intestinal cholecystokinin controls …...cholecystokinin (cck) is a gut peptide involved in the...

113
INTESTINAL CHOLECYSTOKININ CONTROLS GLUCOSE PRODUCTION THROUGH A NEURONAL NETWORK by Grace Wing Chee Cheung A thesis submitted in conformity with the requirements for the degree of MASTER OF SCIENCE DEPARTMENT OF PHYSIOLOGY UNIVERSITY OF TORONTO © Grace Wing Chee Cheung Copyright 2010

Upload: others

Post on 10-Jul-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

INTESTINAL CHOLECYSTOKININ CONTROLS GLUCOSE PRODUCTION THROUGH A NEURONAL NETWORK

by

Grace Wing Chee Cheung

A thesis submitted in conformity with the requirements for the degree of MASTER OF SCIENCE

DEPARTMENT OF PHYSIOLOGY UNIVERSITY OF TORONTO

© Grace Wing Chee Cheung Copyright 2010

Page 2: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

ii

Title of thesis: Intestinal cholecystokinin controls glucose production through a neuronal

network

Name of candidate: Grace Wing Chee Cheung

Degree: Master of Science

Department of Physiology, University of Toronto

Year of convocation: 2010

GENERAL ABSTRACT

Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by

duodenal lipids via a neuronal network. However, it is unknown whether CCK also regulates

glucose homeostasis through a neuronal network. Using an in vivo rat model, we demonstrated

that duodenal CCK-8 (biologically active form of CCK) can lower glucose production through

the activation of a gut-brain-liver axis via CCK-A receptors, and this glucose-regulatory effect is

physiologically relevant. Since duodenal lipids can also lower glucose production through a gut-

brain-liver axis, we verified that this duodenal-lipid effect is mediated by CCK-A receptor

activation. Lastly, in rats fed on a high-fat diet for three days, duodenal CCK failed to suppress

glucose production, suggesting a state of CCK-resistance. In summary, these findings revealed

that intestinal CCK can regulate glucose homeostasis through a neuronal network and suggest

that intestinal CCK resistance may contribute to hyperglycemia in response to high-fat feeding.

Page 3: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

iii

Acknowledgement

First of all, I would like to express my deepest gratitude to my supervisor, Dr. Tony Lam. He has

given me opportunities to develop in various ways academically. He taught me how to design

experiments, how to write scientific papers, how to present my findings. I am also thankful for

his willingness in guiding my life decisions. Moreover, I would like to thank my supervisory

committee members, Dr. Harvey Anderson and Dr. Adria Giacca. I am grateful to have such

wonderful scientists in my supervisory committee. Dr. Anderson and Dr. Giacca taught me to not

be satisfied with a single answer but to be curious and seek for other explanations. In addition to

the supervisory figures, the laboratory had been an integral part of my graduate experience and it

would not have been so rewarding without these people: Carol Lam, Madhu Chari, Andrea

Kokorovic, Clair Yang, Penny Wang and Teresa Lai.

Aside from the academic support, I would also like to thank the following people. First, I would

like to thank my parents, Edward Cheung and Enid Li, for their unconditional support and

encouragement to pursue my interests. My sister, Bonnie Cheung, for all the joy she has brought

to my life. My significant other, Terry Wong, for standing by my side through all the ups and

downs and providing all the support I ever needed. Last but not least, I would like to thank my

Lord for His continual guidance and all the wonderful plans He made for me, I will continue to

trust Him with all my heart for all the challenges ahead.

“Trust in the Lord with all your heart and lean not on your own understanding; in all

your ways acknowledge him, and he will make your paths straight.” Proverbs 3:5

Page 4: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

iv

Table of Contents

Acknowledgement ......................................................................................................................... iii

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

List of Figures ................................................................................................................................. v

List of Tables ................................................................................................................................. vi

List of Abbreviations .................................................................................................................... vii

Publication that contributed to this thesis ...................................................................................... ix

1 Introduction .................................................................................................................................. 1

1.1 Obesity and Diabetes .................................................................................................... 1

1.2 Regulation of Glucose Homeostasis: The Role of the Small Intestine ......................... 3

1.3 Gastrointestinal Peptides ............................................................................................... 6

1.3.1 Gastric Peptide ............................................................................................... 6

1.3.2 Upper Intestinal Peptide ............................................................................... 10

1.3.3 Lower Intestinal Peptide .............................................................................. 15

1.4 Neuronal Regulation of Glucose Homeostasis by the Small Intestine ....................... 22

1.4.1 Hepatoportal Glucose Sensor ....................................................................... 23

1.4.2 Lipid-induced gut-brain-liver axis ............................................................... 25

2 General Hypothesis and Aims ................................................................................................... 29

3 General Materials and Methods ................................................................................................. 35

4 Results ........................................................................................................................................ 50

5 Discussion .................................................................................................................................. 72

6 Future Directions ....................................................................................................................... 78

7 Conclusion ................................................................................................................................. 81

8 References .................................................................................................................................. 82

Page 5: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

v

List of Figures

Figure 1 Upper intestinal lipids activate a gut-brain-liver axis to suppress glucose production ......................................................................................................................28

Figure 2 Working hypothesis – upper intestinal lipids stimulate CCK/CCK-A receptor signaling to suppress glucose production through a gut-brain-liver axis ......................34

Figure 3 Schematic representation of the working hypothesis – duodenal CCK activates CCK-A receptors to regulate glucose production and experimental design (pharmacological approach). .........................................................................................58

Figure 4 Duodenal CCK activates CCK-A receptors to suppress glucose production ................59

Figure 5 Schematic representation of the working hypothesis – duodenal CCK activates CCK-A receptors to regulate glucose production and experimental design (molecular approach). ....................................................................................................60

Figure 6 Duodenal CCK suppresses glucose production in LETO but not in CCK-A receptor deficient OLETF rats .......................................................................................61

Figure 7 Schematic representation of the working hypothesis – duodenal CCK activates a gut-brain-liver axis to suppress glucose production and experimental design ..............62

Figure 8 Duodenal CCK can activate a gut-brain-liver axis to regulate glucose production ......63

Figure 9 Schematic representation of the working hypothesis – duodenal CCK signaling is downstream of lipid-sensing to regulate glucose production and experimental design .............................................................................................................................64

Figure 10 Duodenal CCK-A receptor activation is required for lipids to lower glucose production ......................................................................................................................65

Figure 11 Pharmacological inhibition of CCK-A receptors in the gut disrupts glucose homeostasis during refeeding ........................................................................................66

Figure 12 Molecular inhibition of CCK-A receptors in the gut disrupts glucose homeostasis during refeeding ............................................................................................................67

Figure 13 Schematic representation of the working hypothesis – duodenal CCK fails to suppress glucose production in response to high fat feeding and experimental design .............................................................................................................................68

Figure 14 Duodenal CCK fails to suppress glucose production following 3-days of high-fat feeding ...........................................................................................................................69

Page 6: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

vi

List of Tables

Table 1 Dietary contents of the regular chow and the lard-oil enriched high fat diet. .................49

Table 2 Plasma insulin and glucose concentrations for SAL and CCK8 during basal and clamp conditions .........................................................................................................................70

Page 7: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

vii

List of Abbreviations AA Arachidonic acid

AMPK Adenosine monophosphate kinase

ANOVA Analysis of variances

ATP Adenosine triphosphate

B Bound fraction

B0 Total binding

cAMP cyclic adenosine monophosphate

CCK Cholecystokinin

CCK-A receptor Cholecystokinin-A receptor

CCK-B receptor Cholecystokinin-B receptor

GHS-R Growth hormone secretagogue receptor

GIP Glucose-dependent insulinotropic polypeptide

GIP-R Glucose-dependent insulinotropic polypeptide receptor

GLP-1 Glucagon-like peptide-1

GLP-1R Glucagon-like peptide-1 receptor

GLUT4 Glucose transporter 4

GPR40 G-protein coupled-receptor 40

GSIS Glucose-stimulated insulin secretions

HFD High-fat diet

ICV Intracerebroventricular

IP Intraperitoneal

IP3 Inositol triphosphate

Page 8: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

viii

IV Intravenous

KATP channels ATP-sensitive potassium channels

Kv channels voltage-dependent potassium channels

LCFA Long chain fatty acid

LETO rat Long-Evans Tokushima Otsuka rat

NMDA receptor N-methyl-d-aspartate receptor

NTS Nucleus of the solitary tract

OLETF rat Otsuka Long-Evans Tokushima Fatty rat

OXM Oxyntomodulin

PKA Protein kinase A

PKC Protein kinase C

PLA2 Phospholipase A2

PLC Phospholipase C

PYY Peptide YY

Ra Rate of appearance

Rd Rate of disappearance

RIA Radioimmunoassay

SC Standard chow

SD rat Sprague Dawley rat

Page 9: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

ix

Publication that contributed to this thesis

- Cheung GW

, Kokorovic A, Lam CK, Chari M and Lam TK. Intestinal cholecystokinin

controls glucose production through a neuronal network. Cell Metab. 10: 99-109, 2009.

Page 10: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

1

1 Introduction

1.1 Obesity and Diabetes

According to the World Health Organization, approximately 1.6 billion adults (age 15+; body

mass index ≥ 25 and <30) were overweight, with at least 400 million adults obese (BMI ≥ 30) in

2005. Obesity is associated with type 2 diabetes, cardiovascular disease and various cancers

[1;2]. It has been proposed that elevated plasma free fatty acids may serve as a causative link

between obesity, insulin resistance and diabetes [3;4]. While diabetes is a disease characterized

by hyperglycemia due to impairments in the regulation of glucose homeostasis, it has been

demonstrated that free fatty acids can induce insulin resistance [5], strongly suggesting a link

between obesity and type 2 diabetes.

In 2000, approximately 170 million individuals were affected by diabetes, with the affected

population expected to double by 2030 [6]. There are two forms of diabetes mellitus: type 1 and

type 2. Type 1 diabetes is an autoimmune disorder in which there is near total deficiency in

insulin secretions due to the destruction of insulin-producing β-cells [7]. On the other hand, type

2 diabetes is the more common form, affecting more than 90% of the patients with diabetes, and

it is linked to a combination of insulin resistance and deficient insulin secretion [8]. Insulin is

responsible for (i) stimulating glucose uptake into muscle and fat tissues (ii) suppressing glucose

production in the liver [8]. Hence, insulin resistance can lead to excessive glucose in the

circulation resulting in chronic hyperglycemia. Various complications can result from chronic

hyperglycemia [9;10], including retinopathy [11], kidney failure [12], increased risks of

cardiovascular disease [13] and stroke [9]. As such, it is essential to dissect the mechanisms

Page 11: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

2

involved in the regulation of glucose homeostasis and identify possible interventions to restore

the regulation.

Page 12: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

3

1.2 Regulation of Glucose Homeostasis: The Role of the Small Intestine

The regulation of glucose homeostasis can be distinguished in two different states. In the fasting

basal state, plasma glucose levels are derived endogenously. There are primarily two hormones

involved in the regulation of glucose homeostasis: glucagon and insulin [14]. Glucagon is

secreted from pancreatic α-cells to prevent hypoglycemia through stimulating hepatic glucose

production [15]. In contrast, insulin prevents hyperglycemia through stimulating glucose uptake

and suppressing glucose production [8]. In the fasting state, glucagon secretion rises while

insulin secretion falls to ensure that there is sufficient glucose circulating.

Upon meal intake, glucose regulation changes from the fasting state to the fed state. In the fed

state, there are both exogenous and endogenous sources of glucose. Exogenous glucose is

absorbed from the gastrointestinal tract as nutrients become available for absorption in the small

intestine. Hence, glucose absorption rate depends on the rate at which nutrients are emptied from

the stomach to the small intestine, known as the gastric emptying rate [16]. As exogenous

glucose arrives in the circulation, a biphasic insulin secretion response is stimulated. In response

to an intravenous (iv) glucose challenge, insulin is rapidly secreted to reach an initial peak in 5 to

7 minutes [17], and this early-phase of insulin release lasts about 10 to 15 minutes. On the other

hand, the late-phase is characterized by a more sustained insulin secretion lasting several hours

[17]. In addition to the responses in insulin, glucagon secretion is suppressed. Together,

endogenous glucose production is suppressed, while glucose uptake is augmented to allow

plasma glucose concentrations to return to baseline values such that the fasting state resumes.

Page 13: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

4

Since the small intestine is one of the first sites of contact of the body to incoming nutrients, the

small intestine is at a strategic location to exert actions on the regulation of glucose homeostasis

during feeding. The regulation of glucose homeostasis by the small intestine can largely be

distinguished into two types of mechanisms: (i) indirect actions through gastrointestinal peptides

(ii) direct neuronal pathway.

For the indirect actions mediated by gastrointestinal peptides, the effects are mainly exerted

through modulating the gastric emptying rate and pancreatic hormone levels in response to the

entry of nutrients as will be described in more details later. Through the modulation of gastric

emptying rate, the rate at which glucose is delivered to the small intestine can be controlled to

affect the rise of glucose in the blood [16]. On the other hand, the secretion of insulin and

glucagon can also be potentiated or inhibited by gastrointestinal peptides to regulate plasma

glucose levels differentially.

For the neuronal regulation of glucose homeostasis by the small intestine, a hepatoportal sensor

[18;19] and a gut-brain-liver [20] neuronal axis induced by glucose and lipid respectively have

been identified. While it seems that the mechanisms of action for the gastrointestinal peptides are

unrelated to the neuronal mechanisms, gastrointestinal peptides have been shown to regulate

food intake through neuronal actions [21]. Hence, it is possible that gastrointestinal peptides are

involved in the activation of the neuronal networks in the regulation of glucose homeostasis.

In the following section, we will first focus on the gastrointestinal peptides that are modulated by

duodenal nutrients (duodenum is the first segment of the small intestine) and the role of each in

Page 14: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

5

the regulation of glucose homeostasis, and where applicable, the neuronal network responsible

for modulating food intake by each gastrointestinal peptide. The gastrointestinal peptides will be

presented according to their anatomical origin.

Page 15: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

6

1.3 Gastrointestinal Peptides 1.3.1 Gastric Peptide

1.3.1.1 Ghrelin Ghrelin was isolated in 1999 as an acylated 28-amino acid peptide with the acylation required for

its bioactivity [22]. Ghrelin is predominantly secreted in the stomach and duodenum [22-25] and

its levels are usually increased prior to eating, while reduced in response to nutrient entry

[26;27]. In particular, intraduodenal administration of glucose [28-31], amino acids [31], or

lipids [31-34] can all suppress ghrelin secretion to discontinue the fasting signal. With respect to

the regulation of secretion by lipids, lipids in the form of triglycerides must first be broken down

by lipases to release fatty acids [33]. Following the release of fatty acids, the accumulation of

long chain fatty acids (LCFA) [34] in the duodenum leads to the secretion of another

gastrointestinal peptide called cholecystokinin (CCK) and the binding of CCK to cholecystokinin

A (CCK-A) receptors is necessary for lipids to suppress ghrelin secretion [32].

Since ghrelin is mostly secreted during the fasting state, it is not surprising that ghrelin plays a

significant role in the maintenance of fasting blood glucose levels through modulating insulin

secretion. In humans, ghrelin increases glucose while decreasing plasma insulin levels [35].

Ghrelin is a natural ligand for the growth hormone secretagogue receptor (GHS-R) [22;36;37],

the blockade of GHS-R resulted in reduced fasting blood glucose concentrations [38] and

increased plasma insulin levels [39]. This suggests that ghrelin plays a significant role in the

regulation of fasting blood glucose through playing an inhibitory role on insulin secretion.

Interestingly, the source of ghrelin for the effects on insulin may not originate from the stomach

Page 16: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

7

or duodenum [40], but from the pancreas [38]. After undergoing gastrectomy to eliminate the

secretion of ghrelin by the stomach, it was found that the administration of ghrelin-antagonist

could still augment glucose-stimulated insulin secretions (GSIS), strongly suggesting that a local

source of ghrelin in the pancreas is responsible for the GSIS effects. However, the regulation of

secretion for pancreatic ghrelin remains to be determined. Nonetheless, GHS-R have been

located in pancreatic islets [25;38;41-44], suggesting that GHS-R in the pancreas may mediate

the effects of ghrelin on pancreatic insulin secretions. Indeed, in isolated rat islets of GHSR-

knockout mice, GSIS was found to be significantly greater than that of control mice [39],

supporting the notion that endogenous ghrelin acts on the GHS-R of pancreatic islets to regulate

GSIS. At the level of the islets, ghrelin failed to suppress insulin secretion in the presence of

pertussis toxin or voltage-dependent potassium (Kv) channel blockers [39], suggesting that

ghrelin can activate a specific isoform of G-proteins, which can lead to the activation of Kv

channels. In particular, it was confirmed with a molecular antisense approach that the isoform of

G-protein involved is the G-alpha i2 form [39]. Importantly, when the activity of Kv channels

was prevented, the rise in intracellular calcium levels associated with insulin release was

attenuated [39]. On the basis of these findings, ghrelin binds to the GHS-R on the islets to

decrease the activity of glucose-stimulated action potentials of the islet membranes marked by

decreased frequency and amplitude of firings [39]. As the activity of action potentials reduces,

the duration of the bursting potentials stimulated by glucose is shortened, preventing further

calcium-dependent insulin exocytosis. In turn, the suppression of GSIS allows the body to

restrain insulin secretion in the fasting state whereas the restraining effect on insulin can be lifted

in the presence of duodenal nutrients through the inhibition of ghrelin. Conversely, ghrelin does

not affect glucagon secretion [45].

Page 17: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

8

In addition to its effects on insulin secretion, ghrelin may also affect insulin sensitivity. In

humans, ghrelin infusion induced acute insulin resistance [46]. In another study, it was shown

that ghrelin may induce insulin resistance specifically in muscle to reduce glucose disposal while

having no effects on glucose production [47]. On the other hand, the ablation of ghrelin

improved peripheral insulin sensitivity in diabetic mice by affecting insulin action both on

glucose production and glucose disposal [48]. Although there are disagreements regarding the

tissue-specific effects of ghrelin, these studies in general suggest that ghrelin is responsible for

inducing insulin resistance. In contrast, there is also a study showing otherwise, that ghrelin may

improve peripheral insulin sensitivity while hampering hepatic insulin sensitivity [49]. Hence,

the role of ghrelin on insulin sensitivity remains controversial.

Nonetheless, it is commonly accepted that ghrelin can affect glucose homeostasis through its

action on gastric emptying. When ghrelin is infused, there is higher postprandial glucose as a

result of accelerated gastric emptying [50]. By increasing the gastric emptying rate, ghrelin

accelerates the speed at which glucose is delivered to the duodenum for absorption. There are

findings suggesting that ghrelin may accelerate gastric emptying through a neural mechanism.

First, GHS-R has been identified in nerves fibers belonging to the enteric nervous system [51]. In

rats, ghrelin enhanced contractions in strips of the gastric body while tetrodotoxin (neurotoxin)

abolished the effect, supporting that the intrinsic enteric nervous system may be involved [52]. In

addition to the enteric nervous system, in rats pretreated with capsaicin, atropine, or cervical

vagotomy, the ability of ghrelin to accelerate gastric emptying was abrogated [52]. Since there

are ghrelin receptors expressed in vagal afferent neurons [53], ghrelin may stimulate the vagus

Page 18: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

9

nerve directly or through the enteric nervous system to accelerate gastric emptying. The

increased gastric emptying rate can then allow nutrients to be delivered to the duodenum for

absorption as soon as they are available. As nutrients enter the duodenum, the nutrients can

stimulate a negative feedback system on gastric emptying by inhibiting ghrelin secretion,

resulting in the blunting of postprandial hyperglycemia.

As mentioned previously, ghrelin is mostly secreted in the fasting state, it is not surprising that

ghrelin can stimulate feeding in rats [53]. In response to ghrelin, the arcuate nucleus in the

hypothalamus is activated as indicated by fos-staining, and both the feeding and hypothalamic

activation responses are abolished in response to capsaicin treatment, gastric branch vagotomy,

or subdiaphragmatic vagotomy [53]. Hence, a neuronal network connecting the gut and the brain

should exist to mediate the effects of ghrelin on feeding, suggesting that there is a gut-brain

neuronal axis to allow communication between the gut and the brain.

Page 19: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

10

1.3.2 Upper Intestinal Peptide

1.3.2.1 Cholecystokinin CCK is a gut peptide that has been discovered by Ivy and Oldberg in 1928 to stimulate gall

bladder contractions [54]. In the intestine, CCK is synthesized as a 115 amino acid prepro-CCK

polypeptide. The prepro-CCK polypeptide then undergoes multiple posttranslational cleavages to

generate the shortest and the complete biologically active form of CCK-8 [55].

CCK is released basolaterally by I cells lining the mucosa of the duodenum to the surrounding

areas such as the bloodstream and nerve endings. Duodenal glucose, lipids, and proteins have all

been shown to stimulate CCK secretion [29;56-58]. For lipids, the release of LCFAs is essential

for the release of CCK [59-61]. While proteins can stimulate CCK secretions [62], individual

amino acids such as tryptophan [63] and phenylalanine [64] can also stimulate CCK release.

Given that CCK is released in response to nutrients, CCK serves many important functions

pertaining to the digestion and absorption of nutrients, including the stimulation of pancreatic

secretions, excretion of bile into the intestine, and slowing of gastric emptying [65].

In the context of glucose homeostasis regulation, CCK can act through two different mechanisms

to reduce postprandial hyperglycemia. First, there are mixed reports regarding the ability of CCK

to regulate insulin secretion in physiological setting. While some studies demonstrated that CCK

may potentiate GSIS [66-75], this effect was absent in other studies [76-80], questioning the

physiological relevance of this effect. Nonetheless, systemic administration of CCK induced a

biphasic response of insulin secretions [81], with an early rise after a minute, and a slow second

phase, associated with a concomitant drop in plasma glucose. First, CCK may potentiate GSIS in

Page 20: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

11

rat islets via interacting with the alpha q/11 subunit of G-proteins [82]. This subunit of G-protein

has been implicated in the phospholipase C (PLC) pathway, suggesting that CCK may potentiate

GSIS through the PLC system [82] to increase the production of inositol triphosphate (IP3) [82]

and intracellular calcium levels [83] to potentiate calcium-dependent insulin secretions. A

product of PLC activation is diacylglycerol (DAG), which is an activator of protein kinase C

(PKC). Indeed, CCK-8 has been shown to activate PKC through stimulating PLC [84].

Subsequent to PKC activation, CCK may activate phospholipase A2 (PLA2) to form arachidonic

acid (AA) [85] in isolated rat islets. It has been shown that the activation of PLA2 is partly

dependent on protein kinase C (PKC) and independent of calcium for the effects of CCK on

insulin [86]. Nonetheless, although CCK may potentiate GSIS through these mechanisms, these

effects may be pharmacological rather than physiological due to the high dosage used.

