upper small intestinal protein sensing in the regulation ... · upper small intestinal protein...

88
Upper Small Intestinal Protein Sensing in the Regulation of Glucose Homeostasis by Sophie Claire Hamr A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Physiology University of Toronto © Copyright by Sophie Hamr (2016)

Upload: others

Post on 27-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

Upper Small Intestinal Protein Sensing in the Regulation of Glucose Homeostasis

by

Sophie Claire Hamr

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

Department of Physiology University of Toronto

© Copyright by Sophie Hamr (2016)

Page 2: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

ii

Upper small intestinal protein sensing in the regulation of glucose

homeostasis

Sophie Claire Hamr

Master of Science

Department of Physiology University of Toronto

2016

Abstract

High-protein diets improve glucose control in both healthy and type 2 diabetic

individuals. Given that upper intestinal lipids trigger a pre-absorptive, gut-brain-liver

neuronal axis to suppress glucose production (GP), it is urgent to assess the potential

glucoregulatory capacity of intestinal protein sensing. We here demonstrate that in

healthy rodents in vivo, upper small intestinal infusion of protein hydrolysate improved

intravenous glucose tolerance. This was due at least in part to insulin-independent

effects on the liver, given that the same infusion suppressed hepatic GP during a basal-

insulin euglycemic clamp. Co-infusion of tetracaine reversed the ability of protein to

improve glucose tolerance, indicating that a neuronal network is initiated at the level of

the gut to regulate glucose homeostasis. Collectively, these findings show for the first

time that pre-absorptive protein sensing in the upper small intestine improves glucose

tolerance by triggering a neuronal network to lower GP.

Page 3: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

iii

Acknowledgments

I would like to acknowledge my supervisor, Dr. Tony Lam, as well as my MSc

committee members Drs. Nicola Jones and Thomas Wolever, for their guidance and

support throughout my graduate studies. I would also like to thank all of the past and

present members of the Lam lab for their direction and assistance with my studies and

for their unconditional moral support, particularly Dr. Frank Duca, Dr. Brittany

Rasmussen and Paige Bauer for their work on this project.

Page 4: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

iv

Table of Contents Chapter 1 Introduction ................................................................................................... 1

1.1 Obesity and diabetes ............................................................................................ 1 1.2 Dietary protein and metabolic homeostasis .......................................................... 3 1.3 Intestinal nutrient sensing in the regulation of metabolic homeostasis ................. 7

1.3.1 Control of food intake ............................................................................... 11 1.3.2 Regulation of glucose homeostasis ......................................................... 14 1.3.3 Dysregulation in obesity and diabetes ..................................................... 18

1.4 Intestinal protein sensing .................................................................................... 22 Chapter 2 Hypothesis and Aims ................................................................................. 26 Chapter 3 Materials and Methods ............................................................................... 29

3.1 Animals .............................................................................................................. 29 3.2 Surgical Procedure ........................................................................................... 29

3.2.1 Preparation of cannulae ........................................................................... 29 3.2.2 Cannulation surgery ................................................................................. 30

3.3 Intestinal infusions and treatments ................................................................ 32 3.4 Intravenous glucose tolerance test ................................................................. 33 3.5 Pancreatic (basal insulin) euglycemic clamp ................................................. 34 3.6 Biochemical analysis ........................................................................................ 35

3.6.1 Plasma glucose ........................................................................................ 35 3.6.2 Plasma amino acids ................................................................................. 35 3.6.3 Plasma [3-3H]-glucose specific activity ..................................................... 37

3.7 Tracer-dilution calculations ............................................................................. 37 3.8 Statistical analysis ............................................................................................ 38

Chapter 4 Results ......................................................................................................... 40 Chapter 5 Discussion ................................................................................................... 54 Chapter 6 References ................................................................................................... 65

Page 5: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

v

List of Tables

Table 1: Typical amino acid content of Casein enzymatic hydrolysate from Sigma-

Aldrich (Product

22090).................................................................................................51

Table 2: Body weight and cumulative post-surgical food intake of the groups of rats that

underwent either the intravenous glucose tolerance test or clamp

protocol...................52

Table 3: Plasma glucose concentrations during the basal insulin euglycemic

clamp.....53

Page 6: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

vi

List of Figures

Figure 1: Intestinal nutrient sensing in the regulation of metabolic

homeostasis............25

Figure 2: Schematic of the working

hypothesis..............................................................28

Figure 3: Gut infusion-IVGTT protocol to evaluate the effects of upper small intestinal

protein sensing on glucose

tolerance.............................................................................47

Figure 4: Upper small intestinal infusion of protein improves glucose

tolerance............48

Figure 5. Upper small intestinal casein hydrolysate increases the glucose infusion rate

during the basal insulin euglycemic

clamp.....................................................................49

Figure 3: Gut infusion-IVGTT protocol to evaluate the effects of upper small intestinal

protein sensing on glucose

tolerance.............................................................................50

Page 7: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

vii

List of Abbreviations ACS Acyl-CoA synthetase AUC Area under the curve CCK Cholecystokinin CD36 Cluster of differentiation 36 DPP-IV Dipeptidyl Peptidase-4 DVC Dorsal vagal complex EEC Enteroendocrine cell ENS Enteric nervous system FXR Farnesoid X receptor GI Gastrointestinal GLP-1 Glucagon-like peptide 1 GIP Glucose-dependent insulinotropic peptide GIR Glucose infusion rate GPCR G-protein coupled receptor HbA1c Glycated hemoglobin IVGTT Intravenous glucose tolerance test i.p. Intraperitoneal LCFA Long-chain fatty acid NMDA N-methyl-D-aspartate NTS Nucleus of the solitary tract OGTT Oral glucose tolerance test OLETF Otsuka-Long-Evans-Tokushima Fatty PepT1 Peptide transporter 1 PLC Phospholipase C POMC Pro-opiomelanocortin PKA Protein kinase A PKC Protein kinase C PYY Peptide YY Ra Rate of glucose appearance Rd Rate of glucose disappearance SD Sprague Dawley SGLT-1 Sodium-glucose cotransporter 1

Page 8: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

viii

Published Manuscripts that contributed to this thesis

Hamr SC, Wang B, Swartz TD and Duca FA. Does nutrient sensing determine how we

“see” food? Curr Diab Rep 15(6):604 (2015).

Duca FA, Bauer PV, Hamr SC and Lam TK. Glucoregulatory relevance of small

intestinal nutrient sensing in physiology, bariatric surgery and pharmacology. Cell Metab

22(3):367-80 (2015).

Bauer PV, Hamr SC and Duca FA. Regulation of energy balance by a gut-brain axis

and involvement of the gut microbiota. Cell Mol Life Sci 73(4):737-55 (2016).

Page 9: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

1

Chapter 1

Introduction

1.1 Obesity and diabetes

Over 700 million individuals are obese, and over 300 million have type 2 diabetes

worldwide, rendering a state of global epidemic1,2. Given that obesity and diabetes are

associated with a number of life-threatening co-morbidities, and collectively cost the

Canadian healthcare system over $20 billion per year3,4, there is an urgent need to

understand their pathologies in hopes to develop successful treatments.

The etiologies of these two diseases are complex and interconnected given that

approximately 80% of type 2 diabetic individuals are overweight or obese5. Obesity and

diabetes are linked to a number of genetic and environmental factors, which collectively

lead to a dysregulation in the homeostatic control of energy balance and glucose

homeostasis. In healthy individuals, energy homeostasis is tightly regulated by a

multitude of interconnected signaling networks, which integrate signals of energy status

and regulate exogenous nutrient intake and endogenous nutrient metabolism,

accordingly. The classical view of glucose regulation and of diabetes development is an

islet-centered view, whereby blood glucose levels are primarily regulated by the

pancreatic hormones insulin and glucagon. Insulin resistance develops at the level of

peripheral tissues, leading to a compensatory increase in pancreatic insulin secretion.

Page 10: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

2

As insulin resistance worsens, β-cell exhaustion occurs and insulin secretion no longer

compensates, resulting in hyperglycemia and a diagnosis of type 2 diabetes6. However,

50% of glucose tolerance is accounted for by insulin-independent mechanisms7, which

remain incompletely understood. Schwartz et al. recently proposed the importance of a

“brain-centered glucoregulatory system,” whereby the brain senses circulating nutrients

to regulate plasma glucose independently of insulin8. Indeed, nutrient sensing

mechanisms are an important form of insulin-independent regulation on energy

homeostasis.

Interestingly, more recent evidence brings to light the importance of nutrient sensing by

the gastrointestinal (GI) tract, indicating that nutrients generate critical negative

feedback on energy homeostasis upon initial contact with the body9. The impact of GI

or “gut” nutrient sensing on glucose homeostasis is highlighted by the fact that

metformin, the most widely prescribed drug for the treatment of type 2 diabetes, has

been found in both rodents and humans to exert effects on glycaemia via a gut-

localized site of action10,11. Further, when the GI tract is surgically re-arranged to treat

obesity, redirecting nutrient flux and implicating distal intestinal nutrient sensing, a drop

in plasma glucose levels occurs almost immediately, prior to any weight loss12,13.

Finally, pharmacological treatments increasing levels of the nutrient-stimulated gut

hormone glucagon-like peptide 1 (GLP-1) have proven effective in regulating glycaemia

in type 2 diabetic individuals14. While the mechanisms underlying these therapies

remain incompletely understood, they collectively implicate an urgent need to further

Page 11: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

3

elucidate intestinal nutrient signaling in the regulation of glucose homeostasis and

pathogenesis of obesity and diabetes.

1.2 Dietary protein and metabolic homeostasis

In comparison to the increasing knowledge of how carbohydrates and lipids regulate

glucose homeostasis, the effects of dietary proteins remain less clear. Numerous

studies show that intravenous infusion of amino acids in a hyperinsulinemic,

euglycemic condition induces acute insulin resistance at the level of glucose uptake by

peripheral tissues15-19. In vitro studies in isolated myocytes indicate that amino acids

inhibit insulin-stimulated glucose uptake likely via competitive inhibition of glucose

oxidation (glycolysis)20-22, activation of mTOR23, and inhibition of the insulin signaling

pathway24. Interestingly, however, these findings are not consistent, given that others

found that elevating plasma amino acids had no effect on glucose uptake during a

hyperinsulinemic clamp in healthy men25, and actually increased glucose uptake to

lower glucose levels during unclamped hyperinsulinemia26. Thus, in order to examine

how dietary protein affects glucose turnover in the context of high-protein feeding,

Rossetti et al pair-fed rats either a high-protein (60% protein) or low-protein (18%

protein) diet27. Following 10 days of dietary intervention, rats fed the high-protein diet

had increased fasting glucose levels and decreased insulin sensitivity during a

hyperinsulinemic clamp at the levels of both hepatic glucose production and peripheral

glucose uptake27. This study suggests that acutely increasing dietary protein intake

may be detrimental to glucose homeostasis, due to amino acid-induced insulin

Page 12: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

4

resistance. However, as described below, longer dietary interventions in fact show that

high protein feeding improves energy balance and glucose control, suggesting that

dietary proteins may exert other, yet to be identified effects that positively impact

metabolic homeostasis.

High-protein diets have been widely prescribed as a dietary intervention in overweight

individuals, and over a 6-month test period, high-protein diets have more successful

weight loss outcomes than conventional, high-carbohydrate weight-loss diets28-30.

These high-protein diet-treated individuals not only experience a greater overall weight

loss, but also display decreased fat mass and reduced fasting plasma free fatty-acids,

triglycerides and cholesterol29,30, indicating an overall improvement in lipid

homeostasis. These results were confirmed in healthy rodents, where a 42-day high-

protein diet reduced body weight and fat mass accumulation compared to a low-protein

diet31. Interestingly, individuals fed a high-protein weight-loss diet often have

corresponding increases in insulin sensitivity and lowering of glycated hemoglobin

(HbA1c)28,29,32,33, suggesting that increasing dietary proteins can improve glucose

homeostasis in addition to their weight-loss properties. Indeed, when Samaha and

colleagues followed up after 1 year of high-protein versus high-carbohydrate weight-

loss dieting, the high-protein group displayed lower levels HbA1c despite no persisting

effects on body weight34. However, improvements in glucose homeostasis following

long-term high-protein feeding may be secondary to the initial weight-loss, given that

modest weight loss per se is associated with improved glycemic control35. Thus, it is

important to look at the effects of short-term high-protein feeding independent of any

Page 13: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

5

weight loss. On this note, when subjects were fed a weight-maintenance diet for 5

weeks, a high-protein diet improved glucose tolerance and HbA1c levels compared to a

high-carbohydrate diet, independent of any change in body weight36. Along with an

epidemiological analysis linking higher dietary amino acid intake with a lower risk of

type 2 diabetes37, these findings highlight the importance of looking more closely at the

effects of high-protein feeding to regulate blood glucose.

Paralleling these human studies, rodents fed a high milk-protein diet for 6 months

exhibited lower fasting plasma glucose and improved glucose tolerance as assessed

by an oral glucose tolerance test (OGTT) compared to rodents fed a normal-protein

diet38. However, it is important to note that in this study, rats were fed the diets ad

libitum, resulting in a 13% lower total food intake in the high-protein diet group38.

Indeed, both rodents and humans commonly exhibit lower food intake in response to a

high-protein diet31,33,39. Thus, in a follow up study, rats were pair-fed to look at the

effects of high-protein feeding independent of effects on food intake. In this context,

high-protein feeding no longer decreased fat mass, however the high-protein-fed rats

displayed persistent improvements in glucose tolerance and fasting plasma glucose40.

This study provides strong evidence that dietary protein can regulate glucose

homeostasis independent of effects on food intake and body weight.

In an acute setting, rats fasted for 24 hours and fed either a standard mixed meal or an

equivalent caloric load of protein hydrolysate experienced a much lower glucose

Page 14: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

6

response to the protein meal41. However, in this case, it is impossible to distinguish the

glucose-lowering effects of the protein per se from the coinciding lack of dietary

carbohydrate contributing to the glucose response. Indeed, this variable must also be

considered in the case of the high-protein diet models described earlier, given that

protein was always substituted for carbohydrates in these diets. Thus, a model to look

at the glucose-lowering effects of protein per se is presented in studies where protein is

administered in addition to a standard meal. In one such study, overnight-fasted human

subjects consumed a standard maltodextrin beverage, with or without the addition of

protein hydrolysates42. The addition of protein lowered the glucose response to the

maltodextrin beverage, despite the equal contribution of carbohydrates and the higher

caloric load42. This phenomenon is mirrored by two similar studies where protein was

administered in the form of a whey-protein beverage either directly or 30 minutes prior

to a standard meal, while control groups did not receive a pre-load. In both cases, pre-

loading with a whey protein bolus lowered the glucose response to the subsequent

meal43,44. It is interesting to note that although amino acids stimulate insulin secretion,

the lower glucose response observed in the study by Akhavan et al was not associated

with higher insulin levels43. Thus, it would seem that the additional dietary protein load

exerts insulin-independent effects on glucose regulation in order to improve glucose

tolerance during the subsequent meal.

Our laboratory recently investigated this insulin-independent, protein-induced regulation

of glucose homeostasis. Healthy, male Sprague-Dawley rats were overnight-fasted and

fed a 19 kcal meal with either 20% or 60% calories from protein (milk-derived casein).

Page 15: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

7

While the total calorie intake, rate of food intake, and insulin response were indeed

identical between groups, the 60%, high-protein diet resulted in a lower plasma glucose

response from 30-60 minutes post meal initiation45. Interestingly, although the insulin

response was the same, the high-protein meal increased plasma glucagon whereas the

low-protein meal did not. Indeed, glucagon signaling was partially responsible for the

glucose-lowering effects of protein, as hindbrain glucagon receptor blockade reversed

the improvement in glucose response seen at 60 minutes45. This is consistent with the

previously documented glucagon-stimulating effects of protein and glucose-lowering

effects of brain glucagon signaling45-47. However, it is important to note that glucagon

receptor blockade did not reverse the glucose-lowering effects of high-protein feeding

during the earlier time-points, indicating that there are additional, potentially insulin- and

glucagon-independent mechanisms at play. Given that this glucose-lowering response

is robust within just 30 minutes of feeding, one could speculate that negative feedback

mechanisms arise at the level of the gut upon interaction with incoming nutrients. This

theory is in fact not unprecedented given that, as described below, sensing of nutrients

such as protein by the GI tract can initiate rapid and potent negative feedback on

whole-body metabolic homeostasis.

1.3 Intestinal nutrient sensing in the regulation of metabolic homeostasis

The GI tract is the primary site of interaction between incoming nutrients and the body,

providing a site for early negative feedback on metabolic homeostasis. The extensive

Page 16: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

8

neural and humoral connectivity of the gut wall to other important energy-regulating

areas, such as the brain, allows it to effectively relay information about incoming

nutrients in order to prevent energy excess upon nutrient absorption into the circulation.

The upper small intestine, where the majority of nutrient absorption occurs, is the major

feedback site responsible for energy regulation48.

Nutrient signaling is mediated by the release of gut peptide hormones from

enteroendocrine cells (EECs). EECs, expressed throughout the gut epithelial mucosa,

projecting their apical surfaces into the intestinal lumen, allowing them to act as

chemoreceptors, detecting nutrients predominantly via cell-surface G-protein coupled

receptor (GPCRs) or solute transporters49. In response to the detection of nutrients,

EECs release gut peptides from their basal surfaces into intestinal capillaries. Thus, the

EEC is proposed as the gut’s nutrient-sensing cell, directly sensing luminal nutrients

and generating humoral signals in response. Gut peptide hormones are thought to be

associated with individual EEC types that differ in the gut peptides they produce and

their localization along the GI tract. The proximal small intestine expresses “I cells,”

which produce the peptide cholecystokinin (CCK), and “K-cells” that produce glucose-

dependent insulinotropic peptide (GIP), while the more distal “L cells” produce GLP-1

and Peptide YY (PYY). However, there is accumulating evidence for co-expression of

gut peptides thought to be produced in distinct cell types, suggesting that the EEC may

in fact be a single cell type that produces varying spectra of peptides depending on its

environment50,51, though this is continually debated52.

Page 17: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

9

Specific and independent sensory and signal transduction mechanisms mediate gut

peptide release in response to individual types of nutrients. EECs express a number of

nutrient-specific GPCRs such as the fatty-acid receptors GPR4053,54 and GPR12054,55

and the amino acid and peptide-receptive calcium sensing receptor (CaSR)56, as well

as solute transporters such as sodium-glucose transporter 1 (SGLT-1)57 and peptide

transporter 1 (PepT1)58. Via these chemosensory machineries, luminal nutrients

promote the exocytosis of gut peptides through intracellular metabolism, second-

messenger signaling cascades and/or membrane depolarization59. Indeed, chemical

inhibition or genetic knockout of one of these nutrient-sensing receptors or transporters

will results in impaired CCK, GLP-1 or GIP release in response to feeding53,60-63.

