFP cell line, which is a pure population of representative NPY

neurons from the entire hypothalamus. We used this cell line to study the effects of palmitate

on inflammatory status, AgRP gene expression and insulin signaling in representative

NPY/AgRP neurons.

It was therefore hypothesized that palmitate would alter cellular function in

NPY/AgRP neurons. Treatment of mHypoA-NPY/GFP cells with palmitate would: (i)

induce a state of inflammation, (ii) alter AgRP gene expression and (iii) alter insulin

signal transduction.

Aim #1: Determine whether palmitate can alter inflammatory status and AgRP gene

expression in mHypoA-NPY/GFP neurons. Whether changes (if any) in AgRP gene

expression are palmitate metabolism-dependent will also be determined. These results are

presented in section 3.1 and 3.3.

Aim #2: Determine whether TNF-α, a pro-inflammatory surrogate of palmitate, can

alter inflammatory status and AgRP gene expression in mHypoA-NPY/GFP neurons. These

results are presented in section 3.2.

Aim #3: Determine whether palmitate induces the phosphorylation/activation of

ERK1/2, JNK and/or p38 MAPK in mHypoA-NPY/GFP neurons. These results are presented

in section 3.4.

Aim #4: Determine whether prolonged exposure to insulin or palmitate will have an

effect on insulin signaling in mHypoA-NPY/GFP neurons. These results are presented in

section 3.5.

Page 42: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

31

Table 1.1 Characterization of the mHypoA-NPY/GFP cell line via immunofluorescence

and semi-quantitative PCR.

(Screening data courtesy of Jennifer Chalmers and Prasad Dalvi)

mRNA mHypoA-NPY/GFP

Enzyme mRNA

CPT1a and c +

Na+/K

+ ATPase +

Transporter, Channel and Receptor mRNA

GLUT 1,3,4,5,8 +

GLUT 2 -

IR +

GPR120 +

ObRb +

TLR4 +

Neuropeptide and Cytokine mRNA

AgRP +

NPY +

POMC -

TNF-α +

IL-6 +

Page 43: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

32

Chapter 2

Materials and Methods

Page 44: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

33

2. Materials and Methods

2.1 Cell Culture and Reagents

The hypothalamic cell line from the adult mouse, mHypoA-NPY/GFP, was isolated

and immortalized as previously described (103). mHypoA-NPY/GFP cells were grown to

confluency and maintained in Dulbecco’s Modified Eagle Medium (DMEM) (Sigma, St.

Louis MO, USA) supplemented with 5% fetal bovine serum (HyClone Laboratories, Logan,

UT) and 1% penicillin/streptomycin (Life Technologies Inc., Burlington, Canada) at 37°C in

an atmosphere of 5% CO2. Cells were then seeded onto 60 mm culture plates 24 hours prior

to treatments. Sodium palmitate, TNF-α and methyl palmitate were obtained from Sigma-

Aldrich (Oakville, Canada). Insulin was purchased from Novo Nordisk Canada Inc.

(Mississauga, Canada).

2.2 Palmitate Preparation

A 25 mM stock of palmitate was first prepared by dissolving approximately 6.9 mg of

sodium palmitate (Sigma) in 1 mL of Hypure water (Thermo Scientific, Rockford, IL, USA)

and heated to 60°C. Upon treatment, the media in each 60 mm culture plate was replaced

entirely with 3 mL of fresh treatment media. To achieve a working concentration of 25 μM

palmitate in the treatment media (DMEM, 5% FBS, 1% penicillin/streptomycin), 3 μL of

stock palmitate was added per 3 mL treatment media.

2.3 TNF-α Preparation

Stock TNF-α powder (10 µg) was dissolved in 1 mL of Hypure H2O. 50 uL aliquots

of 10 µg/mL TNF-α were then stored in a -20°C freezer. The day before treating cells with

TNF-α, media was changed to 2.5 mL low glucose, 5% FBS, 1% penicillin/streptomycin

Page 45: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

34

media. To achieve a working concentration of 50 ng/mL TNF-α in the culture plates, 15 μL

of stock TNF-α was added per 500 μL serum-free DMEM and added to each culture plate on

the day of treatment.

2.4 Insulin Preparation

Insulin (600 µM) was first diluted with 1x PBS to a stock concentration of either 100

µM or 10 µM in 0.6 mL eppendorf tubes. Working concentrations of 100 nM or 10 nM

insulin were achieved by adding 3 μL of stock insulin per 500 μL serum-free media and

added to cell culture dishes already containing 2.5 mL serum-free media.

2.5 Quantitative RT-PCR

Total RNA was isolated from mHypoA-NPY/GFP cells at the indicated time points

using the guanidinium thiocyanate phenol chloroform extraction method, as previously

described (104). RNA concentrations, and their accompanying purity ratios, were determined

using a NanoDrop 2000c spectrophotometer (Thermo Scientific, Nepean, Ontario, Canada).

Contaminating DNA was removed from all RNA samples using Turbo DNAase (Ambion,

Austin, TX, USA) treatment (1 hr, 37°C). RNA was then reverse-transcribed using the High

Capacity cDNA Reverse Transcriptase kit (Applied Biosystems, Streetsville, Ontario,

Canada). 50 ng of cDNA was loaded per sample well and amplified using an Applied

Biosystems Prism 7000 Real-Time PCR machine together with gene-specific primers (Cell

Signaling Technology Inc., Danvers, MA, USA) and SYBR green master mix. Each sample

was run in triplicate according to the following cycle sequence: 2 minutes at 50°C, 10

minutes at 95°C; 40 cycles of 15 seconds at 95°C, 1 minute at 60°C; 15 seconds at 95°C, 15

Page 46: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

35

seconds at 60°C, and 15 seconds at 95°C. Quantitative reverse transcriptase PCR data was

then quantified using the standard curve method and normalized to histone 3a.

2.6 Western Blot Analysis

Total protein was harvested using 1x cell lysis buffer (Cell Signaling) supplemented

with 1 mM PMSF, 1% phosphatase inhibitor cocktail 2 (Sigma) and 1% protease inhibitor.

The soluble fraction was isolated after centrifugation at 14,000 rpm for 10 minutes at 4°C.

Protein was then quantified using a BCA protein assay kit (Thermo Scientific) according to

the manufacturer’s instructions, and lysates were resolved on 8% polyacrylamide gels and

transferred onto Immobilon-P PVDF membrane (Bio-Rad, Hercules, CA, USA). Membranes

were blocked in 5% milk in Tris-buffered saline with 0.1% Tween-20 (TBS-T) for 1 hour,

followed by primary antibody incubation overnight at 4°C. Membranes were washed with

TBS-T before and after incubation with goat anti-rabbit HRP secondary antibody (Cell

Signaling) for one hour. Protein was visualized using a Kodak Image Station 2000R

(Eastman Kodak Company, Rochester, NY, USA) and band intensity was quantified using

Kodak 1D Image Analysis Software 3.6. Primary antibodies used for western blotting include

Gβ, phospho-AKT, AKT, phospho-p44/42 MAPK (ERK1/2), phospho-JNK and phospho-

p38 MAPK. All primary antibodies were obtained from Cell Signaling.

In section 3.4, mHypoA-NPY/GFP neurons were treated with palmitate (25 µM) or

vehicle alone over a 60 minute time course and protein was harvested at 5, 15, 30 and 60

minute time points. For insulin resistance experiments, cells were pre-treated with either 100

nM insulin or 25 µM palmitate for 24 hours before insulin treatment media was changed to

serum-free DMEM for one hour. Cells were then re-challenged with 10 nM insulin and

Page 47: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

36

protein was isolated after 15 minutes. In the insulin resistance studies, phospho-proteins were

normalized to total protein. However, for the signaling experiments, phospho-proteins were

normalized to the loading control Gβ. Although total protein comparisons are ideal, our

laboratory has found in previous studies that Gβ is a reliable indicator of loading status.

2.7 Statistical Analysis

Data are presented as the mean ± the standard error of the mean (SEM). All statistics

were calculated using Graphpad Prism 6.0 (San Diego, CA, USA). Groups were compared

using two-way analysis of variance (ANOVA) with Tukey’s test for post hoc comparisons. A

minimum of three experimental replicates were performed for each experiment. Significance

is denoted by *p <0.05, **p <0.01 and ***p <0.001.

Page 48: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

37

Chapter 3

Results

Page 49: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

38

3. Results

3.1 Palmitate elicits an inflammatory response and upregulates Agrp gene expression in

mHypoA-NPY/GFP neurons

Animal models of diet-induced obesity exhibit inflammation in hypothalamic areas

critical for energy homeostasis. This is generally defined as the activation of pro-

inflammatory signaling within neurons and glia (56). While systemic levels of pro-

inflammatory cytokines, such as IL-6 and TNF-α, are also elevated in the obese state (105),

hypothalamic inflammation has been shown to occur weeks before peripheral cytokines are

even produced (63). Long-chain saturated fatty acids, in particular, have been shown to

activate toll-like receptor 4 signaling to induce local cytokine expression in these neurons

(65). Indeed, mice with neuron-specific deletion of IKKβ are resistant to the effects of a HFD

to cause obesity and central insulin resistance (60). Such findings suggest a role for insulin

resistance induced by hypothalamic inflammation in the mechanism whereby HF feeding

leads to obesity. However, the specific hypothalamic neuronal cell types that trigger this

inflammation in response to HF feeding remain unknown.

To determine whether the saturated fatty acid palmitate can elicit an inflammatory

response in mHypoA-NPY/GFP neurons, qRT-PCR was used to assess relative changes in

mRNA levels of NF-κB (p105 subunit) and IkBα following treatment with 25 µM palmitate.

Treatment with 25 µM palmitate for 2 hours was sufficient to increase NF-κB mRNA levels

relative to control (n=3; P<0.05; Fig. 3.1A) in this cell line. Within 4 hours, this dose of

palmitate also significantly increased IkBα mRNA levels (n=3; P<0.01; Fig. 3.1B).

Page 50: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

39

While changes in AgRP gene expression are likely following the establishment of

insulin resistance in NPY/AgRP neurons, the ability of fatty acids to directly modulate AgRP

gene expression is unknown. Therefore, to determine whether AgRP gene expression could

be modulated by the saturated fatty acid palmitate in mHypoA-NPY/GFP neurons, qRT-PCR

was used to assess the regulation of AgRP mRNA levels upon exposure to palmitate.

Treatment of mHypoA-NPY/GFP neurons with palmitate (25-50 µM) led to an increase in

AgRP mRNA levels at 24 hours (n=3-4; P<0.05; Fig. 3.1 E). However, high levels of

saturated fatty acids can be toxic to the neurons. To this end, we evaluated the toxicity of the

doses used. In particular, we examined the susceptibility of the mHypoA-NPY/GFP neurons

to apoptosis by measuring the ratio of Bax/Bcl2 mRNA. While 50 µM palmitate resulted in a

significant increase in Bax/Bcl2 mRNA at all time points examined (n=3; P<0.05; Fig.

3.1D), 25 µM palmitate did not alter this ratio at any time (n=4; Fig. 3.1C). Since the

susceptibility to apoptosis was minimized with 25 µM palmitate, it would be the

concentration used in all subsequent studies.