Secondly, a widely accepted mechanism by which CCK can regulate glucose homeostasis is

through the modulation of gastric emptying rate. CCK can reduce postprandial hyperglycemia

[87] by slowing the delivery of glucose to the duodenum in humans [80;88]. There are two types

of CCK receptors: CCK-A and CCK-B receptors, which reside in the periphery (predominantly

in the gastrointestinal system) and brain respectively [89]. For the regulation of gastric emptying

by CCK, CCK-A receptors are essential to stimulate the contraction of the pyloric sphincter [90-

95]. Interestingly, CCK may not need to bind to CCK-A receptors on the pyloric sphincter to

regulate gastric emptying [96]. Rather, CCK may activate CCK-A receptors on capsaicin-

sensitive vagal afferent C-fibers [97], which then regulate gastric emptying through the gastric

branch of the vagus nerve [98]. Hence, CCK regulates postprandial glucose homeostasis through

Page 21: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

12

the regulation of gastric emptying, and it may also potentiate GSIS when used

pharmacologically.

Moreover, CCK is the first gut peptide implicated in the control of food intake [99]. Specifically,

CCK has been shown to inhibit food intake in rats [100;101], humans [102], and mice [103]. The

effect of CCK on food intake is mediated by CCK-A receptors [104] and it is abolished in rats

subjected to abdominal vagotomy or gastric branch vagotomy [101], indicating that the vagus

nerve is required. Lastly, there is evidence that peripheral CCK can stimulate activation of

different brain areas via capsaicin-sensitive vagal afferent [105], strongly supporting that there is

direct communication between the gut and the brain.

Page 22: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

13

1.3.2.2 Glucose-dependent Insulinotropic Polypeptide Glucose-dependent insulinotropic polypeptide (GIP) is a 42-amino acid hormone [106]

expressed by K cells [107] with the highest concentration in the duodenum and jejunum in

humans [108]. GIP release can be induced by intraduodenal administration of glucose [58;109-

112], lipids [113-115], proteins [116] and amino acids [115]. For lipids in particular, long-chain

triglycerides are more potent than medium-chain triglyceride in stimulating GIP release in dogs

[115]. Interestingly, it has recently been found that free fatty acids can also stimulate the

secretion of GIP through a G-protein coupled receptor known as G-protein coupled-receptor 40

(GPR40) [117].

GIP is most commonly known as an “incretin”. Incretins are hormones that are responsible for

producing a greater insulin response to oral glucose than an equivalent amount of glucose

administered intravenously. Hence, one of the most important functions of GIP is its ability to

potentiate GSIS. It has been found that GIP has hypoglycemic effects through potentiating GSIS

in rats [118] and humans [119], with a specific effect on the early-phase insulin release [120]. In

studies using isolated rat pancreatic islets [121] and rat pancreas [122], it is suggested that GIP

acts directly on pancreatic islets to exert its potentiating effects. In the pancreas, the expression

of a receptor for GIP (GIP-R) has been identified [123], suggesting that GIP may activate the

GIP-R on pancreatic islets for its potentiating effect. Indeed, in GIP-R null mice, GIP failed to

augment GSIS [124]. Following the stimulation by GIP, there is increased cyclic adenosine

monophosphate (cAMP) production [123], and cAMP can lead to the activation of cAMP-

guanine nucleotide exchange factor (GEF) II (Epac2) and protein kinase A (PKA). Importantly,

treatment of pancreatic β-cells with either antisense oligodeoxynucleotides against Epac2 or the

Page 23: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

14

PKA antagonist H-89 demonstrated that both Epac2 and PKA are involved in the potentiating

effects of GIP [125]. Moreover, the closure of adenosine triphosphate (ATP)-sensitive potassium

(KATP) channels has also been found to be critical for the potentiation of insulin secretion by

cAMP [126] and GIP-induced potentiation of GSIS [127]. The closing of KATP channels alters

the membrane potential such that voltage-dependent calcium channels can be opened to increase

intracellular calcium levels. Indeed, the voltage-dependent calcium channels to allow the influx

of extracellular calcium [128] is essential as there is an absence of increases in intracellular

calcium levels in the presence of ethylene glycol tetraacetic acid to chelate extracellular calcium

[128]. In brief, GIP decreases KATP channel activity through the actions of PKA and Epac2 to

depolarize the membranes. As the membrane depolarizes, the opening of voltage-dependent

calcium channels allows the influx of calcium to increase calcium-dependent exocytosis of

insulin.

While it is clear that GIP potentiates GSIS, GIP appears to stimulate glucagon secretions in both

rats [118] and humans [129]. Since glucagon would exert an upregulating effect on circulating

glucose and GIP has been shown to lower glucose levels, it is unclear how this stimulatory effect

on glucagon influences the regulation of glucose by GIP.

Contrary to the hormones already discussed, GIP does not inhibit gastric emptying [130] in

humans. Hence, the main mechanism by which GIP regulates glucose homeostasis is through the

potentiation of GSIS.

GIP has no effects on food intake [131].

Page 24: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

15

1.3.3 Lower Intestinal Peptide

1.3.3.1 Glucagon-like peptide-1 Glucagon-like peptide-1 (GLP-1) is expressed in L cells [132] found in the ileum and colon of

rats, pigs and humans [133]. There are two active forms of GLP-1: GLP-17-37 and GLP-17-36-NH2

[134]. In humans and animal models, the secretion of GLP-1 demonstrates a biphasic profile

with an acute first peak of secretion within 15-30 minutes of nutrient ingestion and a second

sustained peak within 60-90 minutes [135]. However, nutrients normally do not reach the ileum

or colon within 15-30 minutes to account for the first peak of GLP-1 secretion [136], suggesting

that neural mechanisms originating from the proximal gut may play a role in the stimulation of

GLP-1.

Indeed, intraduodenal administration of glucose [58;137-139] and fat [140-144] can both

stimulate the release of GLP-1. With respect to duodenal glucose-stimulated GLP-1 release, the

concomitant transport of sodium ions and glucose via the sodium-glucose cotransporter-1 is

required while glucose metabolism is not necessary [138]. On the other hand, the mechanism(s)

by which duodenal fats stimulate GLP-1 release appears to be more complex. Upon entry of

lipids into the duodenum, the release of LCFAs through fat hydrolysis is essential to stimulate

GLP-1 [59;140]. Following the release of LCFAs, both CCK and GIP have been implicated in

mediating the stimulation of GLP-1 release. When the LCFA sodium oleate was coadministered

with a CCK-A receptor blocker, the rise of GLP-1 was abolished, indicating the importance of

CCK signaling in GLP-1 release in humans [140]. On the other hand, the involvement of GIP in

the stimulation of GLP-1 secretion may be species-dependent. In rats, GIP has been suggested to

mediate the stimulation of GLP-1 by duodenal fat [142], however, GIP had no effects on GLP-1

Page 25: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

16

secretion in humans [145;146]. GIP can stimulate GLP-1 secretion through the hepatic branch of

the vagus nerve in rats, and stimulating the distal end of the celiac branch of the

subdiaphragmatic vagus nerve also results in the secretion of GLP-1 [144]. In light of these

findings, it is probable that a neural mechanism exists between the proximal and distal intestine

for duodenal nutrients to stimulate GLP-1 secretion. Two neurotransmitters are proposed to be

involved: gastrin-releasing peptide (GRP) [143] and acetylcholine [147]. In the presence of the

GRP antagonist, the GLP-1 response to duodenal fat was abolished [143]. Additionally,

cholinergic innervations are also important as atropine or the M1 muscarinic-receptor antagonist

pirenzepine completely abolished fat-induced GLP-1 secretion in rats [147]. Hence, a complex

network exists to mediate the release of GLP-1 in response to duodenal nutrients.

In addition to GIP, GLP-1 is also known for its function as an incretin to regulate postprandial

glucose levels [148] through its potentiating effects on GSIS. As GLP-1 is secreted, it can bind to

GLP-1 receptors on β-cells [149;150] to induce depolarization of pancreatic β-cells [151]. The

mechanism by which GLP-1 induces depolarization requires the closure of KATP channels [151].

With cAMP levels increased in response to GLP-1 receptor activation [152], both PKA and

Epac2 activation were found to be required to potentiate insulin secretion [125]. In particular,

PKA activation is required in mediating the closure of KATP channels [153;154]. Additionally,

other pathways such as a calmodulin-mediated pathway have also been implicated. In the

presence of the calmodulin inhibitor, the inhibitory actions of GLP-1 on the membrane KATP

conductance and the resultant membrane depolarization were completely reversed [155]. At the

level of the KATP channels, GLP-1 seems to potentiate ATP sensitivity to KATP channels such that

less ATP is required to close the KATP channels [151]. Subsequent to membrane depolarization,

Page 26: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

17

the activation of L-type voltage-dependent calcium channels allows intracellular calcium to

increase [156;157] leading to calcium-dependent exocytosis of insulin. Finally, membrane

repolarization is controlled by the activity of voltage-dependent potassium (Kv) channels. In

particular, the inhibition of Kv channels has been shown to be dependent on both the PKA [152],

and the PKC-ζ pathways [158] to enhance (larger and prolonged) action potentials in the β-cells

[152]. Hence, GLP-1 can potentiate GSIS through modulating membrane potential via complex

mechanisms.

Moreover, GLP-1 has also been shown to suppress endogenous glucagon secretion, which may

contribute to its hypoglycemic effects [159-162]. Importantly, the suppressive effect of GLP-1

on glucagon secretion is glucose-dependent to prevent hypoglycemia from the lack of glucagon

[163]. Interestingly, GLP-1 receptors are not expressed in α-cells, meaning that GLP-1 should

regulate glucagon through an indirect mechanism [150]. Indeed, GLP-1 may act on the GLP-1

receptors on the δ-cells [149;150] to stimulate the secretion of somatostatin, then somatostatin

can act on the somatostatin receptor-2 to suppress glucagon secretion [164].

While GLP-1 can exert insulinotropic and glucagonostatic effects, GLP-1 can also regulate

glucose through altering the gastric emptying rate. Specifically, it has been shown that GLP-1

can inhibit gastric emptying to attenuate rises in glucose [127;165-167].

In summary, GLP-1 can regulate glucose homeostasis through acting on insulin and glucagon

secretions and gastric emptying. Interestingly, it has been found that GLP-1 can activate

neuronal mechanisms to increase insulin secretion.

Page 27: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

18

Despite that GLP-1 can exert its incretin effects through direct interaction with β-cells, GLP-1 is

rapidly degraded by DPP-IV [168], thus it is unclear whether the short half-life of GLP-1 would

allow it to act on the pancreas directly. Hence, a rapid neural mechanism may actually be

responsible for the actions of GLP-1 on insulin secretion. As GLP-1 is released, it can enter the

portal vein to stimulate GLP-1 receptors expressed on nodose ganglions to improve glucose

disposal rate [169] through augmenting insulin secretion [169;170]. Importantly, when the

ganglionic blocker chlorisondamine is coinfused with GLP-1 into the portal vein, the insulin

secretion response is abolished [169]. Therefore, a neural mechanism is proposed to mediate the

portal GLP-1 effects on insulin secretion. Indeed, it has been found that administration of GLP-1

into the portal vein increased the firing rates of the hepatic afferent and pancreatic efferent

nerves [171], suggesting that a vagal hepatopancreatic reflex pathway may be involved in the

mediation of the insulinotropic effects of GLP-1 in addition to the endocrine pathway.

Moreover, GLP-1 can also activate a neuronal network to regulate food intake. In both humans

[172] and rats [173], GLP-1 has been shown to reduce food intake. Importantly, the satiety effect

is abolished in rats given subdiaphragmatic vagotomy or transection of a brainstem-

hypothalamic pathway [174]. These findings together suggest that GLP-1 can stimulate a gut-

brainstem-hypothalamic neuronal axis to regulate food intake, supporting the existence of a gut-

brain neuronal axis.

Page 28: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

19

1.3.3.2 Oxyntomodulin Oxyntomodulin (OXM) is a 37-amino acid peptide expressed [175] throughout the

gastrointestinal tract [176]. OXM is expressed in L cells and the expression of OXM increases

from the duodenum to the ileum [176]. While OXM is mainly expressed in the lower intestine,

the release of OXM has only been directly assessed in a study which subjected received

intraduodenal administrations of a liquid meal or oleic acid [177]. Since OXM is also a product

of the proglucagon gene expressing GLP-1, OXM is believed to be co-secreted with GLP-1

[178].

Despite that OXM has been isolated and sequenced in 1994 [179], its action on glucose

homeostasis remains poorly studied. In response to glucose tolerance test, OXM administration

lowered blood glucose without affecting gastric emptying in mice [180], whereas OXM has been

shown to reduce gastric emptying rate in humans [181]. Although the effects of OXM on gastric

emptying rate remains to be clarified, it has been suggested that OXM can stimulate insulin

release in a glucose-dependent manner [180;182]. Interestingly, an oxyntomodulin receptor has

not been clearly demonstrated [183], whereas it has been shown that the action of OXM on

insulin secretion is dependent on GLP-1R [180]. In β-cells, cAMP formation is increased in

response to OXM [180], suggesting that OXM may bind to GLP-1 receptors on β-cells, and

potentiate GSIS through a cAMP-dependent mechanism.

Although OXM can reduce food intake [184], the mechanism of action remains largely

unknown.

Page 29: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

20

1.3.3.3 Peptide YY Peptide YY (PYY) is synthesized in the body as a 36-amino acid peptide (PYY1-36) [185]. After

being released, dipeptidyl peptidase IV (DPP-IV) cleaves the N-terminal tyrosine-proline

residues to form PYY3-36 [186]. In humans, PYY is expressed in L cells in the lower part of the

ileum, colon, and rectum [187]. Similar to GLP-1, PYY is released in two phases, with the early

phase occurring when the nutrients are in the proximal gut [188], and have not reached the ileum

or colon yet. Indeed, PYY can be stimulated by the infusion of fat into the duodenum in humans

[32;33;58;189]. In response to fat, fat hydrolysis [32;33] must occur to release LCFAs

[34;60;190] for PYY secretion to be stimulated. Given the short timeframe for the first phase of

PYY release, a neural mechanism connecting the proximal gut and the distal gut is highly likely

to be in place. CCK has been suggested to mediate the release of GLP-1 stimulated by duodenal

nutrients, and there is evidence to suggest that CCK-A receptor signaling may be responsible for

the release of PYY stimulated by duodenal lipids [32].

In the circulation, both forms of PYY are present, but they can bind to different sets of receptors.

PYY1-36 binds to the Y1, Y2, Y4 and Y5 receptors, and PYY3-36 predominantly binds to the Y2 and

Y5 receptor [191]. Since DPP-IV is very widespread in the body, PYY3-36 is the dominant form

in the circulation, and PYY3-36 seems to play an important role in the regulation of glucose

homeostasis. In contrast to the other gastrointestinal peptides that are released postprandially to

lower glucose levels, PYY3-36 infusion increases postprandial glucose concentrations [192]. The

rather different effect of PYY3-36 on postprandial glucose levels from the other gastrointestinal

peptides may be attributed to its inhibitory effect on GSIS [193], preventing the secretion of

insulin to lower glucose. Moreover, PYY3-36 has no effects on insulin sensitivity under basal

Page 30: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

21

conditions [194]. Hence, PYY3-36 may be responsible for balancing the effects of other

gastrointestinal peptides to prevent hypoglycemia as a result of the augmented postprandial

insulin secretions.

In addition to increasing postprandial glucose levels, both PYY1-36 and PYY3-36 can inhibit

gastric emptying with PYY3-36 being more effective [195;196]. These findings suggest that

PYY3-36 can also exert suppressive effects on glucose levels secondary to gastric emptying.

Similar to GLP-1, PYY3-36 has been shown to reduce food intake in rats [174]. In response to

PYY3-36, the arcuate nucleus of the hypothalamus is activated and both the food intake and brain

activation effects are abolished in rats subjected to subdiaphragmatic vagotomy or transection of

a brainstem-hypothalamus pathway [174]. These findings suggest that PYY3-36 can regulate

energy homeostasis through a neuronal network, supporting the existence of a gut-brain neuronal

axis.

Page 31: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

22

1.4 Neuronal Regulation of Glucose Homeostasis by the Small Intestine

As mentioned, the small intestine can regulate glucose homeostasis through the actions of

gastrointestinal peptides as well as the activation of neuronal networks by nutrients. While it

appears that the mechanisms of action for gastrointestinal peptides are largely endocrine in

nature, some gastrointestinal peptides can also activate neuronal networks. For example, portal

GLP-1 can stimulate a hepatopancreatic reflex pathway to stimulate insulin secretion [169].

Similarly, portal administration of glucose can also stimulate hepatic glucose uptake and

peripheral glucose utilization [18] via neural communications.

Moreover, while some gastrointestinal peptides can regulate energy homeostasis through a gut-

brain neuronal axis, this axis is also involved in the inhibition of food intake by upper intestinal

lipids [197]. Recently, our laboratory has discovered a gut-brain-liver axis induced by duodenal

lipids to suppress glucose production to regulate glucose homeostasis [20].

In the following section, the neuronal axes involved in the regulation of glucose homeostasis

activated in the gut will be discussed.

Page 32: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

23

1.4.1 Hepatoportal Glucose Sensor

As glucose is absorbed from the intestine, glucose can enter the portal vein. It has been suggested

that as glucose enters the portal vein, the body can adopt appropriate mechanisms to compensate

for the incoming glucose. Indeed, a hepatoportal glucose sensor has been identified to stimulate

hepatic glucose uptake and peripheral glucose utilization in independent studies.

Hepatic glucose uptake

Studies on the effect of portal glucose on hepatic glucose uptake were mainly carried out by the

group of Cherrington AD. In brief, they discovered that hepatic glucose uptake is increased when

glucose is delivered intraportally in comparison to peripheral vein administration in dogs [198].

Importantly, the increase in hepatic glucose uptake was blocked when the liver was denervated

[199], suggesting that neural communications is required for the effect. The neural component of

this effect was proposed to be of parasympathetic nature based on two observations. First, it was

found that afferent discharges in the hepatic branch of the vagus nerve can be modulated by

portal glucose in guinea pigs [200]. Second, acetylcholine administration in the portal vein was

also able to stimulate hepatic glucose uptake in dogs [201]. In the liver, the glucose taken up is

disposed as glycogen in dogs [202] and rats [203]. Hence, a vagally-mediated mechanism exists

to mediate communication between the portal vein and the liver to upregulate hepatic glucose

uptake in response to portal signals.

Page 33: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

24

Peripheral glucose utilization

As mentioned, a hepatoportal glucose sensor exists to stimulate peripheral (muscle) glucose

utilization in response to portal glucose infusion in mice [18]. Since the hepatoportal glucose

sensor is connected to the hypothalamus and the nucleus of the solitary tract through hepatic

vagal afferents [204-207], a vagus-mediated neuronal network is likely in place between the

portal vein and the brain. Moreover, the effect of intraportal glucose infusion was abolished in

denervated muscles, suggesting that neuronal communication between the brain and oxidative

tissues is also necessary to increase glucose utilization [18].

While these findings formulate a basis for the portal-brain-muscle neuronal axis, the mechanism

of activation at the level of the portal vein appears to require the mediation by GLP-1 since the

effect of this neuronal axis is absent in GLP-1 receptor knockout mice [208]. At the level of the

muscle, the increase in glucose utilization is attenuated in mice lacking the expression of glucose

transporter-4 (GLUT4) or mice expressing the dominant negative form of adenosine

monophosphate kinase (AMPK) [209], suggesting that the activation of AMPK and the presence

of GLUT4 are required to increase glucose utilization by the muscles. In contrast, muscle-

specific inactivation of the insulin receptor gene had no effects on the stimulation of glucose

utilization, implying that the effect operates in an insulin-independent manner [209].

Nonetheless, a glucose-induced portal-brain-muscle axis exists to regulate glucose homeostasis

through enhancing glucose utilization by oxidative muscles.

Page 34: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

25

1.4.2 Lipid-induced gut-brain-liver axis

Recently, our laboratory has demonstrated that intraduodenal administration of lipids can trigger

a gut-brain-liver axis to suppress glucose production [20].

At the level of the duodenum, the lipid-sensing mechanism is dependent on lipid metabolism. As

lipids in the form of triglycerides enter the duodenum, the triglycerides are hydrolyzed by lipases

to release fatty acids. As fatty acids, specifically LCFAs, are released, the LCFAs are converted

into long-chain fatty acyl-coenzyme A (LCFA-CoA) by acyl-coenzyme A synthetase (ACS), and

the accumulation of LCFA-CoA in the duodenum is required to lower glucose production. This

is evident by the fact that when lipids are co-infused with the ACS inhibitor, triascin C, the

ability of upper intestinal lipids to lower glucose production was abolished. Hence, lipid-sensing

in the duodenum requires the esterification of lipid metabolites.

Further experiments demonstrated that a neuronal axis connecting the gut to the brain, then the

brain to the liver is involved. Briefly, duodenal lipids failed to lower glucose production in the

presence of tetracaine (a local anesthetic which inhibits neuronal activation), indicating that

duodenal lipids regulate glucose production in the preabsorptive state by activating

neurotransmissions. It was identified that the vagal afferents innervating the gut is responsible

for mediating the signals because intraduodenal lipid infusion in rats that received vagal

deafferentations did not lower glucose production. The critical role of the vagus nerve in

mediating the lipid-induced effect was further confirmed when glucose production remained

unchanged in response to intraduodenal administration of lipids in rats that received a

Page 35: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

26

subdiaphragmatic vagotomy. A hindbrain region known as the nucleus of the solitary tract (NTS)

was identified to be the receiver of the vagal signals [210-213]. Importantly, the principal

neurotransmitter released from vagal afferent terminals in the NTS is glutamate [214] [215].

Glutamate can act on at least 4 different classes of receptors: NMDA, α-amino-3-hydroxy-5-

methyl-4-isoxazolproprionic acid (AMPA), kainite, and metabotropic receptors [216]. In the

NTS, NMDA receptors have been localized [217], and a previous study has demonstrated that

the administration of MK-801 (N-methyl D-aspartate (NMDA) receptor blocker) in the NTS

extended meal duration [218]. As such, MK-801 was administered in the NTS concomitantly

with the administration of lipids in the duodenum to assess whether NMDA receptors in the NTS

play a role in the lipid-induced glucose production suppressive effect. It was found that duodenal

lipids had no effects on glucose production, suggesting that the gut sends signals to the NTS

through the activation of NMDA-receptors in response to lipids. Lastly, intraduodenal

administration of lipids into rats that received a hepatic vagotomy failed to lower glucose

production, demonstrating the essentiality of the hepatic vagus nerve in mediating the effect.