However, refuting this classical model of EEC nutrient sensing, there is accumulating

evidence for indirect mechanisms through which nutrients may alternatively stimulate

gut peptide release. For example, numerous studies show that lipid absorption and

packaging into chylomicrons, and the subsequent release of Apolipoprotein A-IV from

enterocytes, is required for dietary lipids to stimulate the release of CCK and GIP64-66.

Bile acids, components of bile that are released form the liver into the intestinal lumen

in response to nutrient ingestion to aid with the solubilization and digestion of lipids,

have also been implicated in stimulating gut peptide release. Bile acids themselves

may act directly on EECs to regulate gut peptide secretion via their endogenous

receptor, the farnesoid X receptor (FXR), though the evidence to date is

contradictory67,68. Nonetheless, whether nutrients signal directly on or within EECs, or

indirectly via other cell types, the resulting release of gut peptide hormones is the first

step in the initiation of nutrient-induced negative feedback axes.

Page 18: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

10

Upon exocytosis from EECs, gut peptides are secreted into the portal circulation to act

directly on target organs in an endocrine fashion. For example, systemic plasma CCK

levels increase in the postprandial state in both humans and rodents69,70, and the brain

expresses CCK receptors that are implicated in metabolic regulation71. The brain also

expresses GLP-1 receptors72,73, however unlike CCK, GLP-1 is rapidly degraded in

circulation by the enzyme Dipeptidyl Peptidase-4 (DPP-IV), resulting in less than 10%

of GLP-1 reaching the systemic circulation following secretion74. GLP-1 may instead

reach the brain via lymph vessels, given that in the postprandial state, lower levels of

DPP-IV and higher levels of GLP-1 are found in the lymph than in the portal vein75-77.

The gut peptide PYY is likely to signal predominantly in an endocrine fashion, given

that circulating PYY levels peak 1.5 hours after feeding and remain elevated for several

hours78. Alternatively, CCK and GLP-1, which display more transient peaks, may in fact

be more important in paracrine signaling with the gut.

The proximal small intestine is densely innervated by the vagus nerve, with afferent

fibers (projecting to the brain) far outnumbering efferent fibers79,80. Extensive evidence

indicates that the acute regulation of food intake and plasma glucose in response to

intestinal nutrients is mediated by these vagal afferents. Indeed, vagal afferents

express receptors for CCK, GLP-1 and PYY, and activation of these vagal gut peptide

receptors leads to afferent firing81. Vagal afferent dendrites innervate the lamina propria

of the gut mucosa, rather than EECs directly, indicating that gut peptides likely diffuse

to nearby vagal targets in a paracrine fashion once released81. Alternatively, neurons of

the enteric nervous system (ENS), which also express gut peptide receptors and are

Page 19: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

11

situated in between EECs and vagal afferents, may act as an intermediate neuronal

relay82-84. One study found that the nutrient-sensing receptor GPR40 is expressed by

vagal afferent neurons themselves, suggesting that in some cases nutrients diffuse

through the gut epithelium to activate vagal afferents directly85. Nonetheless, nutrient

influx results in the activation and firing of vagal afferent neurons, which subsequently

relay a signal to the nucleus of the solitary tract (NTS) in the dorsal vagal complex

(DVC) of the hindbrain, where vagal afferents terminate86. Indeed, intraduodenal

nutrient infusion leads to c-fos activation of NTS neurons, and this response is blocked

by treatment of gut vagal afferents with the neurotoxin capsaicin87. Gut-derived signals

are integrated by the NTS, which relays to other brain areas to regulate metabolic

homeostasis accordingly88. This EECàvagal afferentàhindbrain axis mediates a wide

range of feedback systems activated by nutrients in the intestine, including important

regulation on food intake and glucose homeostasis (Figure 1).

1.3.1 Control of food intake

Fasted rats decrease their rate of feeding within minutes of meal initiation89, indicating

the presence of negative feedback on food intake before nutrients reach the circulation

(the term “pre-absorptive” hereafter refers to effects generated by nutrients prior to the

release of nutrients into the portal and systemic circulation, but not necessarily prior to

absorption by enterocytes). This pre-absorptive feedback is initiated within the small

intestine, given that a suppression of food intake is entirely absent during sham

feeding, when food is drained from gastric fistulas, never reaching the duodenum90.

Page 20: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

12

Alternatively, infusion of a nutrient solution directly into the lumen of the duodenum

results in a rapid suppression of sham feeding91. Surgical ablation of the

subdiaphragmatic vagus or gut capsaicin treatment completely reverses the

suppressive effects of intraduodenal proteins, carbohydrates or lipids on sham

feeding92,93. Likewise, capsaicin treatment of the hindbrain blocks the effect of intestinal

nutrients to suppress sham feeding94. Together, these results indicate that the acute

feedback on food intake in response to duodenal nutrients is entirely mediated by a

vagal gutàhindbrain neuronal axis.

CCK, often referred to as the “satiety hormone,” has long been associated with acute

control of food intake in both rodents and humans95,96. In humans, intravenous infusion

of a CCK receptor antagonist delays feelings of “fullness” during a meal97, and reverses

the suppression of ad libitum food intake by intraduodenal lipid infusion98. In rodents,

CCK receptor knockout, or co-infusion of a CCK receptor antagonist, reverse vagal

afferent firing and suppression of food intake in response to intraduodenal nutrients99-

101. Likewise, peripheral (intraperitoneal; i.p.) CCK-8 (the biologically active form of

CCK) injection activates brainstem neurons to suppress feeding, and this effect is

completely abolished by intestinal capsaicin, subdiaphragmatic vagotomy, or hindbrain

capsaicin94,102,103. This CCK-mediated gutàbrainstem axis requires CCK-A (CCK-1),

but not CCK-B receptors, which are indeed expressed by vagal afferent fibers101,104,105.

Together, these results suggest that CCK produced by the gut regulates satiety entirely

via vagal CCK-1 receptors. While CCK receptors are expressed in the brain, and

intracerebroventricular CCK administration lowers feeding106, central receptors may

Page 21: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

13

mediate pathways by centrally-produced, and not intestinally-produced CCK103, given

that central CCK-1 receptor inhibition does not block feeding suppression by intestinal

nutrients107.

Postprandial GLP-1 levels correlate with lower hunger scores in humans108, suggesting

a role for GLP-1, in addition to CCK, in the suppression of appetite. Indeed, in rodents,

i.p. GLP-1 injection suppresses food intake, and this effect is blocked by vagotomy or

capsaicin, implicating a parallel role for vagal afferent GLP-1 action in nutrient-induced

satiety109,110. In humans, intravenous GLP-1 infusion dose-dependently suppresses

energy intake during an ad libitum meal111. Patients that have undergone truncal

vagotomy surgery as a treatment for duodenal ulcers provide a comparative human

model to subdiaphragmatic vagotomized rodents, and these individuals are

characterized by impaired suppression of feeding in response to intravenous GLP-1

infusion112. Thus, there is extensive evidence that like CCK, GLP-1 released in

response to luminal nutrients acts on local, vagal afferents to trigger a gut-brain axis to

suppress feeding.

At the level of the hindbrain, the activation of NTS neurons by vagal afferents and

subsequent neuronal relay is incompletely understood, but involves N-methyl-D-

aspartate (NDMA) receptor transmission and subsequent phosphorylation of ERK 1/2

and synapsin I within the NTS113-115. The distinct neuronal populations within the NTS

that are activated by vagal afferent signaling appear to include anorexigenic pro-

Page 22: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

14

opiomelanocortin (POMC)-expressing neurons116. Indeed, blockade of NTS

melanocortin receptor (MC4R) signaling reverses the activation of NTS ERK1/2 and

suppression of feeding in response to peripheral CCK117. NTS neurons then project to

other brainstem nuclei which contain neuronal motor output that controls the

behaviours involved in feeding, supporting a possible brainstem-isolated reflex to

control food intake. Indeed, intestinal nutrients suppress food intake in decerebrate rats

with only the brainstem intact118. However, NTS neurons also project to many higher-

order brain regions involved in the control of feeding, including the hypothalamic

arcuate and paraventricular nuclei. Indeed, Otsuka-Long-Evans-Tokushima Fatty

(OLETF) rats, which lack the CCK-1 receptor and are subsequently hyperphagic and

mildly obese, display altered hypothalamic melanocortin neuropeptides119, supporting a

role for the hypothalamus in regulation of feeding by gut peptides.

1.3.2 Regulation of glucose homeostasis

While the suppression of food intake by gut peptide-mediated nutrient sensing

mechanisms has been studied extensively, far less is known about the role of nutrient

signaling in the regulation of blood glucose. However, accumulating evidence supports

a role for the GI tract in acutely regulating whole-body glucose metabolism. The first

observation that the gut could regulate blood glucose was the discovery of the “incretin”

effect, whereby oral glucose administration results in roughly double the insulin

response compared to delivery of the same amount of glucose intravenously120.

Indeed, the gut peptides GIP and GLP-1, both secreted in response to intestinal

Page 23: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

15

nutrients, act directly on pancreatic β-cells to stimulate glucose-dependent insulin

secretion, subsequently improving glucose tolerance121-123. Interestingly, when GLP-1

is infused intravenously, glucose tolerance improves not only through an increase in

insulin secretion but also by insulin-independent increases in glucose disposal124. In

another study, intravenous GLP-1 infusion during a basal insulin pancreatic clamp

suppressed hepatic glucose production independent of any pancreatic action125.

Although the mechanisms are unknown, these results suggest that the glucoregulatory

capacity of gut-derived signals is not in fact limited to the incretin effect. Brain GLP-1

signaling may play a role, given that central GLP-1 infusion stimulates muscle glucose

uptake independent of muscle insulin receptors126, and hypothalamic GLP-1 infusion

suppresses hepatic glucose production127. These studies suggest that gut-derived

GLP-1 may reach the brain to signal via central GLP-1 receptors to regulate blood

glucose, though again, this signaling axis could be more relevant to centrally-produced

GLP-1.

Given the implication of vagal afferent gut peptide receptors in the regulation of food

intake, vagal GLP-1 receptor signaling within the gut might also play a role in the

regulation of glucose homeostasis by GLP-1. To investigate this hypothesis, a rat

model was developed with specific knockdown of GLP-1 receptors in vagal afferent

neurons through delivery of an shRNA lentiviral vector into the nodose ganglia128. In

this rat model, a 50% knockdown of vagal afferent GLP-1 receptors resulted in

increased post-meal glucose levels following an overnight fast128, confirming that

paracrine, vagal GLP-1 signaling plays a physiological role in glucose regulation.

Page 24: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

16

Furthermore, a low oral dose of DPP-IV inhibitor, which selectively inhibits intestinal

DPP-IV activity, increases vagal afferent firing and improves glucose tolerance, and

this effect is absent in GLP-1-/- mice129, further supporting the role of vagal GLP-1

signaling within the intestine to regulate glucose homeostasis. Of note, in addition to

innervating the gut wall, vagal afferent fibers innervate the portal vein and this is a

second site at which nutrients may trigger a gut-brain axis prior to reaching the

systemic circulation. Indeed, infusion of glucose directly into the portal vein increases

whole-body GU to lower plasma glucose, and this is blocked by co-infusion of a GLP-1

receptor antagonist130, implicating a potential role for hepatoportal GLP-1 receptors in

regulating whole-body glucose homeostasis via a gut-brain axis. Nevertheless, further

work is required to dissect intestinal and hepatoportal GLP-1 signaling in the regulation

of glucose homeostasis via a gut-brain axis.

Gut peptide regulation of glucose homeostasis is in fact not limited to the well-known

“incretin hormones” GLP-1 and GIP, as intravenous infusion of CCK increases insulin

secretion and lowers plasma glucose levels131. Indeed, β-cells express the CCK

receptor132, and CCK-/- mice display impaired insulin response to i.p. glucose

injection133. In vitro studies show that like GLP-1 and GIP, CCK stimulates insulin

secretion from β-cells by enhancing GSIS134-136. In addition, CCK-1 receptor signaling

within the intestine contributes to the glucoregulatory effects of CCK. Intraduodenal

infusion of a CCK-1 receptor inhibitor dysregulates the glucose response to mixed-meal

feeding in healthy rodents137, indicating that intestinal CCK-1 receptor signaling is in

fact required for normal glucose homeostasis during digestion.

Page 25: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

17

In order to dissect the mechanisms through which intestinal nutrient sensing might

regulate glucose homeostasis via CCK, we examined the effect of intraduodenal

nutrient infusion on glucose kinetics during a basal insulin euglycemic pancreatic

clamp. During the clamp, intraduodenal infusion of Intralipid (a lipid emulsion) rapidly

and potently suppressed hepatic glucose production, but did not alter glucose

uptake138. This effect on glucose production was mediated by a gut-brain-liver neuronal

axis, involving vagal afferents, NMDA receptor transmission in the NTS, and the

hepatic vagal efferent138. At the level of the gut, the effect of Intralipid to lower glucose

production required esterification of long-chain fatty acids (LCFA) to LCFA-CoA by

acyl-CoA synthetase (ACS), implicating cellular uptake and subsequent metabolism of

fatty-acids, rather than activation of cell-surface receptors, in the stimulation of gut

peptide release. Co-infusion of a CCK-1 receptor antagonist or genetic knockout of the

CCK-1 receptor reversed the effects of intraduodenal lipids to suppress glucose

production, while intraduodenal infusion of CCK-8 per se suppressed glucose

production137, indicating that a release of CCK in response to intestinal LCFA-CoA

mediates this gut-brain-liver axis. Blocking duodenal PKC-δ negated the ability of

intraduodenal lipids to lower glucose production139, indicating that LCFA-CoA

stimulates the release of CCK via activation of PKC-δ, a signaling pathway that has

been confirmed in vitro140,141. Of note, intraduodenal infusion of a PKC-δ inhibitor

resulted in a dysregulation of glucose response during an un-clamped feeding

paradigm, proving the physiological relevance of this duodenal lipid-sensing pathway.

The CCK-1 receptor, a GPCR, activates both phospholipase C (PLC)- and protein

kinase A (PKA)-dependent pathways, both of which have been shown to be necessary

Page 26: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

18

for activation of vagal afferent neurons by CCK142. During the clamp, activation of PKA

was required for intraduodenal lipids or CCK-8 to stimulate vagal afferent firing and

suppress hepatic glucose production143. Thus, a LCFAàLCFA-CoAàPKC-δ

àCCKàCCK1RàPKA signaling axis within the upper small intestine activates a gut-

brain-liver neuronal axis to suppress hepatic glucose production and maintain glucose

homeostasis in healthy rodents in vivo144. This confirms that in parallel to a gut-brain

axis to regulate food intake, pre-absorptive lipids within the intestine activate a neuronal

gut-brain-liver axis to regulate glucose homeostasis (Figure 1). Whether a similar

negative feedback axis to regulate glucose homeostasis is initiated by upper intestinal

carbohydrates or protein remains to be investigated.

1.3.3 Dysregulation in obesity and diabetes

Obesity, subsequent dysregulation of glucose homeostasis and the onset of diabetes

develop from a combination of genetic and environmental factors that disrupt that

homeostatic control of food intake and glucose homeostasis. One such environmental

factor is the overconsumption of a highly palatable “western diet” that is high in sugar

and fat. Evidence suggests that the consumption of a high-fat diet alters intestinal

nutrient-sensing pathways that, as described above, are important factors in the

regulation of energy homeostasis, and that this may in fact be an important contributor

to the development of obesity and diabetes. Understanding the precise mechanisms of

this disruption to nutrient sensing has the potential to unveil novel therapeutic targets to

restore homeostasis in disease.

Page 27: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

19

Following 21 days of high-fat diet feeding in rats, the reduction of food intake in

response to intraduodenal fatty acid infusion is attenuated compared to rats maintained

on a low-fat diet145. Likewise, humans maintained on a high-fat diet for just 14 days are

no longer sensitive to the satiating properties of intraduodenal lipid infusion146. In

another study, 14 day high-fat diet-fed humans consuming an ad libitum meal reported

lower feelings of “fullness” and ate more than when fed a balanced diet for 14 days

beforehand147. Thus, it is evident that high-fat feeding leads to a disruption in intestinal

nutrient sensing mechanisms, at least in the regulation of food intake, but it is unclear

which precise mechanism(s) in the feedback axis might be disrupted.

There is evidence that high-fat feeding alters the expression of the GPCR’s and

transporters through which gut epithelial cells sense luminal nutrients. EECs isolated

from the small intestines of high-fat diet-fed mice have lower expression of the nutrient-

sensing transporters SGLT-1 and PepT1 as well as the fatty-acid receptor GPR120

compared to those of healthy mice148. Likewise, overweight and obese humans have

lower duodenal expression of the fatty-acid receptor GPR11954. However, in the same

study, the authors identified higher expression of duodenal cluster of differentiation 36

(CD36; a lipid transporter implicated in gut peptide release) and GPR12054,149. This is

consistent with another study in rodents, where rats fed a high-fat diet for 10 weeks

displayed higher expression of the fatty-acid receptors GPR40, GPR41 and

GPR120150. Increased fatty-acid receptor expression suggests that the disruption in

Page 28: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

20

nutrient sensing may occur downstream of lipid receptor activation, leading to a

compensatory increase in receptor expression.

Indeed, the same obese individuals who showed increased duodenal CD36 and

GPR120 displayed decreased numbers of CCK- and GLP-1-expressing cells54.

Similarly, rats fed a high-fat diet for 10-weeks displayed lower intestinal CCK and GLP-

1 protein expression150. This suggests that high-fat feeding can lead to a decrease in

CCK and GLP-1 production, which may at least in part contribute to the disruption in

the ability of nutrient sensing to suppress food intake described above. However, of

note, the impaired satiation by oral or intraduodenal nutrients in 14-day high-fat diet-fed

humans was not in fact associated with lower release of CCK or GLP-1146,147. This

suggests that in high-fat feeding, even if nutrients are able to stimulate the release of

sufficient CCK or GLP-1, the ability of gut hormones to initiate the gut-brain feedback

axis may be impaired.

Indeed, rats fed a high-fat diet for 3 weeks do not suppress food intake in response to

i.p. CCK-8 injection at a dose that significantly reduces food intake in low-fat diet-fed

rats151. Likewise, we found that in rats fed a 3-day high-fat diet, neither intraduodenal

Intralipid nor CCK-8 infusion suppresses hepatic glucose production during the

pancreatic clamp137,138. The inability of CCK to initiative negative feedback on food

intake or glucose production implicates a potential role for resistance at the level of

vagal afferent gut peptide receptors. Indeed, 3-week high-fat diet-fed rats display

Page 29: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

21

reduced c-fos activation of neurons in both the nodose ganglion (vagal afferents) and

the NTS in response to i.p. CCK injection or intraduodenal lipid infusion152-154,

implicating reduced neuronal sensitivity to CCK. We found that the resistance for CCK

to suppress hepatic glucose production occurred at the level of the activation of PKA

and subsequently vagal afferent firing by intraduodenal CCK143. As such, bypassing

CCK receptor activation and infusing a PKA activator directly into the duodenum

restored vagal afferent firing and suppression of hepatic glucose production143. Thus,

through unclear mechanisms, high-fat feeding disrupts the ability of CCK activation of

the CCK-1 receptor to stimulate PKA signaling within the gut (Figure 1). More work is

required to understand the precise mechanisms through which high-fat feeding disrupts

the gut-brain axis to regulate various physiological functions, and the potential to

restore intestinal nutrient signaling pathways to treat obesity and diabetes.