3.2 TNF-α, a pro-inflammatory surrogate of palmitate, also upregulates AgRP gene

expression in mHypoA-NPY/GFP neurons

Given palmitate’s ability to activate TLR4 and downstream IKKβ/NF-κB signaling, it is

likely that palmitate also induces expression of the pro-inflammatory cytokine TNF-α. TNF-

α is a pro-inflammatory cytokine that drives tissue inflammation, at least in part, through

activation of the classic IKKβ/NF-κB inflammatory signaling pathway. Activation of the

transcription factor NF-κB leads to further expression of pro-inflammatory cytokines, such as

TNF-α, IL-1β and IL-6. However, it is not known whether TNF-α can directly regulate AgRP

gene expression in NPY/AgRP neurons. Therefore, mHypoA-NPY/GFP neurons were

Page 51: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

40

3.3 Palmitate-mediated regulation of AgRP gene expression is metabolism-dependent

Time (h)

Bax/B

cl2

/His

ton

e m

RN

A

4h 24h0.0

0.5

1.0

1.5

2.0

Time (h)

Bax/B

cl2

/His

ton

e m

RN

A

4h 24h0.0

0.5

1.0

1.5

2.0

*

*

Time (h)

NF

-kB

/His

ton

e m

RN

A

1 2 4 240.0

0.5

1.0

1.5 *

Time (h)

IkB

/His

ton

e m

RN

A

1 2 4 240.0

0.5

1.0

1.5

2.0

2.5**

4h 24h0.0

0.5

1.0

1.5

2.0

Time (h)

Ag

RP

/His

ton

e m

RN

A

H2O

25 uM Palmitate*50 uM Palmitate

*

Fig. 3.1 Saturated fatty acid palmitate upregulates pro-inflammatory and Agrp gene expression.

Top: mHypoA-NPY/GFP neurons were treated with 25 µM palmitate (grey) or vehicle (black), RNA was isolated from 1 to 24

hours and analyzed using quantitative real-time PCR with primers for NF-κB (A) or IκBα (B). Treatment with 25 µM palmitate

resulted in a significant increase in NF-κB transcript levels at 2 hours, and a significant increase in IκBα transcript levels at 4

hours. Data was normalized to histone 3a and shown as mean values ± SEM (n=3 independent experiments). *P<0.05;

**P<0.01; ***P<0.001, as per two-way ANOVA with Tukey’s post-hoc test.

Bottom: To determine the optimal dose of palmitate in mHypoA-NPY/GFP neurons, dose curve analysis was performed and

relative changes in Bax/Bcl2 mRNA were assessed. 50 µM palmitate significantly increased Bax/Bcl2 mRNA at 4 and 24 hours

following treatment (D). However, 25 µM palmitate did not significantly alter this ratio (C). Both 25 and 50 µM palmitate

significantly upregulated AgRP transcript levels at 24 hours (E), but only a 25 µM dose of palmitate was used for the remaining

studies. Data was normalized to histone 3a and shown as mean values ± SEM (n=3-4 independent experiments). *P<0.05;

**P<0.01; ***P<0.001, as per two-way ANOVA with Tukey’s post-hoc test.

A B

C D

E

Page 52: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

41

exposed to 50 ng/mL TNF-α for up to 24 hours and gene expression of AgRP and certain

pro-inflammatory markers (IkBα, NF-κB and IL-6) was assessed using qRT-PCR. Analysis

of the results indicates that in the mHypoA-NPY/GFP neurons, AgRP gene expression was

significantly increased by TNF-α at 4, 8 and 24 hours following treatment (n=3; P<0.05; Fig.

3.2A). At 2 to 4 hours following treatment, TNF-α also upregulated mRNA levels of NF-κB

(n=3; P<0.05; Fig. 3.2B), IkBα (n=3; P<0.05; Fig. 3.2C), and IL-6 (n=3; P<0.05; Fig. 3.2D).

These findings demonstrate that TNF-α, like palmitate, can regulate the expression of AgRP

while inducing/exacerbating an inflammatory state in mHypoA-NPY/GFP neurons.

3.3 Palmitate-mediated regulation of AgRP gene expression is metabolism-dependent

Current literature has demonstrated the importance of fatty acid metabolism to overall energy

homeostasis. For example, DIO mice treated with myriocin (an inhibitor of de novo ceramide

synthesis) show a complete reversal of glucose intolerance and peripheral insulin resistance

(106). Moreover, central administration of inhibitors of β-oxidation reduce food intake (54).

However, whether fatty acid metabolism plays a role in palmitate-mediated regulation of

AgRP gene expression is unknown. Therefore, to determine whether palmitate-mediated

regulation of AgRP gene expression is palmitate metabolism-dependent, the mHypoA-

NPY/GFP neurons were treated with methyl palmitate. Methyl palmitate cannot be

metabolized, as the additional methyl group prevents activation of the fatty acid to palmitoyl-

CoA by the long chain fatty acyl-CoA synthetase enzyme. Treating our neurons with

different doses of methyl palmitate (25, 50 and 75 µM) for 24 hours did not result in similar

upregulation of AgRP mRNA as seen following 25 µM palmitate treatment (n=4; Fig. 3.3).

This result suggests that palmitate-mediated upregulation of AgRP gene expression is

palmitate metabolism-dependent.

Page 53: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

42

Time (h)

Ag

RP

/His

ton

e m

RN

A

0 1 2 4 8 240.0

0.5

1.0

1.5

2.0

2.5H2O

50 ng/mL TNF-

*

****

*

2 40.0

0.5

1.0

1.5

Time (h)

NF

-kB

/His

ton

e m

RN

A

*

Time (h)

IkB

/His

ton

e m

RN

A

2 40.0

0.5

1.0

1.5

2.0*

2 40.0

0.5

1.0

1.5

Time (h)

IL-6

/His

ton

e m

RN

A

H2O

50 ng/mL TNF-*

A

B C

D

Fig. 3.2 TNF-α upregulates pro-inflammatory and Agrp gene expression.

Top: mHypoA-NPY/GFP neurons were treated with 50 ng/mL TNF-α (grey) or vehicle (black), RNA was isolated from 1 to 24

hours and analyzed using quantitative real-time PCR with a primer for AgRP (A). Treatment with 50 ng/mL TNF-α resulted in a

significant increase in AgRP transcript levels at 4, 8 and 24 hours. Data was normalized to histone 3a and shown as mean values

± SEM (n=3 independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-way ANOVA with Tukey’s post-hoc test.

Bottom: mHypoA-NPY/GFP neurons were treated with 50 ng/mL TNF-α (grey) or vehicle (black), RNA was isolated from 1 to

4 hours and analyzed using quantitative real-time PCR with primers for NF-κB (B), IκBα (C) and IL-6 (D). Treatment with 50

ng/mL TNF-α resulted in a significant increase in NF-κB transcript levels at 4 hours, a significant increase in IκBα transcript

levels at 2 hours and a significant increase in IL-6 transcript levels at 4 hours. Data was normalized to histone 3a and shown as

mean values ± SEM (n=3 independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-way ANOVA with Tukey’s

post-hoc test.

Page 54: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

43

25 uM 50 uM 75 uM0.0

0.5

1.0

1.5

Treatment Dose

Ag

RP

/His

ton

e m

RN

A

H2O

Methyl Palmitate

mHypoA-NPY/GFP, AgRP, 24 hrs

Fig. 3.3 Methyl palmitate does not regulate Agrp gene expression.

mHypoA-NPY/GFP neurons were treated with 25, 50 or 75 µM methyl palmitate (grey) or vehicle (black), RNA was

isolated at 24 hours and analyzed using quantitative real-time PCR with a primer for AgRP (A). Methyl palmitate treatment

did not result in similar upregulation of AgRP mRNA as seen with 25 µM palmitate treatment. Data was normalized to

histone 3a and shown as mean values ± SEM (n=4 independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-

way ANOVA with Tukey’s post-hoc test.

Page 55: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

44

3.4 Palmitate triggers the phosphorylation of p38 MAPK in mHypoA-NPY/GFP

neurons

In peripheral tissues, palmitate has been shown to induce the phosphorylation and

activation of ERK1/2, JNK and p38 MAPK pathways. For example, palmitate induces

phosphorylation/activation of the MEK-ERK-IKK axis and pro-inflammatory cytokine

expression in skeletal muscle (107). In HepG2 cells, palmitate induces hepatic insulin

resistance through activation of JNK and p38 MAPK pathways (108). Yet, the direct effects

of palmitate on signaling events in specific hypothalamic neurons remain relatively

unknown. Recent work by Mayer et al. has shown that palmitate induces JNK

phosphorylation in a hypothalamic, neuronal cell model, mHypoE-44 (109). To test whether

any of the aforementioned pathways become activated in response to palmitate exposure in

mHypoA-NPY/GFP neurons, Western Blot analysis was used to analyze the phosphorylation

status of these key signaling kinases. Although palmitate did not induce phosphorylation of

ERK1/2 or JNK, it induced phosphorylation of p38 MAPK after 30 minutes of exposure

(n=3; P<0.05; Fig. 3.4A). This result may provide some insight into the mechanism by which

palmitate mediates its regulation of AgRP gene expression.

3.5 Palmitate pre-treatment dampens the mHypoA-NPY/GFP neurons’ response to

insulin

High-fat diets and elevated levels of saturated fatty acids have been shown to result in

impaired insulin signaling, which can be defined as a state of insulin resistance. Saturated

fatty acids are TLR4 agonists, and a study by Shi et al. demonstrated that TLR4 is essential

for acute lipid-induced insulin resistance (110). While many studies have begun to determine

Page 56: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

45

Time (min)

Rela

tive p

38 M

AP

K P

ho

sp

ho

ryla

tio

n

5 15 30 600.0

0.5

1.0

1.5H2O

25 uM Palmitate

* mHypoA-NPY/GFP, 30 min

- +

p-

p38MAPK

Gbeta

A B

Fig. 3.4 Palmitate induces phosphorylation of p38 MAPK.

mHypoA-NPY/GFP neurons were treated with 25 µM palmitate (grey) or vehicle (black), and protein was harvested

after 5, 15, 30 or 60 minutes. Relative phospho-p38 MAPK was measured using Western Blot analysis and normalized

to Gβ, which served as a loading control. Results indicate that 25 µM palmitate significantly increased p38 MAPK

phosphorylation at 30 minutes (A). Representative blots are shown in B. Data are shown as mean values ± SEM (n=3

independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-way ANOVA with Tukey’s post-hoc test.

Page 57: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

46

the consequences of dyslipidemia in peripheral tissues, little is known about the effects of

excess lipids in the brain. Recently, Mayer et al. demonstrated that prolonged exposure to

palmitate impairs insulin activation in the mHypoE-44 hypothalamic neuronal cell model, as

assessed by phosphorylation of Akt (109). Whether palmitate has similar effects on insulin

signaling in representative NPY/AgRP neurons is unknown. To determine the effects of

prolonged palmitate exposure on neuronal insulin signaling, we pre-treated mHypoA-

NPY/GFP neurons with 25 µM palmitate for 24 hours. After palmitate pre-treatment, cells

were washed with PBS and subsequently serum-starved for one hour. Cells were then re-

challenged with 10 nM of insulin to determine neuronal responsiveness. Using Western Blot

analysis, it was determined that 24 hours of palmitate pre-treatment was sufficient to

significantly decrease insulin-mediated Akt phosphorylation in mHypoA-NPY/GFP neurons

(n=4; P<0.05; Fig. 3.5A).