Hence, after the NTS receives the signals from the gut, the signals are relayed to the liver to

lower glucose production via the hepatic vagus nerve. Ultimately, these experiments revealed a

gut-brain-liver axis activated by upper intestinal lipids to regulate glucose homeostasis (Figure

1). This neuronal axis represents one of the first lines of metabolic defenses against nutrient

excess to provide metabolic balance by lowering glucose production upon nutrient exposure.

Model of Diet-Induced Insulin Resistance

Since it is clear that a gut-brain-liver axis exists to regulate glucose production in response to

duodenal lipids in normal rodents, it was further evaluated whether the effect is present in a

Page 36: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

27

model of diet-induced insulin resistance. It has previously been demonstrated that Sprague

Dawley (SD) rats can develop hepatic insulin resistance in response to three days of high fat

feeding [219]. In contrast to rodents fed on standard chow, intraduodenal administration of lipids

failed to lower glucose production in this model of diet-induced insulin resistance. Therefore, it

is essential to investigate the downstream mechanisms involved in mediating the activation of

the gut-brain-liver axis by duodenal lipids to uncover possible ways to restore the functionality

of this axis.

Page 37: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

28

Figure 1 Upper intestinal lipids activate a gut-brain-liver axis to suppress glucose production. As lipids enter the duodenum (first segment of the small intestine), lipids in the form of triglycerides are broken down by lipases to release long-chain fatty acids in the lumen. Long chain fatty acids (LCFA) are converted to long-chain fatty acyl-coenzyme A (LCFA-CoA) and the accumulation of LCFA-CoA leads to the activation of vagal afferents innervating the duodenum. The vagus nerve sends the signals to the nucleus of the solitary tract (NTS) through activating the N-methyl-D-aspartate (NMDA) receptors. The NTS then acts as a relay center and send signals through the hepatic vagus nerve to suppress glucose production.

lumen

Lamina propria

Vagus

DUODENUM lipid

Lipase

LCFA

ACS

CoA LCFA-CoA

NTS NMDA receptors

Glucose Production

Vagus

Page 38: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

29

2 General Hypothesis and Aims Cholecystokinin

Here, we identify CCK as a possible candidate for the downstream signaling events underlying

the duodenal lipid-sensing process that leads to the suppression of glucose production via a gut-

brain-liver axis. While lipids are known to inhibit food intake, there is now accumulating

evidences suggesting that lipid-induced CCK mediates the satiety effect through a neuronal

network in the preabsorptive state, suggesting that CCK can activate vagal afferents to send

signals to the brain [220;221]. In food intake studies, it has been shown that the activation of

duodenal CCK-A receptors is responsible for mediating the lipid-induced satiation.

Pharmacological blockade of CCK-A receptors by its specific antagonist MK-329 has been

shown to reduce lipid-induced satiation [222-225]. Additionally, duodenal lipids fail to lower

food intake in CCK-A receptor deficient (OLETF) rats [226;227]. Together, studies utilizing

either pharmacological or genetic means to prevent the activation of CCK-A receptors in the

presence of duodenal lipids demonstrated that CCK and CCK-A receptors play a significant role

in mediating duodenal lipid-sensing mechanisms.

Duodenal CCK-induced gut-brain-liver axis

The general hypothesis of this thesis is that duodenal CCK can activate a gut-brain-liver axis to

regulate glucose homeostasis (Figure 2). First it will be assessed whether CCK-A receptors play

a role in the regulation of glucose production. Secondly, duodenal lipids have been shown to

activate a gut-brain-liver axis to suppress glucose production while the duodenal lipid-sensing

Page 39: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

30

mechanisms remain unknown. Since CCK-A receptors have been located on capsaicin-sensitive

vagal afferents [228;229] and the duodenal lipid-induced glucose production lowering effect is

dependent on vagal afferent signaling, it is plausible that duodenal CCK/CCK-A receptor

signaling is downstream of lipid-sensing. In order for us to be able to examine whether duodenal

CCK can regulate glucose homeostasis independent of changes in glucose-regulatory hormones,

we will be utilizing clamp studies in which we will fix insulin levels at near-basal levels.

OLETF – CCK-A receptor deficient rats

One model that is available to evaluate whether CCK-A receptors are involved in the regulation

of glucose homeostasis is a rat strain known as Otsuka Long Evans Tokushima Fatty (OLETF)

rats. OLETF rats are an outbred strain of Long-Evans rats that congenitally lack a 6847 base pair

segment of the promoter region of the gene encoding CCK-A receptors [230]. Consequently,

OLETF rats are deficient in CCK-A receptors. Importantly, CCK-A receptors are involved in the

regulation of feeding behaviour, thus OLETF rats are hyperphagic from birth. When placed on a

regular chow ad libitum, OLETF rats develop glucose intolerance by week 5, obesity by week

10, and diabetes by week 24 [231-233] as compared to the LETO control rats. Of relevance to

this study, it has been shown that at week 8, OLETF rats have a higher basal hepatic glucose

production than LETO rats when fed ad libitum [234]. This finding supports our hypothesis that

duodenal CCK signaling is necessary to negatively regulate glucose production.

Since OLETF rats tend to be obese by week 10, in order to evaluate whether duodenal CCK

regulates glucose production in OLETF rats independent of weight gain, we will pair-feed the

Page 40: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

31

OLETF rats to LETO rats once we receive them from Tokushima Institute, Otsuka

Pharmaceutical at 4 weeks of age. Subsequently, we will be able to evaluate whether duodenal

CCK-A receptor plays a role in the regulation of glucose production by using OLETF rats that

are pair-fed with LETO rats.

Physiological relevance

After confirming the role of duodenal CCK on the regulation of glucose homeostasis using

clamp studies, we will evaluate whether duodenal CCK regulates plasma glucose levels in

response to fasting-refeeding in rats to address the physiological relevance of our findings. Since

we are hypothesizing that CCK acts downstream of duodenal lipid-sensing to regulate glucose

homeostasis, feeding will provide a physiological means to introduce nutrients into the

duodenum. By performing a fasting/refeeding protocol, we will be able to assess whether

blocking duodenal CCK signaling will impair the regulation of plasma glucose levels in

physiological settings. Altogether, these experiments will allow us to determine whether

duodenal CCK can regulate glucose production in normal settings.

High-fat diet model

Moreover, we will also evaluate whether duodenal CCK can regulate glucose production in the

high-fat diet-induced model of early onset insulin resistance. Based on the findings from feeding

studies, upper intestinal lipids fail to regulate energy homeostasis following adaptation to high

fat feeding [235], consistent with our previous findings in regards to glucose homeostasis. It has

Page 41: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

32

been proposed that the dysregulation of energy homeostasis by upper intestinal lipids is due to a

defect in CCK signaling. In both rats and humans, high-fat diets have been associated with

elevated levels of circulating CCK [236;237]. This observation leads to the speculation that

individuals who consume high-fat diets chronically may develop CCK insensitivity, such that an

upregulated level of CCK is required to elicit satiation in response to upper intestinal lipids. This

hypothesis is further supported by the finding that exogenous CCK reduced food intake

significantly less in rats fed with high-fat diets for 3 weeks compared with rats fed with low-fat

diets [238-240]. Hence, these findings suggest that CCK insensitivity develop following the

adaptation to high-fat feeding. In regards to the regulation of glucose homeostasis, we

hypothesize that CCK-resistance will impair the regulation of glucose homeostasis by duodenal

CCK in response to high-fat feeding.

Page 42: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

33

Hypothesis: Duodenal CCK can suppress glucose production through the activation of a gut-

brain-liver axis.

Aim 1: To determine whether the activation of CCK-A receptors in the duodenum can suppress

glucose production.

Aim 2: To examine whether duodenal CCK can stimulate a gut-brain-liver axis to suppress

glucose production.

Aim 3: To investigate whether CCK/CCK-A receptor signaling is required for duodenal lipids

to lower glucose production.

Aim 4: To confirm whether the regulation of glucose homeostasis by duodenal CCK is

physiologically relevant.

Aim 5: To determine whether duodenal CCK can lower glucose production in a model of diet-

induced insulin resistance.

Page 43: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

34

Figure 2 Working hypothesis – upper intestinal lipids stimulate CCK/CCK-A receptor signaling to suppress glucose production through a gut-brain-liver axis As lipids in the form of triglycerides enter the lumen of the duodenum as part of the ingested food, CCK is released and it binds to CCK-A receptors (CCK-AR) on the vagus nerve to send signals to the NTS by activating the NMDA receptors. The NTS acts as a relay center and send signals through the hepatic vagus nerve back to the periphery to suppress glucose production.

Vagus

lumen

Lamina propria

DUODENUM lipid

Lipase

LCFA

ACS

CoA LCFA-CoA

NTS NMDA receptors

Glucose Production

Vagus

CCK-AR CCK

Page 44: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

35

3 General Materials and Methods

*Note: All the animal study protocols were reviewed and approved by the Institutional Animal

Care and Use Committee of the University Health Network.

Chemicals

CCK-8 (Sulfated), tetracaine, and NMDA receptor blocker MK-801 were obtained from Sigma.

CCK-A receptor antagonist MK-329 was obtained from Tocris Bioscience. 20% Intralipid was

obtained from Baxter Healthcare Corporation. Stock solution of CCK-8 and MK-801 were

prepared in saline whereas stock solutions of tetracaine and MK-329 were prepared in

dimethylsulfoxide (DMSO). All stock solutions were diluted in 0.9% NaCl to the desired

concentrations.

Models

Male Sprague-Dawley Rats

9-week old Sprague-Dawley (SD) rats, weighing between 250 – 300g (Charles River

Laboratories, Montreal QC) were used for our studies. Rats were housed individually and

maintained on a standard 12-12h light-dark cycle with access to rat chow (Harlan Teklad 6%

Mouse/Rat Diet; composition: 52% carbohydrate, 31% protein and 17% fat; total calories

provided by digestible nutrients: 3.83 kcal/g) and water ad libitum. The rats were allowed to

acclimatize for 5 days upon arrival and the appropriate surgeries were then performed.

Page 45: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

36

Male LETO/OLETF Rats

4-week old Long Evans Tokushima Otsuka (LETO) and Otsuka Long Evans Tokushima Fatty

(OLETF) rats were obtained as a generous gift from Dr. Kawano (Tokushima Research Institute,

Otsuka Pharmaceuticals, Tokushima, Japan). Rats were housed individually and maintained on a

standard 12-12h light-dark cycle with water ad libitum. In order to avoid the results from being

confounded with the effects of obesity, OLETF rats were pair-fed with LETO rats to prevent the

development of obesity as described [241]. All LETO rats had access to standard chow (Harlan

Teklad 6% Mouse/Rat Diet as described) ad libitum. At 8AM each day, body weights and food

intakes of both LETO and OLETF rats were measured to ensure that the body weights were

similar. After daily food intakes of LETO rats were measured, OLETF rats were supplied with

the amount of chow equal to the prior day’s average daily chow intake of LETO rats maintained

on ad libitum access. The OLETF rats were pair-fed until they were 11-12 weeks old (280-300g),

and the appropriate surgeries were then performed.

Surgical Procedures

Duodenal and Intravenous Cannulations

Rats were first anesthetized with intraperitoneal (ip) ketamine (Ketalean; Bimeda-MTC,

Cambridge, Ontario) and xylazine (Rompun; Bayer). Duodenal cannulation surgeries were

performed as described [20;227]. Laparotomy incisions were made on the ventral midline and

the abdominal muscle wall to expose the gastrointestinal tract in the peritoneum. The pyloric

sphincter was identified and the proximal 1.5cm of the duodenum was then isolated. A 25-gauge

Page 46: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

37

needle was used to make a small puncture would on the ventral aspect of the duodenum in a

region where the vascular arcade was as sparse as possible to minimize bleeding. A saline-filled

catheter made of silicone tubing (0.040 in. ID, 0.085 in. OD; Sil-Tec, Technical Products, USA)

with a 0.5cm extension of a smaller silicone tubing (0.025 in. ID, 0.037 in. OD; Sil-Tec,

Technical Products, USA) was inserted into the duodenum at a position approximately 2cm

downstream of the pyloric sphincter for intraduodenal infusions [20]. The cannula was flushed

with saline to ensure that it was inserted into the lumen of the duodenum. The cannula was

anchored to the outer serosal surface of the proximal duodenum around the puncture wound with

a drop of 3M adhesives (Vetbond) and a 0.5-cm2 piece of Marlex mash sewn to the serosal

surface with a 6-0 silk suture. The proximal portion of the cannula exited to the abdominal cavity

through the site of the laparotomic incision and the abdominal wall incision was closed with a 4-

0 silk suture. A 2-cm midline incision was made on the skin of the back of the neck, just rostral

to the interscapular area, and the proximal portion of the duodenal cannula was then tunneled

subcutaneously to exit through the incision. All the skin incisions were closed with 4-0 silk

sutures and the proximal end of the duodenal cannula was closed with a metal pin.

For rats that were to undergo the pancreatic (basal insulin) – euglycemic clamp experiments,

immediately following the duodenal cannulation surgery, indwelling catheters were also inserted

into the right internal jugular vein and left carotid artery for infusion and blood sampling

purposes. Catheters were made of polyethylene catheters (PE 50, Clay Adams) with a 15mm

cuff-extension of Silastic tubing (Corning). Both catheters were tunneled subcutaneously and

exteriorized. The catheters were filled with 10% heparinzed saline to maintain potency of the

vascular cannula, and then closed at the end with a metal pin.

Page 47: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

38

Recovery from surgery was monitored by measuring daily food intake and weight gain for 4-6

days after surgery and the duodenal catheter was flushed with saline daily to ensure potency.

Stereotaxic Surgery

For a subgroup of rats which required stereotaxic surgeries, rats were stereotaxically implanted

with indwelling cannula (Plastics One Inc., Roanoke, VA) according to the atlas of the rat brain

as previously described [20]. In brief, rats were anesthetized with ip ketamine and xylazine then

fixed in a stereotaxis apparatus (David Kopf Instruments, Tujunga, CA) with ear bars and a nose

piece set at +5.0mm. 26-gauge stainless steel double guide cannulae were used for implantations

in the nucleus of the solitary tract with the following coordinates [20]: 0.0mm on occipital crest,

0.4mm lateral to midline, 7.9mm below skull surface. Rats were given a week of recovery time

post-stereotaxic surgery in individual cages, maintained on a standard 12h-12h light-dark cycle

with access to standard rat chow and water ad libitum, followed by duodenal and vascular

cannulation surgeries.

Selective Hepatic Branch Vagotomy

For a subgroup of rats which were subjected to hepatic branch vagotomy, the surgeries were

performed as previously described [20]. A laparotomy incision was made on the ventral midline,

followed by a second incision to open the abdominal muscle wall, exposing the gastrointestinal

tract in the peritoneum. A gastrohepatic ligament was severed using fine forceps and the stomach

was gently retracted into sterile saline soaked cotton gauze to reveal descending esophagus and

Page 48: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

39

ventral subdiaphragmatic vagal trunk. The hepatic branch of the ventral subdiaphragmatic vagal

trunk was transected by microcautery in between the two sutures, severing and cauterizing

hepatic vagus. Transecting the hepatic branch of the vagal nerve disrupts neural communications

between the liver and the brain. This also results in slightly decreased innervations to the

intestine as there are minor innervations of the hepato-duodenal sub-branch which supplies a

small portion of the intestine. Following the vagotomy surgery, duodenal and vascular

cannulation surgeries were immediately performed.

High-Fat Feeding in Male SD Rats

A subgroup of male SD rats, with duodenal and vascular catheters implanted, was placed on a

lard-oil enriched high-fat diet ad libitum for 3 days (see Table 1 for diet composition of standard

chow and high-fat diet). It was first confirmed that the rats on high fat diet consumed at least the

same amount of calories as the average calories consumed for the rats on regular chow which

were used for other clamp experiments. A pancreatic basal insulin clamp was then performed on

these rats to address whether intestinal CCK regulates glucose production in the early onset of

diet-induced insulin resistance in rodents.

Page 49: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

40

Intraduodenal Infusions and Treatments

The following substances were continuously infused to the duodenum through the duodenal

catheter at t = 150 – 200 min (0.01 mL/min):

1.) saline

2.) CCK-8 (35 pmol/kg/min; 100 pmol/kg/min)

3.) CCK-8 (35 pmol/kg/min) + tetracaine (0.01 mg/min)

4.) CCK-8 (35 pmol/kg/min) + CCK-A receptor antagonist MK-329 (1.6 µg/kg/min; 3.2

µg/kg/min)

5.) 20% Intralipids (0.03 kcal/min)

6.) 20% Intralipids (0.03 kcal/min) + MK-329 (1.6 µg/kg/min).

Since this is one of the first studies to examine the biological function of duodenal CCK-8 in

rats, we used an infusion rate that is analogous to the ip dose of CCK-8 at 1750 pmol/kg which

was found to suppress food intake in SD rats [242]. It was determined that CCK-8 was to be

infused at a rate of 35 pmol/kg/min (or 1750 pmol/kg in 50 min) as this rate is analogous to the

ip dose of CCK-8 at 1750 pmol/kg which was found to suppress food intake in SD rats [55]. This

infusion was further justified by a previous study which indicated that intraduodenal CCK-8

administered at 30 or 100 pmol/kg/min stimulates pancreatic secretion in calves independent of

changes in circulating CCK levels [243].

Page 50: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

41

Fasting-Refeeding Experiments

For male SD rats, five days post-duodenal surgery, rats whose daily food intake and body weight

had recovered back to baseline underwent the fasting-refeeding protocol. Rats were fasted for 40

hours (fast began at 5:00 PM on Day 5 until 9:00 AM on Day 7). Ten minutes prior to the

completion of the 40-hour fast (i.e. 8:30 AM on Day 7, t = -10), a continuous intraduodenal

infusion (Harvard Apparatus PHD 2000 infusion pumps) of either (i) saline or (ii) MK-329 (3.2

µg/kg/min) was initiated, which lasted until t = 20. Upon completion of the 40-hour fast (i.e.

9:00 AM on Day 7, t = 0), rats were allowed to consume a regular chow diet ad libitum. Blood

glucose levels and food intake were measured at t = -10, 0, 10, 20 min.

On the other hand, male LETO and OLETF rats did not receive any surgeries. Similar to the

male SD rats, they were fasted for 40 hours. Upon completion of the 40-hour fast, LETO and

OLETF rats were allowed to consume a regular chow diet ad libitum. Blood glucose levels and

food intake were measured at t = 0, 10, 20 min.

Clamp Procedure

Rats were restricted to ~58 kcal of caloric intake the night prior to the in vivo infusion

experiments. The infusion experiments spanned a total of 200 minutes. At the onset of the

experiment, a primed continuous infusion of [3-3H]-glucose (bolus 40 µCi; 0.4 µCi/min; all

infusion performed with Harvard Apparatus PHD 2000 infusion pumps) was initiated (t = 0 min)

and maintained throughout the protocol to assess glucose kinetics based on the tracer-dilution

methodology.

Page 51: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

42

At t = 90 min, a pancreatic (basal insulin) clamp was initiated by providing a continuous infusion

of insulin (0.8 mU/kg/min) and somatostatin (3 µg/kg/min) to inhibit endogenous insulin and

glucagon secretions. To maintain euglycemia, 25% glucose was provided at a variable rate to

maintain plasma glucose levels that are comparable to basal (t = 60 – 90 min), and the rates were

adjusted at 10-min intervals from t = 120 – 200 min. For experiments which required the

administration of MK-801 (0.03 ng/min; CMA/400 syringe microdialysis infusion pumps) to the

NTS, the infusion began at t = 90 min and lasted until t = 200 min. Intraduodenal infusions were

initiated at t = 150 min and lasted until t = 200 min to determine the effects of different duodenal

treatments on glucose kinetics. Blood samples were taken at 10-min intervals to determine the

specific activity of [3-3H]-glucose between t = 60 – 90 min (basal) and t = 180 – 200 min

(clamp) to assess glucose kinetics. Additional blood samples were collected at t = 90 min (basal)

and t = 180 and 200 min to determine plasma insulin levels. All the blood samples were

subjected to centrifugation at 6000 rpm to separate the plasma and the biochemical analyses were

performed as described below.

Peripheral and Portal CCK Measurements

A commercially available radioimmunoassay kit (Alpco) was used for the measurement of CCK

concentrations in the peripheral and portal circulation. According to the manufacturer’s

specifications, 1.0 mL of plasma was required to obtain a precise CCK measurement. Therefore,

a separate set of experiments was required to collect sufficient plasma for the measurement of

CCK. In this set of experiments, SD rats underwent both the duodenal and vascular cannulation

surgeries. Following a recovery period, they received either saline or CCK-8 in the duodenum to

Page 52: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

43

determine whether duodenal saline or CCK-8 has any effect on peripheral and portal CCK

concentrations. The infusion lasted for 50 minutes to mimic the protocol used in the clamp

studies. At t = 50 min, 2.0 mL of blood was collected. The rats were anesthetized immediately

using ketamine (5 mg IV) and 2.0 mL of portal blood was extracted from the portal vein. The

blood samples were put in tubes containing 1 unit of ethylenediaminetetraacetic acid (EDTA;

(Phoenix Pharmaceuticals) and 1 unit of aprotinin (Phoenix Pharmaceuticals) and plasma was

separated by centrifugation at 6000 rpm for 5 minutes at 4ºC. Plasma samples were stored in -

20ºC until the radioimmunoassay was performed.

Biochemical Analyses

Plasma Glucose

Plasma glucose concentrations were measured with the use of a glucose analyzer (Glucose

Analyzer GM9, Analox Instruments, Lunenbertg, MA). The analyzer was calibrated before

usage in each experiment. Blood samples of rat (~0.1mL) were centrifuged at 6000 rpm to

separate the plasma. A 10 µL sample of plasma was immediately injected into the glucose

analyzer to determine plasma glucose concentration. The glucose analyzer determines glucose

concentrations by the glucose oxidase method, which is based on the following reaction:

Under the assay conditions, the rate of oxygen consumption is directly proportional to the plasma

glucose concentration. Therefore, oxygen consumption is measured in the glucose analyzer with

a polarographic oxygen sensor to determine the plasma glucose concentration. Specifically,

Clark-type amperometric oxygen electrodes are immersed in the sample with a potential applied

β-D-glucose + O2 D-Gluconic acid + H2O2 Glucose oxidase

Page 53: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

44

between them that is sufficient to reduce dissolved oxygen at the working electrode. Through

this, the partial pressure of oxygen in the sample can be measured given that it is proportional to

the limiting current.