Evidently, intestinal nutrient sensing plays an important role in the acute regulation of

both food intake and blood glucose during feeding, and these regulatory mechanisms

are critical for the body to achieve energy homeostasis. Further, disruption of gut

nutrient sensing during high-fat feeding likely contributes to the development of obesity

and diabetes. Thus, it is crucial to further dissect the mechanisms through which

individual nutrients stimulate this gut-brain axis, and how they may contribute to the

pathology of metabolic diseases. To date, the most is known about intestinal lipid-

sensing mechanisms in the control of glucose homeostasis. Interestingly, however,

numerous studies have shown that intestinal protein is more potent than equicaloric

amounts of lipid or carbohydrate to stimulate gut peptide release155 and suppress

Page 30: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

22

feeding91,156. In light of this, it is vital to further understand the role of dietary protein in

the gut-brain axis.

1.4 Intestinal protein sensing

High-protein diets increase circulating levels of both CCK and GLP-1157,158, and indeed,

luminal protein is a potent stimulus of the gut hormones CCK, GLP-1 and PYY158-161.

Intraduodenal infusion of protein hydrolysates or individual amino acids potently

suppresses feeding in rodents and humans91,92,161-163, the suppression of food intake by

intestinal protein requires CCK-dependent vagal afferent firing164, and high-protein diet

feeding is associated with activation of anorexigenic, CCK-responsive NTS neurons165.

However, the reduction of food intake by high-protein feeding is also associated with a

rise in GLP-1, and the potential role of GLP-1 is less clear158,166.

At the level of the intestinal epithelium, various sensory mechanisms have been

proposed to link luminal amino acids and proteins to the release of gut peptides. Gut

mucosal cells express a number of GPCRs sensitive to amino acids, oligopeptides, or

both, as well as transporters for either free amino acids or oligopeptides. While many of

these protein sensors have been linked to gut peptide release in vitro, the relative

importance of each sensor in the physiological regulation of the gut-brain axis remains

unclear.

Page 31: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

23

T1R1-T1R3, the α-gustucin-coupled “umami” receptor that senses amino acids in taste

buds of the tongue, is also expressed throughout the small intestine, specifically by

CCK-expressing EECs, and in vitro, blocking T1R1 blunts CCK secretion in response

to a number of aliphatic amino acids167,168. Individual amino acids are taken up by an

array of transporters with varying specificities, many of which remain to be identified.

However, one such transporter, B0AT1, has been cloned in both rodents and humans

and found to transport the bulk of neutral amino acids169. B0AT1 knockout mice have

decreased intestinal amino acid signaling and insulin release in response to feeding170,

and intestinal B0AT1 expression is altered in obesity171, suggesting that this transporter

could play a role in nutrient sensing, though more work is required to fully elucidate its

role. GPR93 is a GPCR expressed throughout the gut mucosal epithelium, and

specifically by CCK-expressing cells, which senses both individual amino acids and

small peptides and has been implicated in CCK release in vitro172,173. Likewise, CaSR

is a GPCR that was originally identified as a receptor for calcium, but that also senses

amino acids and small peptides174. CaSR is expressed by primary duodenal I-cells

isolated from mice, and mediates aromatic amino acid-induced CCK release56. CaSR

has also been shown to mediate peptide-induced CCK and GLP-1 release in vitro175-

177. While each of these protein sensors has been implicated in cell lines, future studies

are required to address their physiological role in gut peptide release in vivo.

A number of the aforementioned receptors sense both amino acids and oligopeptides,

and it remains unclear whether luminal proteins usually signal in the form of free amino

Page 32: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

24

acids, small peptides, or both, to stimulate gut peptide release during digestion. An

intraluminal free amino acid solution stimulates vagal afferent firing in rodents178 and

induces gut peptide release in humans179, suggesting that individual amino acids can

induce gut-brain signaling. However, intraduodenal protein hydrolysate infusion also

stimulates CCK-dependent vagal afferent firing180,181, and one study showed that co-

infusion of the proteolytic enzyme trypsin decreased the CCK response to protein

hydrolysates by 60%182, suggesting that proteins signal at least partially in the form of

oligopeptides. Further, one in vitro study showed that removing the lower molecular

weight fractions from protein hydrolysates, which includes free amino acids, has no

effect on its ability to induce CCK release from STC-1 cells, and that a free amino acid

solution does not in fact induce CCK release from these cells176. Along with the fact

that non-metabolizable peptidomimetics (synthetic peptides) activate EECs and

stimulate CCK release183-185, these results suggest that luminal protein may in fact

signal mainly in the form of di- and tripeptides, prior to further lysis into free amino

acids, to initiate a gut-brain negative feedback axis.

Unlike amino acid transport, the uptake of di- and tripeptides in the small intestine is

solely mediated by one molecule, PepT1. In addition to mediating enterocyte peptide

absorption, PepT1 has been localized on CCK-secreting cells and is able to elicit

voltage-gated calcium entry in response to the uptake of peptides177. Indeed, non-

metabolizable PepT1 substrates stimulate CCK release from STC-1 cells in vitro58.

Further, PepT1 inhibition blocks firing of CCK-responsive vagal afferents in response to

Page 33: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

25

intraduodenal protein hydrolysate infusion ex vivo181, further highlighting its likely role in

the initiation of gut-brain signaling in response to luminal protein.

Page 34: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

26

Figure 1: Intestinal nutrient sensing in the regulation of metabolic homeostasis. Luminal nutrients signal via cell-surface receptors and transporters to stimulate the

release of gut peptide hormones from enteroendocrine cells in the gut mucosa, via

receptor-induced membrane depolarization, second-messenger signaling cascades or

intracellular nutrient metabolism. Once released, gut peptides act on receptors

expressed locally by vagal afferent fibers innervating the lamina propria to induce vagal

afferent firing. Vagal afferents terminate in the NTS of the hindbrain, where neuronal

signals are relayed via the hypothalamus and/or vagal efferents to suppress food intake

and hepatic glucose production to maintain energy homeostasis. High-fat diet feeding

disrupts this nutrient-signaling axis, most likely at the level of gut peptide receptor

signaling on vagal afferents. NTS, nucleus of the solitary tract; HYP, hypothalamus.

Page 35: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

26

Chapter 2

Hypothesis and Aims

Upper small intestinal nutrient sensing mechanisms are implicated in the regulation of

food intake and whole-body glucose homeostasis, and are disrupted in high-fat diet

feeding, potentially contributing to the development of obesity and diabetes. More

specifically, upper intestinal lipids stimulate the release of CCK, which acts on CCK-1

receptors expressed by vagal afferent neurons to signal via a gut-brain axis to

suppress feeding and a gut-brain-liver axis to suppress hepatic glucose production.

Alternatively, little is known about the role of intestinal protein sensing mechanisms to

regulate metabolic homeostasis, despite the fact that acute dietary protein intake

improves glucose homeostasis in both rodents and humans. Given that upper intestinal

protein suppresses food intake via CCK-mediated vagal afferent firing, we propose that

while in the pre-absorptive state, dietary proteins trigger a neuronal network to regulate

whole-body glucose homeostasis.

General Hypothesis: Upper small intestinal protein sensing triggers a neuronal

network to lower hepatic glucose production and maintain glucose homeostasis in

healthy rodents in vivo (Figure 2).

Page 36: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

27

Aims:

Aim 1: To evaluate whether upper small intestinal protein sensing regulates glucose

homeostasis.

Aim 2: To investigate whether upper small intestinal protein-sensing mechanisms

regulate glucose homeostasis through direct effects on glucose uptake or glucose

production.

Aim 3: To evaluate whether a neuronal network is required for upper small intestinal

protein sensing to regulate glucose homeostasis.

Page 37: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

28

Figure 2: Schematic of the working hypothesis. Luminal protein is sensed by the

gastrointestinal tract prior to absorption into the portal circulation, signalling to activate

a neuronal network within the gut wall. This signal is then relayed via a gut-brain-liver

neuronal axis to directly suppress hepatic glucose production, improving whole-body

glucose tolerance. NTS, Nucleus of the solitary tract; NMDA; N-methyl D-aspartate;

DVC, dorsal vagal complex.

Page 38: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

29

29

Chapter 3

Materials and Methods «

3.1 Animals

For all studies, male Sprague-Dawley rats were obtained from Charles River

Laboratories (Montreal, QC, Canada) at 8-weeks of age (250-270g). Rats were

individually housed and maintained for six days with a standard 12-hour light-dark cycle

and ad libitum access to water and rat chow (Harlan Teklad; % calories: 16% fat/60%

carbohydrate/24% protein; total caloric value: 3.1 kcal/g) in order to acclimatize to our

facilities prior to surgical manipulation. All animal protocols were reviewed and

approved by the Institutional Animal Care and Use Committee at the University Health

Network.

3.2 Surgical Procedure

3.2.1 Preparation of cannulae

Duodenal cannula: 18 cm of 0.04 inner-diameter/0.085 outer-diameter silicon tubing

(Sil-Tec) was overlapped with 1 cm of smaller, 0.025 inner diameter/0.037 outer-

diameter silicon tubing (Sil-Tec). A 1-cm2 piece of polypropylene mesh (Bard Davol)

Page 39: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

30

30

was glued to the cannula at the intersection of the two tubing sizes, such that the small

tubing passed through a hole in the center of the mesh square. Vascular cannulae: 15

cm of PE50 polyethylene tubing (Clay Adams) was fitted with a 1.5 cm cuff extension of

15 mm inner-diameter Silastic silicon tubing (Dow Corning).

3.2.2 Cannulation surgery

Rats were anesthetized with an intraperitoneal injection of a cocktail of ketamine (60

mg/kg; Vetalar, Bioniche) and xylazine (8 mg/kg; Rompun, Bayer). The abdominal and

neck regions were shaved and sanitized with 70% ethanol and 10% providine-iodine

solution (Betadine). A 4 cm laparotomic incision was made along the ventral midline

through both the skin and abdominal muscle wall, exposing the GI tract within the

peritoneal cavity. The duodenum was isolated 6 cm distal to the pyloric sphincter. A

small hole was made in the intestinal wall using a 21-guage needle in an area with the

least vascularization to minimize bleeding, and the intestinal cannula was inserted into

this hole and anchored to the outer surface of the duodenum by applying tissue

adhesive (3M Vetbond) to the surrounding mesh. The duodenal cannula was flushed

with saline to ensure that the infusion flowed into the lumen of the duodenum with no

leakage. The duodenum was then repositioned within the peritoneal cavity and the

abdominal wall was closed with 4-0 silk sutures such that the cannula exited through

the abdominal wall between 2 continuous sutures. A 1.5 cm incision was made in the

back of the neck and the cannula was tunneled subcutaneously from the abdomen,

exiting the incision in the neck. The neck and abdominal incisions were closed with 4-0

silk sutures and the end of the cannula, which now exited through the back of the neck,

was closed with a metal pin.

Page 40: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

31

A 3 cm incision was then made in the front of the neck, and through blunt dissection of

fat, connective tissue and muscle, the right jugular vein was isolated. The vessel was

ligated at the cranial end and loosely tied at the caudal end using 4-0 sutures, and the

ligatures were pulled taught to prevent blood flow in the isolated segment of the vessel.

A small incision was made in the vessel wall using microscissors, and a vascular

cannula filled with 1% heparinized saline was inserted into the vessel lumen until it

reached a position where blood freely flowed into the cannula when an attached

sampling syringe was pulled back. The cannula was then secured in place by

tightening the loosely tied ligature around the vessel and cannula. This procedure was

repeated with the right carotid artery. The two cannulae were then tunneled

subcutaneously with a 16-guage needle so that they exited the back of the neck on

either side of the intestinal cannula. The neck incision was closed with 4-0 sutures, and

the vascular cannulae were filled with 10% heparin solution to prevent clotting and

closed with metal pins. 10 mL of 0.9% saline was injected subcutaneously to help with

recovery and rats were returned to their cages.

Rats were given 4 days to recover from surgery with ad libitum access to water and

regular chow. The intestinal cannula was flushed with saline daily to prevent clogging.

Food intake and body weight were monitored daily and rats recovered to at least 90%

of their pre-surgical body weight prior to experiments.

Page 41: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

32

3.3 Intestinal infusions and treatments

The following treatments were infused into the upper small intestine via the indwelling

cannula during the in vivo experiments, at a rate of 0.12 ml/min for 1 minute to fill the

dead space of the cannula followed by 0.01 ml/min to a total of 50 minutes: 1) 0.9%

saline, 2) 8% (w/v) casein hydrolysate 3) tetracaine (0.01 mg/mL) and 4) 8% (w/v)

casein hydrolysate + tetracaine (0.01 mg/mL). All of the treatments were adjusted to pH

5, which is the native pH of the rat upper small intestine186.

Casein was selected for its broad amino acid composition and because it is a common

dietary protein source. The hydrolyzed form of casein protein is composed of small

peptides and free amino acids and most closely resembles the partially digested form

in which dietary proteins reach the upper small intestine during digestion. The typical

amino acid analysis of casein hydrolysate (Sigma-Aldrich) can be seen in Table 1. The

dose of 8% (w/v) casein hydrolysate (0.32 kcal/mL) was selected because

intraduodenal 8% peptone stimulated vagal firing and suppressed food intake in

rodents in vivo181,187. 3.2 g of casein hydrolysate (Sigma-Aldrich) was solubilized in 40

mL of saline by lowering the pH to 1 through titration with 10 M HCl and subsequently

raising the pH to 5 through titration with 10 M NaOH.

The tetracaine dose was based on a previous study where 0.01 mg/min tetracaine

blocked intraduodenal lipid infusion to regulate glucose homeostasis138. Tetracaine

Page 42: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

33

(Sigma-Aldrich) was dissolved in DMSO in a stock solution of 200 mg/mL, and 22.5 µL

of this stock solution was added to 3 mL of either saline or 8% casein hydrolysate.

3.4 Intravenous glucose tolerance test

Food was removed at 5 pm the night before the glucose tolerance test for an overnight

fast. Experiments took place at 10:00 AM. The experimental protocol can be seen in

Figure 3A. The intraintestinal infusion was initiated at t=-15 at a rate of 0.12 mL/min for

1 minute to fill the dead space of the intestinal cannula, followed thereafter by a rate of

0.01 mL/min, administered using a Harvard Apparatus PHD 2000 syringe pump. At t=0,

the glucose injection was administered (0.25g/kg, 20% glucose solution), over 10

seconds, via the jugular vein cannula. 0.4 mL blood samples were drawn from the

carotid artery cannula at t=-15, t=0 (prior to the glucose injection), t=2, t=5, and every 5

minutes thereafter until t=35, for plasma glucose and amino acid analysis (see below).

The experiment was terminated at t=35, or the end of 50 minutes of gut infusion. At the

end of the 50-minute experiment, rats were anesthetized via intravenous administration

of ketamine, and a 0.5 mL portal vein blood sample was drawn for plasma amino acid

analysis.

Page 43: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

34

3.5 Pancreatic (basal insulin) euglycemic clamp

Rats were restricted to 15 g (~47 kcal) of food the night prior to the clamp experiment,

which took place at 9:00 AM. The clamp protocol can be seen in Figure 4A. An

intravenous infusion of [3-3H]-glucose (40 µCi bolus; 0.4 µCi/min; Perkin Elmer) was

initiated at t=0 and continued for the duration of the 200-minute experiment. From t=60

to t=90, once the [3-3H]-glucose has reached steady-state in the rat, 0.2 mL blood

samples were drawn every 10 minutes from the carotid artery for plasma glucose

analysis (see below) in order to obtain basal plasma glucose readings. At t=90, the

pancreatic clamp was initiated through continuous intravenous infusion of somatostatin

(3 µg/kg/min; Bachem) and insulin (1.2 mU/kg/min; porcine insulin; Sigma-Aldrich).

Somatostatin was infused to inhibit all endogenous insulin secretion while the

exogenous insulin infusion replaced insulin at basal levels. An intravenous infusion of

exogenous glucose (25% solution) was also initiated at t=90. Plasma glucose readings

were obtained every 10 minutes from t=90-200, and the exogenous glucose infusion

rate was adjusted as required to maintain euglycemia according to the basal glucose

readings. During the final 50 minutes of the experiment (t=150-200), the upper small

intestinal infusions were administered as previously described above. Additional 50 µL

plasma samples were obtained at all of the 10-minute time points for later plasma

specific activity analysis. At the end of the experiment, at time t=200, rats were

anesthetized via intravenous administration of ketamine, and a 0.5 mL portal vein blood

sample was drawn for plasma amino acid analysis. All infusions were administered

using a Harvard Apparatus PHD 2000 syringe pump.

Page 44: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

35

3.6 Biochemical analysis

3.6.1 Plasma glucose

During in vivo experiments, blood samples were collected into heparinized Eppendorf

tubes and centrifuged (6000 rpm, 1 min) to separate the plasma. 10 µL of plasma was

then analyzed using a GM9 Glucose Analyzer (Analox Instruments), which utilizes the

glucose oxidase method, in order to obtain plasma glucose readings in situ.

3.6.2 Plasma amino acids

A protocol to measure plasma amino acid concentrations using the colorimetric

ninhydrin reaction was adapted from 188. Briefly, the assay is based on the principle that

ninhydrin reacts with equal affinity to the α-amino nitrogen groups of all amino acids,

with the exception of profile, producing a purple colour. This colour change can be

measured using a spectrophotometer and compared to known concentrations of α-

amino acids, resulting in a measure of total α-amino acids in the sample.

The specific protocol was as follows: The plasma samples were first deproteinized such

that the α-amine groups in the samples would be indicative of only free α-amino acids.