Obesity is also associated with hyperinsulinemia (111). This increase in plasma

insulin levels is thought to be a contributing factor to the development and/or perpetuation of

insulin resistance. Therefore, to determine the effects of prolonged insulin exposure on

neuronal insulin signaling, we pre-treated mHypoA-NPY/GFP neurons with 100 nM insulin

for 24 hours. After insulin pre-treatment, cells were washed with PBS and subsequently

serum-starved for one hour. Cells were then re-challenged with 10 nM of insulin to

determine neuronal responsiveness. Using Western Blot analysis, it was determined that 24

hours of insulin pre-treatment was sufficient to significantly decrease insulin-mediated Akt

phosphorylation in mHypoA-NPY/GFP neurons (n=3; P<0.001; Fig. 3.5C). Collectively,

these data indicate that prolonged exposure to either palmitate or insulin impairs insulin

signaling in an NPY/AgRP neuronal cell model.

Page 58: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

47

Time (min)

Time (min)

pAKT/AKT, 15 min, mHypoA-NPY/GFP

25 uM Palmitate Pre-Treatment

pA

KT

/AK

T (

A.U

.)

- +0

1

2

3

4PBS

10 nM Insulin

*

pAKT/AKT, 15 min, mHypoA-NPY/GFP

100 nM Insulin Pre-Treatment

pA

KT

/AK

T (

A.U

.)

- +0

1

2

3PBS

10 nM Insulin

***

pAKT –

AKT –

Pre-Treatment

mHypoA-NPY/GFP, 15 min

- + - +

pAKT –

AKT –

Pre-Treatment

mHypoA-NPY/GFP, 15 min

- + - +

B A

C D

Fig. 3.5 Prolonged palmitate or insulin exposure dampens the insulin-mediated increase in phospho-

Akt.

Top: mHypoA-NPY/GFP neurons were pre-treated with 25 µM palmitate (+) or vehicle (-) for 24 hours. Cells were

then washed and placed in serum-free media. Following 1 hour serum starvation, cells were re-challenged with 10

nM of insulin (grey) or vehicle (black) for 15 minutes. Relative phospho-Akt was measured using Western Blot

analysis and normalized to total Akt. Results indicate that 25 µM palmitate pre-exposure significantly decreased

insulin-mediated phosphorylation of Akt at 15 minutes (A). Representative blots are shown in B. Data are shown as

mean values ± SEM (n=4 independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-way ANOVA with

Tukey’s post-hoc test.

Bottom: mHypoA-NPY/GFP neurons were pre-treated with 100 nM insulin (+) or vehicle (-) for 24 hours. Cells were

then washed and placed in serum-free media. Following 1 hour serum starvation, cells were re-challenged with 10

nM of insulin (grey) or vehicle (black) for 15 minutes. Relative phospho-Akt was measured using Western Blot

analysis and normalized to total Akt. Results indicate that 100 nM insulin pre-exposure significantly decreased

insulin-mediated phosphorylation of Akt at 15 minutes (C). Representative blots are shown in D. Data are shown as

mean values ± SEM (n=3 independent experiments). *P<0.05; **P<0.01; ***P<0.001, as per two-way ANOVA with

Tukey’s post-hoc test.

Page 59: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

48

Chapter 4

Discussion

Page 60: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

49

4. Discussion

4.1 General Discussion

Two functionally opposing neuronal populations in the arcuate nucleus of the

hypothalamus act as sensors for body energy stores and coordinate the activity of a complex

network of neurons that regulates feeding behavior and overall energy homeostasis. One

population expresses the orexigenic neuropeptides NPY and AgRP, while the other expresses

the anorexigenic POMC (α-MSH). These first-order neurons express receptors and have

intracellular molecular systems for detecting changes in the levels of hormones or nutrients

in the bloodstream (112). During fasting, the expression of NPY and AgRP are induced,

while POMC is inhibited. This coordinated response is in part the result of simultaneous

sensing of decreased nutrient availability and reduced levels of circulating insulin.

Conversely, in the postprandial state, NPY/AgRP neurons are inhibited and POMC neurons

are active. In this scenario, nutrient availability and insulin levels are increased. Therefore,

under normal physiological conditions, the hypothalamus acts under the control of peripheral

factors to maintain energy homeostasis (112).

When the balance is lost between food intake and energy expenditure, body adiposity

can no longer be maintained at a physiological level. This may lead to disturbances at the

hypothalamic level, which can cause metabolic disease. Excessive caloric intake is a primary

risk factor for the development of obesity, and a number of epidemiological studies have

shown that populations consuming diets high in fat are particularly prone to gaining weight

(55). According to Milanski et al., the long-chain saturated fatty acids are most harmful, and

can activate signal transduction through the toll-like receptor family leading to activation of

an inflammatory response in the hypothalamus (66). Another mechanism involved in the

Page 61: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

50

breakdown of energy homeostasis is the development of resistance to the anorexigenic

effects of insulin (59). In studying the molecular mechanism(s) underlying this resistance,

many groups found that hypothalamic inflammation led to activation of intracellular

signaling pathways that promote negative cross-talk with insulin signaling pathways (113).

While classical in vivo approaches have enhanced our basic understanding of energy

homeostasis and highlighted the importance of NPY/AgRP and POMC neurons in the

maintenance of whole body energy homeostasis, such approaches cannot establish the direct

actions of nutrients (particularly fatty acids) on signaling events or neuropeptide gene

expression in specific hypothalamic neurons. Therefore, the purpose of this study was to

investigate the effects of saturated fatty acid palmitate on inflammatory state, neuropeptide

gene expression and signal transduction events in a representative NPY/AgRP cell model.

Here, the mHypoA-NPY/GFP cell line was used.

4.2 Plasma Non-Esterified Fatty Acids and the Determination of Palmitic Acid

Concentrations in the Brain

More than 95% of all fatty acids are stored as triacylglycerol fatty acids within

adipose tissue. Adipose tissue releases these stored fatty acids via lipolysis into the

circulation, where NEFAs become the major circulating lipid fuel (114). Lipolysis is a highly

regulated process and generally provides the exact amount of lipids to fat-oxidizing tissues.

For example, physiological increases in plasma insulin can reduce plasma NEFA

concentrations from their normal concentration of approximately 500 µM to 10-20 µM (115).

Conversely, under conditions of fasting, plasma NEFAs may increase to concentrations in

excess of 3000 µM (116). Excess fatty acid availability, however, can lead to increased

Page 62: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

51

systemic NEFA release. This increase in systemic NEFA availability may contribute to the

adverse health consequences of obesity. Indeed, plasma concentrations of free fatty acids are

significantly elevated in obesity because 1) the enlarged adipose tissue mass releases more

FFA and 2) FFA clearance may be reduced (117). The saturated fatty acid palmitate (C16:0)

is one of the most pre-dominant NEFAs in the plasma of healthy individuals (118), and its

concentration nearly doubles in obese individuals.

In the brain, fatty acids serve both as an energy source and as components of

membrane lipids which maintain the structure and function of neuronal membranes. Fatty

acids are specifically sensed by hypothalamic neurons and, along with being involved in the

regulation of energy homeostasis, are required by the brain for regulation of hepatic glucose

production. Fatty acids cross the blood-brain barrier mainly by simple diffusion in the

unbound form, such that the concentration of fatty acids entering the brain is generally

proportional to their plasma concentration (119). However, longer chain fatty acids require a

facilitated uptake process to enter the cell. Fatty acid translocase (FAT/CD36) is one integral

membrane glycoprotein that has high affinity for long-chain fatty acids. This protein

mediates fatty acid dissociation from albumin and its “flip-flop” transport across the

phospholipid bilayer. Fatty acid binding proteins (FABPs) exist on the intracellular side of

the membrane, and provide additional means for the uptake of fatty acids.

In selecting a concentration of palmitate that could be used for treating the mHypoA-

NPY/GFP neurons, it was important to consider the physiological levels of palmitate that

would be seen by the brain. While little work has been done to determine exact

concentrations of palmitate in the brain, there are a handful of studies that enable us to make

a more accurate estimate. Given that the average total fatty acid concentration in the blood is

Page 63: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

52

approximately 500 µM (115) and that palmitate comprises ~30% of total plasma NEFA

(152), we can say that the average concentration of palmitate circulating in the plasma is 150

µM. Determining how much circulating palmitate actually enters the brain required further

investigation of the literature. In 2000, Smith and Nagura calculated that the single-pass

extraction of radiolabelled palmitate from blood into brain was approximately 2% (64).

Recent work by Karmi et al. suggests that brain fatty acid uptake of radiolabelled palmitate

may even be increased in obese individuals compared to healthy controls (120). Combined

with the observation that plasma palmitate concentrations nearly double in obese individuals,

we can estimate that at least 6 µM of palmitate enters the brain. Accounting for an increased

uptake of palmitate in obese individuals, this number may be even higher. Therefore, the 25

µM dose of palmitate used to treat the mHypoA-NPY/GFP neurons reflects a roughly

physiological concentration that may be seen by the brain.

As the accumulation of lipids may lead to a lipotoxic response in the mHypoA-

NPY/GFP neurons, it was also important to consider the toxicity of the dose used.

Accumulation of lipids in non-adipose tissues (such as the brain) can lead to cellular

dysfunction and/or death. This may be the result of stimulating apoptotic pathways or

initiating cellular stress responses in the endoplasmic reticulum (ER stress). Mayer et al.

demonstrated the lipotoxic effects of palmitate on hypothalamic neurons, where doses greater

than 0.2 mM were sufficient to activate the MAPK JNK pathway, EIF2 (an ER stress

marker) and caspase-3 (a common cell death effector) in neuronal cells (109). In this thesis,

however, the Bax to Bcl-2 mRNA ratio was used to determine the neurons’ susceptibility to

apoptosis upon treatment with different doses of palmitate. Bcl-2 is a 26-kD protein that

blocks programmed cell death without affecting cellular proliferation, while Bax is a protein

Page 64: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

53

that promotes apoptosis. Therefore, increases in the Bax to Bcl-2 ratio would indicate an

increase in susceptibility to apoptosis and vice-versa (121). Upon treatment with 50 µM

palmitate for 4 or 24 hours, there was a significant increase in the Bax to Bcl-2 ratio in

mHypoA-NPY/GFP neurons, suggesting an increase in susceptibility to apoptosis with this

dose. This assertion was confirmed by visual inspection using a light microscope, where

considerable cell death could be seen. However, treatment with 25 µM palmitate for 4 or 24

hours did not cause a significant change in the Bax to Bcl-2 ratio in mHypoA-NPY/GFP

neurons. This was one reason why the 25 µM palmitate dose was used for all subsequent

experiments. Interestingly, I could only treat my neuronal cell line with a maximum of 25

µM palmitate for 24 hours before cells began to die. In adipocytes, myocytes and β-cell lines,

investigators have used 500, 750 and 1000 µM doses of palmitate, respectively, for up to 48

hours (122-124). This implies that neurons may be more susceptible to the toxic nature of

saturated fatty acid palmitate.

4.3 Transcriptional Effects of Palmitate on mHypoA-NPY/GFP Neurons

While diet-induced obesity is associated with low-grade inflammation in peripheral

tissues, a similar process occurs in the hypothalamus. In 2005, evidence first emerged that

inflammatory changes could be detected in the brains of HFD-fed animals. In this study, 20

weeks of high-fat feeding increased reactive oxygen species and prostaglandin E2

production, along with upregulation of NF-κB signaling in the rat brain (58). In the

hypothalamus, De Souza et al. demonstrated that immune-related molecules (i.e. TNF-α, IL-

6) represent the largest class of genes with altered expression following an extended period

of high-fat feeding (59). Underlying this inflammatory response is the activation of both JNK

Page 65: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

54

(59) and the classic IKKβ/NF-κB pathway, and the induction of endoplasmic reticulum stress

(66).