Plasma Glucose Tracer Specific Activity

The specific activity of the [3-3H] in plasma was determined using 50-µL samples of plasma.

The plasma samples were deproteinized with Ba(OH)2 and ZnSO4, and centrifuged at 6000 rpm

for 7 minutes at 4ºC. The protein-free supernatant was kept. Since tritium on the C-3 position of

glucose is lost to water during glycolysis, the supernatant was evaporated to dryness to remove

the tritiated water. Scintillation fluid was added to the dried sample and the liquid scintillation

counting would represent radioactivity from the [3-3H]-glucose in the plasma only.

Plasma Insulin

Plasma insulin levels were determined by radioimmunoassay (RIA) using a 2-days commercial

rat insulin RIA kit (100% specificity) from Linco Research (St. Charles, MO). The principle of

RIA is based on antigen-antibody binding. In brief, insulin from the plasma samples or standards

competes with the labeled tracer antigen (125I-labeled insulin) to bind with the antibodies raised

against insulin (guinea pig anti-rat insulin antibody). As the reaction occurs, 125I-labeled insulin

binds in a reverse proportion to the concentration of insulin in standards and samples. Antibody-

bound labeled tracer antigens (125I-labeled insulin) are separated from the unbound fraction using

double antibody solid phase. The radioactivity of the bound fraction is measured in a gamma

Page 54: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

45

counter. The radioactivity counts (B) for the samples and the standards are expressed as a

percentage of the mean counts of the total binding reference tubes (B0):

The percent activity bound for each standard is plotted against the known concentration to

construct a standard curve. Finally, the concentration of insulin in the samples can be determined

by interpolation.

Specifically, a 2-days protocol provided by the supplier was used. 125I-labeled insulin (50 µL)

and rat insulin antibody (50 µL) were added to 50 µL of experimental samples and standards in a

range of concentrations (0.1, 0.2, 0.5, 1.0, 2.0, 5.0 and 10.0 ng/mL), followed by vortexing. After

an overnight incubation at 4ºC, 1.0mL of precipitating reagent was added to each tube followed

by vortexing and incubation at 4ºC for 20 minutes. The samples were centrifuged to pellet the

bound insulin. The radioactivity of the pellet was counted by a gamma counter (Perkin Elmer

1470). A standard curve was constructed using the method as described previously and the

concentration of insulin in the samples was determined by interpolation.

Plasma CCK

Peripheral and portal CCK levels were measured using a commercially available

radioimmunoassay kit (Alpco). This RIA kit follows the same principle as described in the

plasma insulin section. Briefly, CCK from the peripheral and portal plasma samples or standards

competes with the labeled tracer antigen (125I-labeled CCK) to bind with the antibodies raised

against CCK (rabbit anti-CCK antibody). As the reaction occurs, 125I-labeled CCK binds in a

% total binding = % = x 100% B0 B

Sample or standard B0

Page 55: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

46

reverse proportion to the concentration of CCK in standards and samples. Antibody-bound

labeled tracer antigens (125I-labeled CCK) are separated from the unbound fraction using double

antibody solid phase. The radioactivity of the bound fraction is measured in a gamma counter.

The radioactivity counts (B) for the samples and the standards are expressed as a percentage of

the mean counts of the total binding reference tubes (B0). A standard curve is constructed by

plotting the percent activity bound for each standard against the known concentration. Finally,

the concentration of CCK in the samples is determined by interpolation.

Prior to the performance of the RIA, CCK was first extracted from the plasma samples to

eliminate non-specific interference from plasma proteins. 2.00 mL of 96% ethanol was added to

the plasma samples, vortexed. The samples were allowed to incubate for 10 minutes, followed by

centrifugation at 1,700g for 15 minutes. The CCK-containing supernatant was kept and allowed

to dry by using a Speed Vac (Savant SPD 131 DDA) overnight. The dry extracts were dissolved

back into solution by adding 1.00 mL of diluent. The dissolved samples were vortexed and

incubated for 30 minutes in room temperature. A recovery control for the extraction procedure

was also included by adding 200 µL of CCK-standard (50 pmol/L) to 800 µL to separate plasma

samples, making the final concentration 10 pmol/L. 200 µL of diluent was added to another 800

µL of plasma sample and both of these samples underwent the same extraction procedure and

RIA with the experimental samples concurrently. The inclusion of the recovery controls allowed

for the determination of the recovery rate of the extraction procedure, which could be calculated

by the following formula:

Page 56: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

47

After calculating the % recovery from the recovery controls, the % recovery was used to correct

the concentration of CCK in the samples as determined by the RIA.

For the RIA, a 7-days protocol provided by the supplier was used. 500 µL of 125I-labeled CCK

and 500 µL of anti-CCK were added to 200 µL experimental samples and standards in a range of

concentrations (0.78, 1.56, 3.12, 6.25, 12.5, 25 pmol/L). The tubes were vortexed and allowed to

incubate for 4 days at 4ºC. 100 µL of precipitating reagent was added to each tube followed by

vortexing and incubation at 4ºC for an hour. The samples were centrifuged to pellet the bound

CCK. The radioactivity of the pellet was counted by a gamma counter (Perkin Elmer 1470). A

standard curve was constructed and the final concentration of CCK in the samples was

determined by interpolation and accounting for the % recovery.

Calculations

As described previously, a radioactive [3-3H]-glucose tracer is infused during the clamp

experiments in order to determine glucose production and uptake in our experimental animals

using the steady state formula. The [3-3H]-glucose tracer was infused at a constant rate into the

rat and an hour was given to allow for equilibration of the tracer glucose with the glucose in the

body. Under steady-state basal condition, the rate of glucose uptake (Rd) equals the rate of

glucose appearance (Ra), which is the same as the rate of the endogenous glucose production.

Therefore, using the steady state formula, the rate of glucose uptake and glucose appearance can

% recovery = x 100% pmol/L found with addition – pmol/L found without addition

10 pmol/L

Page 57: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

48

be determined by dividing the [3-3H]-glucose infusion rate by the specific activity of the plasma

[3-3H]-glucose:

During the pancreatic clamp settings where exogenous glucose was infused to maintain

euglycemia, the rate of endogenous glucose production was obtained from the difference

between Rd and the rate of glucose infusion.

Statistical Analysis

Data were presented as means ± standard errors of the mean. Statistical differences between

groups were determined by either analysis of variance (ANOVA) followed by Tukey’s test or the

unpaired Student’s t-test as appropriate with a probability of p < 0.05 accepted as significant. For

pancreatic clamp experiments, the time period t = 60 – 90 min and t = 180 – 200 min were

averaged for the basal and clamp condition respectively.

Ra = Rd = Constant tracer infusion rate (µCi/min)

Specific activity (µCi/mg)

Page 58: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

49

Calories Provided Regular Chow Diet High-Fat Diet

Carbohydrate (%) 52 45

Protein (%) 31 22

Fat (%) 17 33

Saturated 6.2 14.1

Monounsaturated 7.1 13.2

Polyunsaturated 3.5 6.1

Total calorie provided (kcal/g) 3.83 5.14

Table 1 Dietary contents of the regular chow and the lard-oil enriched high fat diet.

Page 59: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

50

4 Results

Duodenal CCK-A receptor activation can suppress glucose

production

During the clamp studies (180 – 200 min), when peripheral circulating plasma insulin was

maintained at near basal levels, intraduodenal CCK-8 administration (Figure 3A, B) significantly

increased the exogenous glucose infusion rate required to maintain euglycemia in comparison to

saline (p < 0.05) (Figure 4A). Based on the steady-state tracer data (180-200 min), this was

selectively attributed to a significant reduction in glucose production (Figure 4B,C) in rats that

received CCK-8 (p < 0.001) with no changes in glucose uptake (Figure 4D). In another set of

rats, we performed intraduodenal CCK-8 (35 pmol/kg/min) administration for 50 min and found

plasma CCK levels did not change at 50 min. Portal vein samples were then taken soon after

giving anesthesia at 50 min. It was found that CCK levels in the portal vein (5.1 ± 0.6 pM) was

~2.2-fold higher than the plasma CCK levels obtained in duodenal saline-infused rodents,

without reaching statistical significance. Importantly, portal CCK levels of duodenal CCK-8

infused rats (3.4 ± 0.6 pM) were comparable to those found in the saline-infused rats. Thus, a

primary increase of CCK-8 in the duodenum lowers glucose production independent of changes

in circulating CCK or insulin levels (Table 2).

Next, we co-administered the CCK-A receptor inhibitor MK-329 (3.2 μg/kg/min) with CCK-8

into the duodenum (Figure 3A) to inhibit CCK-A receptors in the gut in the presence of CCK

activation. In the presence of MK-329, the effects of duodenal CCK-8 administration on glucose

Page 60: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

51

infusion rate and glucose production were abolished, with no effects on glucose uptake.

Importantly, MK-329 (a competitive inhibitor for CCK-A receptors) alone also had no effects on

glucose infusion rate, glucose production, and glucose uptake when given alone (Figure 4A-D).

The abolishment of the glucose production suppression effect of duodenal CCK in the presence

of the CCK-A receptor inhibitor suggests that CCK-A receptor activation is required for the

potent control of gut CCK-8 on glucose production.

Duodenal CCK fails to lower glucose production in CCK-A receptor

deficient rats

After identifying that CCK-A receptor activation can lower glucose production via a

pharmacological approach, we wished to confirm this finding in OLETF rats, which are rats with

congenital CCK-A receptor deficiency (Figure 5A). Since these rats are known to be

hyperphagic, actions were taken in order to evaluate whether duodenal CCK-8 regulate glucose

production in OLETF rats independent of weight gain. Upon the arrival of the OLETF rats from

Tokushima Institute, Otsuka Pharmaceutical at 4 weeks of age, we began pair-feeding the

OLETF rats with the LETO rats. As shown in Figure 6A, we successfully maintained similar

body weights between LETO and OLETF rats by pair-feeding (Figure 6B). After ensuring that

the OLETF rats were not obese in comparison to LETO rats, we performed the same set of

experiments in the OLETF and LETO rats as in SD rats.

Initially, we administered CCK-8 to the duodenum of LETO rats at 35 pmol/kg/min (same rate

as in SD rats; Figure 5A,B). However, there were no changes in glucose infusion rate, glucose

Page 61: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

52

production and glucose uptake. Therefore, we doubled the dosage of CCK-8 to 70 pmol/kg/min,

and there were still no effects on glucose infusion rate, glucose production and glucose uptake.

Finally, we administered CCK-8 to the duodenum of LETO rats at 100 pmol/kg/min, and an

increased glucose infusion rate was required to maintain euglycemia as compared to

intraduodenal saline-treated LETO rats (p < 0.01) (Figure 6C). This was accounted for by a

reduction in glucose production (p < 0.05) (Figure 6D,E), with no changes in glucose uptake

(Figure 6F). In contrast, intraduodenal CCK-8 administrations in OLETF rats had no effects on

the glucose infusion rate required to maintain euglycemia (Figure 6C). There were also no

changes in glucose production (Figure 6D,E) and glucose uptake (Figure 6F) in response to

intraduodenal CCK-8 administrations in OLETF rats.

It is unclear why LETO rats required a higher dosage of CCK-8 to have an effect on glucose

production than SD rats. However, a previous study demonstrated that in the duodenum, the

level of CCK-A receptor gene expression is significantly higher in SD rats than LETO rats [244]

indicating that LETO rats may have a lower protein expression of CCK-A receptors in the

duodenum. Hence, a possible reason for the increased dosage of CCK-8 required for LETO rats

to lower glucose production as compared to SD rats is that there may be potentially reduced

availability of CCK-A receptors, suggesting that LETO rats may be less sensitive to CCK-8 in

the duodenum than SD rats.

Nonetheless, both the pharmacological loss-of-function in SD rats and genetic loss-of-function

experiments in CCK-A receptor-deficient rats indicate that CCK-A receptor activation is

sufficient for CCK action in the duodenum to lower glucose production.

Page 62: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

53

Duodenal CCK can activate a gut-brain-liver axis to regulate

glucose production

Subsequently, we examined whether a gut-brain-liver axis is involved in duodenal CCK

signaling. First, we co-infused CCK-8 with tetracaine to determine whether duodenal innervation

is required for duodenal CCK to lower glucose production (Figure 7A,B). As shown in Figure

8A-D, duodenal CCK-8 administration had no effects on glucose infusion rate, glucose

production and glucose uptake in the presence of tetracaine. Importantly, the administration of

tetracaine alone had no effects on all parameters. Taken together, these results confirmed our

prediction that duodenal CCK regulates glucose production in the preabsorptive state by

activating neurotransmissions.

Next, we examined whether NMDA receptor activation in the NTS is necessary for duodenal

CCK to lower glucose production. We found that the effects of duodenal CCK-8 administration

on glucose infusion rate and glucose production were completely abolished when NMDA

receptor activation in the NTS is prevented by the administration of MK-801 (Figure 8A-C).

Consistently, glucose uptake was unchanged with concomitant administrations of CCK-8 in the

duodenum and MK-801 in the NTS (Figure 8D). MK-801 administration in the NTS alone had

no effects on all of the parameters. Hence, these data suggest that NMDA receptors in the NTS

play a role in mediating the glucose production suppression effect elicited by duodenal CCK.

After confirming that signals are transmitted from the gut to the brain, we examined whether the

hepatic vagus nerve plays a role in transmitting signals from the brain to the liver to lower

glucose production. We observed that the effects of duodenal CCK-8 administration on glucose

Page 63: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

54

infusion rate and glucose production were abolished in rats that received a hepatic vagotomy

(Figure 8A-C), with no changes in glucose uptake in comparison to other groups (Figure 8D).

Notably, hepatic vagotomy had no effects on all of the parameters (Figure 8A-D). Together, our

results indicate that duodenal CCK activates a gut-brain-liver axis to regulate glucose

production.

Duodenal CCK-A receptor activation is required for lipids to trigger

a gut-brain-liver axis to regulate glucose production

Since CCK-A receptors have been implicated to play a role in mediating the lipid-induced gut-

brain axis to regulate energy homeostasis, we proposed that CCK-A receptors might also play a

role in mediating the lipid-induced gut-brain-liver axis to regulate glucose homeostasis (Figure

9A,B). First, we confirmed that duodenal lipids upregulated the glucose infusion rate required to

maintain euglycemia (p < 0.01) (Figure 10A), which is fully accounted by a suppression in

glucose production (p < 0.01) (Figure 10B,C) with no effects on glucose uptake (Figure 10D)

within the same timeframe as seen in CCK-infused experiments. We then co-infused lipids with

MK-329 into the duodenum to determine whether CCK-A receptor activation is required for

lipids to lower glucose production. In the presence of the CCK-A receptor inhibitor, duodenal

lipids had no effects on glucose infusion rate, glucose production and glucose uptake (Figure

10A-D). Hence, these data support the hypothesis that duodenal CCK-A receptor activation is

essential in mediating the lipid-induced gut-brain-liver axis to suppress glucose production.

Page 64: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

55

Duodenal CCK-A receptors regulate plasma glucose levels in

physiological settings

To this point, we have demonstrated that duodenal lipid sensing can result in a reduction of

glucose production through the release of CCK which would activate a gut-brain-liver axis

through binding to duodenal CCK-A receptors. It is now important to evaluate whether these

findings are physiologically relevant by determining if CCK-A receptors are involved in the

regulation of plasma glucose levels in response to nutrient consumption.

To address this question, we performed a fasting-refeeding protocol, which allowed nutrients to

be delivered to the duodenum through a physiological mechanism to stimulate CCK release. Five

days after duodenal cannulation surgeries, the rats were subjected to a 40-hour fast (Figure 11A).

Following the fasting, we began an intraduodenal infusion of either saline or MK-329 at t = -10

min, during which the plasma glucose levels were at ~110 mg/dL (Figure 11B). At time 0 min,

we allowed the rats to feed ad libitum on regular chow, and plasma glucose levels remained the

same (Figure 11B). In response to feeding, both plasma glucose level and cumulative food intake

(Figure 11B,C) increased in 10 and 20 min of feeding for the intraduodenal saline-infused rats.

Interestingly, the plasma glucose levels in rats that received intraduodenal CCK-A receptor

blocker MK-329 were significantly higher than saline-treated rats by ~20 mg/dL at t = 20 min (p

< 0.01) (Figure 11B) despite a similar cumulative food intake (Figure 11C). Therefore, the

results of these pharmacological experiments suggest that duodenal CCK-A receptors in the

duodenum are involved in the regulation of glucose homeostasis in physiological settings.

Page 65: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

56

Furthermore, we performed the same set of fasting-refeeding experiments in the CCK-A receptor

deficient OLETF rats and their corresponding LETO controls (Figure 12A). Similar to the

previous set of experiments, OLETF rats had significantly higher plasma glucose levels (Figure

12B) in comparison to LETO rats in response to 20 minutes of feeding by ~20 mg/dL (p < 0.01)

despite comparable cumulative food intakes in both groups (Figure 12C). Hence, these results

from genetic loss-of-function experiments further confirm that the activation of duodenal CCK-

A receptors physiologically regulate glucose homeostasis.

Duodenal CCK fails to lower glucose production in response to

high-fat feeding

Previously our laboratory has demonstrated that duodenal lipids fail to activate the gut-

brain-liver axis to lower glucose production in a model of early-onset diet-induced insulin

resistance. On the other hand, we have demonstrated in the current study that the activation of

CCK-A receptor is downstream of gut lipids to lower glucose production via the gut-brain-liver

axis in normal rodents. Therefore, it is now of interest to determine whether the activation of

CCK-A receptors directly through intraduodenal administration of CCK-8 can restore the

activity of the gut-brain-liver axis to regulate glucose production in the same disease model

(Figure 13A,B). Consistent with the findings above, intraduodenal CCK-8 administration

increased the glucose infusion rate (p < 0.05) (Figure 14A) and lowered glucose production (p <

0.001) (Figure 14B,C) in comparison to saline-treated rats when fed on a regular chow diet. In

contrast, intraduodenal CCK-8 administration had no effects on glucose infusion rate (Figure

14A) and glucose production (Figure 14B,C) in comparison to saline-treated rats when fed on a

Page 66: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

57

high-fat diet for 3 days. In all treatment groups, glucose uptake remained unchanged (Figure

14D). Hence, these data suggest that high-fat feeding induces duodenal CCK-resistance, leading

to a disruption in the regulation of glucose homeostasis.

Page 67: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

58

Figure 3 Schematic representation of the working hypothesis – duodenal CCK activates CCK-A receptors to regulate glucose production and experimental design (pharmacological approach). (A) Proposed model for a duodenal CCK-A receptor pathway to lower hepatic glucose production. CCK-8 is an activator of CCK-A receptors and MK-329 is an antagonist of CCK-A receptors that prevents the activation of the receptors. Intraduodenal administration of CCK-8 lowers glucose production while MK-329 abolishes the effect. (B) Schematic representation of experimental design: intravenous, intra-arterial and intraduodenal catheters were implanted on male Sprague Dawley rats (~250 – 300g). Rats were given 7 recovery days until clamp studies upon which intraduodenal infusion of saline, CCK-8 ± MK-329 were given.

clamp

Intraduodenal SAL or

CCK-8 ± MK-329

Day 1 7

0

90 150 200

[3-3H]-Glucose (0.4 μCi/min)

Insulin (0.8 mU/kg/min)

SRIF (3 μg/kg/min)

Glucose (as needed)

vascular & duodenal catheters

B

A CCK-AR

SAL or CCK-8 ± MK-329

Vagal afferent

MK-329

CCK-8

GP

Page 68: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

59

Figure 4 Duodenal CCK activates CCK-A receptors to suppress glucose production (A and B) During the pancreatic clamp (180-200 min), intraduodenal CCK-8 (35 pmol/kg/min; n = 6) infusion increased the glucose infusion rate required to maintain euglycemia (A; *p < 0.05 versus SAL [n = 7]) which was associated with a reduction in glucose production (B; *p < 0.001 versus SAL, MK-329 [n = 7] and CCK-8 + MK-329 [n = 6]). In contrast, coinfusion with MK-329 abolished the effects of CCK-8 on glucose infusion rate (A) and glucose production (B). (C) Suppression of glucose production during the clamp period (180-200 min) expressed as the percentage reduction from basal steady state (60-90 min) glucose production. (*p < 0.01 versus all other groups ). (D) Glucose uptake was unchanged in all groups. Values are shown as mean ± SEM.

A *

*

*

B

C D

Page 69: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

60

Figure 5 Schematic representation of the working hypothesis – duodenal CCK activates CCK-A receptors to regulate glucose production and experimental design (molecular approach). (A) Proposed model for a duodenal CCK-A receptor pathway to lower hepatic glucose production. CCK-8 is an activator of CCK-A receptors. OLETF rats are CCK-A receptor deficient such that intraduodenal administration of CCK-8 fails to lower glucose production. (B) Schematic representation of experimental design: intravenous, intra-arterial and intraduodenal catheters were implanted on male LETO and OLETF rats (~250 – 300g). Rats were given 7 recovery days until clamp studies upon which intraduodenal infusion of saline or CCK-8 were given.

CCK-8

CCK-AR

GP

SAL or CCK-8

Vagal afferent

CCK-AR deficient rat (OLETF)

clamp

Intraduodenal SAL or CCK-8

Day 1 7

0

90 150 200

[3-3H]-Glucose (0.4 μCi/min)

Insulin (0.8 mU/kg/min)

SRIF (3 μg/kg/min)

Glucose (as needed)

vascular & duodenal catheters

B

A

Page 70: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

61

Figure 6 Duodenal CCK suppresses glucose production in LETO but not in CCK-A receptor deficient OLETF rats (A and B) The body weight of OLETF rats (n = 19) was kept at the same as LETO rats (n = 20) through pair-feeding. (C and D) Intraduodenal CCK-8 infusion (100 pmol/kg/min) increased the glucose infusion rate required to maintain euglycemia during the clamp in LETO rats but not in OLETF rats (C; *p < 0.01 versus all groups) (LETO: SAL [n = 5], CCK-8 [n = 6]; OLETF: SAL [n = 4], CCK-8 [n = 5]). No changes were observed in the glucose infusion rate (C) and glucose production (D) for OLETF rats in response to intraduodenal CCK-8 infusion. (E) Suppression of glucose production during the clamp period (180-200 min) expressed as the percentage reduction from basal (60-90 min) glucose production (*p < 0.01 versus other groups). (F) Glucose uptake was unchanged in all groups. Values are shown as means ± SEM

A

C

B

D

* *

E F

*

Page 71: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

62

Figure 7 Schematic representation of the working hypothesis – duodenal CCK activates a gut-brain-liver axis to suppress glucose production and experimental design (A) Proposed model for a gut-brain-liver axis activated by duodenal CCK to lower hepatic glucose production. CCK-8 is an activator of CCK-A receptors. Tetracaine is a local anesthetic that prevents neuronal activations. MK-801 is a potent NMDA receptor antagonist that prevents the activation of the receptors. A separate group of rats received hepatic vagotomy. Intraduodenal administration of CCK-8 fails to lower glucose production in the presence of tetracaine in the duodenum, MK-801 in the NTS, or rats subjected to a hepatic vagotomy. (B) Schematic representation of experimental design: Stereotaxic surgeries were performed on male SD rats (~250 – 300g) 7 days prior to vascular and duodenal cannulations. Rats were given 7 recovery days after the vascular and duodenal cannulation surgeries until clamp studies upon which intraduodenal infusion of saline or CCK-8 ± tetracaine/NTS MK-801 were given.