50 µL of plasma in 350 µL of ddH2O was deproteinized through the addition of 50 µL of

Page 45: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

36

10% (w/v) sodium tungstate (Sigma-Aldrich) and 50 µL of 2N HCl, with successive

mixing. After standing for 5 minutes, samples were centrifuged (13000 rpm, 5 min,

4°C). 250 µL of supernatant was then combined with 125 µL of cyanide-acetate buffer

(pH 5.2) containing: 114.65 g sodium acetate (Sigma-Aldrich), 500 mL ddH2O, 36 mL

glacial acetic acid, 5g disodium EDTA (BioShop) and 20 mL cyanide solution (490 mg

sodium cyanide [Sigma-Aldrich] in 1 L ddH2O). 125 µL of ninhydrin reagent (3% (w/v)

ninhydrin [Sigma-Aldrich] in 2-methoxyethanol [Sigma-Aldrich]) was added. Tubes were

mixed and capped, and then heated in a boiling water bath for 15 minutes. Immediately

upon removal from the water bath, 3.75 mL cold diluent (50% isopropanol in ddH2O)

was added. Samples were mixed thoroughly by inversion, and transferred to 1 cm

cuvettes. In addition to the plasma samples, a “blank” sample was prepared as

described above but with 50 µL ddH2O replacing the 250 µL of deproteinized plasma,

and a “standard” sample was prepared using 50 µL of stock amino acid solution (26.8

mg glycine [BioShop] in 100 mL ddH2O) replacing the deproteinized plasma. All

cuvettes were read in a spectrophotometer (Genesys 10S VIS, Thermo Scientific) at

570 nm. Given that the standard sample contains a known concentration of 5 mg α-

nitrogen/100 mL, the following formula was then used to calculate the concentration of

α-nitrogen in the samples based on their absorbencies (E):

Plasma α-N (mg/100 mL) = (E test) – (E blank) X 5 (E standard) – (E blank)

Page 46: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

37

3.6.3 Plasma [3-3H]-glucose specific activity

50 µL plasma samples were deproteinized through the addition of 100 µL Ba(OH)2 and

100 µL ZnSO4, vortexing, and centrifugation (13200 rpm, 5 min, 4°C). The supernatant

was then transferred to scintillation vials without lids and left for 48 hours to evaporate

under a fumehood. Evaporation rids the samples of water containing [3H] as a product

of glycolysis, such that only [3-3H]-glucose will contribute to the radioactivity in the

sample. 7.5 mL of Bio-Safe Scintillation Cocktail (Research Products International

Corp) was then added to the dry samples and the vials were counted in a LS6500

Multipurpose Scintillation Counter (Beckman Coulter).

3.7 Tracer-dilution calculations

The constant, radioactive [3-3H]-glucose tracer infusion reaches steady state in the

plasma and tissues of the rat within one hour. At this point, a steady state formula can

be applied to calculate glucose uptake and glucose production, whereby the rate of

glucose disappearance (Rd) is equal to the rate of endogenous glucose appearance

(Ra). The specific activity of the plasma samples collected during the basal period of

the clamp (t=60-90) can therefore be used to calculate glucose uptake and glucose

production at each time point as follows:

Page 47: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

38

Ra = Rd = Tracer infusion rate (µCi/min) = Rates of Glucose production Specific activity (µCi/mg) and uptake (mg/kg/min)

During the clamp, when the exogenous glucose infusion is introduced, the rate of

glucose production must be calculated by subtracting the exogenous glucose infusion

rate (GIR), as follows:

Ra + GIR = Rd Ra = Rd – GIR = Rate of glucose production (mg/kg/min)

3.8 Statistical analysis

For the intravenous glucose tolerance test, the glucose concentration over time was

compared using a two-way anaylsis of variance (ANOVA) and was followed by a

Tukey’s post-hoc test when comparing more than two groups. The area under the

curve (AUC) was calculated with Prism software (GraphPad) as compared using an

unpaired Student’s t-test when comparing two groups or a one-way ANOVA when

comparing more than two groups. For the pancreatic clamp/tracer-dilution data

analysis, an average from t=60-90 was used as the “basal” values and an average from

t=180-200 was used as the “clamp” values. Comparisons between basal versus clamp

Page 48: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

39

glucose uptake and glucose production were made using an unpaired Student’s t-test.

For plasma amino acid concentrations, the concentration at t=50 was compared

between saline versus casein hydrolysate treated rats using an unpaired Student’s t-

test. Prism software (GraphPad Software Inc.) was used for all statistical calculations.

A probability of P < 0.05 was accepted as significant. Values were presented as mean

± SEM.

Page 49: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

40

Chapter 4

Results «

Upper small intestinal protein sensing improves glucose tolerance

To investigate the effects of upper small intestinal protein sensing mechanisms on

whole-body glucose homeostasis, we first developed a protocol incorporating a

simultaneous upper intestinal protein infusion and intravenous glucose tolerance test

(IVGTT) in healthy rodents in vivo. Sprague-Dawley rats were chosen as the rodent

model to allow for future comparisons between healthy and high-fat diet-fed rodents.

Upon high-fat feeding, Sprague-Dawley rats are susceptible to the development of

hyperphagia, insulin resistance and obesity, with less variability than other rat

strains189. Accordingly, only male rats were chosen as male Sprague-Dawley rats are

more susceptible than females to high-fat diet-induced hyperphagia189, and their

metabolism is not affected by variable sexual hormone cycles. All male Sprague

Dawley rats were obtained from Charles River Laboratories at 8 weeks of age, and

given exactly six days to acclimatize to our facilities prior to surgical manipulation. It is

now well understood that environmental history influences rodents’ metabolism and

their response to dietary challenges and should therefore be controlled190. On the

seventh day in our facilities, rats underwent jugular, carotid and small intestinal

cannulation surgeries. After 4 days

Page 50: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

41

of recovery time during which they were fed an ad libitum regular chow diet, rats were

subjected to an overnight fast (food removed at 5 pm on day 4) followed by the gut-

infusion IVGTT experiment on the morning of day 5 (Figure 3A).

The IVGTT was selected to evaluate whole-body glucose homeostasis during an upper

small intestinal protein infusion, while circulating glucoregulatory hormones change at

will. The OGTT, while commonly utilized, is confounded by the rates of intestinal

glucose absorption and gastric emptying, thus producing more variable data than an

IVGTT191, which evaluates the effects of a treatment specifically on the tolerance of

circulating glucose, controlled only by glucose uptake and glucose production.

Additionally, the IVGTT provides the possibility of future mathematical modeling of our

results using the “minimal model” system developed by Bergman and colleagues to

evaluate the relative roles of insulin-dependent and insulin–independent effects7.

Finally, and most importantly, intravenous administration of glucose bypasses the

upper small intestine and does not interfere with our intestinal treatments. Rats were

fully awake throughout the IVGTT experiments, as general anesthetics can have both

direct and indirect effects on metabolic studies192. The IVGTT protocol is summarized

in Figure 3A: Briefly, the intravenous glucose injection was administered after 15

minutes of a 50-minute intestinal infusion of the given treatment, and blood samples

taken frequently to assess plasma glucose levels. The upper intestinal treatments were

administered directly into the lumen via the small intestinal cannula, first at a rate of

0.12 mL/min for one minute to fill the dead space of the cannula with the treatment and

then at a rate of 0.01 mL/min for a total of 50 minutes. Infusion of 8% casein

Page 51: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

42

hydrolysate for 50 minutes at this rate did not increase portal or systemic free amino

acids (Figure 3B). This is consistent with previous findings that 20% intralipid

administered into the duodenum at the same rate does not increase portal or systemic

free fatty acids138. Thus, we successfully developed an in vivo model to evaluate the

effects of pre-absorptive protein-signaling mechanisms in the upper small intestine on

the regulation of whole-body glucose homeostasis.

As assessed by the IVGTT, upper small intestinal infusion of casein hydrolysate

significantly improved glucose tolerance compared to the control group receiving

intraintestinal saline, where plasma glucose levels were significantly lower from times

10-25 (Figure 4A). Further, the integrated area under the curve (AUC) of the glucose

collapsed over time was significantly reduced in protein-treated rats compared to saline

(Figure 4B). These results were independent of any differences in body weight or

cumulative post-surgical food intake (Table 2). Given that the protein treatment did not

raise circulating amino acids, our data demonstrate that upper small intestinal protein

sensing regulates whole body glucose homeostasis to improve glucose tolerance.

Page 52: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

43

Upper small intestinal protein sensing suppresses glucose production

An improvement in glucose tolerance during an IVGTT can be accounted for entirely by

an increase in peripheral glucose uptake and/or a suppression of endogenous glucose

production. Furthermore, the effects of intestinal nutrient sensing on either glucose

uptake or glucose production may be either dependent on, or independent of, an

incretin effect to increase insulin secretion. Thus, our lab has developed a protocol

utilizing a pancreatic basal-insulin euglycemic clamp technique in conjunction with

tracer-dilution methodology to assess the direct effects of intestinal nutrient infusions

on whole-body glucose kinetics (glucose uptake and glucose production)138 (Figure

5A). To evaluate the mechanisms through which intestinal protein improves glucose

tolerance, we administered the 50-minute upper intestinal casein hydrolysate treatment

in a clamp setting, in a new group of rats that underwent the same handling, surgery

and recovery protocol as those receiving the IVGTT. The clamp experiment took place

the morning of day 5 post-surgery, and rats were restricted to 15 g (~47 kcal) of food

the night before to ensure a similar post-absorptive status. The 200-minute clamp

protocol is summarized in Figure 5A: Briefly, the [3-3H]-glucose is infused and allowed

to reach steady state, somatostatin is infused to inhibit endogenous insulin secretion

and insulin is replaced to basal levels. In the final 50 minutes of the clamp, the 50-

minute gut infusion is administered at the same rate as during the IVGTT, and an

exogenous glucose infusion is administered as required to maintain euglycemia. At

basal circulating free amino acid and insulin levels (Figure 3B), upper small intestinal

infusion of casein hydrolysate increased the exogenous glucose infusion rate (GIR)

Page 53: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

44

required to maintained euglycemia 5-fold compared to a control saline infusion (Figure

5B). Analysis of the tracer-dilution data revealed that this increase in GIR was entirely

accounted for by a 48% suppression of hepatic glucose production (Figure 6A,B), as

glucose uptake was unaltered (Figure 6C). There were no differences in body weight or

food intake between groups (Table 2). Thus, upper small intestinal protein sensing

regulates glucose homeostasis through directly suppressing hepatic glucose

production, independent of changes in circulating insulin.

Upper small intestinal protein sensing requires local, neuronal signaling to

regulate glucose homeostasis

Given that upper intestinal lipids suppress glucose production via a gut-brain-liver

neuronal axis138, we next evaluated the role of a neuronal axis to mediate the effects of

protein sensing on whole-body glucose homeostasis. Gut-peptides released in

response to luminal nutrients act on locally expressed receptors to stimulate vagal

afferent neuron firing9, and co-infusion of tetracaine, a topical anesthetic, with

intraintestinal nutrients negates the ability of nutrient sensing to suppress food intake

and lower glucose production by blocking afferent neuronal signaling to the brain138,193.

We therefore co-infused tetracaine with upper small intestinal casein hydrolysate during

the IVGTT in a new group of rats, to assess whether a gut-brain neuronal axis is

required for intestinal protein sensing to improve glucose tolerance. Infusion of

tetracaine alone (1 mg/mL, 0.01 mg/min) had no effect on glucose tolerance compared

Page 54: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

45

to saline, while co-infusion of tetracaine with 8% casein hydrolysate reversed the

improvement in glucose tolerance by casein hydrolysate (Figure 7A). The integrated

AUC was reversed to that of the saline-treated rats (Figure 7B). Therefore, intestinal

protein activates local, neuronal signaling in order to regulate glucose homeostasis and

improve glucose tolerance, further supporting that the signaling mechanism of protein

sensing is localized within the gut.

Page 55: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

46

A B

Figure 3: Gut infusion-IVGTT protocol to evaluate the effects of upper small intestinal protein sensing on glucose tolerance. A, Experimental protocol for the

intravenous glucose tolerance test with upper intestinal infusions. IVGTT, intravenous

glucose tolerance test; SI, small intestine. B, Systemic and portal vein plasma free

amino acid levels in response to the 50-minute upper intestinal saline (n=10) or casein

hydrolysate (n=10) infusion. Values expressed as mean ± SEM. No differences among

groups as determined by unpaired Student’s t-test.

Page 56: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

47

A B

Figure 4: Upper small intestinal infusion of protein improves glucose tolerance. A, Plasma glucose concentrations over time during the intravenous glucose tolerance

test in rats receiving an upper small intestinal infusion of saline (n=10) or casein

hydrolysate (n=10). B, The integrated area under the curve (AUC). Values expressed

as mean ± SEM. * p < 0.05, ** p < 0.005 compared to saline as determined by two-way

ANOVA.

Page 57: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

48

A B

Figure 5. Upper small intestinal casein hydrolysate increases the glucose infusion rate during the basal insulin euglycemic clamp. A, Experimental protocol

for the basal insulin euglycemic clamp. B, The glucose infusion rate required to

maintain euglycemia during the clamp with upper small intestinal infusion of saline

(n=7) or casein hydrolysate (n=7). Values expressed as mean ± SEM. * P < 0.05

compared to saline as determined by unpaired Student’s t-test.

Page 58: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

49

A B

C

m

Figure 6. Upper small intestinal casein hydrolysate suppresses hepatic glucose production. A, B, C, Rate of glucose production (A), suppression of glucose

production expressed as the percent decrease from basal (B) and clamp rate of

glucose uptake (C) with upper small intestinal infusion of saline (n=7) or casein

hydrolysate (n=7). Values expressed as mean ± SEM. * P < 0.05 compared to saline as

determined by unpaired Student’s t-test.

Page 59: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

50

A

B

Figure 7: Upper small intestinal protein requires local, neuronal signaling to regulate glucose tolerance. A, Plasma glucose concentrations over time during the

intravenous glucose tolerance test with upper small intestinal infusion of saline (n=10),

casein hydrolysate (n=10), tetracaine (n=8) or casein hydrolysate with tetracaine (n=8).

B, The integrated area under the curve (AUC). Values expressed as mean ± SEM. * P

< 0.05 compared to saline, # P < 0.05 compared to tetracaine, t P < 0.05 compared to

Page 60: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

51

casein hydrolysate + tetracaine, as determined by two-way ANOVA and Tukey post

hoc test.

Table 1: Typical amino acid content of Casein enzymatic hydrolysate from Sigma-Aldrich (Product 22090).

Amino acid Typical concentration (%w/w)

Glutamic acid 16.5

Proline 8.5

Leucine 6.5

Lysine 6.4

Aspartic acid 5.5

Valine 5.3

Isoleucine 4.5

Serine 4.5

Phenylalanine 3.8

Threonine 3.6

Arginine 2.9

Alanine 2.4

Methionine 2.4

Histidine 2.1

Tyrosine 1.8

Glycine 1.6

Tryptophan 0.95

Cystine 0.67

Page 61: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

52

Table 2: Body weight and cumulative post-surgical food intake of the groups of rats that underwent either the intravenous glucose tolerance test or clamp protocol.

  SAL  (IVGTT)  

CAS  (IVGTT)  

TET  (IVGTT)  

CAS  +  TET  

(IVGTT)  

SAL  (clamp)  

CAS  (clamp)  

Body  weight  (g)   279  ±  6.0   280  ±  3.9   274  ±  4.9   263  ±  7.9   295  ±  6.7   298  ±  10.4  

Food  intake  (g)   26.1  ±  3.5   23.3  ±  3.2   21  ±  2.9   18  ±  3   30.0  ±  2.9   29.2  ±  2.6  

Values expressed as mean ± SEM. Saline (SAL). Casein hydrolysate (CAS).

Tetracaine (TET). Intravenous glucose tolerance test (IVGTT). Basal insulin euglycemic

clamp (clamp).

Page 62: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

53

Table 3: Plasma glucose concentrations during the basal insulin euglycemic clamp.

Basal Saline Casein hydrolysate

Glucose (mg/dL) 142.4 ± 6.5 137.8 ± 6.3

Clamp Saline Casein hydrolysate

Glucose (mg/dL) 159.3 ± 5.5 147.2 ± 2.8

Values expressed as mean ± SEM.

Page 63: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

54

Chapter 5

Discussion

To date, the role of dietary protein in the regulation of glucose homeostasis remains

unclear. Although there is evidence that the resulting increase in plasma amino acids

may contribute to gluconeogenesis and induce insulin resistance27,194, many studies

show that high-protein diet feeding in fact lowers fasting glucose and HbA1c levels and

improves glucose tolerance28,29,32-34. The mechanisms of this improvement in glucose

control are unknown, but may involve acute negative feedback by intestinal protein

sensing. Indeed, using a model of healthy, male, Sprague-Dawley rats, we here

demonstrate that upper small intestinal protein sensing regulates whole-body glucose

homeostasis. Specifically, a 50-minute upper small intestinal infusion of casein

hydrolysate improved glucose tolerance as assessed by an IVGTT in comparison to

rats given a control infusion of saline, independent of any differences in food intake or

body weight.

Analysis using the ninhydrin method indicated that this improvement in glucose

tolerance occurred independent of a rise in either portal vein or systemic free amino

acid levels. Circulating amino acids can act on other targets to regulate glucose

homeostasis, such as in the hypothalamus to lower hepatic glucose production via a

brain-liver neuronal network195. In addition, vagal afferents innervating the portal vein

respond to portal amino acid infusion196,197, implying the presence of portal amino acid

Page 64: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

55

sensors. However, the fact that amino acid levels were not raised by the 50-minute

casein hydrolysate infusion suggests that protein acts via gut-localized sensing

mechanisms to regulate whole-body glucose homeostasis during the IVGTT.

It is, however, important to note that the plasma samples were deproteinized, and the

ninhydrin assay measured only free amino acids, not accounting for those that make up

oligopeptides. It can therefore not be ruled out that di- and tripeptides absorbed by the

gut epithlium intact are reaching the portal vein within 50 minutes. During digestion of a

mixed meal, 90% of oligopeptides absorbed by epithelial cells intact are hydrolyzed

intracellularly and reach the portal vein as free amino acids198. However, given that

apical peptide transporters are upregulated by the presence of luminal oligopeptides199,

our intraintestinal casein hydrolysate infusion may lead to increased flux of peptide

absorption, causing release of excess peptides into the circulation, which are missed

by the ninhydrin assay, and which can act on peripheral targets to regulate glucose

homeostasis. Therefore, future studies should employ a control group where the 8%

casein hydrolysate treatment is infused at the same rate directly into the portal vein

(assuming 100% leakage), to rule out post-absorptive effects improving glucose

tolerance. Nevertheless, the present results suggest that dietary proteins act locally

within the upper small intestine to improve whole-body glucose tolerance in healthy

rodents.

Page 65: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

56

These findings are in line with previous studies showing that acute protein intake can

improve glucose tolerance. Consumption of a protein hydrolysate beverage lowers the

glucose response to a subsequent mixed meal43,200, and the addition of protein

hydrolysates to a standard carbohydrate beverage lowers the glucose response42.

Furthermore, consumption of a 5g high-protein diet meal results in a lower glucose

response than a 5g low-protein meal as early as 30 minutes after the initiation of

feeding45. One proposed mechanism of this lowered glucose response is a protein-

induced lowering of gastric emptying rate201, which would then slow the appearance of

glucose into the circulation. However, our current findings indicate that when the

confounding variable of gastric emptying is removed by intravenous injection of the

glucose bolus, intestinal protein signals to improve whole-body glucose tolerance, and

gut protein sensing may therefore contribute to the improvement in glucose response

seen in these acute feeding studies.