Recent research has focused on determining how HFD induces hypothalamic

inflammation. One potential mechanism is the ability of saturated, but not unsaturated, fatty

acids to activate toll-like receptor 4 (TLR4)/NF-κB signaling. Indeed, Milanski et al. found

that long-chain saturated fatty acids exert the most potent inflammatory stimulus within the

hypothalamus and that this inflammation is dependent on TLR4 activation (66). Palmitate, a

16-carbon saturated fatty acid, is capable of inducing NF-κB signaling through TLR4

activation both in neuronal cell culture and following direct infusion into the brain (65).

Therefore, we wanted to determine whether palmitate could elicit such an inflammatory

response in the mHypoA-NPY/GFP cell line. To do this, we assessed relative changes in

mRNA levels of NF-κB and IκBα following treatment with 25 µM palmitate using qRT-

PCR. Treatment with 25 µM palmitate was sufficient to increase NF-κB and IκBα mRNA

levels by 2 and 4 hours, respectively. However, it is unlikely that these changes in NF-κB

and IκBα mRNA were mediated at the transcriptional level. More likely, these changes were

due to changes in mRNA stability. Indeed, palmitate has been shown to activate miRNAs in

peripheral tissues (134), and these miRNAs may regulate mRNA transcript stability.

However, this cannot be confirmed until an RNA stability assay is performed (see future

directions). Nonetheless, these results suggest that the classic IKKβ/NF-κB inflammatory

signaling pathway is conserved in the mHypoA-NPY/GFP cell line and becomes activated in

response to the saturated fatty acid palmitate. This acute increase in pro-inflammatory

markers by palmitate may underlie the early-onset hypothalamic inflammation seen in diet-

induced obesity.

Page 66: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

55

It is understood that fatty acids act on the central nervous system to modulate glucose

metabolism and overall energy homeostasis (52). For example, central administration of the

monounsaturated fatty acid oleate inhibits food intake and glucose production in rats (53).

These effects are accompanied by a decrease in expression of hypothalamic NPY after short-

term overfeeding, which suggests that small changes in plasma FA concentrations may serve

as a satiety signal to the CNS. Yet, little is known about the direct effects of fatty acids (such

as palmitate) on neuropeptide gene expression in specific hypothalamic neurons. Therefore,

we treated the mHypoA-NPY/GFP neurons with 25 µM palmitate over the course of 24

hours and assessed the regulation of AgRP mRNA levels using qRT-PCR. Treatment of

mHypoA-NPY/GFP neurons with 25 µM palmitate led to an increase in AgRP mRNA levels

at 24 hours. Unlike the changes in NF-κB and IκBα mRNA levels, the changes in AgRP

mRNA levels occurred after a longer time course. It is more likely, then, that the regulation

of AgRP mRNA levels by palmitate is mediated at the level of transcription. However, since

changes in mRNA levels may not accurately reflect changes in protein levels, AgRP protein

levels must be quantified in response to palmitate treatment before definitive conclusions can

be made. Regardless, this novel finding suggests that the saturated fatty acid palmitate

upregulates AgRP gene expression in the short-term, which may lead to increased feeding.

Indeed, this idea would fall in line with the observation that mice fed a high-fat diet tend to

have higher energy intake than normal-diet fed mice (125).

To begin to understand the mechanism(s) underlying palmitate’s acute regulation of

AgRP gene expression, we analyzed the AgRP gene promoter for potential transcription

factor (TF) binding sites using the online TF binding prediction tool Alibaba 2.1. The

program revealed potential binding sites for a host of transcription factors, including: NF-κB,

Page 67: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

56

CREB and Elk-1. Given palmitate’s ability to activate the IKKβ/NF-κB inflammatory

signaling pathway and NF-κB gene expression in mHypoA-NPY/GFP neurons, it is

reasonable to believe that palmitate may modulate AgRP gene expression directly through

activation of NF-κB. While palmitate has not previously been shown to activate CREB in the

hypothalamus, it has been shown to phosphorylate and activate CREB in primary

hepatocytes (126). In the study by Collins et al., CREB phosphorylation by palmitate was

blocked by the inhibition of p38 MAPK. Indeed, p38 MAPK activation can lead to indirect

activation of CREB through MSK1 and thus, palmitate may modulate AgRP gene expression

through this pathway of CREB activation in hypothalamic NPY/AgRP neurons. Finally, Elk-

1 is a transcription factor that can become activated downstream of ERK1/2, JNK and/or p38

MAPK activation. Each of these MAP kinases has been shown to be activated by palmitate in

different peripheral tissues, making Elk-1 a candidate transcription factor of the AgRP gene.

For example, palmitate was shown to increase intramyocellular diacylglycerol (DAG) and

induce phosphorylation/activation of the MEK-ERK-IKK axis in skeletal muscle (107). In

HepG2 cells, palmitate-induced hepatic insulin resistance was found to be mediated through

JNK and p38 MAPK pathways (108). Though little is known about how palmitate modulates

intracellular signaling in the brain, it is plausible that palmitate may regulate AgRP gene

expression through activation of any of these MAP kinases.

4.4 Transcriptional Effects of TNF-α on mHypoA-NPY/GFP Neurons

As stated previously, De Souza et al. found that immune related molecules (i.e. TNF-

α and IL-6) were the largest class of genes with altered hypothalamic gene expression

following an extended period of high-fat feeding (59). Long-chain saturated fatty acids, such

as palmitate, appear to be the most potent inflammatory stimulus in the hypothalamus and

Page 68: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

57

mediate their inflammatory effects via activation of the TLR4/NF-κB pathway. Downstream

targets of the transcription factor NF-κB include genes encoding for pro-inflammatory

cytokines, such as TNF-α. Cytokines (i.e. TNF-α) can then act as positive feedback signals to

enhance the inflammatory response by further activating inflammatory pathways through

receptors such as TNFR. Indeed, 50 ng/mL TNF-α significantly upregulated NF-κB, IκBα

and IL-6 mRNA levels within 4 hours of treatment in mHypoA-NPY/GFP neurons.

However, it is not known whether TNF-α (serving as a pro-inflammatory surrogate of

palmitate) can directly regulate AgRP gene expression. Therefore, mHypoA-NPY/GFP

neurons were treated with 50 ng/mL TNF-α for up to 24 hours and AgRP gene expression

was assessed using qRT-PCR. The results indicate that AgRP gene expression was

significantly increased by TNF-α at 4, 8 and 24 hours following treatment. Palmitate, being

able to upregulate NF-κB gene expression and its downstream targets (i.e. TNF-α), may thus

act to increase AgRP gene expression in mHypoA-NPY/GFP neurons by first upregulating

TNF-α gene expression.

4.5 Palmitate Metabolism and Signaling Dynamics in mHypoA-NPY/GFP neurons

While palmitate may indeed regulate AgRP gene expression through activation of the

TLR4/NF-κB pathway in mHypoA-NPY/GFP neurons, its effects may also be mediated by

its metabolites. The hypothalamus receives and processes nutrients that reflect the energy

status of the individual, and these “signals” can trigger the expression and secretion of

orexigenic and anorexigenic neuropeptides that regulate food intake and energy expenditure.

The idea that an intermediate of fatty acid metabolism could play a role in the regulation of

energy homeostasis has been supported by findings brought forth in previous studies. For

example, DIO mice treated with myriocin (an inhibitor of de novo ceramide synthesis) show

Page 69: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

58

a complete reversal of glucose intolerance and peripheral insulin resistance (106).

Furthermore, hypothalamic CPT1 inhibition leads to increased intracellular levels of long-

chain fatty acyl CoAs, inhibition of feeding and reduced expression of ARC NPY and AgRP

(54). Yet, whether fatty acid metabolism plays a role in palmitate’s regulation of AgRP gene

expression is unknown. Therefore, to determine whether palmitate-mediated regulation of

AgRP gene expression is palmitate metabolism-dependent, mHypoA-NPY/GFP neurons

were treated with non-metabolizable methyl palmitate. Treating the mHypoA-NPY/GFP

neurons with different doses of methyl palmitate (25, 50 and 75 µM) did not result in a

similar upregulation in AgRP gene expression at 24 hours as seen following 25 µM palmitate

treatment. This finding suggests that palmitate-mediated regulation of AgRP gene expression

is palmitate metabolism-dependent.

Ceramide, a sphingolipid metabolite of palmitate, is one candidate mediator of

palmitate-mediated regulation of AgRP gene expression. A study by Ramirez et al. examined

the importance of ceramide synthesis to the orexigenic actions of ghrelin in terms of NPY

and AgRP gene expression. It was found that central inhibition of ceramide synthesis with

myriocin blocked the orexigenic actions of ghrelin and normalized levels of both NPY and

AgRP (127). Moreover, ceramide has been shown to activate NF-κB in human colon

epithelial HT-29 cells (135). This activation of NF-κB by ceramide may lead to upregulation

of TNF-α gene expression, which (as described in the previous section) may lead to changes

in AgRP gene expression. Nonetheless, it remains to be seen whether ceramides are involved

in palmitate-mediated regulation of AgRP gene expression. Future studies will involve the

treatment of mHypoA-NPY/GFP neurons with C16 ceramide to determine its direct effects

(if any) on AgRP gene expression.

Page 70: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

59

In the periphery, palmitate has been shown to mediate its metabolic effects through

activation of ERK1/2, JNK and/or p38 MAPK. For example, Guo et al. demonstrated that

palmitate could induce apoptosis in both mouse 3T3-L1 and rat primary preadipocytes. In

this study, treatment of preadipocytes with palmitate induced ER stress responses and

increased the phosphorylation/activation of JNK and ERK1/2 (128). Similarly, palmitate

treatment induced ER stress through a JNK-dependent pathway in mHypoE-44 neurons

(109). In HepG2 cells, pamitate-induced hepatic insulin resistance was found to be mediated

through JNK and p38 MAPK pathways (108). Yet, it remains to be seen whether any of the

aforementioned pathways become activated by palmitate in the brain. Therefore, to assess the

signaling dynamics of palmitate in mHypoA-NPY/GFP neurons, we treated the cells with

palmitate for up to 1 hour and assessed the phosphorylation status of ERK1/2, JNK and p38

MAPK by Western Blot. The results indicate that palmitate increased phosphorylation of p38

MAPK, but not ERK1/2 or JNK. Closer examination reveals slight variations in control

(basal) levels of p38 MAPK over the course of this one hour time period, and this may be the

result of circadian clock-dependent oscillations in p38 MAPK activation. Indeed, Goldsmith

observed rhythmic p38 MAPK activation in cells derived from the suprachiasmatic nucleus

and fibroblasts of a mouse. However, in cells that lacked a functional circadian oscillator,

this rhythmic activation in p38 MAPK was lost and overall levels of p38 protein were lower

(137). Additionally, basal levels of p38 MAPK can change with movement. For this reason,

treatments were administered within the incubator in order to avoid excessive movement of

the plates.