CCK-8

CCK-AR

SAL or CCK-8 ± tetracaine

GP

NTS MK-801

B

A

NTS cannula clamp

Day 1 13

Intraduodenal SAL or

CCK-8 ± tetracaine

0

90 150 200

[3-3H]-Glucose (0.4 μCi/min)

Insulin (0.8 mU/kg/min)

SRIF (3 μg/kg/min)

Glucose (as needed)

NTS MK-801

Vascular & duodenal catheters (hepatic vagotomy)

7

Page 72: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

63

Figure 8 Duodenal CCK can activate a gut-brain-liver axis to regulate glucose production (A and B) Intraduodenal CCK-8 infusion increased the glucose infusion rate required to maintain euglycemia (A; *p < 0.01 versus other groups) and decreased glucose production (B; *p < 0.01 versus other groups). Rats that received tetracaine, NTS MK-801 or hepatic vagotomy failed to suppress glucose production in response to intradudoenal CCK-8 infusions. (SAL [n = 5], CCK-8 [n = 6], CCK-8 + tetracaine [n = 5], CCK-8 + NTS MK-801 [n = 5], CCK-8 + HVAG [n = 6]). (C) Suppression of glucose production during the clamp period (180-200 min) expressed as the percentage reduction from basal (60-90 min) glucose production (*p < 0.05 versus other groups). (D) Glucose uptake was unchanged in all groups. Intraduodenal tetracaine (n = 7), intra-NTS MK-801 (n = 5) or HVAG (n = 6) alone did not affect glucose kinetics. Values are shown as means ± SEM

A B

C D

*

*

*

Page 73: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

64

Figure 9 Schematic representation of the working hypothesis – duodenal CCK signaling is downstream of lipid-sensing to regulate glucose production and experimental design (A) Proposed model for the necessity of CCK-A receptor activation in response to duodenal lipids to lower glucose production. MK-329 is a CCK-A receptor blocker that prevents the activation of the receptors. Intraduodenal administration of Intralipids fails to lower glucose production in the presence of MK-329 in the duodenum. (B) Schematic representation of experimental design: vascular and duodenal catheters were implanted on male SD rats (~250 – 300g). Rats were given 7 recovery days until clamp studies upon which intraduodenal infusion of vehicle or Intralipids ± MK-329 were given.

CCK

CCK-AR

GP

lipid

clamp

Intraduodenal SAL or lipid ± MK-329

Day 1 7

0

90 150 200

[3-3H]-Glucose (0.4 μCi/min)

Insulin (0.8 mU/kg/min)

SRIF (3 μg/kg/min)

Glucose (as needed)

vascular & duodenal catheters

B

A

VEH or Lipid ± MK-329

Page 74: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

65

Figure 10 Duodenal CCK-A receptor activation is required for lipids to lower glucose production (A and B) Intraduodenal lipids increased the glucose infusion rate (A, *p < 0.01 versus all other groups), and decreased glucose production rate (B, *p < 0.001 versus all other groups) required to maintain euglycemia. The presence of MK-329 abolished the ability of duodenal lipids to suppress glucose production. (VEH; saline + MK-329 alone [n = 11], lipid [n = 10], lipid + MK-329 [n = 7]). (C) Suppression of glucose production during the clamp period (180-200 min) expressed as the percentage reduction from basal (60-90 min) glucose production (*p < 0.01 versus other groups). (D) Glucose uptake was unchanged in all groups. Values are shown as means ± SEM

A B

C D

*

*

*

Page 75: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

66

Figure 11 Pharmacological inhibition of CCK-A receptors in the gut disrupts glucose homeostasis during refeeding (A) Schematic representation of the experimental design. Duodenal cannulation was performed on male SD rats (~250 – 280g). Rats were given 5 recovery days until they were subjected to a 40-hr fast. Ten minutes prior to the completion of the fast, an intraduodenal administration of either saline or MK-329 was initiated. Rats were refed on regular chow ad libitum at time 0 min, and food intake and blood glucose levels were monitored at -10, 0, 10 and 20 min. (B) Prior to refeeding, plasma glucose levels were comparable in all rats. After 20 min of refeeding, rats that received MK-329 had significantly higher plasma glucose levels compared to those that received saline. (C) Cumulative food intake was comparable in all groups. Saline (n = 7), MK-329 (n = 6). * p < 0.01 versus saline. Values are shown as means ± SEM

Refed

Day 1 Day 7 at 9am

-10 min 0 60

Intraduodenal SAL or MK-329

Day 5 at 5pm

Fasted

Refed

SD rats

Duodenal catheter

A

B C *

Page 76: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

67

Figure 12 Molecular inhibition of CCK-A receptors in the gut disrupts glucose homeostasis during refeeding (A) Schematic representation of the experimental design. Rats were were subjected to a 40-hr fast followed by refeeding. Rats were refed on regular chow ad libitum at time 0 min where food intake and blood glucose levels were monitored at 0, 10 and 20 min. (B) Prior to refeeding, plasma glucose levels were comparable in all rats. After 20 min of refeeding, CCK-A receptor deficient OLETF rats had significantly higher plasma glucose levels compared to LETO rats. (C) Cumulative food intake was comparable in all groups. LETO (n = 6), OLETF (n = 9). * p < 0.01 versus LETO. Values are shown as means ± SEM

Refed

Day 1 at 5pm Day 3 at 9am

0 min 60

Refed

LETO/OLETF rats

fasted

B C

A

*

Page 77: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

68

Figure 13 Schematic representation of the working hypothesis – duodenal CCK fails to suppress glucose production in response to high fat feeding and experimental design (A) Proposed model for determining whether duodenal can regulate glucose production in response to 3 days of high fat feeding. After being placed on a lard-enriched high fat diet, intraduodenal administration of CCK-8 fails to lower glucose production. (B) Schematic representation of experimental design: vascular and duodenal catheters were implanted on male SD rats (~250 – 300g). Rats were placed on the regular chow for 4 days, which was replaced by high-fat diet for three days until clamp studies upon which intraduodenal infusion of saline ± CCK-8 were given.

CCK-8

CCK-AR

SAL or CCK-8

GP

HFD ?

clamp

Intraduodenal SAL ± CCK-8

Day 1 7

0

90 150 200

[3-3H]-Glucose (0.4 μCi/min)

Insulin (0.8 mU/kg/min)

SRIF (3 μg/kg/min)

Glucose (as needed)

vascular & duodenal catheters

HFD

4 B

A

Page 78: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

69

Figure 14 Duodenal CCK fails to suppress glucose production following 3-days of high-fat feeding (A and B) Intraduodenal CCK-8 administration increased the glucose infusion rate (A, * p < 0.01 versus control), and decreased glucose production rate (B, * p < 0.001 versus control) in rats fed with regular chow (RC). Rats placed on a high-fat diet (HFD) for 3 days failed to respond to intraduodenal CCK-8 infusion to increase the glucose infusion rate (A) and glucose production (B) compared to control. RC: SAL (n = 6), CCK-8 (n = 6); HFD: SAL (n = 4), CCK-8 (n = 5). (C) Suppression of glucose production during the clamp period (180-200 min) expressed as the percentage reduction from basal (60-90 min) glucose production (*p < 0.01 versus other groups). (D) Glucose uptake was unchanged in all groups. Values are shown as means ± SEM

*

*

*

RC HFD RC HFD

RC HFD

RC HFD

A B

C D

Page 79: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

70

Basal Insulin (ng/mL) 0.85 ± 0.14 Glucose (mM)

7.6 ± 0.2

SAL CCK-8 Clamp Insulin (ng/mL) 0.73 ± 0.09 0.63 ± 0.13 Glucose (mM) 7.3 ± 0.2 7.2 ± 0.5

Table 2 Plasma insulin and glucose concentrations for SAL and CCK8 during basal and clamp conditions Data are expressed as means ± SEM.

Page 80: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

71

5 Discussion

In this study, we revealed a neuronal mechanism by which duodenal CCK can regulate glucose

homeostasis. Duodenal CCK can activate a gut-brain-liver axis through the mediation by CCK-A

receptors and suppress glucose production without affecting glucose uptake. Importantly, this

CCK/CCK-A receptor signaling pathway is essential for duodenal lipids to suppress glucose

production. While these studies were performed in clamped settings, it allowed us to assess the

role of CCK on glucose homeostasis independent of changes in insulin secretions. Moreover, our

intraduodenal administration approach also eliminated the possible interfering effects of gastric

emptying. In a set of fasting-refeeding experiments, we demonstrated that this neuronal network

may also play a role in regulating prandial glucose levels in physiological settings. Consistently,

it has been observed that CCK-A receptor-deficient rats develop an elevated rate of hepatic

glucose production, diabetes and obesity [231-233]. Together, these findings strengthen the

physiological relevance of our findings that gut CCK controls glucose production through a

neuronal network. Here we uncovered a novel neural mechanism for CCK to regulate glucose

homeostasis. Our results suggest that the CCK may be involved in the first line of defense by the

body to adapt appropriately to the incoming nutrients through interactions between the peripheral

nervous system and the central nervous system. Also, in combination with the recent discoveries

that circulating peptide hormones such as insulin or leptin trigger signaling cascades in the brain

to lower glucose production and regulate glucose homeostasis [245-247], we propose that the

central nervous system plays a vital role to convey peptide hormonal signals initiated in the gut

or brain to regulate hepatic glucose production and maintain glucose homeostasis.

Page 81: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

72

We showed that duodenal CCK can activate a gut-brain-liver axis to lower glucose production as

an adaptive response to incoming nutrients, the identification of the gut-brain-liver axis was

based on the presence of a gut-brain axis for energy homeostasis by upper intestinal lipids, and a

brain-liver axis for glucose homeostasis by hypothalamic fatty acids [197]. As mentioned in the

introduction, as lipids enter the duodenum, lipases release LCFAs through fat hydrolysis. The

accumulation of LCFAs leads to the stimulation of the brain through the vagus nerve resulting in

inhibition of food intake, thereby regulating energy homeostasis. On the other hand,

intrahypothalamic administration of LCFA led to a suppression of glucose production [248]. As

LCFA accumulates in the hypothalamus, it is esterified by acyl-coenzyme A synthetase to

become LCFA-CoA. The accumulation of LCFA-CoA then leads to the activation of KATP-

channels [248]. Eventually, the hepatic branch of the vagus nerve is stimulated to suppress

glucose production, suggesting that there is direct communication between the brain and the liver

through the vagus nerve. Based on these two neuronal axes, it was first tested whether upper

intestinal lipids can activate a neuronal network to suppress glucose production. Indeed,

duodenal lipids were able to suppress glucose production through a gut-brain-liver axis [20].

Given that CCK is secreted in response to duodenal lipids and we demonstrated that CCK-A

receptor signaling is required for duodenal lipid-sensing, we postulate that CCK is released in

response to duodenal lipids to trigger a CCK/CCK-A receptor signaling mechanism. Since CCK-

A receptors have been located on vagal afferents [249;250] and vagal afferents have been found

to be essential in duodenal lipid-sensing [251], it is probable that the activation of duodenal

CCK-A receptors can lead to stimulation of vagal afferents to send signals to the central nervous

system. One of the brain regions that receive signals from the gut is the hindbrain region NTS

and then the NTS sends signals back to the periphery through the hepatic vagus nerve to suppress

Page 82: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

73

glucose production. Hence, our findings also further the fact that the central nervous system can

receive nutrient signals from the gut to regulate glucose homeostasis.

Here, we highlighted the role of the NTS as an integration center in the central nervous system

receiving signals from the peripheral nervous system and sending signals back to the periphery

through the vagus nerve to suppress hepatic glucose production. It has been known that

peripheral CCK can activate CCK-A receptors, leading to the stimulation of the NTS through the

vagus nerve [252-254]. In this study, we specifically showed that duodenal CCK regulates

glucose production through stimulating the NTS via NMDA receptors. As mentioned previously,

the vagus nerve is proposed to be involved in the link between duodenal lipid-sensing and NTS

activation. While peripheral CCK is known to inhibit food intake through activating the NTS, it

has recently been demonstrated that this activation is mediated by the NR2 subunit of NMDA

receptors [255]. NMDA receptors exist as heteromeric complexes composed of NR1, NR2 and

NR3 subunits while MK-801 is only selective for NR1 and NR2 subunits. Since the NR1

subunits are necessary for the NMDA-receptor ion channels to operate, we postulate that the

NR1 subunit should be essential in mediating the effect seen in our study. On the other hand, the

principal neurotransmitter released from vagal afferent terminals in the NTS is glutamate [214]

and glutamate binds with high affinity to the NR2 subunits of the NMDA receptor [256]. These

findings suggest that the NR1 and NR2 subunits may be mediating the effects seen in our study.

Although we were unable to confirm the subunits of the NMDA receptor mediating the effects

seen in our study, we have nonetheless furthered the role of the NTS in mediating glucose-

regulatory effects elicited by duodenal CCK.

Page 83: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

74

While we showed that CCK is downstream of lipid-sensing in the duodenum for the regulation

of glucose homeostasis through the gut-brain-liver axis, it was shown that duodenal lipids fail to

lower glucose production in high-fat fed rodents. Hence, one of the goals of the current study

was to determine whether CCK administration can restore the regulation of glucose homeostasis

by the small intestine in high-fat fed rodents. However, we report that there is an acquired defect

in the CCK-A receptor pathway such that duodenal CCK fails to suppress glucose production in

response to three days of high-fat diet. From these results, we postulate that in addition to hepatic

insulin resistance, duodenal CCK-resistance can develop in response to high-fat feeding. Indeed,

while CCK is known to inhibit food intake in normal conditions, it fails to do so in adaptation to

high-fat diets [257-259] with plasma CCK levels upregulated [260;261]. These evidences

support the notion that there is an acquired defect in duodenal CCK-signaling mechanism in

high-fat fed rodents, and this raises the possibility that the restoration of the CCK-signaling

pathway in the gut can lead to restoration of proper glucose and energy homeostasis in diabetes

and obesity.

Page 84: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

75

Limitations:

While our findings provided interesting advancements in the understanding of the control of

glucose homeostasis by the small intestine, our findings are not without limitations.

1.) Although we were able to measure CCK levels in the portal and circulation, we did not

measure the CCK levels in the duodenal tissues. In our preliminary experiments, the

measurement of CCK-8 in duodenal tissues was met with the problem that the level of CCK

was beyond the linear range of the standard curve, deeming the values unreliable. Indeed, it

was found in a previous study that the amount of CCK in rat intestine mucosa is up to 174

pmol/g [262]. Hence, we will have to identify a proper dilution factor to allow the level of

CCK in our rat duodenal tissues to be within the linear range of the standard curve, until then

we will not be able to conclude whether the CCK levels achieved through our infusion of

CCK-8 in the duodenum is indeed physiological or pharmacological.

2.) For our fasting-refeeding experiments, we demonstrated that the SD rats infused with CCK-

A receptor blocker or the OLETF rats that are deficient in CCK-A receptors had higher

glucose levels in response to refeeding. Based on this observation, we concluded that the

failure to activate the neuronal network is the reason for the higher glucose levels in these

rats and that the regulation of glucose homeostasis through the neuronal network by duodenal

CCK is physiologically relevant. However, as mentioned in the introduction, upon meal

ingestion, a major source of glucose comes from the meal and the rate at which the glucose

appears in the circulation depends on the gastric emptying rate. In turn, CCK-A receptors

have been implicated in the inhibition of gastric emptying and CCK can actually reduce

postprandial hyperglycemia in humans through slowing gastric emptying [58]. Given these

Page 85: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

76

observations, it is possible that the higher glucose observed in the OLETF rats or in response

to CCK-A receptor blocker in SD rats during refeeding was due to a lack of inhibition on

gastric emptying compared to saline, thus allowing glucose to be delivered to the duodenum

for faster absorption. Hence, we are unable to conclude that the higher glucose levels in the

rats abolished of its CCK-A receptor signaling is secondary to accelerated gastric emptying.

A separate set of fasting-refeeding experiments with the same conditions would need to be

performed with an emphasis on the measurement of gastric emptying to confirm that the

differences in glucose levels occurred independent of differences in gastric emptying rate.

3.) Another shortcoming of our study is that we cannot rule out the fact that other gut peptides

may be involved in mediating the observed effects of CCK on glucose production. As

mentioned in the introduction, CCK has been implicated in the secretion of other gut

peptides. For instance, CCK is suggested to be involved in the suppression of ghrelin

secretion and stimulation of GLP-1 and PYY. Hence, to determine whether other gut

peptides are involved, the most convenient way would be to administer CCK to the

duodenum of an overexpression model of ghrelin to assess whether the suppression of

ghrelin is required for CCK to exert its effects on glucose homeostasis. On the other hand, we

will have to infuse CCK to the duodenum of a knockout model of GLP-1 or PYY to

determine whether these gut peptides are involved in the effects of duodenal CCK on glucose

production. However, a knockout model of GLP-1 receptor or PYY is only available in mice

[263;264], such that the CCK-induced gut-brain-liver axis will need to be recapitulated in

mice before it can be confirmed whether GLP-1 receptor is involved in mediating the effects.

Page 86: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

77

6 Future Directions

Our study has successfully demonstrated that CCK lies downstream of lipid-sensing in the

duodenum, mediating the activation of a gut-brain-liver axis to regulate glucose homeostasis. As

mentioned in the introduction, intraduodenal administration of glucose or proteins can also lead

to the release of CCK [29;56-58]. On the basis of these findings, it will be of interest to

determine whether duodenal glucose or protein is able to activate the gut-brain-liver axis as

lipids to regulate glucose homeostasis, and if so, whether CCK is involved. If these proposals are

true, CCK would present as a convergence point in small intestinal nutrient sensing to regulate

glucose homeostasis. This will be important because we showed that upon high-fat feeding, the

ability of CCK to regulate glucose homeostasis is lost, it would be of interest to determine

whether high carbohydrate or high protein diets can also lead to duodenal CCK-resistance. These

studies will provide insights into how a balanced diet is integral to effective regulation of glucose

homeostasis by small intestinal nutrients.

Moreover, as we showed that high-fat feeding leads to CCK-resistance, an important future

direction would be to characterize the nature of the resistance to uncover possible ways to

overcome it. Resistance can occur at multiple levels of the pathway, including the

downregulation of available hormones or receptors, or a defect in the signaling cascade. Since

CCK is secreted in response to fat intake, a maintenance high-fat diet can potentially lead to the

adaptation of the body to increase the activity of degradation enzymes to prevent excess CCK

signaling. As such, CCK-resistance may be a result of reduced availability of the peptide, leading

to a reduction in its signaling. However, it has indeed been found that rats on a high-fat diet

Page 87: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

78

exhibit elevated postprandial CCK concentrations [265], this suggests that increased CCK

degradation may not be the reason for the development of CCK-resistance. Alternatively, the

development of CCK resistance can occur at the level of the receptor. There could be a reduction

in the expression of the CCK-A receptors in the vagal afferents such that there are less CCK-A

receptors available for activation rendering it more difficult to reach the threshold level of

activation required to stimulate the vagal afferent. To confirm whether this is the case, the

protein expression of CCK-A receptors in the duodenum of regular-chow fed rats would need to

be compared with those of high-fat fed rats. Moreover, since CCK-A receptors are G-protein

coupled receptors, they are subjected to desensitization mechanisms including receptor

internalization and phosphorylation [266]. Therefore, another possibility for the development of

CCK resistance is that there is increased sequestration of receptors such that the downstream

signaling events cannot be triggered. To this end, the amount of CCK-A receptors internalized or

phosphorylated will need to be compared between regular-chow fed rats and high-fat fed rats.

Lastly, the development of CCK-resistance may be due to a defect in its downstream signaling

mechanisms. According to exocrine pancreatic secretion studies, which is another extensively

studied area in regards to CCK-A receptor signaling, it has been discovered that PKA [267] and

PLC [268] play significant roles in mediating the actions of CCK-A receptors in the stimulation

of pancreatic secretions. Similarly, CCK can also activate PKA and PLC in central vagal afferent

fibers [269]. On the basis of these findings, it would be of interest to determine whether PKA

and/or PLC are activated in response to CCK-A receptor signaling in the duodenum to suppress

glucose production. Upon verifying their roles in the duodenum, it would be imperative to

determine whether the activation of these pathways can restore the glucose-regulatory function

of the small intestines in high-fat fed rodents.

Page 88: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

79

Furthermore, another future direction stemming from this study is to evaluate the possible role of

CCK-58 in the regulation of glucose homeostasis. In this study, we mainly used the CCK-8 form

of CCK for our experiments, yet there are also other forms of CCK in the circulation: CCK-22,

CCK-33, CCK-39 and CCK-58 [270;271]. While CCK-8 is the shortest biologically active form

of CCK for the CCK-A receptors and it is abundant in the circulation, CCK-58 has been

demonstrated to be another major form in dogs, man and rats [272-275]. Interestingly, CCK-58

can also stimulate intestinal afferent nerve fibers via CCK-A receptors [276]. Functionally,

CCK-58 can inhibit food intake in rats [277] similar to CCK-8, but CCK-58 appears to be more

potent and less readily digested by endopeptidase 24:11 than CCK-8 (longer half-life than CCK-

8) [278]. Therefore, it will be of interest to determine whether CCK-58 can also exhibit the same

glucose-regulatory effects as CCK-8 and whether its actions are affected in high-fat fed rodents.

The results from these experiments would contribute to understanding whether the CCK-

resistance demonstrated is specific for shorter forms of CCK or it is of general resistance to

CCK.

Page 89: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

80

7 Conclusion

In this thesis, we examined the possible role of CCK in the regulation of glucose homeostasis

through a neuronal network. Specifically, we demonstrated that, in normal rodents (i) duodenal

CCK can suppress glucose production (ii) this effect requires the activation of CCK-A receptors

(iii) the activation of CCK-A receptors triggers a gut-brain-liver axis (iv) CCK/CCK-A receptor

signaling is required for duodenal lipids to suppress glucose production. However, in rats fed on

three days of high fat diet, duodenal CCK could not suppress glucose production, suggesting that

there is an acquired defect in the CCK-A receptor signaling cascade in response to high fat

feeding. Future studies will need to focus on elucidating the downstream signaling mechanisms

of CCK-A receptors to identify possible ways to restore the regulation of glucose homeostasis by

duodenal lipids.