The improvement in intravenous glucose tolerance induced by small intestinal protein

sensing must be accounted for by a suppression of endogenous glucose production

and/or increase in peripheral glucose uptake. There are multiple mechanisms through

which gut protein sensing may alter either of these factors. The improvement in glucose

turnover may be secondary to a change in circulating glucoregulatory hormones, given

that protein intake stimulates the release of insulin202, and may do so via pre-absorptive

gut peptide-mediated signaling. GLP-1, which is released in response to luminal

protein, stimulates the release of insulin either via direct action on β-cells or a local

neuronal axis mediated by hepatoportal vagal afferents and pancreatic efferents203, in

Page 66: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

57

order to improve glucose tolerance129. Additionally, protein-induced CCK release may

contribute to this incretin effect given that CCK is implicated in stimulating postprandial

insulin secretion both in vitro and in vivo134,204. In many instances, however, acute

protein intake improves glucose homeostasis independently of a rise in insulin

levels43,45. Indeed, protein-induced gut peptide release could regulate glucose

homeostasis independently of insulin, such as via a GLP-1-mediated gut-brain-muscle

neuronal axis to increase muscle uptake and metabolism130,205. However, the most like

likely explanation may be a suppression of hepatic glucose production rather than an

increase in glucose uptake, given that both fatty acid and glucose sensing within the

small intestine suppresses hepatic glucose production via a gut-brain-liver neuronal

axis, independently of any effects on glucose uptake, during a basal insulin pancreatic

clamp13,138.

To investigate whether upper small intestinal protein sensing regulates either glucose

uptake or glucose production, independently of any changes in insulin, the 50-minute

casein hydrolysate infusion was next given during a basal-insulin euglycemic clamp.

During the clamp, upper small intestinal protein infusion increased the glucose infusion

rate required to maintain euglycemic, and this was completely accounted for by a

suppression of hepatic glucose production, and glucose uptake remained unchanged.

Thus, upper small intestinal protein-sensing mechanisms directly regulate hepatic

glucose production, and this likely contributes to the improvement in whole-body

glucose tolerance in response to intestinal protein during the IVGTT. However, the

contribution of this suppression of hepatic glucose production to the improvement in

Page 67: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

58

glucose tolerance seen during the IVGTT remains unknown. In order to address the

contribution of hepatic glucose metabolism in future experiments, metabolic pathways

regulating glucose production in the liver could be blocked during the IVGTT.

Upper small intestinal lipids initiate a gut-brain-liver neuronal axis, mediated by vagal

afferents, NMDA receptor transmission in the DVC and the hepatic vagal efferent to

suppress hepatic glucose production138. Thus, a similar neuronal axis may mediate

intestinal protein to suppress glucose production. Indeed, co-infusion of the local

anesthetic tetracaine completely reversed the improvement in glucose tolerance by

upper intestinal protein infusion, indicating that pre-absorptive protein initiates a

neuronal network at the level of the gut to exert its effects on glucose homeostasis.

Alternatively, upper small intestinal infusion of tetracaine alone did not affect glucose

homeostasis during the IVGTT, indicating that the contribution of basal vagal afferent

firing to glucoregulatory control is negligible, whereas a postprandial increase in firing is

required for nutrients to regulate glucose homeostasis. These findings are consistent

with a recent study investigating the role of gut neuronal signaling in humans. When

implanted in the stomach, the electrical DIAMOND® device senses incoming food and

responds with electrical stimulation of the gut wall. After 5 weeks, this postprandial

neuronal stimulation resulted in an improvement in glycemic control (lower HbA1c) in

type 2 diabetic individuals206. Thus, the present findings are likely to be conserved in

humans and may be important in the context of disease.

Page 68: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

59

Future studies are required to dissect the involvement of a gut-brain or gut-brain-liver

neuronal axis in the regulation of glucose tolerance and glucose production. The ability

of upper small intestinal lipids to suppress hepatic glucose production is attenuated not

only by co-infusion of tetracaine but also by blocking NMDA receptor transmission

within the NTS of the hindbrain or by surgical ablation of the hepatic vagal efferent,

implicating a gut-brain-liver axis mediated by vagal afferents, NMDA transmission in the

NTS and the hepatic vagus138. Pharmacological inhibition of NTS NMDA receptors, by

NTS infusion of the noncompetitive high-affinity antagonist MK-801, and hepatic

vagotomy surgery should thus be utilized during future studies to investigate whether a

similar gut-brain-liver axis mediates intestinal protein to suppress hepatic glucose

production during the pancreatic clamp.

The precise mechanisms through which upper small intestinal casein hydrolysate

initiates a neuronal network regulating glucose homeostasis remain unknown. Casein

hydrolysate is a partially hydrolyzed form of protein, containing both individual amino

acids and oligopeptides, for both of which there are sensory machineries expressed by

GI epithelial cells that are implicated in gut peptide release. A number of amino acid-

specific GPCRs are expressed by EECs and mediate CCK release in response to

amino acids in vitro56,168,172,173, which vary in their affinity for different classes of amino

acids. Casein hydrolysate is composed of a broad range of amino acids, with the

highest proportion being Glutamic acid followed by the aliphatic amino acids Proline,

Leucine and Lysine. Indeed, the gut epithelium expresses glutamate receptors, which

mediate gut peptide release and vagal afferent firing in response to luminal glutamate

Page 69: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

60

infusion and possibly to high protein meals207, one of which is T1R1-T1R3, the gut-

expressed α–gustducin coupled GPCR and “umami” taste receptor168. The role of

aliphatic amino acids such as Leucine in the gut-brain axis is less clear given that

although intraduodenal infusion of protein hydrolysate suppresses feeding, infusion of

Leucine, Arginine or Alanine independently does not suppress feeding162. Alternatively,

infusion of the aromatic amino acids Phenylalanine or Tryptophan alone suppresses

feeding162. Indeed, Phenylalanine and Tryptophan stimulate CCK release and suppress

feeding via a vagal afferent-dependent neural axis in both rodents and humans161,208,

and Phenylalanine infusion is in fact more potent than casein hydrolysate to suppress

feeding in rodents92. These findings indicate that although Tryptophan and

Phenylalanine are in lower proportion than other amino acids in casein hydrolysate,

they may be the signaling molecules through which this protein solution initiates a gut-

brain axis. Future studies should look at whether infusion of either of aromatic amino

acids alone can equally or more potently regulate glucose tolerance and glucose

production in comparison to the casein treatment. In vitro, aromatic amino acids are

sensed by the GPCR CaSR, which is expressed by EECs, in order to stimulate CCK

release56, thus the role of this transporter in protein-induced glucoregulation should

also be investigated. Alternatively, uptake and metabolism of amino acids by gut

epithelial cells may be required, as in the case of intestinal lipid sensing, where

metabolism to LCFA-CoA is required for lipids to stimulate CCK release and lower

glucose production138. Indeed, hypothalamic leucine infusion lowers plasma glucose via

a suppression of hepatic glucose production, and this is dependent upon metabolism to

the intermediate α-ketoisocaproic acid and subsequently to acetyl-coA and malonyl-

CoA195, which is known to increase cytoplasmic accumulation of LCFA-CoA, increasing

Page 70: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

61

flux into the lipid sensing pathway to suppress glucose production. Given that upper

intestinal accumulation of LCFA-CoA activates a CCK-dependent gut-brain-liver axis to

suppress glucose production138, intestinal amino acids may act through a similar

pathway to regulate glucose homeostasis via metabolism to malonyl-CoA.

However, protein hydrolysates or solutions of small peptides are more efficiently

absorbed than amino acid mixtures, suggesting that a large amount of dietary protein is

absorbed in the form of oligopeptides rather than hydrolyzed into amino acids within the

lumen. Therefore, luminal protein hydrolysate may additionally or alternatively signal in

the form of oligopeptides. Indeed, co-infusion of the proteolytic enzyme trypsin

attenuates the CCK response to luminal protein hydrolysate182, and non-metabolizable

oligopeptides stimulate CCK release183. The protein-sensor PepT1 is likely involved in

sensing these oligopeptides, given that this di- and tripeptide-specific transporter

mediates CCK release as well as vagal afferent firing in response to intestinal protein

hydrolysate58,181. In order to evaluate the role of PepT1, future studies will employ co-

infusion of the competitive non-translocated inhibitor 4-aminomethylbenzoic acid (4-

AMBA) with upper small intestinal protein during both the IVGTT and clamp protocols.

To further confirm the role of PepT1, the PepT1 protein will be knocked down in the

upper small intestine with a lentiviral shRNA infection during the gut cannulation

surgery as confirmed in Côté et al209. Finally, upper small intestinal infusion with non-

metabolizable substrates of PepT1 such as the cephalosporin antibiotic Cefaclor or the

synthetic dipeptide Gly-Sar during the IVGTT or clamp would confirm whether substrate

binding and activation of PepT1 per se initiates the neuronal network to regulate

Page 71: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

62

glucose homeostasis or whether subsequent intracellular metabolism of peptides is

required.

Identification of the precise mechanisms through which protein initiates this negative

feedback pathway on glucose metabolism is important to potentially unveil therapeutic

targets at the level of the gut to lower hyperglycemia in disease. Indeed, this gut-brain-

liver axis has recently been demonstrated to have important therapeutic relevance.

Both metformin and resveratrol activate gut-localized signaling pathways to lower

hepatic glucose production via a neuronal gut-brain-liver axis in high-fat diet-fed and

type 2 diabetic rodents10,209. Furthermore, in humans, 12-week treatment with delayed-

release Metformin, which targets the lower bowel where little absorption occurs, is

more effective in lowering glucose levels in type 2 diabetic patients than 12 weeks of

commercially available immediate-release or extended-release Metformin11. These

results confirm those in rodents suggesting that the primary effect of Metformin on

glucose homeostasis may be via gut-localized signaling. These exciting new studies

confirm that gut nutrient signaling pathways may have pharmacological relevance in

disease and that it is urgent to elucidate the precise molecular mechanisms through

which intestinal nutrients signal, and how signaling defects in high-fat feeding may be

bypassed by selectively targeting these pathways.

While we have confirmed that upper small intestinal protein sensing regulates

intravenous glucose tolerance and suppresses glucose production during the

Page 72: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

63

pancreatic clamp, future studies are required to determine the physiological relevance

of protein sensing in the context of high-protein feeding. As previously mentioned, re-

feeding of a high-protein diet following an overnight fast lowers the glucose response in

healthy male Sprague-Dawley rats in comparison to low-protein feeding45.

Interestingly, blocking hindbrain glucagon receptor signaling partially attenuates this

improvement in glucose tolerance in the later time points (60 minutes after re-

feeding)45, but has no effect on the earlier time points. Thus, the same fasting re-

feeding protocol should be employed in future studies to investigate the role of

intestinal protein sensing in mediating this early improvement in glucose control. Once

we have identified the specific mucosal sensing mechanisms that mediate protein to

improve glucose homeostasis, using the techniques described above, this pathway

should be blocked during re-feeding of a high-protein diet through pharmacological

inhibition or viral knockdown. To investigate how intestinal protein sensing might

contribute to the glucoregulatory capacity of longer-term dietary interventions, knockout

mouse models could be employed. For example, PepT1 mice that lack the transporter

in the intestines and kidney have been developed, and do not display any obvious

abnormalities in size, weight fertility or viability210. It would be interesting to investigate

whether the lack of intestinal PepT1 might become important in the context of high

protein diet feeding, whereby PepT1 knockout attenuates the ability of long-term high

protein diet intervention to improve glucose regulation.

In summary, the present study shows for the first time to our knowledge that upper

small intestinal protein initiates a neuronal network to improve glucose tolerance and

Page 73: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

64

suppress hepatic glucose production, likely via pre-absorptive signaling to initiate gut-

brain and/or gut-brain liver signaling axes. These findings demonstrate that the GI tract

and brain have parallel protein-sensing mechanisms to lower hepatic glucose

production given that central amino acid infusion lowers glucose production during the

same basal insulin pancreatic clamp protocol195,211. While blocking neuronal signaling

with tetracaine completely reversed the effects of intraintestinal protein infusion on

glucose tolerance in the context of a dose which does not rise portal or systemic

plasma amino acids, it is important to note that under physiological high-protein feeding

conditions, where amino acids rise in the circulation, other protein-sensing mechanisms

such as in the brain likely become important.

It will be of great interest in future studies to test the efficacy of upper small intestinal

protein infusion to regulate glucose homeostasis in high-fat diet-fed rats. 3 days of

high-fat diet feeding reverses the effects of upper small intestinal lipid infusion to lower

glucose production, by disrupting the lipid-sensing pathway at the level of intestinal

CCK-1 receptor signaling138,143. Interestingly, however, high protein diet feeding

effectively improves glucose homeostasis in diet-induced obese rodents212,213, and

acute protein intake lowers blood glucose levels in type 2 diabetic humans214,215,216.

Thus, we propose that unlike lipid sensing, intestinal protein sensing mechanisms may

in fact remain intact in rodent models of metabolic disease, suggesting that protein-

sensing pathways may differ from lipid sensing, and further justifying the importance to

investigate the potential therapeutic relevance.

Page 74: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

65

Chapter 6

References

1   Scully,  T.  Diabetes  in  numbers.  Nature  485,  S2-­‐3  (2012).  2   Speakman,  J.  R.  &  O'Rahilly,  S.  Fat:  an  evolving  issue.  Dis  Model  Mech  5,  569-­‐573,  

doi:10.1242/dmm.010553  (2012).  3   Association,  C.  D.  Diabetes  Statistics  in  Canada,  <http://www.diabetes.ca/how-you-can-

help/advocate/why-federal-leadership-is-essential/diabetes-statistics-in-canada - _ftn1>  (2016).  

4   Canada,  P.  H.  A.  o.  Obesity  in  Canada,  <http://www.phac-aspc.gc.ca/hp-ps/hl-mvs/oic-oac/index-eng.php>  (2011).  

5   Stagnitti,  M.  N.  &  Medical  Expenditure  Panel  Survey  (U.S.).  in  Statistical  brief  ;  #34        [5]  p.  (Medical  Expenditure  Panel  Survey,  Agency  for  Healthcare  Research  and  Quality,  [Rockville,  Md.,  2004).  

6   Scheen,  A.  J.  Pathophysiology  of  type  2  diabetes.  Acta  Clin  Belg  58,  335-­‐341,  doi:10.1179/acb.2003.58.6.001  (2003).  

7   Best,  J.  D.  et  al.  Role  of  glucose  effectiveness  in  the  determination  of  glucose  tolerance.  Diabetes  Care  19,  1018-­‐1030  (1996).  

8   Schwartz,  M.  W.  et  al.  Cooperation  between  brain  and  islet  in  glucose  homeostasis  and  diabetes.  Nature  503,  59-­‐66,  doi:10.1038/nature12709  (2013).  

9   Hamr,  S.  C.,  Wang,  B.,  Swartz,  T.  D.  &  Duca,  F.  A.  Does  nutrient  sensing  determine  how  we  "see"  food?  Curr  Diab  Rep  15,  604,  doi:10.1007/s11892-­‐015-­‐0604-­‐7  (2015).  

10   Duca,  F.  A.  et  al.  Metformin  activates  a  duodenal  Ampk-­‐dependent  pathway  to  lower  hepatic  glucose  production  in  rats.  Nat  Med  21,  506-­‐511,  doi:10.1038/nm.3787  (2015).  

11   Buse,  J.  B.  et  al.  The  Primary  Glucose-­‐Lowering  Effect  of  Metformin  Resides  in  the  Gut,  Not  the  Circulation:  Results  From  Short-­‐term  Pharmacokinetic  and  12-­‐Week  Dose-­‐Ranging  Studies.  Diabetes  Care  39,  198-­‐205,  doi:10.2337/dc15-­‐0488  (2016).  

12   Schauer,  P.  R.  et  al.  Bariatric  surgery  versus  intensive  medical  therapy  in  obese  patients  with  diabetes.  N  Engl  J  Med  366,  1567-­‐1576,  doi:10.1056/NEJMoa1200225  (2012).  

13   Breen,  D.  M.  et  al.  Jejunal  nutrient  sensing  is  required  for  duodenal-­‐jejunal  bypass  surgery  to  rapidly  lower  glucose  concentrations  in  uncontrolled  diabetes.  Nat  Med  18,  950-­‐955,  doi:10.1038/nm.2745  (2012).  

14   Lovshin,  J.  A.  &  Drucker,  D.  J.  Incretin-­‐based  therapies  for  type  2  diabetes  mellitus.  Nat  Rev  Endocrinol  5,  262-­‐269,  doi:10.1038/nrendo.2009.48  (2009).  

15   Tessari,  P.  et  al.  Hyperaminoacidaemia  reduces  insulin-­‐mediated  glucose  disposal  in  healthy  man.  Diabetologia  28,  870-­‐872  (1985).  

16   Buckspan,  R.  et  al.  alpha-­‐Ketoisocaproate  is  superior  to  leucine  in  sparing  glucose  utilization  in  humans.  Am  J  Physiol  251,  E648-­‐653  (1986).  

Page 75: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

66

17   Flakoll,  P.  J.,  Wentzel,  L.  S.,  Rice,  D.  E.,  Hill,  J.  O.  &  Abumrad,  N.  N.  Short-­‐term  regulation  of  insulin-­‐mediated  glucose  utilization  in  four-­‐day  fasted  human  volunteers:  role  of  amino  acid  availability.  Diabetologia  35,  357-­‐366  (1992).  

18   Pisters,  P.  W.,  Restifo,  N.  P.,  Cersosimo,  E.  &  Brennan,  M.  F.  The  effects  of  euglycemic  hyperinsulinemia  and  amino  acid  infusion  on  regional  and  whole  body  glucose  disposal  in  man.  Metabolism:  Clinical  &  Experimental  40,  59-­‐65  (1991).  

19   Krebs,  M.  et  al.  Mechanism  of  amino  acid-­‐induced  skeletal  muscle  insulin  resistance  in  humans.  Diabetes  51,  599-­‐605  (2002).  

20   Traxinger,  R.  R.  &  Marshall,  S.  Role  of  amino  acids  in  modulating  glucose-­‐induced  desensitization  of  the  glucose  transport  system.  J  Biol  Chem  264,  20910-­‐20916  (1989).  

21   Walajtys-­‐Rode,  E.  &  Williamson,  J.  R.  Effects  of  branched  chain  alpha-­‐ketoacids  on  the  metabolism  of  isolated  rat  liver  cells.  III.  Interactions  with  pyruvate  dehydrogenase.  J  Biol  Chem  255,  413-­‐418  (1980).  