Ultimately, the phosphorylation and activation of p38 MAPK is notable as the

transcription factor CREB can be indirectly activated downstream of p38 MAPK activation

Page 71: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

60

(as described earlier). That palmitate induces phosphorylation of p38 MAPK only further

supports the notion that CREB may be a downstream mediator of palmitate’s regulation of

AgRP gene expression. Whether palmitate’s regulatory actions on AgRP gene expression

require p38 MAPK activation, however, remains to be elucidated (see future directions).

4.6 Palmitate Impairs Insulin Signaling in mHypoA-NPY/GFP Neurons

Central insulin action is critical for normal energy homeostasis. The orexigenic

NPY/AgRP and anorexigenic POMC neurons are responsive to insulin, and the coordinated

regulation of these two populations of neurons helps to maintain normal energy homeostasis.

However, many obese individuals develop insulin resistance and subsequent type 2 diabetes

mellitus. The excess circulating lipids found in obese individuals is a major contributing

factor to this insulin resistant state. One mechanism mediating this insulin resistance involves

serine phosphorylation and inhibition of insulin receptor substrate-1 (IRS-1). Saturated fatty

acid activation of IKKβ/NF-κB can lead to insulin resistance through this mechanism as

IKKβ becomes activated and serine phosphorylates IRS-1 (129). Serine phosphorylation of

IRS-1 can also occur through palmitate-induced activation of PKCθ (130). Moreover,

palmitate metabolites play a role, as ceramide can activate protein phosphatase 2A which

dephosphorylates pAKT and further contributes to this insulin resistant state (96). Therefore,

we wanted to assess the impact of palmitate on insulin signaling in the mHypoA-NPY/GFP

cell line. Our data suggest that prolonged palmitate pre-treatment (24 hours) significantly

dampens Akt phosphorylation in mHypoA-NPY/GFP neurons after 15 minutes of insulin re-

challenge. Indeed, the impairment of normal insulin signaling in these hypothalamic neurons

may have significant physiological impact, as it could lead to a dysregulation of neuropeptide

gene expression (AgRP, NPY) through impaired FoxO1 activity. Additionally, amino acids

Page 72: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

61

were found to inhibit AgRP gene expression through an mTOR-dependent mechanism in the

GT1-7 hypothalamic cell line. By inhibiting mTOR signaling with rapamycin, cells exposed

to even high amino acid levels responded by increasing AgRP mRNA levels (138). Thus, it is

possible that dampening of Akt phosphorylation could also lead to a dampening of mTOR

activation. This may relieve the mTOR-dependent inhibition of AgRP gene expression,

contributing to further increases in AgRP gene expression.

Hyperinsulinemia is another hallmark feature of obesity. A study by Genuth found

that obese persons exhibited three fold increases in both fasting and postprandial insulin

levels (131). This increase in basal plasma insulin levels is thought to be a contributing factor

to the development or perpetuation of insulin resistance. It is likely, then, that continual

exposure of hypothalamic neurons to high levels of insulin may cause or contribute to the

development of an insulin resistant state. Therefore, we assessed the impact of prolonged

insulin exposure on insulin signaling in the mHypoA-NPY/GFP cell line. Our data suggest

that, like prolonged palmitate pre-treatment, lengthy insulin pre-exposure significantly

dampens Akt phosphorylation in mHypoA-NPY/GFP neurons.

4.8 Limitations

The hypothalamus consists of a multitude of fully differentiated neuronal cell types

regulating many bodily functions, including energy homeostasis. Hypothalamic neurons

express neuropeptides and may be controlled by both internal and external inputs. Due to the

complex nature of the hypothalamus, mechanistic studies of neuropeptide gene regulation

and elucidation of signaling events in these neurons is difficult to perform in vivo. Moreover,

classical in vivo approaches cannot be used to investigate the direct effects of a particular

Page 73: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

62

agent on specific hypothalamic neuronal subtypes, largely due to the number of inputs

received from afferent neurons. Yet, cell lines immortalized from primary culture provide an

individualized model that lacks the complexity and integrated network of neuronal inputs

which makes studying these neurons difficult in vivo. Additional benefits of immortalized

cell lines include the fact that they can be maintained in a relatively controlled environment

with fewer uncontrolled variables, and different neuronal phenotypes can be used to

investigate the expression of specific genes or proteins (132). However, the use of

immortalized hypothalamic cell lines also has its limitations.

Although cell lines allow for a closer examination of the molecular processes at work

within a cell, the immortalizing gene SV40 T-antigen can interfere with these same cellular

processes. This may affect the pathways being studied. Initial studies conducted in our

laboratory using short-hairpin RNA knockdown of SV40 T-antigen demonstrate that T-

antigen expression can increase the basal activity of intracellular signaling kinases such as

Akt, Jak2 and STAT3 (Belsham, unpublished data). Therefore, I have attempted to reduce

the basal activity of such kinases using a combination of serum starvation and low glucose

during experimentation.

Another inherent limitation of this study is the physiological relevancy of the doses

used for treatments. In this thesis, the doses of TNF-α and palmitate used were chosen

primarily based on their degree of toxicity such that apoptosis was minimized in the

mHypoA-NPY/GFP neurons. Indeed, little research has been done to determine typical

concentrations of TNF-α or palmitate in the hypothalamus of obese versus healthy

individuals. While we can make estimates based on currently available literature regarding

the topic, it is still difficult to select physiologically (or pathologically) accurate doses for

Page 74: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

63

treatments. Moreover, this study did not address the effects of other fatty acids (specifically

unsaturated fatty acids) on neuropeptide gene expression in mHypoA-NPY/GFP neurons.

Cintra et al. found that substitution of oleic acid (C18:1) into the diet of DIO mice was

sufficient to correct hypothalamic inflammation, hypothalamic insulin resistance and body

adiposity in these animals (136). How these fatty acids influence AgRP gene expression and

either potentiate or dampen palmitate’s effects on AgRP gene expression in mHypoA-

NPY/GFP neurons remain unknown.

Additionally, a long period of continuous passaging may change the cell

characteristics such that they become phenotypically different from the single cell population

of lower passage cells (132). Therefore, cell phenotype was continually monitored and a

maximum of 30 passages was performed. Furthermore, as with all in vitro studies, there are

limitations to their translatability to the in vivo setting. Immortalized neuronal cell models are

removed from the complex architecture and afferent cellular connections present in the intact

brain. Thus, it is challenging to account for the interneuronal communications within the

hypothalamus that are known to play important roles in feeding behavior and energy

homeostasis. For this reason, conclusions based on the results found here cannot be

generalized to all neurons or the hypothalamus itself.

While methyl palmitate was used to determine whether intracellular metabolism of

palmitate was required for palmitate-mediated regulation of AgRP gene expression, the

functional aspects of fatty acid methyl esters still remain largely unknown. Currently, it is

difficult to detect the localization of a specific fatty acid due to the lack of specific

radioactive or antibody labeling for a particular fatty acid chain. Thus, the only currently

available method of detecting the presence of fatty acid methyl esters is by using mass

Page 75: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

64

spectrometry. However, this cannot be used to directly determine fatty acid localization

within specific tissue types. As such, whether methyl palmitate actually enters the cell cannot

be firmly established. Nonetheless, it is believed that the esterification of palmitate by a

methyl group in methyl palmitate prevents its activation by CoA. Lack of CoA activation

therefore abrogates any downstream metabolism of the fatty acid (150). Yet, recent literature

has suggested a possible anti-inflammatory role for methyl palmitate in different

experimental rat models (151). In light of these findings, the use of methyl palmitate may be

insufficient. Future studies will utilize another nonmetabolizable analog of palmitate, 2-

bromopalmitate, to further confirm that palmitate-mediated changes in AgRP gene

expression are palmitate metabolism-dependent.

Despite these limitations, the value of immortalized hypothalamic cell lines cannot be

underestimated. History demonstrates that cell models are reflective of natural physiology.

Importantly, the information gained from the study of these cell models will lead to a more

focused approach in the whole animal.

4.8 Future Directions

The thesis herein outlines the direct effects of saturated fatty acid palmitate on AgRP

gene expression. In addition, the experiments presented demonstrate palmitate’s ability to

induce a state of inflammation, phosphorylate/activate p38 MAPK and impair insulin

signaling in a hypothalamic neuronal cell model representative of NPY/AgRP neurons.

However, key questions remain. For one, whether palmitate-mediated changes in NF-κB and

IκBα mRNA levels are due to changes in mRNA stability remains unknown. Future studies

may employ an RNA stability assay to assess the stability of such mRNAs following

Page 76: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

65

palmitate treatment. Moreover, the exact palmitate metabolite(s) that act to regulate AgRP

gene expression in mHypoA-NPY/GFP neurons have yet to be determined. Therefore, future

studies will utilize inhibitors for β-oxidation (etomoxir), protein palmitoylation (2-

bromopalmitate) and de novo ceramide synthesis (myriocin and/or fumonisin B1) to

determine which metabolite(s) may be mediating palmitate’s effects on AgRP gene

expression.

Additionally, while palmitate was found to induce the phosphorylation/activation of

p38 MAPK in mHypoA-NPY/GFP neurons, it was not determined whether this activation

was necessary for palmitate-mediated regulation of AgRP gene expression. Future studies

will therefore employ the use of a p38 inhibitor (such as SB239063) prior to palmitate

treatment to determine whether p38 MAPK activation is required for downstream regulation

of AgRP gene expression. In pursuing the idea that p38 MAPK activation may lead to

activation of the transcription factor CREB as a means of altering AgRP gene expression,

experiments will also be performed to: 1) determine the phosphorylation status of CREB

following palmitate treatment in mHypoA-NPY/GFP neurons and 2) utilize an inhibitor of

CREB prior to palmitate treatment to determine the effect of such inhibition on AgRP gene

expression.

Finally, the presence of functional GPR120 and adequate omega-3 fatty acids for its

activation is sufficient to lower systemic inflammation in mice (133). Likewise, genetic

disruption of GPR120 eliminates the ability of dietary omega-3 FAs to improve energy

homeostasis in obese mice (133). The anti-inflammatory properties of GPR120 activation by

omega-3 FAs have been demonstrated in adipocytes and macrophages, but little is known

about their actions in other areas of the body. Therefore, it would be of interest to examine

Page 77: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

66

the effects of DHA pre-treatment on palmitate-mediated inflammation in mHypoA-

NPY/GFP neurons. Given the association between inflammation and insulin resistance,

additional studies will also be conducted to determine whether DHA pre-treatment can

prevent palmitate-mediated induction of insulin resistance.

4.9 Conclusion

The current thesis addresses the direct effects of the saturated fatty acid palmitate on

neuropeptide gene expression, signal transduction and insulin signaling in an immortalized,

hypothalamic cell model. Herein, palmitate has been shown to upregulate gene expression of

certain pro-inflammatory markers and AgRP in the mHypoA-NPY/GFP cell line. While

current literature has focused primarily on palmitate’s ability to activate TLR4/NF-κB

signaling, it was found that palmitate-mediated regulation of AgRP gene expression was

palmitate metabolism-dependent in the mHypoA-NPY/GFP neurons. Moreover, we provide

the first evidence that palmitate induces the phosphorylation/activation of p38 MAPK in a

distinct population of hypothalamic neurons. Finally, lengthy pre-exposure to palmitate or

insulin leads to impairment of normal insulin signaling in our cell model.