Page 90: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

81

8 References

[1] Kopelman PG (2000) Obesity as a medical problem. Nature, 404, 635-643.

[2] Visscher TL & Seidell JC (2001) The public health impact of obesity. Annu. Rev. Public Health, 22, 355-375.

[3] Boden G (1997) Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes, 46, 3-10.

[4] Lewis GF, Carpentier A, Adeli K, & Giacca A (2002) Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr. Rev., 23, 201-229.

[5] Lam TK, Carpentier A, Lewis GF, van de Werve G, Fantus IG, & Giacca A (2003) Mechanisms of the free fatty acid-induced increase in hepatic glucose production. Am. J. Physiol Endocrinol. Metab, 284, E863-E873.

[6] Wild S, Roglic G, Green A, Sicree R, & King H (2004) Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care, 27, 1047-1053.

[7] Skyler JS (2007) Prediction and prevention of type 1 diabetes: progress, problems, and prospects. Clin. Pharmacol. Ther., 81, 768-771.

[8] DeFronzo RA (2004) Pathogenesis of type 2 diabetes mellitus. Med. Clin. North Am., 88, 787-835, ix.

[9] Calcutt NA, Cooper ME, Kern TS, & Schmidt AM (2009) Therapies for hyperglycaemia-induced diabetic complications: from animal models to clinical trials. Nat. Rev. Drug Discov., 8, 417-429.

[10] Turner RC & Holman RR (1995) Lessons from UK prospective diabetes study. Diabetes Res. Clin. Pract., 28 Suppl, S151-S157.

[11] Kempen JH, O'Colmain BJ, Leske MC, Haffner SM, Klein R, Moss SE, Taylor HR, & Hamman RF (2004) The prevalence of diabetic retinopathy among adults in the United States. Arch. Ophthalmol., 122, 552-563.

[12] (1993) The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N. Engl. J. Med., 329, 977-986.

[13] Mooradian AD (2009) Dyslipidemia in type 2 diabetes mellitus. Nat. Clin. Pract. Endocrinol. Metab, 5, 150-159.

Page 91: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

82

[14] Wahren J & Ekberg K (2007) Splanchnic regulation of glucose production. Annu. Rev. Nutr., 27, 329-345.

[15] Quesada I, Tuduri E, Ripoll C, & Nadal A (2008) Physiology of the pancreatic alpha-cell and glucagon secretion: role in glucose homeostasis and diabetes. J. Endocrinol., 199, 5-19.

[16] Horowitz M, Edelbroek MA, Wishart JM, & Straathof JW (1993) Relationship between oral glucose tolerance and gastric emptying in normal healthy subjects. Diabetologia, 36, 857-862.

[17] Del PS (2003) Loss of early insulin secretion leads to postprandial hyperglycaemia. Diabetologia, 46 Suppl 1, M2-M8.

[18] Burcelin R, Dolci W, & Thorens B (2000) Portal glucose infusion in the mouse induces hypoglycemia: evidence that the hepatoportal glucose sensor stimulates glucose utilization. Diabetes, 49, 1635-1642.

[19] Moore MC, Cherrington AD, & Wasserman DH (2003) Regulation of hepatic and peripheral glucose disposal. Best. Pract. Res. Clin. Endocrinol. Metab, 17, 343-364.

[20] Wang PY, Caspi L, Lam CK, Chari M, Li X, Light PE, Gutierrez-Juarez R, Ang M, Schwartz GJ, & Lam TK (2008) Upper intestinal lipids trigger a gut-brain-liver axis to regulate glucose production. Nature, 452, 1012-1016.

[21] Chaudhri OB, Salem V, Murphy KG, & Bloom SR (2008) Gastrointestinal satiety signals. Annu. Rev. Physiol, 70, 239-255.

[22] Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, & Kangawa K (1999) Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature, 402, 656-660.

[23] Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS, Suganuma T, Matsukura S, Kangawa K, & Nakazato M (2000) Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology, 141, 4255-4261.

[24] Ariyasu H, Takaya K, Tagami T, Ogawa Y, Hosoda K, Akamizu T, Suda M, Koh T, Natsui K, Toyooka S, Shirakami G, Usui T, Shimatsu A, Doi K, Hosoda H, Kojima M, Kangawa K, & Nakao K (2001) Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans. J. Clin. Endocrinol. Metab, 86, 4753-4758.

[25] Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, Bhattacharya S, Carpenter R, Grossman AB, & Korbonits M (2002) The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J. Clin. Endocrinol. Metab, 87, 2988.

Page 92: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

83

[26] Tschop M, Smiley DL, & Heiman ML (2000) Ghrelin induces adiposity in rodents. Nature, 407, 908-913.

[27] Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, & Weigle DS (2001) A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes, 50, 1714-1719.

[28] Parker BA, Doran S, Wishart J, Horowitz M, & Chapman IM (2005) Effects of small intestinal and gastric glucose administration on the suppression of plasma ghrelin concentrations in healthy older men and women. Clin. Endocrinol. (Oxf), 62, 539-546.

[29] Little TJ, Doran S, Meyer JH, Smout AJ, O'Donovan DG, Wu KL, Jones KL, Wishart J, Rayner CK, Horowitz M, & Feinle-Bisset C (2006) The release of GLP-1 and ghrelin, but not GIP and CCK, by glucose is dependent upon the length of small intestine exposed. Am. J. Physiol Endocrinol. Metab, 291, E647-E655.

[30] Cukier K, Pilichiewicz AN, Chaikomin R, Brennan IM, Wishart JM, Rayner CK, Jones KL, Horowitz M, & Feinle-Bisset C (2008) Effect of small intestinal glucose load on plasma ghrelin in healthy men. Am. J. Physiol Regul. Integr. Comp Physiol, 295, R459-R462.

[31] Overduin J, Frayo RS, Grill HJ, Kaplan JM, & Cummings DE (2005) Role of the duodenum and macronutrient type in ghrelin regulation. Endocrinology, 146, 845-850.

[32] Degen L, Drewe J, Piccoli F, Grani K, Oesch S, Bunea R, D'Amato M, & Beglinger C (2007) Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am. J. Physiol Regul. Integr. Comp Physiol, 292, R1391-R1399.

[33] Feinle-Bisset C, Patterson M, Ghatei MA, Bloom SR, & Horowitz M (2005) Fat digestion is required for suppression of ghrelin and stimulation of peptide YY and pancreatic polypeptide secretion by intraduodenal lipid. Am. J. Physiol Endocrinol. Metab, 289, E948-E953.

[34] Feltrin KL, Patterson M, Ghatei MA, Bloom SR, Meyer JH, Horowitz M, & Feinle-Bisset C (2006) Effect of fatty acid chain length on suppression of ghrelin and stimulation of PYY, GLP-2 and PP secretion in healthy men. Peptides, 27, 1638-1643.

[35] Broglio F, Arvat E, Benso A, Gottero C, Muccioli G, Papotti M, van der Lely AJ, Deghenghi R, & Ghigo E (2001) Ghrelin, a natural GH secretagogue produced by the stomach, induces hyperglycemia and reduces insulin secretion in humans. J. Clin. Endocrinol. Metab, 86, 5083-5086.

[36] Arvat E, Maccario M, Di VL, Broglio F, Benso A, Gottero C, Papotti M, Muccioli G, Dieguez C, Casanueva FF, Deghenghi R, Camanni F, & Ghigo E (2001) Endocrine activities of ghrelin, a natural growth hormone secretagogue (GHS), in humans: comparison and interactions with hexarelin, a nonnatural peptidyl GHS, and GH-releasing hormone. J. Clin. Endocrinol. Metab, 86, 1169-1174.

Page 93: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

84

[37] Arvat E, Di VL, Broglio F, Papotti M, Muccioli G, Dieguez C, Casanueva FF, Deghenghi R, Camanni F, & Ghigo E (2000) Preliminary evidence that Ghrelin, the natural GH secretagogue (GHS)-receptor ligand, strongly stimulates GH secretion in humans. J. Endocrinol. Invest, 23, 493-495.

[38] Dezaki K, Hosoda H, Kakei M, Hashiguchi S, Watanabe M, Kangawa K, & Yada T (2004) Endogenous ghrelin in pancreatic islets restricts insulin release by attenuating Ca2+ signaling in beta-cells: implication in the glycemic control in rodents. Diabetes, 53, 3142-3151.

[39] Dezaki K, Kakei M, & Yada T (2007) Ghrelin uses Galphai2 and activates voltage-dependent K+ channels to attenuate glucose-induced Ca2+ signaling and insulin release in islet beta-cells: novel signal transduction of ghrelin. Diabetes, 56, 2319-2327.

[40] Dezaki K, Sone H, Koizumi M, Nakata M, Kakei M, Nagai H, Hosoda H, Kangawa K, & Yada T (2006) Blockade of pancreatic islet-derived ghrelin enhances insulin secretion to prevent high-fat diet-induced glucose intolerance. Diabetes, 55, 3486-3493.

[41] Volante M, Allia E, Gugliotta P, Funaro A, Broglio F, Deghenghi R, Muccioli G, Ghigo E, & Papotti M (2002) Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J. Clin. Endocrinol. Metab, 87, 1300-1308.

[42] Date Y, Nakazato M, Hashiguchi S, Dezaki K, Mondal MS, Hosoda H, Kojima M, Kangawa K, Arima T, Matsuo H, Yada T, & Matsukura S (2002) Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes, 51, 124-129.

[43] Wierup N, Svensson H, Mulder H, & Sundler F (2002) The ghrelin cell: a novel developmentally regulated islet cell in the human pancreas. Regul. Pept., 107, 63-69.

[44] Wierup N, Yang S, McEvilly RJ, Mulder H, & Sundler F (2004) Ghrelin is expressed in a novel endocrine cell type in developing rat islets and inhibits insulin secretion from INS-1 (832/13) cells. J. Histochem. Cytochem., 52, 301-310.

[45] Salehi A, Dornonville dlC, Hakanson R, & Lundquist I (2004) Effects of ghrelin on insulin and glucagon secretion: a study of isolated pancreatic islets and intact mice. Regul. Pept., 118, 143-150.

[46] Vestergaard ET, Gormsen LC, Jessen N, Lund S, Hansen TK, Moller N, & Jorgensen JO (2008) Ghrelin infusion in humans induces acute insulin resistance and lipolysis independent of growth hormone signaling. Diabetes, 57, 3205-3210.

[47] Vestergaard ET, Djurhuus CB, Gjedsted J, Nielsen S, Moller N, Holst JJ, Jorgensen JO, & Schmitz O (2008) Acute effects of ghrelin administration on glucose and lipid metabolism. J. Clin. Endocrinol. Metab, 93, 438-444.

Page 94: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

85

[48] Sun Y, Asnicar M, Saha PK, Chan L, & Smith RG (2006) Ablation of ghrelin improves the diabetic but not obese phenotype of ob/ob mice. Cell Metab, 3, 379-386.

[49] Heijboer AC, van den Hoek AM, Parlevliet ET, Havekes LM, Romijn JA, Pijl H, & Corssmit EP (2006) Ghrelin differentially affects hepatic and peripheral insulin sensitivity in mice. Diabetologia, 49, 732-738.

[50] Falken Y, Hellstrom PM, Sanger GJ, Dewit O, Dukes G, Gryback P, Holst JJ, & Naslund E (2010) Actions of prolonged ghrelin infusion on gastrointestinal transit and glucose homeostasis in humans. Neurogastroenterol. Motil..

[51] Dass NB, Munonyara M, Bassil AK, Hervieu GJ, Osbourne S, Corcoran S, Morgan M, & Sanger GJ (2003) Growth hormone secretagogue receptors in rat and human gastrointestinal tract and the effects of ghrelin. Neuroscience, 120, 443-453.

[52] Fukuda H, Mizuta Y, Isomoto H, Takeshima F, Ohnita K, Ohba K, Omagari K, Taniyama K, & Kohno S (2004) Ghrelin enhances gastric motility through direct stimulation of intrinsic neural pathways and capsaicin-sensitive afferent neurones in rats. Scand. J. Gastroenterol., 39, 1209-1214.

[53] Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, Kangawa K, & Nakazato M (2002) The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology, 123, 1120-1128.

[54] Ivy AC & Oldberg E (1928) A HORMONE MECHANISM FOR GALL-BLADDER CONTRACTION AND EVACUATION. Am J Physiol, 86, 599-613.

[55] Ritter RC & Ladenheim EE (1985) Capsaicin pretreatment attenuates suppression of food intake by cholecystokinin. Am. J. Physiol, 248, R501-R504.

[56] Baum F, Nauck MA, Ebert R, Cantor P, Hoffmann G, Choudhury AR, Schmidt WE, & Creutzfeldt W (1992) Role of endogenously released cholecystokinin in determining postprandial insulin levels in man: effects of loxiglumide, a specific cholecystokinin receptor antagonist. Digestion, 53, 189-199.

[57] Matzinger D, Gutzwiller JP, Drewe J, Orban A, Engel R, D'Amato M, Rovati L, & Beglinger C (1999) Inhibition of food intake in response to intestinal lipid is mediated by cholecystokinin in humans. Am. J. Physiol, 277, R1718-R1724.

[58] Pilichiewicz AN, Chaikomin R, Brennan IM, Wishart JM, Rayner CK, Jones KL, Smout AJ, Horowitz M, & Feinle-Bisset C (2007) Load-dependent effects of duodenal glucose on glycemia, gastrointestinal hormones, antropyloroduodenal motility, and energy intake in healthy men. Am. J. Physiol Endocrinol. Metab, 293, E743-E753.

[59] Feltrin KL, Little TJ, Meyer JH, Horowitz M, Smout AJ, Wishart J, Pilichiewicz AN, Rades T, Chapman IM, & Feinle-Bisset C (2004) Effects of intraduodenal fatty acids on appetite, antropyloroduodenal motility, and plasma CCK and GLP-1 in humans vary with their chain length. Am. J. Physiol Regul. Integr. Comp Physiol, 287, R524-R533.

Page 95: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

86

[60] Feltrin KL, Little TJ, Meyer JH, Horowitz M, Rades T, Wishart J, & Feinle-Bisset C (2008) Comparative effects of intraduodenal infusions of lauric and oleic acids on antropyloroduodenal motility, plasma cholecystokinin and peptide YY, appetite, and energy intake in healthy men. Am. J. Clin. Nutr., 87, 1181-1187.

[61] Little TJ, Feltrin KL, Horowitz M, Smout AJ, Rades T, Meyer JH, Pilichiewicz AN, Wishart J, & Feinle-Bisset C (2005) Dose-related effects of lauric acid on antropyloroduodenal motility, gastrointestinal hormone release, appetite, and energy intake in healthy men. Am. J. Physiol Regul. Integr. Comp Physiol, 289, R1090-R1098.

[62] Reidelberger RD, Heimann D, Kelsey L, & Hulce M (2003) Effects of peripheral CCK receptor blockade on feeding responses to duodenal nutrient infusions in rats. Am. J. Physiol Regul. Integr. Comp Physiol, 284, R389-R398.

[63] Nawrot-Porabka K, Jaworek J, Leja-Szpak A, Szklarczyk J, Kot M, Mitis-Musiol M, Konturek SJ, & Pawlik WW (2007) Involvement of vagal nerves in the pancreatostimulatory effects of luminal melatonin, or its precursor L-tryptophan. Study in the rats. J. Physiol Pharmacol., 58 Suppl 6, 81-95.

[64] Yox DP, Brenner L, & Ritter RC (1992) CCK-receptor antagonists attenuate suppression of sham feeding by intestinal nutrients. Am. J. Physiol, 262, R554-R561.

[65] Ritter RC, Covasa M, & Matson CA (1999) Cholecystokinin: proofs and prospects for involvement in control of food intake and body weight. Neuropeptides, 33, 387-399.

[66] Kimura I, Nakashima N, Komori T, Kameda Y, & Kimura M (1994) Dependence of cholecystokinin-8-stimulated insulin release on high glucose levels is evidenced by pseudo-alpha-D-glucose in rat pancreas and islets. Jpn. J. Pharmacol., 64, 103-107.

[67] Malm D, Giaever A, Vonen B, & Florholmen J (1993) Cholecystokinin and somatostatin modulate the glucose-induced insulin secretion by different mechanisms in pancreatic islets. A study on phospholipase C activity and calcium requirement. Scand. J. Clin. Lab Invest, 53, 671-676.

[68] Otsuki M, Okabayashi Y, Ohki A, Oka T, Fujii M, Nakamura T, Sugiura N, Yanaihara N, & Baba S (1986) Action of cholecystokinin analogues on exocrine and endocrine rat pancreas. Am. J. Physiol, 250, G405-G411.

[69] Sakamoto C, Otsuki M, Ohki A, Yuu H, Maeda M, Yamasaki T, & Baba S (1982) Glucose-dependent insulinotropic action of cholecystokinin and caerulein in the isolated perfused rat pancreas. Endocrinology, 110, 398-402.

[70] Schmid R, Schusdziarra V, Schulte-Frohlinde E, Maier V, & Classen M (1989) Effect of CCK on insulin, glucagon, and pancreatic polypeptide levels in humans. Pancreas, 4, 653-661.

Page 96: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

87

[71] Shikado F, Miyasaka K, Funakoshi A, & Kitani K (1990) Necessity of hyperglycemia for effects of endogenous cholecystokinin on insulin and pancreatic exocrine secretion in conscious rats. Jpn. J. Physiol, 40, 383-391.

[72] Verspohl EJ, Ammon HP, Williams JA, & Goldfine ID (1986) Evidence that cholecystokinin interacts with specific receptors and regulates insulin release in isolated rat islets of Langerhans. Diabetes, 35, 38-43.

[73] Verspohl EJ, Breuning I, Ammon HP, & Mark M (1987) Significance of Ca2+, Rb+ fluxes, of cAMP and cGMP for the CCK8-modulated insulin release. Regul. Pept., 17, 229-241.

[74] Zawalich W, Takuwa N, Takuwa Y, Diaz VA, & Rasmussen H (1987) Interactions of cholecystokinin and glucose in rat pancreatic islets. Diabetes, 36, 426-433.

[75] Zawalich WS, Cote SB, & Diaz VA (1986) Influence of cholecystokinin on insulin output from isolated perifused pancreatic islets. Endocrinology, 119, 616-621.

[76] Jensen SL, Rehfeld JF, Holst JJ, Nielsen OV, Fahrenkrug J, & Schaffalitzky de Muckadell OB (1981) Secretory effects of cholecystokinins on the isolated perfused porcine pancreas. Acta Physiol Scand., 111, 225-231.

[77] Jensen SL, Holst JJ, Nielsen OV, & Rehfeld JF (1981) Effect of sulfation of CCK-8 on its stimulation of the endocrine and exocrine secretion from the isolated perfused porcine pancreas. Digestion, 22, 305-309.

[78] Reimers J, Nauck M, Creutzfeldt W, Strietzel J, Ebert R, Cantor P, & Hoffmann G (1988) Lack of insulinotropic effect of endogenous and exogenous cholecystokinin in man. Diabetologia, 31, 271-280.

[79] Rushakoff RJ, Goldfine ID, Carter JD, & Liddle RA (1987) Physiological concentrations of cholecystokinin stimulate amino acid-induced insulin release in humans. J. Clin. Endocrinol. Metab, 65, 395-401.

[80] Liddle RA, Rushakoff RJ, Morita ET, Beccaria L, Carter JD, & Goldfine ID (1988) Physiological role for cholecystokinin in reducing postprandial hyperglycemia in humans. J. Clin. Invest, 81, 1675-1681.

[81] Balkan B, Steffens AB, Strubbe JH, & Bruggink JE (1990) Biphasic insulin secretion after intravenous but not after intraportal CCK-8 infusion in rats. Diabetes, 39, 702-706.

[82] Verspohl EJ & Herrmann K (1996) Involvement of G proteins in the effect of carbachol and cholecystokinin in rat pancreatic islets. Am. J. Physiol, 271, E65-E72.

[83] Karlsson S & Ahren B (1991) CCKA receptor antagonism inhibits mechanisms underlying CCK-8-stimulated insulin release in isolated rat islets. Eur. J. Pharmacol., 202, 253-257.

Page 97: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

88

[84] Karlsson S & Ahren B (1991) Cholecystokinin-stimulated insulin secretion and protein kinase C in rat pancreatic islets. Acta Physiol Scand., 142, 397-403.

[85] Simonsson E, Karlsson S, & Ahren B (1996) Involvement of phospholipase A2 and arachidonic acid in cholecystokinin-8-induced insulin secretion in rat islets. Regul. Pept., 65, 101-107.

[86] Simonsson E, Karlsson S, & Ahren B (1998) Ca2+-independent phospholipase A2 contributes to the insulinotropic action of cholecystokinin-8 in rat islets: dissociation from the mechanism of carbachol. Diabetes, 47, 1436-1443.

[87] Rushakoff RA, Goldfine ID, Beccaria LJ, Mathur A, Brand RJ, & Liddle RA (1993) Reduced postprandial cholecystokinin (CCK) secretion in patients with noninsulin-dependent diabetes mellitus: evidence for a role for CCK in regulating postprandial hyperglycemia. J. Clin. Endocrinol. Metab, 76, 489-493.

[88] Phillips WT, Schwartz JG, & McMahan CA (1993) Reduced postprandial blood glucose levels in recently diagnosed non-insulin-dependent diabetics secondary to pharmacologically induced delayed gastric emptying. Dig. Dis. Sci., 38, 51-58.

[89] Cummings DE & Overduin J (2007) Gastrointestinal regulation of food intake. J. Clin. Invest, 117, 13-23.

[90] Borovicka J, Kreiss C, Asal K, Remy B, Mettraux C, Wells A, Read NW, Jansen JB, D'Amato M, Delaloye AB, Fried M, & Schwizer W (1996) Role of cholecystokinin as a regulator of solid and liquid gastric emptying in humans. Am. J. Physiol, 271, G448-G453.

[91] Fried M, Erlacher U, Schwizer W, Lochner C, Koerfer J, Beglinger C, Jansen JB, Lamers CB, Harder F, Bischof-Delaloye A, & . (1991) Role of cholecystokinin in the regulation of gastric emptying and pancreatic enzyme secretion in humans. Studies with the cholecystokinin-receptor antagonist loxiglumide. Gastroenterology, 101, 503-511.