22   Chang,  T.  W.  &  Goldberg,  A.  L.  Leucine  inhibits  oxidation  of  glucose  and  pyruvate  in  skeletal  muscles  during  fasting.  J  Biol  Chem  253,  3696-­‐3701  (1978).  

23   Tremblay,  F.  &  Marette,  A.  Amino  acid  and  insulin  signaling  via  the  mTOR/p70  S6  kinase  pathway.  A  negative  feedback  mechanism  leading  to  insulin  resistance  in  skeletal  muscle  cells.  J  Biol  Chem  276,  38052-­‐38060,  doi:10.1074/jbc.M106703200  (2001).  

24   Patti,  M.  E.,  Brambilla,  E.,  Luzi,  L.,  Landaker,  E.  J.  &  Kahn,  C.  R.  Bidirectional  modulation  of  insulin  action  by  amino  acids.  J  Clin  Invest  101,  1519-­‐1529,  doi:10.1172/JCI1326  (1998).  

25   Boden,  G.  &  Tappy,  L.  Effects  of  amino  acids  on  glucose  disposal.  Diabetes  39,  1079-­‐1084  (1990).  

26   Tappy,  L.  et  al.  Effects  of  infused  amino  acids  on  glucose  production  and  utilization  in  healthy  human  subjects.  Am  J  Physiol  262,  E826-­‐833  (1992).  

27   Rossetti,  L.,  Rothman,  D.  L.,  DeFronzo,  R.  A.  &  Shulman,  G.  I.  Effect  of  dietary  protein  on  in  vivo  insulin  action  and  liver  glycogen  repletion.  Am  J  Physiol  257,  E212-­‐219  (1989).  

28   Foster,  G.  D.  et  al.  A  randomized  trial  of  a  low-­‐carbohydrate  diet  for  obesity.  N  Engl  J  Med  348,  2082-­‐2090,  doi:10.1056/NEJMoa022207  (2003).  

29   Samaha,  F.  F.  et  al.  A  low-­‐carbohydrate  as  compared  with  a  low-­‐fat  diet  in  severe  obesity.  N  Engl  J  Med  348,  2074-­‐2081,  doi:10.1056/NEJMoa022637  (2003).  

30   Skov,  A.  R.,  Toubro,  S.,  Ronn,  B.,  Holm,  L.  &  Astrup,  A.  Randomized  trial  on  protein  vs  carbohydrate  in  ad  libitum  fat  reduced  diet  for  the  treatment  of  obesity.  International  Journal  of  Obesity  &  Related  Metabolic  Disorders  23,  528-­‐536  (1999).  

31   Pichon,  L.,  Huneau,  J.  F.,  Fromentin,  G.  &  Tome,  D.  A  high-­‐protein,  high-­‐fat,  carbohydrate-­‐free  diet  reduces  energy  intake,  hepatic  lipogenesis,  and  adiposity  in  rats.  J  Nutr  136,  1256-­‐1260  (2006).  

32   Arciero,  P.  J.  et  al.  Moderate  protein  intake  improves  total  and  regional  body  composition  and  insulin  sensitivity  in  overweight  adults.  Metabolism:  Clinical  &  Experimental  57,  757-­‐765,  doi:10.1016/j.metabol.2008.01.015  (2008).  

33   Boden,  G.,  Sargrad,  K.,  Homko,  C.,  Mozzoli,  M.  &  Stein,  T.  P.  Effect  of  a  low-­‐carbohydrate  diet  on  appetite,  blood  glucose  levels,  and  insulin  resistance  in  obese  patients  with  type  2  diabetes.  Ann  Intern  Med  142,  403-­‐411  (2005).  

Page 76: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

67

34   Stern,  L.  et  al.  The  effects  of  low-­‐carbohydrate  versus  conventional  weight  loss  diets  in  severely  obese  adults:  one-­‐year  follow-­‐up  of  a  randomized  trial.  Ann  Intern  Med  140,  778-­‐785  (2004).  

35   Wing,  R.  R.  et  al.  Benefits  of  modest  weight  loss  in  improving  cardiovascular  risk  factors  in  overweight  and  obese  individuals  with  type  2  diabetes.  Diabetes  Care  34,  1481-­‐1486,  doi:10.2337/dc10-­‐2415  (2011).  

36   Gannon,  M.  C.,  Nuttall,  F.  Q.,  Saeed,  A.,  Jordan,  K.  &  Hoover,  H.  An  increase  in  dietary  protein  improves  the  blood  glucose  response  in  persons  with  type  2  diabetes.  Am  J  Clin  Nutr  78,  734-­‐741  (2003).  

37   Nagata,  C.  et  al.  Branched-­‐chain  amino  acid  intake  and  the  risk  of  diabetes  in  a  Japanese  community:  the  Takayama  study.  Am  J  Epidemiol  178,  1226-­‐1232,  doi:10.1093/aje/kwt112  (2013).  

38   Lacroix,  M.  et  al.  A  long-­‐term  high-­‐protein  diet  markedly  reduces  adipose  tissue  without  major  side  effects  in  Wistar  male  rats.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  287,  R934-­‐942,  doi:10.1152/ajpregu.00100.2004  (2004).  

39   Faipoux,  R.  et  al.  Yeast  proteins  enhance  satiety  in  rats.  J  Nutr  136,  2350-­‐2356  (2006).  

40   Blouet,  C.  et  al.  The  reduced  energy  intake  of  rats  fed  a  high-­‐protein  low-­‐carbohydrate  diet  explains  the  lower  fat  deposition,  but  macronutrient  substitution  accounts  for  the  improved  glycemic  control.  J  Nutr  136,  1849-­‐1854  (2006).  

41   Gannon,  M.  C.  &  Nuttall,  F.  Q.  Acute  effects  of  ingestion  of  carbohydrate,  protein,  or  fat  on  cardiac  glycogen  metabolism  in  rats.  Metabolism:  Clinical  &  Experimental  36,  595-­‐600  (1987).  

42   Claessens,  M.,  Calame,  W.,  Siemensma,  A.  D.,  van  Baak,  M.  A.  &  Saris,  W.  H.  The  effect  of  different  protein  hydrolysate/carbohydrate  mixtures  on  postprandial  glucagon  and  insulin  responses  in  healthy  subjects.  Eur  J  Clin  Nutr  63,  48-­‐56,  doi:10.1038/sj.ejcn.1602896  (2009).  

43   Akhavan,  T.,  Luhovyy,  B.  L.,  Brown,  P.  H.,  Cho,  C.  E.  &  Anderson,  G.  H.  Effect  of  premeal  consumption  of  whey  protein  and  its  hydrolysate  on  food  intake  and  postmeal  glycemia  and  insulin  responses  in  young  adults.  Am  J  Clin  Nutr  91,  966-­‐975,  doi:10.3945/ajcn.2009.28406  (2010).  

44   Gunnerud,  U.  J.,  Heinzle,  C.,  Holst,  J.  J.,  Ostman,  E.  M.  &  Bjorck,  I.  M.  Effects  of  pre-­‐meal  drinks  with  protein  and  amino  acids  on  glycemic  and  metabolic  responses  at  a  subsequent  composite  meal.  PLoS  One  7,  e44731,  doi:10.1371/journal.pone.0044731  (2012).  

45   LaPierre,  M.  P.,  Abraham,  M.  A.,  Yue,  J.  T.,  Filippi,  B.  M.  &  Lam,  T.  K.  Glucagon  signalling  in  the  dorsal  vagal  complex  is  sufficient  and  necessary  for  high-­‐protein  feeding  to  regulate  glucose  homeostasis  in  vivo.  EMBO  Rep  16,  1299-­‐1307,  doi:10.15252/embr.201540492  (2015).  

46   Day,  J.  L.  et  al.  Factors  governing  insulin  and  glucagon  responses  during  normal  meals.  Clin  Endocrinol  (Oxf)  9,  443-­‐454  (1978).  

47   Mighiu,  P.  I.  et  al.  Hypothalamic  glucagon  signaling  inhibits  hepatic  glucose  production.  Nat  Med  19,  766-­‐772,  doi:10.1038/nm.3115  (2013).  

48   Ritter,  R.  C.  Gastrointestinal  mechanisms  of  satiation  for  food.  Physiology  &  Behavior  81,  249-­‐273,  doi:10.1016/j.physbeh.2004.02.012  (2004).  

Page 77: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

68

49   Tolhurst,  G.,  Reimann,  F.  &  Gribble,  F.  M.  Intestinal  sensing  of  nutrients.  Handb  Exp  Pharmacol,  309-­‐335,  doi:10.1007/978-­‐3-­‐642-­‐24716-­‐3_14  (2012).  

50   Habib,  A.  M.  et  al.  Overlap  of  endocrine  hormone  expression  in  the  mouse  intestine  revealed  by  transcriptional  profiling  and  flow  cytometry.  Endocrinology  153,  3054-­‐3065,  doi:10.1210/en.2011-­‐2170  (2012).  

51   Svendsen,  B.  et  al.  An  analysis  of  cosecretion  and  coexpression  of  gut  hormones  from  male  rat  proximal  and  distal  small  intestine.  Endocrinology  156,  847-­‐857,  doi:10.1210/en.2014-­‐1710  (2015).  

52   Svendsen,  B.  et  al.  GLP1-­‐  and  GIP-­‐producing  cells  rarely  overlap  and  differ  by  bombesin  receptor-­‐2  expression  and  responsiveness.  J  Endocrinol  228,  39-­‐48,  doi:10.1530/JOE-­‐15-­‐0247  (2016).  

53   Edfalk,  S.,  Steneberg,  P.  &  Edlund,  H.  Gpr40  is  expressed  in  enteroendocrine  cells  and  mediates  free  fatty  acid  stimulation  of  incretin  secretion.  Diabetes  57,  2280-­‐2287,  doi:10.2337/db08-­‐0307  (2008).  

54   Little,  T.  J.  et  al.  Characterization  of  duodenal  expression  and  localization  of  fatty  acid-­‐sensing  receptors  in  humans:  relationships  with  body  mass  index.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  307,  G958-­‐967,  doi:10.1152/ajpgi.00134.2014  (2014).  

55   Iwasaki,  K.  et  al.  Free  fatty  acid  receptor  GPR120  is  highly  expressed  in  enteroendocrine  K  cells  of  the  upper  small  intestine  and  has  a  critical  role  in  GIP  secretion  after  fat  ingestion.  Endocrinology  156,  837-­‐846,  doi:10.1210/en.2014-­‐1653  (2015).  

56   Liou,  A.  P.  et  al.  The  extracellular  calcium-­‐sensing  receptor  is  required  for  cholecystokinin  secretion  in  response  to  L-­‐phenylalanine  in  acutely  isolated  intestinal  I  cells.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  300,  G538-­‐546,  doi:10.1152/ajpgi.00342.2010  (2011).  

57   Reimann,  F.  et  al.  Glucose  sensing  in  L  cells:  a  primary  cell  study.  Cell  Metab  8,  532-­‐539,  doi:10.1016/j.cmet.2008.11.002  (2008).  

58   Liou,  A.  P.  et  al.  Protein  hydrolysate-­‐induced  cholecystokinin  secretion  from  enteroendocrine  cells  is  indirectly  mediated  by  the  intestinal  oligopeptide  transporter  PepT1.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  300,  G895-­‐902,  doi:10.1152/ajpgi.00521.2010  (2011).  

59   Psichas,  A.,  Reimann,  F.  &  Gribble,  F.  M.  Gut  chemosensing  mechanisms.  J  Clin  Invest  125,  908-­‐917,  doi:10.1172/JCI76309  (2015).  

60   Parker,  H.  E.  et  al.  Predominant  role  of  active  versus  facilitative  glucose  transport  for  glucagon-­‐like  peptide-­‐1  secretion.  Diabetologia  55,  2445-­‐2455,  doi:10.1007/s00125-­‐012-­‐2585-­‐2  (2012).  

61   Gorboulev,  V.  et  al.  Na(+)-­‐D-­‐glucose  cotransporter  SGLT1  is  pivotal  for  intestinal  glucose  absorption  and  glucose-­‐dependent  incretin  secretion.  Diabetes  61,  187-­‐196,  doi:10.2337/db11-­‐1029  (2012).  

62   Geraedts,  M.  C.  et  al.  Transformation  of  postingestive  glucose  responses  after  deletion  of  sweet  taste  receptor  subunits  or  gastric  bypass  surgery.  American  Journal  of  Physiology:  Endocrinology  &  Metabolism  303,  E464-­‐474,  doi:10.1152/ajpendo.00163.2012  (2012).  

63   Liou,  A.  P.  et  al.  The  G-­‐protein-­‐coupled  receptor  GPR40  directly  mediates  long-­‐chain  fatty  acid-­‐induced  secretion  of  cholecystokinin.  Gastroenterology  140,  903-­‐912,  doi:10.1053/j.gastro.2010.10.012  (2011).  

Page 78: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

69

64   Lu,  W.  J.  et  al.  Chylomicron  formation  and  secretion  is  required  for  lipid-­‐stimulated  release  of  incretins  GLP-­‐1  and  GIP.  Lipids  47,  571-­‐580,  doi:10.1007/s11745-­‐011-­‐3650-­‐1  (2012).  

65   Whited,  K.  L.,  Lu,  D.,  Tso,  P.,  Kent  Lloyd,  K.  C.  &  Raybould,  H.  E.  Apolipoprotein  A-­‐IV  is  involved  in  detection  of  lipid  in  the  rat  intestine.  J  Physiol  569,  949-­‐958,  doi:10.1113/jphysiol.2005.097634  (2005).  

66   Lo,  C.  C.  et  al.  Apolipoprotein  AIV  requires  cholecystokinin  and  vagal  nerves  to  suppress  food  intake.  Endocrinology  153,  5857-­‐5865,  doi:10.1210/en.2012-­‐1427  (2012).  

67   Brighton,  C.  A.  et  al.  Bile  Acids  Trigger  GLP-­‐1  Release  Predominantly  by  Accessing  Basolaterally  Located  G  Protein-­‐Coupled  Bile  Acid  Receptors.  Endocrinology  156,  3961-­‐3970,  doi:10.1210/en.2015-­‐1321  (2015).  

68   Trabelsi,  M.  S.  et  al.  Farnesoid  X  receptor  inhibits  glucagon-­‐like  peptide-­‐1  production  by  enteroendocrine  L  cells.  Nat  Commun  6,  7629,  doi:10.1038/ncomms8629  (2015).  

69   Koide,  M.,  Okabayashi,  Y.  &  Otsuki,  M.  Role  of  endogenous  bile  on  basal  and  postprandial  CCK  release  in  humans.  Digestive  Diseases  &  Sciences  38,  1284-­‐1290  (1993).  

70   Brenner,  L.,  Yox,  D.  P.  &  Ritter,  R.  C.  Suppression  of  sham  feeding  by  intraintestinal  nutrients  is  not  correlated  with  plasma  cholecystokinin  elevation.  Am  J  Physiol  264,  R972-­‐976  (1993).  

71   Lo,  C.  C.  et  al.  Intraperitoneal  CCK  and  fourth-­‐intraventricular  Apo  AIV  require  both  peripheral  and  NTS  CCK1R  to  reduce  food  intake  in  male  rats.  Endocrinology  155,  1700-­‐1707,  doi:10.1210/en.2013-­‐1846  (2014).  

72   Larsen,  P.  J.,  Tang-­‐Christensen,  M.,  Holst,  J.  J.  &  Orskov,  C.  Distribution  of  glucagon-­‐like  peptide-­‐1  and  other  preproglucagon-­‐derived  peptides  in  the  rat  hypothalamus  and  brainstem.  Neuroscience  77,  257-­‐270  (1997).  

73   Campos,  R.  V.,  Lee,  Y.  C.  &  Drucker,  D.  J.  Divergent  tissue-­‐specific  and  developmental  expression  of  receptors  for  glucagon  and  glucagon-­‐like  peptide-­‐1  in  the  mouse.  Endocrinology  134,  2156-­‐2164,  doi:10.1210/endo.134.5.8156917  (1994).  

74   Holst,  J.  J.  The  physiology  of  glucagon-­‐like  peptide  1.  Physiol  Rev  87,  1409-­‐1439,  doi:10.1152/physrev.00034.2006  (2007).  

75   D'Alessio,  D.  et  al.  Fasting  and  postprandial  concentrations  of  GLP-­‐1  in  intestinal  lymph  and  portal  plasma:  evidence  for  selective  release  of  GLP-­‐1  in  the  lymph  system.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  293,  R2163-­‐2169,  doi:10.1152/ajpregu.00911.2006  (2007).  

76   Ohlsson,  L.,  Kohan,  A.  B.,  Tso,  P.  &  Ahren,  B.  GLP-­‐1  released  to  the  mesenteric  lymph  duct  in  mice:  effects  of  glucose  and  fat.  Regul  Pept  189,  40-­‐45,  doi:10.1016/j.regpep.2014.02.001  (2014).  

77   Kohan,  A.,  Yoder,  S.  &  Tso,  P.  Lymphatics  in  intestinal  transport  of  nutrients  and  gastrointestinal  hormones.  Ann  N  Y  Acad  Sci  1207  Suppl  1,  E44-­‐51,  doi:10.1111/j.1749-­‐6632.2010.05753.x  (2010).  

78   Batterham,  R.  L.  et  al.  Gut  hormone  PYY(3-­‐36)  physiologically  inhibits  food  intake.  Nature  418,  650-­‐654,  doi:10.1038/nature02666  (2002).  

79   Berthoud,  H.  R.  Anatomical  demonstration  of  vagal  input  to  nicotinamide  acetamide  dinucleotide  phosphate  diaphorase-­‐positive  (nitrergic)  neurons  in  rat  fundic  stomach.  J  Comp  Neurol  358,  428-­‐439,  doi:10.1002/cne.903580309  (1995).  

Page 79: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

70

80   Prechtl,  J.  C.  &  Powley,  T.  L.  The  fiber  composition  of  the  abdominal  vagus  of  the  rat.  Anatomy  &  Embryology  181,  101-­‐115  (1990).  

81   Dockray,  G.  J.  Enteroendocrine  cell  signalling  via  the  vagus  nerve.  Curr  Opin  Pharmacol  13,  954-­‐958,  doi:10.1016/j.coph.2013.09.007  (2013).  

82   Amato,  A.  et  al.  Peripheral  motor  action  of  glucagon-­‐like  peptide-­‐1  through  enteric  neuronal  receptors.  Neurogastroenterology  &  Motility  22,  664-­‐e203,  doi:10.1111/j.1365-­‐2982.2010.01476.x  (2010).  

83   Patterson,  L.  M.,  Zheng,  H.  &  Berthoud,  H.  R.  Vagal  afferents  innervating  the  gastrointestinal  tract  and  CCKA-­‐receptor  immunoreactivity.  Anat  Rec  266,  10-­‐20  (2002).  

84   Ritter,  R.  C.  A  tale  of  two  endings:  modulation  of  satiation  by  NMDA  receptors  on  or  near  central  and  peripheral  vagal  afferent  terminals.  Physiology  &  Behavior  105,  94-­‐99,  doi:10.1016/j.physbeh.2011.02.042  (2011).  