Physiologically, these findings may provide a molecular basis through which high fat

diets elicit their negative effects on feeding and overall energy homeostasis. Indeed, high fat

diet-induced obesity leads to increased feeding in mice over the long term, and this may be

due to continual upregulation of AgRP gene expression in NPY/AgRP neurons by saturated

fatty acids consumed in the diet. The finding that palmitate impairs insulin signaling in these

same neurons may only exacerbate the resultant increase in feeding. In beginning to elucidate

the signaling mechanisms through which palmitate acts in representative NPY/AgRP

Page 78: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

67

neurons, we may also begin to identify potential therapeutic targets for the treatment of

obesity.

Taken together, these results add to our understanding of how fatty acids modulate

energy homeostasis within the hypothalamus. The knowledge gained through the analysis of

the mHypoA-NPY/GFP cell line will complement in vivo studies, and lead to a better

understanding of NPY/AgRP neuronal function in metabolic disorders such as obesity. This

study also indicates the importance of further neuronal cell line studies to better understand

the mechanism(s) underlying palmitate’s effects on neuropeptide gene expression and central

insulin signaling.

Page 79: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

68

References

1. Schwartz MW, Baskin DG, Kaiyala KJ, Woods SC. Model for the regulation of

energy balance and adiposity by the central nervous system. Am J Clin Nutr.

[1999;69(4):584-596.

2. Drenick EJ, Johnson D. Weight reduction by fasting and semistarvation in morbid

obesity: long-term follow-up. Int J Obes. [1978;2:123–32.

3. Garner DM, Wooley SC. Confronting the failure of behavorial and dietary treatments

for obesity. Clin Psychol Rev. [1991;11:729–80.

4. Bernstein IL, Lotter EC, Kulkosky PJ. Effect of force-feeding upon basal insulin

levels in rats. Proc Soc Exp Biol Med. [1975;150:546–8.

5. Morton GJ, Schwartz MW. The NPY/AgRP neuron and energy homeostasis. Int J

Obes Relat Metab Disord. [2001;25(Suppl 5):S56-62.

6. Woods SC, D’Alessio DA. Central control of body weight and appetite. J Clin

Endocrinol Metab. [2008;93(11 Suppl 1):S37-50.

7. Konner AC, Klockener T, Bruning JC. Control of energy homeostasis by insulin and

leptin: targeting the arcuate nucleus and beyond. [2009;97(5):632-638.

8. Porte DJ, Woods SC. Regulation of food intake and body weight in insulin.

Diabetologia. [1981;20 Suppl:274-280.

9. Polonsky KS, Given BD, Van Cauter E. Twenty-four-hour profiles and pulsatile

patterns of insulin secretion in normal and obese subjects. J Clin Invest.

[1988;81:442-448.

10. Kahn CR. Banting lecture: insulin action, diabetogenes, and the cause of type II

diabetes. Diabetes. [1994;43:1066-1084.

11. White MF. Insulin signaling in health and disease. Science. [2003;302:1710-1711.

12. Plum L, Belgardt BF, Bruning JC. Central insulin action in energy and glucose

homeostasis. J Clin Invest. [2006;116:1761-1766.

13. Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, Mendelsohn

FA. Localization and characterization of insulin receptors in rat brain and pituitary

gland using in vitro autoradiography and computerized densitometry. Endocrinology.

[1987;121:1562-1570.

14. Corp ES, Woods SC, Porte D, Jr., Dorsa DM, Figlewicz DP, Baskin DG. Localization

of 125I-insulin binding sites in the rat hypothalamus by quantitative autoradiography.

Neurosci Lett. [1986;70:17-22.

15. Banks WA. Blood-brain-barrier and energy balance. Obesity. [2006;14:234S-237S.

Page 80: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

69

16. Burbach JPH. Neuropeptides from concept to online database www.neuropeptides.nl.

Eur J Pharmacol. [2010;626:27-48.

17. Parker JA, Bloom SR. Hypothalamic neuropeptides and the regulation of appetite.

Neuropharmacology. [2012;63:18-30.

18. Hetherington AW, Ranson SW. Hypothalamic lesions and adiposity in the rat. Anat

Rec. [1940;78:149-172.

19. Gao Q, Horvath TL. Neurobiology of feeding and energy expenditure. Annu Rev

Neurosci. [2007;30:367-398.

20. Benoit S, Schwartz M, Baskin D, Woods SC, Seeley RJ. CNS melanocortin system

involvement in the regulation of food intake. Horm. Behav. [2000;37:299–78.

21. Obici S, Rossetti L. Minireview: nutrient sensing and the regulation of insulin action

and energy balance. Endocrinology. [2003;144:5172–78.

22. Cone RD. Anatomy and regulation of the central melanocortin system. Nat Neurosci.

[2005;8:571-578.

23. Chronwall BM, DiMaggio DA, Massari VJ, Pickel VM, Ruggiero DA, O’Donohue

TL. The anatomy of neuropeptide-Y-containing neurons in rat brain. Neuroscience.

[1985;15:1159-1181.

24. Morris BJ. Neuronal localisation of neuropeptide Y gene expression in rat brain. J

Comp Neurol. [1989;290:358-368.

25. Broberger C, Johansen J, Johansson C, Schalling M, Hokfelt T. The neuropeptide

Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and

monosodium glutamate-treated mice. Proc Natl Acad Sci USA. [1998;95:15043-

15048.

26. Roseberry AG, Liu H, Jackson AC, Cai X, Friedman JM. Neuropeptide Y-mediated

inhibition of proopiomelanocortin neurons in the arcuate nucleus shows enhanced

desensitization in ob/ob mice. Neuron. [2004;41:711-722.

27. Cabrele C, Langer M, Bader R, Wieland HA, Doods HN, Zerbe O, Beck-Sickinger

AG. The first selective agonist for the neuropeptide YY5 receptor increases food

intake in rats. J Biol Chem. [2000;275:36043-36048.

28. Kask A, Rago L, Harro J. Evidence for involvement of neuropeptide Y receptors in

the regulation of food intake: studies with Y1-selective antagonist BIBP3226. Br J

Pharmacol. [1998;124:1507-1515.

29. Rossi M, Kim MS, Morgan DG, Small CJ, Edwards CM, Sunter D, Abusnana S,

Goldstone AP, Russell SH, Stanley SA, Smith DM, Yagaloff K, Ghatei MA, Bloom

SR. A C-terminal fragment of Agouti-related protein increases feeding and

antagonizes the effect of alpha-melanocyte stimulating hormone in vivo.

Endocrinology. [1998;139:4428-4431.

Page 81: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

70

30. Kim MS, Rossi M, Abusnana S, Sunter D, Morgan DG, Small CJ, Edwards CM,

Heath MM, Stanley SA, Seal LJ, Bhatti JR, Smith DM, Ghatei MA, Bloom SR.

Hypothalamic localization of the feeding effect of agouti-related peptide and alpha-

melanocyte-stimulating hormone. Diabetes. [2000;49:177-182.

31. Ollmann MM, Wilson BD, Yang YK, Kerns JA, Chen Y, Gantz I, Barsh GS.

Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related

protein. Science. [1997;278:135-138.

32. Nijenhuis WA, Oosterom J, Adan RA. AgRP (83-132) acts as an inverse agonist on

the human-melanocortin-4 receptor. Mol Endocrinol. [2001;15:164-171.

33. Kitamura T, Feng Y, Kitamura YI, Chua SC Jr, Xu AW, Barsh GS, Rossetti L, Accili

D. Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake.

Nat Med. [2006;12: 534-540.

34. Vogt MC, Bruning JC. CNS insulin signaling in the control of energy homeostasis

and glucose metabolism – from embryo to old age. Trends Endocrinol Metab.

[2013;24:76-84.

35. Plum L, Belgardt BF, Bruning JC. Central insulin action in energy and glucose

homeostasis. J Clin Invest. [2006;116:1761-1766.

36. Stanley BG, Kyrkouli S, Lampert S, Leibowitz S. Neuropeptide Y chronically

injected into the hypothalamus: a powerful neurochemical inducer of hyperphagia and

obesity. Peptides. [1986;7:1189-1192.

37. Hagan MM, Rushing PA, Pritchard LM, Schwartz MW, Strack AM, Van Der Ploeg

LH, Woods SC, Seeley RJ. Long-term orexigenic effects of AgRP-(83-132) involve

mechanisms other than melanocortin receptor blockade. Am J Physiol Regul Integr

Comp Physiol. [2000;279:R47-52.

38. Makimura H, Mizuno TM, MAstaitis JW, Agami R, Mobbs CV. Reducing

hypothalamic AGRP by RNA interference metabolic rate and decreases body weight

without influencing food intake. BMC Neurosci. [2002;3:18.

39. Gropp E, Shanabrough M, Borok E, Xu AW, Janoschek R, Buch T, Plum L,

Balthasar N, Hampel B, Waisman A, Barsh GS, Horvath TL, Bruning JC. Agouti-

related peptide-expressing neurons are mandatory for feeding. Nat Neurosci.

[2005;8:1289-1291.

40. Luquet S, Perez FA, Hnasko TS, Palmiter RD. NPY/AgRP neurons are essential for

feeding in adult mice but can be ablated in neonates. Science. [2005;310:683-685.

41. Dole VP. A relation between non-esterified fatty acids in plasma and the metabolism

of glucose. J Clin Invest. [1956;35:150-154.

Page 82: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

71

42. Gordon RS Jr. Unesterified fatty acid in human blood plasma. II. The transport

function of unesterified fatty acid. J Clin Invest. [1957;36:810-815.

43. Eaton RP, Berman M, Steinberg D. Kinetic studies of plasma free fatty acid and

triglyceride metabolism in man. J Clin Invest. [1969;48:1560–1579.

44. Veerkamp JH, Zimmerman AW. Fatty acid-binding proteins of nervous tissue. J Mol

Neurosci. [2001;16:133-142.

45. Lafontan M, Langin D. Lipolysis and lipid mobilization in human adipose tissue.

Prog Lipid Res. [2009;48:275-297.

46. Evans K, Burdge GC, Wootton SA, Clark ML, Frayn KN. Regulation of dietary fatty

acid entrapment in subcutaneous adipose tissue and skeletal muscle. Diabetes.

[2002;51:2684-2690.

47. Heimberg M, Dunn GD, Wilcox G. The derivation of plasma-free fatty acids from

dietary neutral fat in man. J Lab Clin Med. [1974;83:393-402.

48. Karpe F, Dickmann JR, Frayn KN. Fatty acids, obesity, and insulin resistance: time

for a reevaluation. Diabetes. [2011;60:2441-2449.

49. Hamilton JA, Johnson RA, Corkey B, Kamp F. Fatty acid transport; the diffusion

mechanism in model and biological membranes. J Mol Neurosci. [2001;16:99-108.

50. Abumrad N, Coburn C, Ibrahimi A. Membrane proteins implicated in long-chain fatty

acid uptake by mammalian cells: CD36, FATP and FABPm. Biochim Biophys Acta.

[1999;1441:4-13.

51. Watkins PA. Fatty acid activation. Prog Lipid Res. [1997;36:55-83.

52. Lam TK, Schwartz GJ, Rossetti L. Hypothalamic sensing of fatty acids. Nat

Neurosci. [2005;8:579-584.

53. Obici S, Feng Z, Morgan K, Stein D, Karkanias G, Rossetti L. Central administration

of oleic acid inhibits glucose production and food intake. Diabetes. [2002;51:271-

275.