[92] Fried M, Schwizer W, Beglinger C, Keller U, Jansen JB, & Lamers CB (1991) Physiological role of cholecystokinin on postprandial insulin secretion and gastric meal emptying in man. Studies with the cholecystokinin receptor antagonist loxiglumide. Diabetologia, 34, 721-726.

[93] Hidalgo L, Clave P, Estorch M, Rodriguez-Espinosa J, Rovati L, Greeley GH, Capella G, & Lluis F (2002) Effect of cholecystokinin-A receptor blockade on postprandial insulinaemia and gastric emptying in humans. Neurogastroenterol. Motil., 14, 519-525.

[94] Kisfalvi K, Racz G, Zsirka-Klein A, Pelosini I, Scarpignato C, & Varga G (2001) Different affinity states of CCK(1) receptors on pancreatic acini and gastric smooth muscle in the rat. J. Physiol Paris, 95, 391-398.

Page 98: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

89

[95] Moran TH, Kornbluh R, Moore K, & Schwartz GJ (1994) Cholecystokinin inhibits gastric emptying and contracts the pyloric sphincter in rats by interacting with low affinity CCK receptor sites. Regul. Pept., 52, 165-172.

[96] Smith GP, Falasco J, Moran TH, Joyner KM, & Gibbs J (1988) CCK-8 decreases food intake and gastric emptying after pylorectomy or pyloroplasty. Am. J. Physiol, 255, R113-R116.

[97] Raybould HE & Tache Y (1988) Cholecystokinin inhibits gastric motility and emptying via a capsaicin-sensitive vagal pathway in rats. Am. J. Physiol, 255, G242-G246.

[98] Schwartz GJ, Berkow G, McHugh PR, & Moran TH (1993) Gastric branch vagotomy blocks nutrient and cholecystokinin-induced suppression of gastric emptying. Am. J. Physiol, 264, R630-R637.

[99] Gibbs J, Young RC, & Smith GP (1973) Cholecystokinin decreases food intake in rats. J. Comp Physiol Psychol., 84, 488-495.

[100] Gibbs J, Young RC, & Smith GP (1973) Cholecystokinin decreases food intake in rats. J. Comp Physiol Psychol., 84, 488-495.

[101] Smith GP, Jerome C, Cushin BJ, Eterno R, & Simansky KJ (1981) Abdominal vagotomy blocks the satiety effect of cholecystokinin in the rat. Science, 213, 1036-1037.

[102] Kissileff HR, Pi-Sunyer FX, Thornton J, & Smith GP (1981) C-terminal octapeptide of cholecystokinin decreases food intake in man. Am. J. Clin. Nutr., 34, 154-160.

[103] Weatherford SC, Chiruzzo FY, & Laughton WB (1992) Satiety induced by endogenous and exogenous cholecystokinin is mediated by CCK-A receptors in mice. Am. J. Physiol, 262, R574-R578.

[104] Moran TH, Ameglio PJ, Schwartz GJ, & McHugh PR (1992) Blockade of type A, not type B, CCK receptors attenuates satiety actions of exogenous and endogenous CCK. Am. J. Physiol, 262, R46-R50.

[105] Monnikes H, Lauer G, & Arnold R (1997) Peripheral administration of cholecystokinin activates c-fos expression in the locus coeruleus/subcoeruleus nucleus, dorsal vagal complex and paraventricular nucleus via capsaicin-sensitive vagal afferents and CCK-A receptors in the rat. Brain Res., 770, 277-288.

[106] McIntosh CH, Widenmaier S, & Kim SJ (2009) Glucose-dependent insulinotropic polypeptide (Gastric Inhibitory Polypeptide; GIP). Vitam. Horm., 80, 409-471.

[107] Buchan AM, Polak JM, Capella C, Solcia E, & Pearse AG (1978) Electronimmunocytochemical evidence for the K cell localization of gastric inhibitory polypeptide (GIP) in man. Histochemistry, 56, 37-44.

Page 99: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

90

[108] Thomas FB, Shook DF, O'Dorisio TM, Cataland S, Mekhjian HS, Caldwell JH, & Mazzaferri EL (1977) Localization of gastric inhibitory polypeptide release by intestinal glucose perfusion in man. Gastroenterology, 72, 49-54.

[109] Chaikomin R, Doran S, Jones KL, Feinle-Bisset C, O'Donovan D, Rayner CK, & Horowitz M (2005) Initially more rapid small intestinal glucose delivery increases plasma insulin, GIP, and GLP-1 but does not improve overall glycemia in healthy subjects. Am. J. Physiol Endocrinol. Metab, 289, E504-E507.

[110] Kuo P, Chaikomin R, Pilichiewicz A, O'Donovan D, Wishart JM, Meyer JH, Jones KL, Feinle-Bisset C, Horowitz M, & Rayner CK (2008) Transient, early release of glucagon-like peptide-1 during low rates of intraduodenal glucose delivery. Regul. Pept., 146, 1-3.

[111] Lavin JH, Wittert GA, Andrews J, Yeap B, Wishart JM, Morris HA, Morley JE, Horowitz M, & Read NW (1998) Interaction of insulin, glucagon-like peptide 1, gastric inhibitory polypeptide, and appetite in response to intraduodenal carbohydrate. Am. J. Clin. Nutr., 68, 591-598.

[112] McCullough AJ, Miller LJ, Service FJ, & Go VL (1983) Effect of graded intraduodenal glucose infusions on the release and physiological action of gastric inhibitory polypeptide. J. Clin. Endocrinol. Metab, 56, 234-241.

[113] Jorde R, Burhol PG, & Waldum HL (1981) The effect of atropine on plasma gastric inhibitory polypeptide (GIP), serum insulin, and blood glucose after intraduodenal infusion of fat. Scand. J. Gastroenterol., 16, 331-336.

[114] Krarup T, Holst JJ, & Larsen KL (1985) Responses and molecular heterogeneity of IR-GIP after intraduodenal glucose and fat. Am. J. Physiol, 249, E195-E200.

[115] O'Dorisio TM, Cataland S, Stevenson M, & Mazzaferri EL (1976) Gastric inhibitory polypeptide (GIP). Intestinal distribution and stimulation by amino acids and medium-chain triglycerides. Am. J. Dig. Dis., 21, 761-765.

[116] Lucey MR, Fairclough PD, Wass JA, Kwasowski P, Medbak S, Webb J, & Rees LH (1984) Response of circulating somatostatin, insulin, gastrin and GIP, to intraduodenal infusion of nutrients in normal man. Clin. Endocrinol. (Oxf), 21, 209-217.

[117] Edfalk S, Steneberg P, & Edlund H (2008) Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes, 57, 2280-2287.

[118] Szecowka J, Lins PE, & Efendic S (1982) Effects of cholecystokinin, gastric inhibitory polypeptide, and secretin on insulin and glucagon secretion in rats. Endocrinology, 110, 1268-1272.

[119] Elahi D, McAloon-Dyke M, Fukagawa NK, Meneilly GS, Sclater AL, Minaker KL, Habener JF, & Andersen DK (1994) The insulinotropic actions of glucose-dependent

Page 100: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

91

insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (7-37) in normal and diabetic subjects. Regul. Pept., 51, 63-74.

[120] Lewis JT, Dayanandan B, Habener JF, & Kieffer TJ (2000) Glucose-dependent insulinotropic polypeptide confers early phase insulin release to oral glucose in rats: demonstration by a receptor antagonist. Endocrinology, 141, 3710-3716.

[121] Schauder P, Brown JC, Frerichs H, & Creutzfeldt W (1975) Gastric inhibitory polypeptide: effect on glucose-induced insulin release from isolated rat pancreatic islets in vitro. Diabetologia, 11, 483-484.

[122] Pederson RA & Brown JC (1976) The insulinotropic action of gastric inhibitory polypeptide in the perfused isolated rat pancreas. Endocrinology, 99, 780-785.

[123] Usdin TB, Mezey E, Button DC, Brownstein MJ, & Bonner TI (1993) Gastric inhibitory polypeptide receptor, a member of the secretin-vasoactive intestinal peptide receptor family, is widely distributed in peripheral organs and the brain. Endocrinology, 133, 2861-2870.

[124] Miyawaki K, Yamada Y, Yano H, Niwa H, Ban N, Ihara Y, Kubota A, Fujimoto S, Kajikawa M, Kuroe A, Tsuda K, Hashimoto H, Yamashita T, Jomori T, Tashiro F, Miyazaki J, & Seino Y (1999) Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice. Proc. Natl. Acad. Sci. U. S. A, 96, 14843-14847.

[125] Kashima Y, Miki T, Shibasaki T, Ozaki N, Miyazaki M, Yano H, & Seino S (2001) Critical role of cAMP-GEFII--Rim2 complex in incretin-potentiated insulin secretion. J. Biol. Chem., 276, 46046-46053.

[126] Nakazaki M, Crane A, Hu M, Seghers V, Ullrich S, Aguilar-Bryan L, & Bryan J (2002) cAMP-activated protein kinase-independent potentiation of insulin secretion by cAMP is impaired in SUR1 null islets. Diabetes, 51, 3440-3449.

[127] Miki T, Minami K, Shinozaki H, Matsumura K, Saraya A, Ikeda H, Yamada Y, Holst JJ, & Seino S (2005) Distinct effects of glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 on insulin secretion and gut motility. Diabetes, 54, 1056-1063.

[128] Lu M, Wheeler MB, Leng XH, & Boyd AE, III (1993) The role of the free cytosolic calcium level in beta-cell signal transduction by gastric inhibitory polypeptide and glucagon-like peptide I(7-37). Endocrinology, 132, 94-100.

[129] Meier JJ, Gallwitz B, Siepmann N, Holst JJ, Deacon CF, Schmidt WE, & Nauck MA (2003) Gastric inhibitory polypeptide (GIP) dose-dependently stimulates glucagon secretion in healthy human subjects at euglycaemia. Diabetologia, 46, 798-801.

Page 101: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

92

[130] Meier JJ, Goetze O, Anstipp J, Hagemann D, Holst JJ, Schmidt WE, Gallwitz B, & Nauck MA (2004) Gastric inhibitory polypeptide does not inhibit gastric emptying in humans. Am. J. Physiol Endocrinol. Metab, 286, E621-E625.

[131] Asmar M, Tangaa W, Madsbad S, Hare K, Astrup A, Flint A, Bulow J, & Holst JJ (2010) On the role of glucose-dependent insulintropic polypeptide in postprandial metabolism in humans. Am. J. Physiol Endocrinol. Metab, 298, E614-E621.

[132] Theodorakis MJ, Carlson O, Michopoulos S, Doyle ME, Juhaszova M, Petraki K, & Egan JM (2006) Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1 and GIP. Am. J. Physiol Endocrinol. Metab, 290, E550-E559.

[133] Eissele R, Goke R, Willemer S, Harthus HP, Vermeer H, Arnold R, & Goke B (1992) Glucagon-like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and man. Eur. J. Clin. Invest, 22, 283-291.

[134] Drucker DJ (2002) Biological actions and therapeutic potential of the glucagon-like peptides. Gastroenterology, 122, 531-544.

[135] Elliott RM, Morgan LM, Tredger JA, Deacon S, Wright J, & Marks V (1993) Glucagon-like peptide-1 (7-36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J. Endocrinol., 138, 159-166.

[136] BORGSTROM B, DAHLQVIST A, LUNDH G, & SJOVALL J (1957) Studies of intestinal digestion and absorption in the human. J. Clin. Invest, 36, 1521-1536.

[137] Chaikomin R, Jones KL, Feinle-Bisset C, Meyer JH, Horowitz M, & Rayner CK (2009) Effects of intraluminal local anesthetic on upper gastrointestinal motor, sensory, and peptide hormone responses to intraduodenal glucose. Eur. J. Gastroenterol. Hepatol., 21, 258-265.

[138] Moriya R, Shirakura T, Ito J, Mashiko S, & Seo T (2009) Activation of sodium-glucose cotransporter 1 ameliorates hyperglycemia by mediating incretin secretion in mice. Am. J. Physiol Endocrinol. Metab, 297, E1358-E1365.

[139] O'Donovan DG, Doran S, Feinle-Bisset C, Jones KL, Meyer JH, Wishart JM, Morris HA, & Horowitz M (2004) Effect of variations in small intestinal glucose delivery on plasma glucose, insulin, and incretin hormones in healthy subjects and type 2 diabetes. J. Clin. Endocrinol. Metab, 89, 3431-3435.

[140] Beglinger S, Drewe J, Schirra J, Goke B, D'Amato M, & Beglinger C (2010) Role of fat hydrolysis in regulating glucagon-like Peptide-1 secretion. J. Clin. Endocrinol. Metab, 95, 879-886.

[141] Roberge JN & Brubaker PL (1991) Secretion of proglucagon-derived peptides in response to intestinal luminal nutrients. Endocrinology, 128, 3169-3174.

Page 102: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

93

[142] Roberge JN & Brubaker PL (1993) Regulation of intestinal proglucagon-derived peptide secretion by glucose-dependent insulinotropic peptide in a novel enteroendocrine loop. Endocrinology, 133, 233-240.

[143] Roberge JN, Gronau KA, & Brubaker PL (1996) Gastrin-releasing peptide is a novel mediator of proximal nutrient-induced proglucagon-derived peptide secretion from the distal gut. Endocrinology, 137, 2383-2388.

[144] Rocca AS & Brubaker PL (1999) Role of the vagus nerve in mediating proximal nutrient-induced glucagon-like peptide-1 secretion. Endocrinology, 140, 1687-1694.

[145] Kreymann B, Williams G, Ghatei MA, & Bloom SR (1987) Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet, 2, 1300-1304.

[146] Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, & Creutzfeldt W (1993) Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J. Clin. Invest, 91, 301-307.

[147] Anini Y, Hansotia T, & Brubaker PL (2002) Muscarinic receptors control postprandial release of glucagon-like peptide-1: in vivo and in vitro studies in rats. Endocrinology, 143, 2420-2426.

[148] Edwards CM, Todd JF, Mahmoudi M, Wang Z, Wang RM, Ghatei MA, & Bloom SR (1999) Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9-39. Diabetes, 48, 86-93.

[149] Fehmann HC & Habener JF (1991) Functional receptors for the insulinotropic hormone glucagon-like peptide-I(7-37) on a somatostatin secreting cell line. FEBS Lett., 279, 335-340.

[150] Fehmann HC, Janssen M, & Goke B (1995) Interaction of glucagon-like peptide-I (GLP-I) and galanin in insulin (beta TC-1)- and somatostatin (RIN T3)-secreting cells and evidence that both peptides have no receptors on glucagon (INR1G9)-secreting cells. Acta Diabetol., 32, 176-181.

[151] Suga S, Kanno T, Ogawa Y, Takeo T, Kamimura N, & Wakui M (2000) cAMP-independent decrease of ATP-sensitive K+ channel activity by GLP-1 in rat pancreatic beta-cells. Pflugers Arch., 440, 566-572.

[152] MacDonald PE, Salapatek AM, & Wheeler MB (2002) Glucagon-like peptide-1 receptor activation antagonizes voltage-dependent repolarizing K(+) currents in beta-cells: a possible glucose-dependent insulinotropic mechanism. Diabetes, 51 Suppl 3, S443-S447.

[153] Holz GG, Kuhtreiber WM, & Habener JF (1993) Pancreatic beta-cells are rendered glucose-competent by the insulinotropic hormone glucagon-like peptide-1(7-37). Nature, 361, 362-365.

Page 103: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

94

[154] Gromada J, Ding WG, Barg S, Renstrom E, & Rorsman P (1997) Multisite regulation of insulin secretion by cAMP-increasing agonists: evidence that glucagon-like peptide 1 and glucagon act via distinct receptors. Pflugers Arch., 434, 515-524.

[155] Ding WG, Kitasato H, & Matsuura H (2001) Involvement of calmodulin in glucagon-like peptide 1(7-36) amide-induced inhibition of the ATP-sensitive K+ channel in mouse pancreatic beta-cells. Exp. Physiol, 86, 331-339.

[156] Shigeto M, Katsura M, Matsuda M, Ohkuma S, & Kaku K (2008) Low, but physiological, concentration of GLP-1 stimulates insulin secretion independent of the cAMP-dependent protein kinase pathway. J. Pharmacol. Sci., 108, 274-279.

[157] Suga S, Kanno T, Dobashi Y, & Wakui M (1997) GLP-1 (7-36) amide activates L-type Ca2+ channels of pancreatic B-cells through c-AMP signaling. Jpn. J. Physiol, 47 Suppl 1, S13-S14.

[158] MacDonald PE, Wang X, Xia F, El-kholy W, Targonsky ED, Tsushima RG, & Wheeler MB (2003) Antagonism of rat beta-cell voltage-dependent K+ currents by exendin 4 requires dual activation of the cAMP/protein kinase A and phosphatidylinositol 3-kinase signaling pathways. J. Biol. Chem., 278, 52446-52453.

[159] Hare KJ, Knop FK, Asmar M, Madsbad S, Deacon CF, Holst JJ, & Vilsboll T (2009) Preserved inhibitory potency of GLP-1 on glucagon secretion in type 2 diabetes mellitus. J. Clin. Endocrinol. Metab, 94, 4679-4687.

[160] Hare KJ, Vilsboll T, Asmar M, Deacon CF, Knop FK, & Holst JJ (2010) The Glucagonostatic and Insulinotropic Effects of Glucagon-Like Peptide-1 Contribute Equally to its Glucose-Lowering Action. Diabetes.

[161] Ritzel R, Orskov C, Holst JJ, & Nauck MA (1995) Pharmacokinetic, insulinotropic, and glucagonostatic properties of GLP-1 [7-36 amide] after subcutaneous injection in healthy volunteers. Dose-response-relationships. Diabetologia, 38, 720-725.

[162] Vilsboll T, Krarup T, Madsbad S, & Holst JJ (2003) Both GLP-1 and GIP are insulinotropic at basal and postprandial glucose levels and contribute nearly equally to the incretin effect of a meal in healthy subjects. Regul. Pept., 114, 115-121.

[163] Nauck MA, Heimesaat MM, Behle K, Holst JJ, Nauck MS, Ritzel R, Hufner M, & Schmiegel WH (2002) Effects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive functions, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteers. J. Clin. Endocrinol. Metab, 87, 1239-1246.

[164] de HJ, Rasmussen C, Coy DH, & Holst JJ (2008) Glucagon-like peptide-1, but not glucose-dependent insulinotropic peptide, inhibits glucagon secretion via somatostatin (receptor subtype 2) in the perfused rat pancreas. Diabetologia, 51, 2263-2270.

Page 104: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

95

[165] Deane AM, Nguyen NQ, Stevens JE, Fraser RJ, Holloway RH, Besanko LK, Burgstad C, Jones KL, Chapman MJ, Rayner CK, & Horowitz M (2010) Endogenous glucagon-like peptide-1 slows gastric emptying in healthy subjects, attenuating postprandial glycemia. J. Clin. Endocrinol. Metab, 95, 215-221.

[166] Little TJ, Pilichiewicz AN, Russo A, Phillips L, Jones KL, Nauck MA, Wishart J, Horowitz M, & Feinle-Bisset C (2006) Effects of intravenous glucagon-like peptide-1 on gastric emptying and intragastric distribution in healthy subjects: relationships with postprandial glycemic and insulinemic responses. J. Clin. Endocrinol. Metab, 91, 1916-1923.

[167] Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Orskov C, Ritzel R, & Schmiegel WH (1997) Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am. J. Physiol, 273, E981-E988.

[168] Orskov C, Wettergren A, & Holst JJ (1993) Biological effects and metabolic rates of glucagonlike peptide-1 7-36 amide and glucagonlike peptide-1 7-37 in healthy subjects are indistinguishable. Diabetes, 42, 658-661.

[169] Balkan B & Li X (2000) Portal GLP-1 administration in rats augments the insulin response to glucose via neuronal mechanisms. Am. J. Physiol Regul. Integr. Comp Physiol, 279, R1449-R1454.

[170] Nakagawa A, Satake H, Nakabayashi H, Nishizawa M, Furuya K, Nakano S, Kigoshi T, Nakayama K, & Uchida K (2004) Receptor gene expression of glucagon-like peptide-1, but not glucose-dependent insulinotropic polypeptide, in rat nodose ganglion cells. Auton. Neurosci., 110, 36-43.

[171] Nakabayashi H, Nishizawa M, Nakagawa A, Takeda R, & Niijima A (1996) Vagal hepatopancreatic reflex effect evoked by intraportal appearance of tGLP-1. Am. J. Physiol, 271, E808-E813.

[172] Gutzwiller JP, Goke B, Drewe J, Hildebrand P, Ketterer S, Handschin D, Winterhalder R, Conen D, & Beglinger C (1999) Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut, 44, 81-86.

[173] Chelikani PK, Haver AC, & Reidelberger RD (2005) Intravenous infusion of glucagon-like peptide-1 potently inhibits food intake, sham feeding, and gastric emptying in rats. Am. J. Physiol Regul. Integr. Comp Physiol, 288, R1695-R1706.

[174] Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, Ghatei MA, & Bloom SR (2005) The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res., 1044, 127-131.

[175] Bataille D, Tatemoto K, Gespach C, Jornvall H, Rosselin G, & Mutt V (1982) Isolation of glucagon-37 (bioactive enteroglucagon/oxyntomodulin) from porcine jejuno-ileum. Characterization of the peptide. FEBS Lett., 146, 79-86.

Page 105: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

96

[176] Kervran A, Blache P, & Bataille D (1987) Distribution of oxyntomodulin and glucagon in the gastrointestinal tract and the plasma of the rat. Endocrinology, 121, 704-713.

[177] Anini Y, Fu-Cheng X, Cuber JC, Kervran A, Chariot J, & Roz C (1999) Comparison of the postprandial release of peptide YY and proglucagon-derived peptides in the rat. Pflugers Arch., 438, 299-306.

[178] Chaudhri OB, Field BC, & Bloom SR (2008) Gastrointestinal satiety signals. Int. J. Obes. (Lond), 32 Suppl 7, S28-S31.

[179] Collie NL, Walsh JH, Wong HC, Shively JE, Davis MT, Lee TD, & Reeve JR, Jr. (1994) Purification and sequence of rat oxyntomodulin. Proc. Natl. Acad. Sci. U. S. A, 91, 9362-9366.

[180] Maida A, Lovshin JA, Baggio LL, & Drucker DJ (2008) The glucagon-like peptide-1 receptor agonist oxyntomodulin enhances beta-cell function but does not inhibit gastric emptying in mice. Endocrinology, 149, 5670-5678.

[181] Schjoldager B, Mortensen PE, Myhre J, Christiansen J, & Holst JJ (1989) Oxyntomodulin from distal gut. Role in regulation of gastric and pancreatic functions. Dig. Dis. Sci., 34, 1411-1419.