85   Darling,  R.  A.  et  al.  Mercaptoacetate  and  fatty  acids  exert  direct  and  antagonistic  effects  on  nodose  neurons  via  GPR40  fatty  acid  receptors.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  307,  R35-­‐43,  doi:10.1152/ajpregu.00536.2013  (2014).  

86   Norgren,  R.  Projections  from  the  nucleus  of  the  solitary  tract  in  the  rat.  Neuroscience  3,  207-­‐218  (1978).  

87   Zittel,  T.  T.,  De  Giorgio,  R.,  Sternini,  C.  &  Raybould,  H.  E.  Fos  protein  expression  in  the  nucleus  of  the  solitary  tract  in  response  to  intestinal  nutrients  in  awake  rats.  Brain  Res  663,  266-­‐270  (1994).  

88   Cote,  C.  D.,  Zadeh-­‐Tahmasebi,  M.,  Rasmussen,  B.  A.,  Duca,  F.  A.  &  Lam,  T.  K.  Hormonal  signaling  in  the  gut.  J  Biol  Chem  289,  11642-­‐11649,  doi:10.1074/jbc.O114.556068  (2014).  

89   Davis,  J.  D.  &  Smith,  G.  P.  Learning  to  sham  feed:  behavioral  adjustments  to  loss  of  physiological  postingestional  stimuli.  Am  J  Physiol  259,  R1228-­‐1235  (1990).  

90   Gibbs,  J.,  Maddison,  S.  P.  &  Rolls,  E.  T.  Satiety  role  of  the  small  intestine  examined  in  sham-­‐feeding  rhesus  monkeys.  Journal  of  Comparative  &  Physiological  Psychology  95,  1003-­‐1015  (1981).  

91   Burton-­‐Freeman,  B.,  Gietzen,  D.  W.  &  Schneeman,  B.  O.  Meal  pattern  analysis  to  investigate  the  satiating  potential  of  fat,  carbohydrate,  and  protein  in  rats.  Am  J  Physiol  273,  R1916-­‐1922  (1997).  

92   Yox,  D.  P.  &  Ritter,  R.  C.  Capsaicin  attenuates  suppression  of  sham  feeding  induced  by  intestinal  nutrients.  Am  J  Physiol  255,  R569-­‐574  (1988).  

93   Yox,  D.  P.,  Stokesberry,  H.  &  Ritter,  R.  C.  Vagotomy  attenuates  suppression  of  sham  feeding  induced  by  intestinal  nutrients.  Am  J  Physiol  260,  R503-­‐508  (1991).  

94   Yox,  D.  P.,  Stokesberry,  H.  &  Ritter,  R.  C.  Fourth  ventricular  capsaicin  attenuates  suppression  of  sham  feeding  induced  by  intestinal  nutrients.  Am  J  Physiol  260,  R681-­‐687  (1991).  

95   Gibbs,  J.,  Young,  R.  C.  &  Smith,  G.  P.  Cholecystokinin  decreases  food  intake  in  rats.  Journal  of  Comparative  &  Physiological  Psychology  84,  488-­‐495  (1973).  

96   Kissileff,  H.  R.,  Pi-­‐Sunyer,  F.  X.,  Thornton,  J.  &  Smith,  G.  P.  C-­‐terminal  octapeptide  of  cholecystokinin  decreases  food  intake  in  man.  Am  J  Clin  Nutr  34,  154-­‐160  (1981).  

97   Beglinger,  C.,  Degen,  L.,  Matzinger,  D.,  D'Amato,  M.  &  Drewe,  J.  Loxiglumide,  a  CCK-­‐A  receptor  antagonist,  stimulates  calorie  intake  and  hunger  feelings  in  humans.  

Page 80: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

71

American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  280,  R1149-­‐1154  (2001).  

98   Matzinger,  D.  et  al.  Inhibition  of  food  intake  in  response  to  intestinal  lipid  is  mediated  by  cholecystokinin  in  humans.  Am  J  Physiol  277,  R1718-­‐1724  (1999).  

99   Lal,  S.,  Kirkup,  A.  J.,  Brunsden,  A.  M.,  Thompson,  D.  G.  &  Grundy,  D.  Vagal  afferent  responses  to  fatty  acids  of  different  chain  length  in  the  rat.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  281,  G907-­‐915  (2001).  

100   Brenner,  L.  A.  &  Ritter,  R.  C.  Type  A  CCK  receptors  mediate  satiety  effects  of  intestinal  nutrients.  Pharmacology,  Biochemistry  &  Behavior  54,  625-­‐631  (1996).  

101   Covasa,  M.  &  Ritter,  R.  C.  Attenuated  satiation  response  to  intestinal  nutrients  in  rats  that  do  not  express  CCK-­‐A  receptors.  Peptides  22,  1339-­‐1348  (2001).  

102   Rinaman,  L.  Hindbrain  noradrenergic  lesions  attenuate  anorexia  and  alter  central  cFos  expression  in  rats  after  gastric  viscerosensory  stimulation.  J  Neurosci  23,  10084-­‐10092  (2003).  

103   Beinfeld,  M.  C.  An  introduction  to  neuronal  cholecystokinin.  Peptides  22,  1197-­‐1200  (2001).  

104   Moran,  T.  H.,  Ameglio,  P.  J.,  Schwartz,  G.  J.  &  McHugh,  P.  R.  Blockade  of  type  A,  not  type  B,  CCK  receptors  attenuates  satiety  actions  of  exogenous  and  endogenous  CCK.  Am  J  Physiol  262,  R46-­‐50  (1992).  

105   Broberger,  C.,  Holmberg,  K.,  Shi,  T.  J.,  Dockray,  G.  &  Hokfelt,  T.  Expression  and  regulation  of  cholecystokinin  and  cholecystokinin  receptors  in  rat  nodose  and  dorsal  root  ganglia.  Brain  Res  903,  128-­‐140  (2001).  

106   Blevins,  J.  E.,  Stanley,  B.  G.  &  Reidelberger,  R.  D.  Brain  regions  where  cholecystokinin  suppresses  feeding  in  rats.  Brain  Res  860,  1-­‐10  (2000).  

107   Brenner,  L.  A.  &  Ritter,  R.  C.  Intracerebroventricular  cholecystokinin  A-­‐receptor  antagonist  does  not  reduce  satiation  by  endogenous  CCK.  Physiology  &  Behavior  63,  711-­‐716  (1998).  

108   Lavin,  J.  H.  et  al.  Interaction  of  insulin,  glucagon-­‐like  peptide  1,  gastric  inhibitory  polypeptide,  and  appetite  in  response  to  intraduodenal  carbohydrate.  Am  J  Clin  Nutr  68,  591-­‐598  (1998).  

109   Abbott,  C.  R.  et  al.  Blockade  of  the  neuropeptide  Y  Y2  receptor  with  the  specific  antagonist  BIIE0246  attenuates  the  effect  of  endogenous  and  exogenous  peptide  YY(3-­‐36)  on  food  intake.  Brain  Res  1043,  139-­‐144,  doi:10.1016/j.brainres.2005.02.065  (2005).  

110   Hayes,  M.  R.  et  al.  The  common  hepatic  branch  of  the  vagus  is  not  required  to  mediate  the  glycemic  and  food  intake  suppressive  effects  of  glucagon-­‐like-­‐peptide-­‐1.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  301,  R1479-­‐1485,  doi:10.1152/ajpregu.00356.2011  (2011).  

111   Verdich,  C.  et  al.  A  meta-­‐analysis  of  the  effect  of  glucagon-­‐like  peptide-­‐1  (7-­‐36)  amide  on  ad  libitum  energy  intake  in  humans.  Journal  of  Clinical  Endocrinology  &  Metabolism  86,  4382-­‐4389,  doi:10.1210/jcem.86.9.7877  (2001).  

112   Plamboeck,  A.  et  al.  The  effect  of  exogenous  GLP-­‐1  on  food  intake  is  lost  in  male  truncally  vagotomized  subjects  with  pyloroplasty.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  304,  G1117-­‐1127,  doi:10.1152/ajpgi.00035.2013  (2013).  

113   Wright,  J.  et  al.  Reduction  of  food  intake  by  cholecystokinin  requires  activation  of  hindbrain  NMDA-­‐type  glutamate  receptors.  American  Journal  of  Physiology:  

Page 81: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

72

Regulatory,  Integrative  &  Comparative  Physiology  301,  R448-­‐455,  doi:10.1152/ajpregu.00026.2011  (2011).  

114   Campos,  C.  A.,  Wright,  J.  S.,  Czaja,  K.  &  Ritter,  R.  C.  CCK-­‐induced  reduction  of  food  intake  and  hindbrain  MAPK  signaling  are  mediated  by  NMDA  receptor  activation.  Endocrinology  153,  2633-­‐2646,  doi:10.1210/en.2012-­‐1025  (2012).  

115   Campos,  C.  A.,  Shiina,  H.,  Silvas,  M.,  Page,  S.  &  Ritter,  R.  C.  Vagal  afferent  NMDA  receptors  modulate  CCK-­‐induced  reduction  of  food  intake  through  synapsin  I  phosphorylation  in  adult  male  rats.  Endocrinology  154,  2613-­‐2625,  doi:10.1210/en.2013-­‐1062  (2013).  

116   Babic,  T.  et  al.  Phenotype  of  neurons  in  the  nucleus  of  the  solitary  tract  that  express  CCK-­‐induced  activation  of  the  ERK  signaling  pathway.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  296,  R845-­‐854,  doi:10.1152/ajpregu.90531.2008  (2009).  

117   Sutton,  G.  M.,  Duos,  B.,  Patterson,  L.  M.  &  Berthoud,  H.  R.  Melanocortinergic  modulation  of  cholecystokinin-­‐induced  suppression  of  feeding  through  extracellular  signal-­‐regulated  kinase  signaling  in  rat  solitary  nucleus.  Endocrinology  146,  3739-­‐3747,  doi:10.1210/en.2005-­‐0562  (2005).  

118   Seeley,  R.  J.,  Grill,  H.  J.  &  Kaplan,  J.  M.  Neurological  dissociation  of  gastrointestinal  and  metabolic  contributions  to  meal  size  control.  Behav  Neurosci  108,  347-­‐352  (1994).  

119   Moran,  T.  H.  &  Bi,  S.  Hyperphagia  and  obesity  in  OLETF  rats  lacking  CCK-­‐1  receptors.  Philos  Trans  R  Soc  Lond  B  Biol  Sci  361,  1211-­‐1218,  doi:10.1098/rstb.2006.1857  (2006).  

120   Elrick,  H.,  Stimmler,  L.,  Hlad,  C.  J.,  Jr.  &  Arai,  Y.  Plasma  Insulin  Response  to  Oral  and  Intravenous  Glucose  Administration.  Journal  of  Clinical  Endocrinology  &  Metabolism  24,  1076-­‐1082,  doi:10.1210/jcem-­‐24-­‐10-­‐1076  (1964).  

121   Moens,  K.  et  al.  Expression  and  functional  activity  of  glucagon,  glucagon-­‐like  peptide  I,  and  glucose-­‐dependent  insulinotropic  peptide  receptors  in  rat  pancreatic  islet  cells.  Diabetes  45,  257-­‐261  (1996).  

122   Kreymann,  B.,  Williams,  G.,  Ghatei,  M.  A.  &  Bloom,  S.  R.  Glucagon-­‐like  peptide-­‐1  7-­‐36:  a  physiological  incretin  in  man.  Lancet  2,  1300-­‐1304  (1987).  

123   Brown,  J.  C.,  Dryburgh,  J.  R.,  Ross,  S.  A.  &  Dupre,  J.  Identification  and  actions  of  gastric  inhibitory  polypeptide.  Recent  Prog  Horm  Res  31,  487-­‐532  (1975).  

124   D'Alessio,  D.  A.,  Kahn,  S.  E.,  Leusner,  C.  R.  &  Ensinck,  J.  W.  Glucagon-­‐like  peptide  1  enhances  glucose  tolerance  both  by  stimulation  of  insulin  release  and  by  increasing  insulin-­‐independent  glucose  disposal.  J  Clin  Invest  93,  2263-­‐2266,  doi:10.1172/JCI117225  (1994).  

125   Prigeon,  R.  L.,  Quddusi,  S.,  Paty,  B.  &  D'Alessio,  D.  A.  Suppression  of  glucose  production  by  GLP-­‐1  independent  of  islet  hormones:  a  novel  extrapancreatic  effect.  American  Journal  of  Physiology:  Endocrinology  &  Metabolism  285,  E701-­‐707,  doi:10.1152/ajpendo.00024.2003  (2003).  

126   Knauf,  C.  et  al.  Brain  glucagon-­‐like  peptide-­‐1  increases  insulin  secretion  and  muscle  insulin  resistance  to  favor  hepatic  glycogen  storage.  J  Clin  Invest  115,  3554-­‐3563,  doi:10.1172/JCI25764  (2005).  

127   Sandoval,  D.  A.,  Bagnol,  D.,  Woods,  S.  C.,  D'Alessio,  D.  A.  &  Seeley,  R.  J.  Arcuate  glucagon-­‐like  peptide  1  receptors  regulate  glucose  homeostasis  but  not  food  intake.  Diabetes  57,  2046-­‐2054,  doi:10.2337/db07-­‐1824  (2008).  

Page 82: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

73

128   Krieger,  J.  P.  et  al.  Knockdown  of  GLP-­‐1  Receptors  in  Vagal  Afferents  Affects  Normal  Food  Intake  and  Glycemia.  Diabetes  65,  34-­‐43,  doi:10.2337/db15-­‐0973  (2016).  

129   Waget,  A.  et  al.  Physiological  and  pharmacological  mechanisms  through  which  the  DPP-­‐4  inhibitor  sitagliptin  regulates  glycemia  in  mice.  Endocrinology  152,  3018-­‐3029,  doi:10.1210/en.2011-­‐0286  (2011).  

130   Burcelin,  R.,  Da  Costa,  A.,  Drucker,  D.  &  Thorens,  B.  Glucose  competence  of  the  hepatoportal  vein  sensor  requires  the  presence  of  an  activated  glucagon-­‐like  peptide-­‐1  receptor.  Diabetes  50,  1720-­‐1728  (2001).  

131   Balkan,  B.,  Steffens,  A.  B.,  Strubbe,  J.  H.  &  Bruggink,  J.  E.  Biphasic  insulin  secretion  after  intravenous  but  not  after  intraportal  CCK-­‐8  infusion  in  rats.  Diabetes  39,  702-­‐706  (1990).  

132   Sakamoto,  C.,  Goldfine,  I.  D.,  Roach,  E.  &  Williams,  J.  A.  Localization  of  saturable  CCK  binding  sites  in  rat  pancreatic  islets  by  light  and  electron  microscope  autoradiography.  Diabetes  34,  390-­‐394  (1985).  

133   Lo,  C.  M.  et  al.  Impaired  insulin  secretion  and  enhanced  insulin  sensitivity  in  cholecystokinin-­‐deficient  mice.  Diabetes  60,  2000-­‐2007,  doi:10.2337/db10-­‐0789  (2011).  

134   Verspohl,  E.  J.,  Ammon,  H.  P.,  Williams,  J.  A.  &  Goldfine,  I.  D.  Evidence  that  cholecystokinin  interacts  with  specific  receptors  and  regulates  insulin  release  in  isolated  rat  islets  of  Langerhans.  Diabetes  35,  38-­‐43  (1986).  

135   Zawalich,  W.,  Takuwa,  N.,  Takuwa,  Y.,  Diaz,  V.  A.  &  Rasmussen,  H.  Interactions  of  cholecystokinin  and  glucose  in  rat  pancreatic  islets.  Diabetes  36,  426-­‐433  (1987).  

136   Malm,  D.,  Giaever,  A.,  Vonen,  B.  &  Florholmen,  J.  Cholecystokinin  and  somatostatin  modulate  the  glucose-­‐induced  insulin  secretion  by  different  mechanisms  in  pancreatic  islets.  A  study  on  phospholipase  C  activity  and  calcium  requirement.  Scandinavian  Journal  of  Clinical  &  Laboratory  Investigation  53,  671-­‐676  (1993).  

137   Cheung,  G.  W.,  Kokorovic,  A.,  Lam,  C.  K.,  Chari,  M.  &  Lam,  T.  K.  Intestinal  cholecystokinin  controls  glucose  production  through  a  neuronal  network.  Cell  Metab  10,  99-­‐109,  doi:10.1016/j.cmet.2009.07.005  (2009).  

138   Wang,  P.  Y.  et  al.  Upper  intestinal  lipids  trigger  a  gut-­‐brain-­‐liver  axis  to  regulate  glucose  production.  Nature  452,  1012-­‐1016,  doi:10.1038/nature06852  (2008).  

139   Kokorovic,  A.  et  al.  Duodenal  mucosal  protein  kinase  C-­‐delta  regulates  glucose  production  in  rats.  Gastroenterology  141,  1720-­‐1727,  doi:10.1053/j.gastro.2011.06.042  (2011).  

140   Chang,  C.  H.,  Chey,  W.  Y.  &  Chang,  T.  M.  Cellular  mechanism  of  sodium  oleate-­‐stimulated  secretion  of  cholecystokinin  and  secretin.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  279,  G295-­‐303  (2000).  

141   Takahashi,  A.  et  al.  Involvement  of  calmodulin  and  protein  kinase  C  in  cholecystokinin  release  by  bombesin  from  STC-­‐1  cells.  Pancreas  21,  231-­‐239  (2000).  

142   Rogers,  R.  C.  &  Hermann,  G.  E.  Mechanisms  of  action  of  CCK  to  activate  central  vagal  afferent  terminals.  Peptides  29,  1716-­‐1725,  doi:10.1016/j.peptides.2008.06.023  (2008).  

143   Rasmussen,  B.  A.  et  al.  Duodenal  activation  of  cAMP-­‐dependent  protein  kinase  induces  vagal  afferent  firing  and  lowers  glucose  production  in  rats.  Gastroenterology  142,  834-­‐843  e833,  doi:10.1053/j.gastro.2011.12.053  (2012).  

Page 83: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

74

144   Duca,  F.  A.,  Bauer,  P.  V.,  Hamr,  S.  C.  &  Lam,  T.  K.  Glucoregulatory  Relevance  of  Small  Intestinal  Nutrient  Sensing  in  Physiology,  Bariatric  Surgery,  and  Pharmacology.  Cell  Metab  22,  367-­‐380,  doi:10.1016/j.cmet.2015.07.003  (2015).  

145   Covasa,  M.  &  Ritter,  R.  C.  Reduced  sensitivity  to  the  satiation  effect  of  intestinal  oleate  in  rats  adapted  to  high-­‐fat  diet.  Am  J  Physiol  277,  R279-­‐285  (1999).  