54. Obici S, Feng Z, Arduini A, Conti R, Rossetti L. Inhibition of hypothalamic carnitine

palmitoyltransferase-1 decreases food intake and glucose production. Nat Med.

[2003;9:756-761.

55. Damiao R, Castro TG, Cardoso MA, Gimeno SG, Ferreira SR, Japanese-Brazilian

Diabetes Study Group. Dietary intakes associated with metabolic syndrome in a

cohort of Japanese ancestry. Br J Nutr. [2006;96:532-538.

Page 83: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

72

56. Thaler JP, Schwartz MW. Minireview: inflammation and obesity pathogenesis: the

hypothalamus heats up. Endocrinology. [2010;151:4109-15.

57. Fessler MB, Rudel LL, Brown JM. Toll-like receptor signaling links dietary fatty

acids to the metabolic syndrome. Curr Opin Lipidol. [2009;20:379-385.

58. Zhang X, Dong F, Ren J, Driscoll MJ, Culver B. High dietary fat induces NADPH

oxidase-associated oxidative stress and inflammation in rat cerebral cortex. Exp

Neurol. [2005;191:318-325.

59. De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC, Saad

MJ, Velloso LA. Consumption of a fat-rich diet activates a proinflammatory response

and induces insulin resistance in the hypothalamus. Endocrinology. [2005;146:4192-

4199.

60. Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D. Hypothalamic IKKβ/NF-kB

and ER stress link overnutrition to energy imbalance and obesity. Cell. [2008;135:61-

73.

61. Posey KA, Clegg DJ, Printz RL, Byun J, Morton GJ, Vivekanandan-Giri A,

Pennathur S, Baskin DG, Heinecke JW, Woods SC, Schwartz MW, Niswender KD.

Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin

resistance in rats fed a high-fat diet. Am J Physiol Endocrinol Metab.

[2009;296:E1003-1012.

62. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava

F, Tuohy KM, Chabo C, Waget A, Delmee E, Cousin B, Sulpice T, Chamontin B,

Ferrieres J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, Burcelin R.

Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes.

[2007;56:1761-1772.

63. Milanski M, Arruda AP, Coope A, Ignacio-Souza LM, Nunez CE, Roman EA,

Romanatto T, Pascoal LB, Caricilli AM, Torsoni MA, Prada PO, Saad MJ, Velloso

LA. Inhibition of hypothalamic inflammation reverses diet-induced insulin resistance

in the liver. Diabetes. [2012;61:1455-1462.

64. Smith QR, Nagura H. Fatty acid uptake and incorporation in brain: studies with the

perfusion model. J Mol Neurosci. [2001;16:167-172.

65. Kleinridders A, Schenten D, Konner AC, Belgardt BF, Mauer J, Okamura T,

Wunderlich FT, Medzhitov R, Bruning JC. MyD88 signaling in the CNS is required

for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell

Metab. [2009;10:249-259.

Page 84: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

73

66. Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, Tsukumo DM,

Anhe G, Amaral ME, Takahashi HK, Curi R, Oliveira HC, Carvalheira JB, Bordin S,

Saad MJ, Velloso LA. Saturated fatty acids produce an inflammatory response

predominantly through the activation of TLR4 signaling in hypothalamus:

implications for the pathogenesis of obesity. J Neurosci. [2009;29:359-370.

67. Centers for Disease Control and Prevention. National diabetes fact sheet: national

estimates and general information on diabetes and prediabetes in the United States,

2011. Atlanta, GA, U.S. Department of Health and Human Services, Centers for

Disease Control and Prevention, 2011.

68. Zimmet P, Alberti KG, Shaw J. Global and societal implications of the diabetes

epidemic. Nature. [2001;414:782-787.

69. Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease.

Diabetes. [1988;37:1595-1607.

70. Ioannidis I. The road from obesity to type 2 diabetes. Angiology. [2008;59:39S-43S.

71. Lazar MA. How obesity causes diabetes: not a tall tale. Science. [2005;307:373-375.

72. Rajala MW, Scherer PE. Minireview: the adipocyte – at the crossroads of energy

homeostasis, inflammation, and atherosclerosis. Endocrinology. [2003;144:3765-

3773.

73. Ye J. Role of insulin in the pathogenesis of free fatty acid-induced insulin resistance

in skeletal muscle. Endocr Metab Immune Disord Drug Targets. [2007;7:65-74.

74. Rizza RA, Mandarino LJ, Genest J, Baker BA, Gerich JE. Production of insulin

resistance by hyperinsulinaemia in man. Diabetologia. [1985;28:70-75.

75. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM.

Diabetes. [1997;46:3-10.

76. Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y, Roth J. Insulin resistance and

hyperinsulinemia: is hyperinsulinemia the cart or the horse? Diabetes Care.

[2008;31:S262-S268.

77. Clegg DJ, Gotoh K, Kemp C, Wortman MD, Benoit SC, Brown LM, D’Alessio D,

Tso P, Seeley RJ, Woods SC. Consumption of a high-fat diet induces central insulin

resistance independent of adiposity. Physiol Behav. [2011;103:10-16.

78. Benoit SC, Kemp CJ, Elias CF, Abplanalp W, Herman JP, Migrenne S, Lefevre AL,

Cruciani-Guglielmacci C, Magnan C, Yu F, Niswender K, Irani BG, Holland WL,

Page 85: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

74

Clegg DJ. Palmitic acid mediates hypothalamic insulin resistance by altering PKC-

theta subcellular localization in rodents. J Clin Invest. [2009;119:2577-2589.

79. Howard JK, Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins.

Trends Endocrinol Metab. [2006;17:365-371.

80. Hotamisligil GS. Inflammation and metabolic disorders. Nature. [2006;444:860-867.

81. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c-Jun NH(2)-terminal

kinasepromotes insulin resistance during association with insulin receptor substrate-1

and phosphorylation of Ser(307). J Biol Chem. [2000;275:9047-9054.

82. Zabolotny JM, Kim YB, Welsh LA, Kershaw EE, Neel BG, Kahn BB. Protein-

tyrosine phosphatase 1B expression is induced by inflammation in vivo. J Biol Chem.

[2008;283:14230-14241.

83. Bence KK, Delibegovic M, Xue B, Gorgun CZ, Hotamisligil GS, Neel BG, Kahn BB.

Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med.

[2006;12:917-924.

84. Oakes ND, Furler SM. Evaluation of free fatty acid metabolism in vivo. Ann N Y

Acad Sci. [2002;967:158-175.

85. McGarry JD. What if Minkowski had been ageusic? An alternative angle on diabetes.

Science. [1992;258:766-770.

86. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA. Lipid

mediators of insulin resistance. Nutr Rev. [2007;65:S39-S46.

87. Briaud I, Harmon JS, Kelpe CL, Segu VB, Poitout V. Lipotoxicity of the pancreatic

beta-cell is associated with glucose-dependent esterification of fatty acids into neutral

lipids. Diabetes. [2001;50:315-321.

88. Neschen S, Morino K, Hammond LE, Zhang D, Liu ZX, Romanelli AJ, Cline GW,

Pongratz RL, Zhang XM, Choi CS, Coleman RA, Shulman GI. Prevention of hepatic

steatosis and hepatic insulin resistance inmitochondrial acyl-CoA:glycerol-sn-3-

phosphate acyltransferase 1 knockoutmice. Cell Metab. [2005;2:55-65.

89. Adams JM, 2nd

, Pratipanawatr T, Berria R, Wang E, DeFronzo RA, Sullards MC,

Mandarino LJ. Ceramide content is increased in skeletal muscle from obese insulin-

resistant humans. Diabetes. [2004;53:25-31.

90. Summers SA, Garza LA, Zhou H, Birnbaum MJ. Regulation of insulin-stimulated

glucose transporter GLUT4 translocation and Akt kinase activity by ceramide. Mol

Cell Biol. [1998;18:5457-5464.

Page 86: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

75

91. Paz K, Hemi R, LeRoith D, Karasik A, Elhanany E, Kanety H, Zick Y. A molecular

basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and

IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and

impairs their ability to undergo insulin-induced tyrosine phosphorylation. J Biol

Chem. [1997;272:29911-8.

92. Ruvolo PP. Intracellular signal transduction pathways activated by ceramide and its

metabolites. Pharmacol Res. [2003;47:383-392.

93. Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin

signaling through IRS-1 phosphorylation. Biochimie. [2005;87:99-109.

94. Dobrowsky RT, Kamibayashi C, Mumby MC, Hannun YA. Ceramide activates

heterotrimeric protein phosphatase 2A. J Biol Chem. [1993;268:15523-15530.

95. Chavez JA, Knotts TA, Wang LP, Li G, Dobrowsky RT, Florant GL, Summers SA. A

role for ceramide, but not diacylglycerol, in the antagonism of insulin signal

transduction by saturated fatty acids. J Biol Chem. [2003;13:10297-10303.

96. Teruel T, Hernandez R, Lorenzo M. Ceramide mediates insulin resistance by tumor

necrosis factor-alpha in brown adipocytes by maintaining Akt in an inactive

dephosphorylated state. Diabetes. [2001;50:2563-2571.

97. Stratford S, DeWald DB, Summers SA. Ceramide dissociates 3’-phosphoinositide

production from pleckstrin homology domain translocation. Biochem J.

[2001;354:359-368.

98. Mayer CM, Fick LJ, Gingerich S, Belsham DD. Hypothalamic cell lines to

investigate neuroendocrine control mechanisms. Front Neuroendocrinol.

[2009;30:405-23.

99. Earle WR. Production of malignancy in vitro. IV. The mouse fibroblast cultures and

changes seen in the living cells. J National Cancer Inst. [1943;4:165.

100. De Vitry F, Carnier M, Czernichow P, Benda P, Cohen P, Tixier-Vidal A.

Establishment of a clone of mouse hypothalamic neurosecretory cells synthesizing

neurophysin and vasopressin. Proc Natl Acad Sci USA. [1974;71:3575-3579.

101. Cepko CL, Roberts BE, Mulligan RC. Construction and applications of a highly

transmissible murine retrovirus shuttle vector. Cell. [1984;37:1053-1062.

102. Belsham DD, Fick LJ, Dalvi PS, Centeno ML, Chalmers JA, Lee PK, Wang Y,

Drucker DJ, Koletar MM. Ciliary neurotrophic factor recruitment of glucagon-like

peptide-1 mediates neurogenesis, allowing immortalization of adult murine

hypothalamic neurons. FASEB J. [2009;23:4256-4265.

Page 87: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

76

103. Belsham DD, Cai F, Cui H, Smukler SR, Salapatek AM, Shkreta L. Generation of a

phenotypic array of hypothalamic neuronal cell models to study complex

neuroendocrine disorders. Endocrinology. [2004;145:393-400.

104. Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium

thiocyanate-phenol-chloroform extraction. Anal Biochem. [1987;162:156-159 .

105. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose

tissue expression of tumor necrosis factor-alpha in human obesity and insulin

resistance. J Clin Invest. [1995;95:2409-2415.

106. Ussher JR, Koves TR, Cadete VJ, Zhang L, Jaswal JS, Swyrd SJ, Lopaschuk DG,

Proctor SD, Keung W, Muoio DM, Lopaschuk GD. Inhibition of de novo ceramide

synthesis reverses diet-induced insulin resistance and enhances whole-body oxygen

consumption. Diabetes. [2010;59:2453-2464.