[182] Jarrousse C, Bataille D, & Jeanrenaud B (1984) A pure enteroglucagon, oxyntomodulin (glucagon 37), stimulates insulin release in perfused rat pancreas. Endocrinology, 115, 102-105.

[183] Wynne K & Bloom SR (2006) The role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control. Nat. Clin. Pract. Endocrinol. Metab, 2, 612-620.

[184] Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M, Ghatei MA, & Bloom SR (2004) Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology, 145, 2687-2695.

[185] Tatemoto K, Nakano I, Makk G, Angwin P, Mann M, Schilling J, & Go VL (1988) Isolation and primary structure of human peptide YY. Biochem. Biophys. Res. Commun., 157, 713-717.

[186] Ballantyne GH (2006) Peptide YY(1-36) and peptide YY(3-36): Part I. Distribution, release and actions. Obes. Surg., 16, 651-658.

[187] El-Salhy M, Grimelius L, Wilander E, Ryberg B, Terenius L, Lundberg JM, & Tatemoto K (1983) Immunocytochemical identification of polypeptide YY (PYY) cells in the human gastrointestinal tract. Histochemistry, 77, 15-23.

[188] Fu-Cheng X, Anini Y, Chariot J, Voisin T, Galmiche JP, & Roze C (1995) Peptide YY release after intraduodenal, intraileal, and intracolonic administration of nutrients in rats. Pflugers Arch., 431, 66-75.

Page 106: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

97

[189] Pilichiewicz AN, Little TJ, Brennan IM, Meyer JH, Wishart JM, Otto B, Horowitz M, & Feinle-Bisset C (2006) Effects of load, and duration, of duodenal lipid on antropyloroduodenal motility, plasma CCK and PYY, and energy intake in healthy men. Am. J. Physiol Regul. Integr. Comp Physiol, 290, R668-R677.

[190] Feltrin KL, Little TJ, Meyer JH, Horowitz M, Rades T, Wishart J, & Feinle-Bisset C (2007) Effects of lauric acid on upper gut motility, plasma cholecystokinin and peptide YY, and energy intake are load, but not concentration, dependent in humans. J. Physiol, 581, 767-777.

[191] Boey D, Sainsbury A, & Herzog H (2007) The role of peptide YY in regulating glucose homeostasis. Peptides, 28, 390-395.

[192] Sloth B, Holst JJ, Flint A, Gregersen NT, & Astrup A (2007) Effects of PYY1-36 and PYY3-36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am. J. Physiol Endocrinol. Metab, 292, E1062-E1068.

[193] Yoshinaga K, Mochizuki T, Yanaihara N, Oshima K, Izukura M, Kogire M, Sumi S, Gomez G, Uchida T, Thompson JC, & . (1992) Structural requirements of peptide YY for biological activity at enteric sites. Am. J. Physiol, 263, G695-G701.

[194] van den Hoek AM, Heijboer AC, Corssmit EP, Voshol PJ, Romijn JA, Havekes LM, & Pijl H (2004) PYY3-36 reinforces insulin action on glucose disposal in mice fed a high-fat diet. Diabetes, 53, 1949-1952.

[195] Chelikani PK, Haver AC, & Reidelberger RD (2004) Comparison of the inhibitory effects of PYY(3-36) and PYY(1-36) on gastric emptying in rats. Am. J. Physiol Regul. Integr. Comp Physiol, 287, R1064-R1070.

[196] Witte AB, Gryback P, Holst JJ, Hilsted L, Hellstrom PM, Jacobsson H, & Schmidt PT (2009) Differential effect of PYY1-36 and PYY3-36 on gastric emptying in man. Regul. Pept., 158, 57-62.

[197] Cheung GW, Kokorovic A, & Lam TK (2009) Upper intestinal lipids regulate energy and glucose homeostasis. Cell Mol. Life Sci., 66, 3023-3027.

[198] Adkins BA, Myers SR, Hendrick GK, Stevenson RW, Williams PE, & Cherrington AD (1987) Importance of the route of intravenous glucose delivery to hepatic glucose balance in the conscious dog. J. Clin. Invest, 79, 557-565.

[199] Adkins-Marshall B, Pagliassotti MJ, Asher JR, Connolly CC, Neal DW, Williams PE, Myers SR, Hendrick GK, Adkins RB, Jr., & Cherrington AD (1992) Role of hepatic nerves in response of liver to intraportal glucose delivery in dogs. Am. J. Physiol, 262, E679-E686.

[200] Niijima A (1982) Glucose-sensitive afferent nerve fibres in the hepatic branch of the vagus nerve in the guinea-pig. J. Physiol, 332, 315-323.

Page 107: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

98

[201] Cherrington AD (1999) Banting Lecture 1997. Control of glucose uptake and release by the liver in vivo. Diabetes, 48, 1198-1214.

[202] Pagliassotti MJ, Holste LC, Moore MC, Neal DW, & Cherrington AD (1996) Comparison of the time courses of insulin and the portal signal on hepatic glucose and glycogen metabolism in the conscious dog. J. Clin. Invest, 97, 81-91.

[203] Cardin S, Emshwiller M, Jackson PA, Snead WL, Hastings J, Edgerton DS, & Cherrington AD (1999) Portal glucose infusion increases hepatic glycogen deposition in conscious unrestrained rats. J. Appl. Physiol, 87, 1470-1475.

[204] Martin JR, Novin D, & Vanderweele DA (1978) Loss of glucagon suppression of feeding after vagotomy in rats. Am. J. Physiol, 234, E314-E318.

[205] Shimizu N, Oomura Y, Novin D, Grijalva CV, & Cooper PH (1983) Functional correlations between lateral hypothalamic glucose-sensitive neurons and hepatic portal glucose-sensitive units in rat. Brain Res., 265, 49-54.

[206] Schmitt M (1973) Influences of hepatic portal receptors on hypothalamic feeding and satiety centers. Am. J. Physiol, 225, 1089-1095.

[207] Adachi A, Shimizu N, Oomura Y, & Kobashi M (1984) Convergence of hepatoportal glucose-sensitive afferent signals to glucose-sensitive units within the nucleus of the solitary tract. Neurosci. Lett., 46, 215-218.

[208] Burcelin R, Da CA, Drucker D, & Thorens B (2001) Glucose competence of the hepatoportal vein sensor requires the presence of an activated glucagon-like peptide-1 receptor. Diabetes, 50, 1720-1728.

[209] Burcelin R, Crivelli V, Perrin C, Da CA, Mu J, Kahn BB, Birnbaum MJ, Kahn CR, Vollenweider P, & Thorens B (2003) GLUT4, AMP kinase, but not the insulin receptor, are required for hepatoportal glucose sensor-stimulated muscle glucose utilization. J. Clin. Invest, 111, 1555-1562.

[210] Badman MK & Flier JS (2005) The gut and energy balance: visceral allies in the obesity wars. Science, 307, 1909-1914.

[211] Cummings DE & Overduin J (2007) Gastrointestinal regulation of food intake. J. Clin. Invest, 117, 13-23.

[212] Moran TH & Schwartz GJ (1994) Neurobiology of cholecystokinin. Crit Rev. Neurobiol., 9, 1-28.

[213] Murphy KG & Bloom SR (2006) Gut hormones and the regulation of energy homeostasis. Nature, 444, 854-859.

Page 108: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

99

[214] Allchin RE, Batten TF, McWilliam PN, & Vaughan PF (1994) Electrical stimulation of the vagus increases extracellular glutamate recovered from the nucleus tractus solitarii of the cat by in vivo microdialysis. Exp. Physiol, 79, 265-268.

[215] Granata AR & Reis DJ (1983) Release of [3H]L-glutamine acid (L-Glu) and [3H]D-aspartic acid (D-Asp) in the area of nucleus tractus solitarius in vivo produced by stimulation of the vagus nerve. Brain Res., 259, 77-93.

[216] Tzschentke TM (2002) Glutamatergic mechanisms in different disease states: overview and therapeutical implications -- an introduction. Amino. Acids, 23, 147-152.

[217] Aicher SA, Sharma S, & Pickel VM (1999) N-methyl-D-aspartate receptors are present in vagal afferents and their dendritic targets in the nucleus tractus solitarius. Neuroscience, 91, 119-132.

[218] Treece BR, Covasa M, Ritter RC, & Burns GA (1998) Delay in meal termination follows blockade of N-methyl-D-aspartate receptors in the dorsal hindbrain. Brain Res., 810, 34-40.

[219] Wang J, Obici S, Morgan K, Barzilai N, Feng Z, & Rossetti L (2001) Overfeeding rapidly induces leptin and insulin resistance. Diabetes, 50, 2786-2791.

[220] Greenberg D, Smith GP, & Gibbs J (1990) Intraduodenal infusions of fats elicit satiety in sham-feeding rats. Am. J. Physiol, 259, R110-R118.

[221] Monnikes H, Lauer G, Bauer C, Tebbe J, Zittel TT, & Arnold R (1997) Pathways of Fos expression in locus ceruleus, dorsal vagal complex, and PVN in response to intestinal lipid. Am. J. Physiol, 273, R2059-R2071.

[222] Lieverse RJ, Jansen JB, Masclee AA, Rovati LC, & Lamers CB (1994) Effect of a low dose of intraduodenal fat on satiety in humans: studies using the type A cholecystokinin receptor antagonist loxiglumide. Gut, 35, 501-505.

[223] Matzinger D, Gutzwiller JP, Drewe J, Orban A, Engel R, D'Amato M, Rovati L, & Beglinger C (1999) Inhibition of food intake in response to intestinal lipid is mediated by cholecystokinin in humans. Am. J. Physiol, 277, R1718-R1724.

[224] Matzinger D, Degen L, Drewe J, Meuli J, Duebendorfer R, Ruckstuhl N, D'Amato M, Rovati L, & Beglinger C (2000) The role of long chain fatty acids in regulating food intake and cholecystokinin release in humans. Gut, 46, 688-693.

[225] Moran TH, Ameglio PJ, Schwartz GJ, & McHugh PR (1992) Blockade of type A, not type B, CCK receptors attenuates satiety actions of exogenous and endogenous CCK. Am. J. Physiol, 262, R46-R50.

[226] Covasa M & Ritter RC (2001) Attenuated satiation response to intestinal nutrients in rats that do not express CCK-A receptors. Peptides, 22, 1339-1348.

Page 109: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

100

[227] Schwartz GJ, Whitney A, Skoglund C, Castonguay TW, & Moran TH (1999) Decreased responsiveness to dietary fat in Otsuka Long-Evans Tokushima fatty rats lacking CCK-A receptors. Am. J. Physiol, 277, R1144-R1151.

[228] Moran TH, Smith GP, Hostetler AM, & McHugh PR (1987) Transport of cholecystokinin (CCK) binding sites in subdiaphragmatic vagal branches. Brain Res., 415, 149-152.

[229] Moran TH, Norgren R, Crosby RJ, & McHugh PR (1990) Central and peripheral vagal transport of cholecystokinin binding sites occurs in afferent fibers. Brain Res., 526, 95-102.

[230] Funakoshi A, Miyasaka K, Shinozaki H, Masuda M, Kawanami T, Takata Y, & Kono A (1995) An animal model of congenital defect of gene expression of cholecystokinin (CCK)-A receptor. Biochem. Biophys. Res. Commun., 210, 787-796.

[231] Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, & Natori T (1992) Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes, 41, 1422-1428.

[232] Moran TH (2008) Unraveling the obesity of OLETF rats. Physiol Behav., 94, 71-78.

[233] Blumberg S, Haba D, Schroeder M, Smith GP, & Weller A (2006) Independent ingestion and microstructure of feeding patterns in infant rats lacking CCK-1 receptors. Am. J. Physiol Regul. Integr. Comp Physiol, 290, R208-R218.

[234] Shiba Y, Yamasaki Y, Kubota M, Matsuhisa M, Tomita T, Nakahara I, Morishima T, Kawamori R, & Hori M (1998) Increased hepatic glucose production and decreased hepatic glucose uptake at the prediabetic phase in the Otsuka Long-Evans Tokushima fatty rat model. Metabolism, 47, 908-914.

[235] Cheung GW, Kokorovic A, & Lam TK (2009) Upper intestinal lipids regulate energy and glucose homeostasis. Cell Mol. Life Sci..

[236] French SJ, Murray B, Rumsey RD, Fadzlin R, & Read NW (1995) Adaptation to high-fat diets: effects on eating behaviour and plasma cholecystokinin. Br. J. Nutr., 73, 179-189.

[237] Spannagel AW, Nakano I, Tawil T, Chey WY, Liddle RA, & Green GM (1996) Adaptation to fat markedly increases pancreatic secretory response to intraduodenal fat in rats. Am. J. Physiol, 270, G128-G135.

[238] Covasa M, Marcuson JK, & Ritter RC (2001) Diminished satiation in rats exposed to elevated levels of endogenous or exogenous cholecystokinin. Am. J. Physiol Regul. Integr. Comp Physiol, 280, R331-R337.

[239] Savastano DM & Covasa M (2005) Adaptation to a high-fat diet leads to hyperphagia and diminished sensitivity to cholecystokinin in rats. J. Nutr., 135, 1953-1959.

Page 110: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

101

[240] Covasa M & Ritter RC (1998) Rats maintained on high-fat diets exhibit reduced satiety in response to CCK and bombesin. Peptides, 19, 1407-1415.

[241] Bi S, Ladenheim EE, Schwartz GJ, & Moran TH (2001) A role for NPY overexpression in the dorsomedial hypothalamus in hyperphagia and obesity of OLETF rats. Am. J. Physiol Regul. Integr. Comp Physiol, 281, R254-R260.

[242] Ritter RC & Ladenheim EE (1985) Capsaicin pretreatment attenuates suppression of food intake by cholecystokinin. Am. J. Physiol, 248, R501-R504.

[243] Zabielski R, Onaga T, Mineo H, Kato S, & Pierzynowski SG (1995) Intraduodenal cholecystokinin octapeptide (CCK-8) can stimulate pancreatic secretion in the calf. Int. J. Pancreatol., 17, 271-278.

[244] Gulley S, Sharma SK, Mansour M, Sullivan CN, Moran TH, & Sayegh AI (2005) Strain differences in myenteric neuron number and CCK1 receptor mRNA expression may account for differences in CCK induced c-Fos activation. Brain Res., 1058, 109-119.

[245] Kievit P, Howard JK, Badman MK, Balthasar N, Coppari R, Mori H, Lee CE, Elmquist JK, Yoshimura A, & Flier JS (2006) Enhanced leptin sensitivity and improved glucose homeostasis in mice lacking suppressor of cytokine signaling-3 in POMC-expressing cells. Cell Metab, 4, 123-132.

[246] Obici S, Zhang BB, Karkanias G, & Rossetti L (2002) Hypothalamic insulin signaling is required for inhibition of glucose production. Nat. Med., 8, 1376-1382.

[247] Schwartz MW & Porte D, Jr. (2005) Diabetes, obesity, and the brain. Science, 307, 375-379.

[248] Lam TK, Pocai A, Gutierrez-Juarez R, Obici S, Bryan J, Aguilar-Bryan L, Schwartz GJ, & Rossetti L (2005) Hypothalamic sensing of circulating fatty acids is required for glucose homeostasis. Nat. Med., 11, 320-327.

[249] Moran TH, Smith GP, Hostetler AM, & McHugh PR (1987) Transport of cholecystokinin (CCK) binding sites in subdiaphragmatic vagal branches. Brain Res., 415, 149-152.

[250] Moran TH, Norgren R, Crosby RJ, & McHugh PR (1990) Central and peripheral vagal transport of cholecystokinin binding sites occurs in afferent fibers. Brain Res., 526, 95-102.

[251] Sclafani A, Ackroff K, & Schwartz GJ (2003) Selective effects of vagal deafferentation and celiac-superior mesenteric ganglionectomy on the reinforcing and satiating action of intestinal nutrients. Physiol Behav., 78, 285-294.

[252] Fraser KA & Davison JS (1992) Cholecystokinin-induced c-fos expression in the rat brain stem is influenced by vagal nerve integrity. Exp. Physiol, 77, 225-228.

Page 111: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

102

[253] Monnikes H, Lauer G, & Arnold R (1997) Peripheral administration of cholecystokinin activates c-fos expression in the locus coeruleus/subcoeruleus nucleus, dorsal vagal complex and paraventricular nucleus via capsaicin-sensitive vagal afferents and CCK-A receptors in the rat. Brain Res., 770, 277-288.

[254] Chen DY, Deutsch JA, Gonzalez MF, & Gu Y (1993) The induction and suppression of c-fos expression in the rat brain by cholecystokinin and its antagonist L364,718. Neurosci. Lett., 149, 91-94.

[255] Guard DB, Swartz TD, Ritter RC, Burns GA, & Covasa M (2009) NMDA NR2 receptors participate in CCK-induced reduction of food intake and hindbrain neuronal activation. Brain Res., 1266, 37-44.

[256] Cull-Candy S, Brickley S, & Farrant M (2001) NMDA receptor subunits: diversity, development and disease. Curr. Opin. Neurobiol., 11, 327-335.

[257] Covasa M & Ritter RC (1998) Rats maintained on high-fat diets exhibit reduced satiety in response to CCK and bombesin. Peptides, 19, 1407-1415.

[258] Covasa M, Marcuson JK, & Ritter RC (2001) Diminished satiation in rats exposed to elevated levels of endogenous or exogenous cholecystokinin. Am. J. Physiol Regul. Integr. Comp Physiol, 280, R331-R337.

[259] Savastano DM & Covasa M (2005) Adaptation to a high-fat diet leads to hyperphagia and diminished sensitivity to cholecystokinin in rats. J. Nutr., 135, 1953-1959.

[260] French SJ, Murray B, Rumsey RD, Fadzlin R, & Read NW (1995) Adaptation to high-fat diets: effects on eating behaviour and plasma cholecystokinin. Br. J. Nutr., 73, 179-189.

[261] Spannagel AW, Nakano I, Tawil T, Chey WY, Liddle RA, & Green GM (1996) Adaptation to fat markedly increases pancreatic secretory response to intraduodenal fat in rats. Am. J. Physiol, 270, G128-G135.

[262] Eberlein GA, Eysselein VE, & Goebell H (1988) Cholecystokinin-58 is the major molecular form in man, dog and cat but not in pig, beef and rat intestine. Peptides, 9, 993-998.

[263] Boey D, Lin S, Karl T, Baldock P, Lee N, Enriquez R, Couzens M, Slack K, Dallmann R, Sainsbury A, & Herzog H (2006) Peptide YY ablation in mice leads to the development of hyperinsulinaemia and obesity. Diabetologia, 49, 1360-1370.

[264] Scrocchi LA, Hill ME, Saleh J, Perkins B, & Drucker DJ (2000) Elimination of glucagon-like peptide 1R signaling does not modify weight gain and islet adaptation in mice with combined disruption of leptin and GLP-1 action. Diabetes, 49, 1552-1560.

[265] Ritter RC, Covasa M, & Matson CA (1999) Cholecystokinin: proofs and prospects for involvement in control of food intake and body weight. Neuropeptides, 33, 387-399.

Page 112: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

103

[266] Rao RV, Roettger BF, Hadac EM, & Miller LJ (1997) Roles of cholecystokinin receptor phosphorylation in agonist-stimulated desensitization of pancreatic acinar cells and receptor-bearing Chinese hamster ovary cholecystokinin receptor cells. Mol. Pharmacol., 51, 185-192.

[267] Marino CR, Leach SD, Schaefer JF, Miller LJ, & Gorelick FS (1993) Characterization of cAMP-dependent protein kinase activation by CCK in rat pancreas. FEBS Lett., 316, 48-52.

[268] Piiper A, Stryjek-Kaminska D, Klengel R, & Zeuzem S (1997) CCK, carbachol, and bombesin activate distinct PLC-beta isoenzymes via Gq/11 in rat pancreatic acinar membranes. Am. J. Physiol, 272, G135-G140.

[269] Rogers RC & Hermann GE (2008) Mechanisms of action of CCK to activate central vagal afferent terminals. Peptides, 29, 1716-1725.

[270] Eysselein VE, Eberlein GA, Hesse WH, Schaeffer M, Grandt D, Williams R, Goebell H, & Reeve JR, Jr. (1990) Molecular variants of cholecystokinin after endogenous stimulation in humans: a time study. Am. J. Physiol, 258, G951-G957.

[271] Reeve JR, Jr., Eysselein V, Walsh JH, Ben-Avram CM, & Shively JE (1986) New molecular forms of cholecystokinin. Microsequence analysis of forms previously characterized by chromatographic methods. J. Biol. Chem., 261, 16392-16397.

[272] Eberlein GA, Eysselein VE, Hesse WH, Goebell H, Schaefer M, & Reeve JR, Jr. (1987) Detection of cholecystokinin-58 in human blood by inhibition of degradation. Am. J. Physiol, 253, G477-G482.

[273] Eysselein VE, Eberlein GA, Hesse WH, Singer MV, Goebell H, & Reeve JR, Jr. (1987) Cholecystokinin-58 is the major circulating form of cholecystokinin in canine blood. J. Biol. Chem., 262, 214-217.

[274] Reeve JR, Jr., Green GM, Chew P, Eysselein VE, & Keire DA (2003) CCK-58 is the only detectable endocrine form of cholecystokinin in rat. Am. J. Physiol Gastrointest. Liver Physiol, 285, G255-G265.

[275] Rehfeld JF, Sun G, Christensen T, & Hillingso JG (2001) The predominant cholecystokinin in human plasma and intestine is cholecystokinin-33. J. Clin. Endocrinol. Metab, 86, 251-258.

[276] Kreis ME, Zittel TT, Raybould HE, Reeve JR, Jr., & Grundy D (1997) Prolonged intestinal afferent nerve discharge in response to cholecystokinin-58 compared to cholecystokinin-8 in rats. Neurosci. Lett., 230, 89-92.

[277] Glatzle J, Raybould HE, Kueper MA, Reeve JR, Jr., & Zittel TT (2008) Cholecystokinin-58 is more potent in inhibiting food intake than cholecystokinin-8 in rats. Nutr. Neurosci., 11, 69-74.

Page 113: INTESTINAL CHOLECYSTOKININ CONTROLS …...Cholecystokinin (CCK) is a gut peptide involved in the regulation of energy homeostasis by duodenal lipids via a neuronal network. However,

104

[278] Reeve JR, Jr., McVey DC, Bunnett NW, Solomon TE, Keire DA, Ho FJ, Davis MT, Lee TD, Shively JE, & Vigna SR (2002) Differences in receptor binding and stability to enzymatic digestion between CCK-8 and CCK-58. Pancreas, 25, e50-e55.