107. Macrae K, Stretton C, Lipina C, Blachnio-Zabielska A, Baranowski M, Gorski J,

Marley A, Hundal HS. Defining the role of DAG, mitochondrial function, and lipid

deposition in palmitate-induced proinflammatory signaling and its counter-

modulation by palmitoleate. J Lipid Res. [2013;54:2366-2378.

108. Gao D, Nong S, Huang X, Lu Y, Zhao H, Lin Y, Man Y, Wang S, Yang J, Li J. The

effects of palmitate on hepatic insulin resistance are mediated by NADPH Oxidase 3-

derived reactive oxygen species through JNK and p38MAPK pathways. J Biol Chem.

[2010;285:29965-29973.

109. Mayer CM, Belsham DD. Palmitate attenuates insulin signaling and induces

endoplasmic reticulum stress and apoptosis in hypothalamic neurons: rescue of

resistance and apoptosis through adenosine 5’ monophosphate-activated protein

kinase activation. Endocrinology. [2010;151:576-585.

110. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate

immunity and fatty acid-induced insulin resistance. J Clin Invest. [2006;116:3015-

3025.

111. Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu K, Hu X, Botezelli JD,

Asadi A, Hoffman BG, Kieffer TJ, Bamji SX, Clee SM, Johnson JD.

Hyperinsulinemia drives diet-induced obesity independently of brain insulin

production. Cell Metab. [2012;16:723-737.

112. Schwartz MW, Woods SC, Porte DJ, Seeley RJ, Baskin DG. Central nervous system

control of food intake. Nature. [2000;404:661-671.

113. Carvalheira JB, Siloto RM, Ignacchitti I, Brenelli SL, Carvalho CR, Leite A, Velloso

LA, Gontijo JA, Saad MJ. Insulin modulates leptin-induced STAT3 activation in rat

hypothalamus. FEBS Lett. [2001;500:119-124.

Page 88: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

77

114. Heiling VJ, Miles JM, Jensen MD. How valid are isotopic measurements of fatty acid

oxidation? Am J Physiol. [1991;261:E572-577.

115. Jensen MD, Caruso M, Heiling V, Miles JM. Insulin regulation of lipolysis in

nondiabetic and IDDM subjects. Diabetes. [1989;38:1595-1601.

116. Jensen MD, Haymon MW, Gerich JE, Cryer PE, Miles JM. Lipolysis during fasting:

decreased suppression by insulin and increased stimulation by epinephrine. J Clin

Invest. [1987;79:207-213.

117. Bjorntorp P, Bergman H, Varnauskas E. Plasma free fatty acid turnover rate in

obesity. Acta Med Scand. [1969;185:351-356.

118. Hodson L, McQuaid SE, Karpe F, Frayn KN, Fielding BA. Differences in

partitioning of meal fatty acids into blood lipid fractions: a comparison of linoleate,

oleate, and palmitate. Am J Physiol Endocrinol Metab. [2009;296:E64-71.

119. Dhopeshwarkar GA, Mead JF. Uptake and transport of fatty acids into the brain and

the role of the blood-brain barrier system. Adv Lipid Res. [1973;11:109-142.

120. Karmi A, Iozzo P, Viljanen A, Hirvonen J, Fielding BA, Virtanen K, Oikonen V,

Kemppainen J, Viljanen T, Guiducci L, Haaparanta-Solin M, Nagren K, Solin O,

Nuutila P. Increased brain fatty acid uptake in metabolic syndrome. Diabetes.

[2010;59:2171-2177.

121. Gross A, McDonnell JM, Korsmeyer SJ. BCL-2 family members and the

mitochondria in apoptosis. Genes Dev. [1999;13:1899-1911.

122. Davis JE, Gabler NK, Walker-Daniels J, Spurlock ME. The c-Jun N-terminal kinase

mediates the induction of oxidative stress and insulin resistance by palmitate and toll-

like receptor 2 and 4 ligands in 3T3-L1 adipocytes. Horm Metab Res. [2009;41:523-

530.

123. Barma P, Bhattacharya S, Bhattacharya A, Kundu R, Dasgupta S, Biswas A, Roy SS.

Lipid induced overexpression of NF-kappaB in skeletal muscle cells is linked to

insulin resistance. Biochim Biophys Acta. [2009;1792:190-200.

124. Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A. Chronic

palmitate but not oleate exposure induces endoplasmic reticulum stress, which may

contribute to INS-1 pancreatic beta-cell apoptosis. Endocrinology. [2006;147:3398-

3407.

125. Winzell MS, Ahrén B. The high-fat diet-fed mouse: a model for studying mechanisms

and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes.

[2004;53:S215-219.

Page 89: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

78

126. Collins QF, Xiong Y, Lupo Jr EG, Liu H, Cao W. p38 Mitogen-activated protein

kinase mediates free fatty acid-induced gluconeogenesis in hepatocytes. J Biol Chem.

[2006;281:24336-24344.

127. Ramirez S, Martins L, Jacas J, Carrasco P, Pozo M, Clotet J, Serra D, Hegardt FG,

Dieguez C, Lopez M, Casals N. Hypothalamic ceramide levels regulated by CPT1C

mediate the orexigenic effect of ghrelin. Diabetes. [2013;62:2329-2337.

128. Guo W, Wong S, Xie W, Lei T, Luo Z. Palmitate modulates intracellular signaling,

induces endoplasmic reticulum stress, and causes apoptosis in mouse 3T3-L1 and rat

primary preadipocytes. Am J Physiol Endocrinol Metab. [2007;293:E576-586.

129. Gao Z, Hwang D, Bataille F, Lefevre M, York D, Quon MJ, Ye J. Serine

phosphorylation of insulin receptor substrate 1 by inhibitor kB kinase complex. J Biol

Chem. [2002;277:48115-21.

130. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, Littman DR, Birbaum MJ,

Polakiewicz RD. Protein kinase C Theta inhibits insulin signaling by phosphorylating

IRS1 at Ser(1101). J Biol Chem. [2004;279:45304-7.

131. Genuth SM. Plasma insulin and glucose profiles in normal, obese, and diabetic

persons. Ann Intern Med. [1973;79:812-22.

132. Dalvi PS, Nazarians-Armavil A, Tung S, Belsham DD. Immortalized neurons for the

study of hypothalamic function. Am J Physiol Regul Integr Comp Physiol.

[2011;300:R1030-52.

133. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins

SM, Olefsky JM. GPR120 is an omega-3 fatty acid receptor mediating potent anti-

inflammatory and insulin-sensitizing effects. Cell. [2010;142:687-698.

134. Zhu H, Yang Y, Wang Y, Li J, Schiller PW, Peng T. MicroRNA-195 promotes

palmitate-induced apoptosis in cardiomyocytes by down-regulating Sirt1. Cardiovasc

Res. [2011;92:75-84.

135. Colell A, Coll O, Mari M, Fernandez-Checa JC, Garcia-Ruiz C. Divergent role of

ceramide generated by exogenous sphingomyelinases on NF-kappa B activation and

apoptosis in human colon HT-29 cells. FEBS Lett. [2002;526:15-20.

136. Cintra DE, Ropelle ER, Moraes JC, Pauli JR, Morari J, de Souza CT, Grimaldi R,

Stahl M, Carvalheira JB, Saad MJ, Velloso LA. Unsaturated fatty acids revert diet-

induced hypothalamic inflammation in obesity. PLoS One. [2012;7:1-15.

137. Goldsmith, CS. Regulation of the p38 MAPK signaling pathway by the circadian

clock. Doctoral dissertation, Texas A&M University, 2013. Available electronically

from http://hdl.handle.net/1969.1/151390

Page 90: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

79

138. Morrison CD, Xi X, White CL, Ye J, Martin RJ. Amino acids inhibit AgRP gene

expression via an mTOR-dependent mechanism. Am J Physiol Endocrinol Metab.

[2007;293:E165-171.

139. Kalaria RN, Gravina SA, Schmidley JW, Perry G, Harik SI. The glucose transporter

of the human brain and blood-brain barrier. Ann Neurol. [1988;24:757-764.

140. Dunn-Meynell AA, Sanders NM, Compton D, Becker TC, Eiki J, Zhang BB, Levin

BE. Relationship among brain and blood glucose levels and spontaneous and

glucoprivic feeding. J Neurosci. [2009;29:7015-7022.

141. Anand BK, China GS, Sharma KN, Dua S, Singh B. Activity of single neurons in the

hypothalamic feeding centers: effect of glucose. Am J Physiol. [1964;207:1146-1154.

142. Oomura Y, Kimura K, Ooyama H, Maeo T, Iki M, Kuniyoshi N. Reciprocal activities

of the ventromedial and lateral hypothalamic areas of cats. Science. [1964;143:484-5.

143. Song Z, Levin BE, McArdle JJ, Bakhos N, Routh VH. Convergence of pre- and

postsynaptic influences on glucosensing neurons in the ventromedial hypothalamic

nucleus. Diabetes. [2001;50:2673-2681.

144. Dunn-Meynell AA, Routh VH, Kang L, Gaspers L, Levin BE. Glucokinase is the

likely mediator of glucosensing in both glucose-excited and glucose-inhibited central

neurons. Diabetes. [2002;51:2056-2065.

145. Ashford ML, Boden PR, Treherne JM. Glucose-induced excitation of hypothalamic

neurones is mediated by ATP-sensitive K+ channels. Pflugers Arch. [1990;415:479-

483.

146. Ibrahim N, Bosch MA, Smart JL, Qiu J, Rubinstein M, Ronnekleiv OK, Low MJ,

Kelly MJ. Hypothalamic proopiomelanocortin neurons are glucose responsive and

express K(ATP) channels. Endocrinology. [2003;144:1331-1340.

147. Fioramonti X, Lorsignol A, Taupignon A, Penicaud L. A new ATP-sensitive K+

channel-independent mechanism is involved in glucose-excited neurons of mouse

arcuate nucleus. Diabetes. [2004;53:2767-2775.

148. Mountjoy PD, Bailey SJ, Rutter GA. Inhibition by glucose or leptin of hypothalamic

neurons expressing neuropeptide Y requires changes in AMP-activated protein kinase

activity. Diabetologia. [2007;50:168-177.

149. Muroya S, Yada T, Shioda S, Takigawa M. Glucose-sensitive neurons in the rat

arcuate nucleus contain neuropeptide Y. Neurosci Lett. [1999;264:113-116.

150. Oakes ND, Thalen P, Aasum E, Edgley A, Larsen T, Furler SM, Ljung B, Severson

D. Cardiac metabolism in mice: tracer method developments and in vivo application

revealing profound metabolic inflexibility in diabetes. Am J Physiol Endocrinol

Metab. [2006;290:E870-881.

Page 91: The Effects of Saturated Fatty Acid Palmitate on ... · neuropeptide gene expression, signal transduction events and insulin signaling. In the mHypoA-NPY/GFP neuronal cell model,

80

151. Saeed NM, El-Demerdash E, Abdel-Rahman HM, Algandaby MM, Al-Abbasi FA,

Abdel-Naim AB. Anti-inflammatory activity of methyl palmitate and ethyl palmitate

in different experimental rat models. Toxicol Appl Pharmacol. [2012;264:84-93.

152. Hodson L, Skeaff CM, Fielding BA. Fatty acid composition of adipose tissue and

blood in humans and its use as a biomarker of dietary intake. Prog Lipid Res.

[2008;47:348-380.