146   Boyd,  K.  A.  et  al.  High-­‐fat  diet  effects  on  gut  motility,  hormone,  and  appetite  responses  to  duodenal  lipid  in  healthy  men.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  284,  G188-­‐196,  doi:10.1152/ajpgi.00375.2002  (2003).  

147   French,  S.  J.,  Murray,  B.,  Rumsey,  R.  D.,  Fadzlin,  R.  &  Read,  N.  W.  Adaptation  to  high-­‐fat  diets:  effects  on  eating  behaviour  and  plasma  cholecystokinin.  Br  J  Nutr  73,  179-­‐189  (1995).  

148   Richards,  P.  et  al.  High  fat  diet  impairs  the  function  of  glucagon-­‐like  peptide-­‐1  producing  L-­‐cells.  Peptides,  doi:10.1016/j.peptides.2015.06.006  (2015).  

149   Sundaresan,  S.  et  al.  CD36-­‐dependent  signaling  mediates  fatty  acid-­‐induced  gut  release  of  secretin  and  cholecystokinin.  FASEB  J  27,  1191-­‐1202,  doi:10.1096/fj.12-­‐217703  (2013).  

150   Duca,  F.  A.,  Swartz,  T.  D.,  Sakar,  Y.  &  Covasa,  M.  Decreased  intestinal  nutrient  response  in  diet-­‐induced  obese  rats:  role  of  gut  peptides  and  nutrient  receptors.  Int  J  Obes  (Lond)  37,  375-­‐381,  doi:10.1038/ijo.2012.45  (2013).  

151   Savastano,  D.  M.  &  Covasa,  M.  Adaptation  to  a  high-­‐fat  diet  leads  to  hyperphagia  and  diminished  sensitivity  to  cholecystokinin  in  rats.  J  Nutr  135,  1953-­‐1959  (2005).  

152   Covasa,  M.,  Grahn,  J.  &  Ritter,  R.  C.  Reduced  hindbrain  and  enteric  neuronal  response  to  intestinal  oleate  in  rats  maintained  on  high-­‐fat  diet.  Auton  Neurosci  84,  8-­‐18,  doi:10.1016/S1566-­‐0702(00)00176-­‐4  (2000).  

153   Swartz,  T.  D.,  Duca,  F.  A.  &  Covasa,  M.  Differential  feeding  behavior  and  neuronal  responses  to  CCK  in  obesity-­‐prone  and  -­‐resistant  rats.  Brain  Res  1308,  79-­‐86,  doi:10.1016/j.brainres.2009.10.045  (2010).  

154   Covasa,  M.,  Grahn,  J.  &  Ritter,  R.  C.  High  fat  maintenance  diet  attenuates  hindbrain  neuronal  response  to  CCK.  Regul  Pept  86,  83-­‐88  (2000).  

155   van  der  Klaauw,  A.  A.  et  al.  High  protein  intake  stimulates  postprandial  GLP1  and  PYY  release.  Obesity  (Silver  Spring)  21,  1602-­‐1607,  doi:10.1002/oby.20154  (2013).  

156   Rezek,  M.  &  Novin,  D.  Duodenal  nutrient  infusion:  effects  on  feeding  in  intact  and  vagotomized  rabbits.  J  Nutr  106,  812-­‐820  (1976).  

157   Bowen,  J.,  Noakes,  M.  &  Clifton,  P.  M.  Appetite  regulatory  hormone  responses  to  various  dietary  proteins  differ  by  body  mass  index  status  despite  similar  reductions  in  ad  libitum  energy  intake.  Journal  of  Clinical  Endocrinology  &  Metabolism  91,  2913-­‐2919,  doi:10.1210/jc.2006-­‐0609  (2006).  

158   Lejeune,  M.  P.,  Westerterp,  K.  R.,  Adam,  T.  C.,  Luscombe-­‐Marsh,  N.  D.  &  Westerterp-­‐Plantenga,  M.  S.  Ghrelin  and  glucagon-­‐like  peptide  1  concentrations,  24-­‐h  satiety,  and  energy  and  substrate  metabolism  during  a  high-­‐protein  diet  and  measured  in  a  respiration  chamber.  Am  J  Clin  Nutr  83,  89-­‐94  (2006).  

159   Fromentin,  G.  et  al.  Peripheral  and  central  mechanisms  involved  in  the  control  of  food  intake  by  dietary  amino  acids  and  proteins.  Nutr  Res  Rev  25,  29-­‐39,  doi:10.1017/S0954422411000175  (2012).  

Page 84: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

75

160   Moran,  L.  J.  et  al.  The  satiating  effect  of  dietary  protein  is  unrelated  to  postprandial  ghrelin  secretion.  Journal  of  Clinical  Endocrinology  &  Metabolism  90,  5205-­‐5211,  doi:10.1210/jc.2005-­‐0701  (2005).  

161   Steinert,  R.  E.  et  al.  Effects  of  intraduodenal  infusion  of  L-­‐tryptophan  on  ad  libitum  eating,  antropyloroduodenal  motility,  glycemia,  insulinemia,  and  gut  peptide  secretion  in  healthy  men.  Journal  of  Clinical  Endocrinology  &  Metabolism  99,  3275-­‐3284,  doi:10.1210/jc.2014-­‐1943  (2014).  

162   Meyer,  J.  H.,  Hlinka,  M.,  Tabrizi,  Y.,  DiMaso,  N.  &  Raybould,  H.  E.  Chemical  specificities  and  intestinal  distributions  of  nutrient-­‐driven  satiety.  Am  J  Physiol  275,  R1293-­‐1307  (1998).  

163   Ryan,  A.  T.  et  al.  Effects  of  intraduodenal  lipid  and  protein  on  gut  motility  and  hormone  release,  glycemia,  appetite,  and  energy  intake  in  lean  men.  Am  J  Clin  Nutr  98,  300-­‐311,  doi:10.3945/ajcn.113.061333  (2013).  

164   Tome,  D.,  Schwarz,  J.,  Darcel,  N.  &  Fromentin,  G.  Protein,  amino  acids,  vagus  nerve  signaling,  and  the  brain.  Am  J  Clin  Nutr  90,  838S-­‐843S,  doi:10.3945/ajcn.2009.27462W  (2009).  

165   Faipoux,  R.,  Tome,  D.,  Gougis,  S.,  Darcel,  N.  &  Fromentin,  G.  Proteins  activate  satiety-­‐related  neuronal  pathways  in  the  brainstem  and  hypothalamus  of  rats.  J  Nutr  138,  1172-­‐1178  (2008).  

166   Smeets,  A.  J.,  Soenen,  S.,  Luscombe-­‐Marsh,  N.  D.,  Ueland,  O.  &  Westerterp-­‐Plantenga,  M.  S.  Energy  expenditure,  satiety,  and  plasma  ghrelin,  glucagon-­‐like  peptide  1,  and  peptide  tyrosine-­‐tyrosine  concentrations  following  a  single  high-­‐protein  lunch.  J  Nutr  138,  698-­‐702  (2008).  

167   Dyer,  J.,  Salmon,  K.  S.,  Zibrik,  L.  &  Shirazi-­‐Beechey,  S.  P.  Expression  of  sweet  taste  receptors  of  the  T1R  family  in  the  intestinal  tract  and  enteroendocrine  cells.  Biochem  Soc  Trans  33,  302-­‐305,  doi:10.1042/BST0330302  (2005).  

168   Daly,  K.  et  al.  Sensing  of  amino  acids  by  the  gut-­‐expressed  taste  receptor  T1R1-­‐T1R3  stimulates  CCK  secretion.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  304,  G271-­‐282,  doi:10.1152/ajpgi.00074.2012  (2013).  

169   Broer,  S.  The  role  of  the  neutral  amino  acid  transporter  B0AT1  (SLC6A19)  in  Hartnup  disorder  and  protein  nutrition.  IUBMB  Life  61,  591-­‐599,  doi:10.1002/iub.210  (2009).  

170   Broer,  A.  et  al.  Impaired  nutrient  signaling  and  body  weight  control  in  a  Na+  neutral  amino  acid  cotransporter  (Slc6a19)-­‐deficient  mouse.  J  Biol  Chem  286,  26638-­‐26651,  doi:10.1074/jbc.M111.241323  (2011).  

171   Wolff,  B.  S.,  Meirelles,  K.,  Meng,  Q.,  Pan,  M.  &  Cooney,  R.  N.  Roux-­‐en-­‐Y  gastric  bypass  alters  small  intestine  glutamine  transport  in  the  obese  Zucker  rat.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  297,  G594-­‐601,  doi:10.1152/ajpgi.00104.2009  (2009).  

172   Choi,  S.,  Lee,  M.,  Shiu,  A.  L.,  Yo,  S.  J.  &  Aponte,  G.  W.  Identification  of  a  protein  hydrolysate  responsive  G  protein-­‐coupled  receptor  in  enterocytes.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  292,  G98-­‐G112,  doi:10.1152/ajpgi.00295.2006  (2007).  

173   Choi,  S.  et  al.  GPR93  activation  by  protein  hydrolysate  induces  CCK  transcription  and  secretion  in  STC-­‐1  cells.  American  Journal  of  Physiology:  Gastrointestinal  &  Liver  Physiology  292,  G1366-­‐1375,  doi:10.1152/ajpgi.00516.2006  (2007).  

Page 85: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

76

174   Wang,  M.,  Yao,  Y.,  Kuang,  D.  &  Hampson,  D.  R.  Activation  of  family  C  G-­‐protein-­‐coupled  receptors  by  the  tripeptide  glutathione.  J  Biol  Chem  281,  8864-­‐8870,  doi:10.1074/jbc.M512865200  (2006).  

175   Nakajima,  S.,  Hira,  T.,  Sufian,  M.  K.,  Eto,  Y.  &  Hara,  H.  Calcium-­‐sensing  receptor  is  a  sensor  for  dietary  peptides  to  stimulate  cholecystokinin  secretion  in  the  enteroendocrine  cell  line  STC-­‐1.  Regul  Pept  164,  45,  doi:10.1016/j.regpep.2010.07.118  (2010).  

176   Nakajima,  S.,  Hira,  T.  &  Hara,  H.  Calcium-­‐sensing  receptor  mediates  dietary  peptide-­‐induced  CCK  secretion  in  enteroendocrine  STC-­‐1  cells.  Mol  Nutr  Food  Res  56,  753-­‐760,  doi:10.1002/mnfr.201100666  (2012).  

177   Diakogiannaki,  E.  et  al.  Oligopeptides  stimulate  glucagon-­‐like  peptide-­‐1  secretion  in  mice  through  proton-­‐coupled  uptake  and  the  calcium-­‐sensing  receptor.  Diabetologia  56,  2688-­‐2696,  doi:10.1007/s00125-­‐013-­‐3037-­‐3  (2013).  

178   Jeanningros,  R.  Vagal  unitary  responses  to  intestinal  amino  acid  infusions  in  the  anesthetized  cat:  a  putative  signal  for  protein  induced  satiety.  Physiology  &  Behavior  28,  9-­‐21  (1982).  

179   Lindgren,  O.  et  al.  Incretin  effect  after  oral  amino  acid  ingestion  in  humans.  Journal  of  Clinical  Endocrinology  &  Metabolism  100,  1172-­‐1176,  doi:10.1210/jc.2014-­‐3865  (2015).  

180   Eastwood,  C.,  Maubach,  K.,  Kirkup,  A.  J.  &  Grundy,  D.  The  role  of  endogenous  cholecystokinin  in  the  sensory  transduction  of  luminal  nutrient  signals  in  the  rat  jejunum.  Neurosci  Lett  254,  145-­‐148  (1998).  

181   Darcel,  N.  P.,  Liou,  A.  P.,  Tome,  D.  &  Raybould,  H.  E.  Activation  of  vagal  afferents  in  the  rat  duodenum  by  protein  digests  requires  PepT1.  J  Nutr  135,  1491-­‐1495  (2005).  

182   Cuber,  J.  C.  et  al.  Luminal  CCK-­‐releasing  factors  in  the  isolated  vascularly  perfused  rat  duodenojejunum.  Am  J  Physiol  259,  G191-­‐197  (1990).  

183   Nemoz-­‐Gaillard,  E.  et  al.  Regulation  of  cholecystokinin  secretion  by  peptones  and  peptidomimetic  antibiotics  in  STC-­‐1  cells.  Endocrinology  139,  932-­‐938,  doi:10.1210/endo.139.3.5802  (1998).  

184   Murai,  A.,  Noble,  P.  M.,  Deavall,  D.  G.  &  Dockray,  G.  J.  Control  of  c-­‐fos  expression  in  STC-­‐1  cells  by  peptidomimetic  stimuli.  Eur  J  Pharmacol  394,  27-­‐34  (2000).  

185   Cordier-­‐Bussat,  M.  et  al.  Peptones  stimulate  cholecystokinin  secretion  and  gene  transcription  in  the  intestinal  cell  line  STC-­‐1.  Endocrinology  138,  1137-­‐1144,  doi:10.1210/endo.138.3.5023  (1997).  

186   McConnell,  E.  L.,  Basit,  A.  W.  &  Murdan,  S.  Measurements  of  rat  and  mouse  gastrointestinal  pH,  fluid  and  lymphoid  tissue,  and  implications  for  in-­‐vivo  experiments.  Journal  of  Pharmacy  &  Pharmacology  60,  63-­‐70,  doi:10.1211/jpp.60.1.0008  (2008).  

187   Schwartz,  G.  J.  et  al.  The  lipid  messenger  OEA  links  dietary  fat  intake  to  satiety.  Cell  Metab  8,  281-­‐288,  doi:10.1016/j.cmet.2008.08.005  (2008).  

188   Matthews,  D.  M.,  Muir,  G.  G.  &  Baron,  D.  N.  Estimation  of  Alpha-­‐Amino  Nitrogen  in  Plasma  and  Urine  by  the  Colorimetric  Ninhydrin  Reaction.  J  Clin  Pathol  17,  150-­‐153  (1964).  

189   Schemmel,  R.,  Mickelsen,  O.  &  Gill,  J.  L.  Dietary  obesity  in  rats:  Body  weight  and  body  fat  accretion  in  seven  strains  of  rats.  J  Nutr  100,  1041-­‐1048  (1970).  

Page 86: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

77

190   Ussar,  S.  et  al.  Interactions  between  Gut  Microbiota,  Host  Genetics  and  Diet  Modulate  the  Predisposition  to  Obesity  and  Metabolic  Syndrome.  Cell  Metab  22,  516-­‐530,  doi:10.1016/j.cmet.2015.07.007  (2015).  

191   Lozner,  E.  L.,  Winkler,  A.  W.,  Taylor,  F.  H.  &  Peters,  J.  P.  The  Intravenous  Glucose  Tolerance  Test.  J  Clin  Invest  20,  507-­‐515,  doi:10.1172/JCI101243  (1941).  

192   Smith,  D.  et  al.  In  vivo  glucose  metabolism  in  the  awake  rat:  tracer  and  insulin  clamp  studies.  Metabolism:  Clinical  &  Experimental  36,  1167-­‐1174  (1987).  

193   Greenberg,  D.,  Smith,  G.  P.  &  Gibbs,  J.  Intraduodenal  infusions  of  fats  elicit  satiety  in  sham-­‐feeding  rats.  Am  J  Physiol  259,  R110-­‐118  (1990).  

194   Tremblay,  F.,  Lavigne,  C.,  Jacques,  H.  &  Marette,  A.  Role  of  dietary  proteins  and  amino  acids  in  the  pathogenesis  of  insulin  resistance.  Annu  Rev  Nutr  27,  293-­‐310,  doi:10.1146/annurev.nutr.25.050304.092545  (2007).  

195   Su,  Y.  et  al.  Hypothalamic  leucine  metabolism  regulates  liver  glucose  production.  Diabetes  61,  85-­‐93,  doi:10.2337/db11-­‐0857  (2012).  

196   Tanaka,  K.,  Inoue,  S.,  Nagase,  H.,  Takamura,  Y.  &  Niijima,  A.  Amino  acid  sensors  sensitive  to  alanine  and  leucine  exist  in  the  hepato-­‐portal  system  in  the  rat.  J  Auton  Nerv  Syst  31,  41-­‐46  (1990).  

197   Niijima,  A.  &  Meguid,  M.  M.  An  electrophysiological  study  on  amino  acid  sensors  in  the  hepato-­‐portal  system  in  the  rat.  Obes  Res  3  Suppl  5,  741S-­‐745S  (1995).  

198   Hellier,  M.  D.,  Holdsworth,  C.  D.,  McColl,  I.  &  Perrett,  D.  Dipeptide  absorption  in  man.  Gut  13,  965-­‐969  (1972).  

199   Thamotharan,  M.,  Bawani,  S.  Z.,  Zhou,  X.  &  Adibi,  S.  A.  Mechanism  of  dipeptide  stimulation  of  its  own  transport  in  a  human  intestinal  cell  line.  Proc  Assoc  Am  Physicians  110,  361-­‐368  (1998).  

200   Anderson,  G.  H.  et  al.  Relation  between  estimates  of  cornstarch  digestibility  by  the  Englyst  in  vitro  method  and  glycemic  response,  subjective  appetite,  and  short-­‐term  food  intake  in  young  men.  Am  J  Clin  Nutr  91,  932-­‐939,  doi:10.3945/ajcn.2009.28443  (2010).  

201   Raybould,  H.  E.,  Zittel,  T.  T.,  Holzer,  H.  H.,  Lloyd,  K.  C.  &  Meyer,  J.  H.  Gastroduodenal  sensory  mechanisms  and  CCK  in  inhibition  of  gastric  emptying  in  response  to  a  meal.  Digestive  Diseases  &  Sciences  39,  41S-­‐43S  (1994).  

202   Rabinowitz,  D.,  Merimee,  T.  J.,  Maffezzoli,  R.  &  Burgess,  J.  A.  Patterns  of  hormonal  release  after  glucose,  protein,  and  glucose  plus  protein.  Lancet  2,  454-­‐456  (1966).  

203   Nakabayashi,  H.,  Nishizawa,  M.,  Nakagawa,  A.,  Takeda,  R.  &  Niijima,  A.  Vagal  hepatopancreatic  reflex  effect  evoked  by  intraportal  appearance  of  tGLP-­‐1.  Am  J  Physiol  271,  E808-­‐813  (1996).  

204   Schmid,  R.,  Schusdziarra,  V.,  Schulte-­‐Frohlinde,  E.,  Maier,  V.  &  Classen,  M.  Effect  of  CCK  on  insulin,  glucagon,  and  pancreatic  polypeptide  levels  in  humans.  Pancreas  4,  653-­‐661  (1989).  

205   Knauf,  C.  et  al.  Role  of  central  nervous  system  glucagon-­‐like  Peptide-­‐1  receptors  in  enteric  glucose  sensing.  Diabetes  57,  2603-­‐2612,  doi:10.2337/db07-­‐1788  (2008).  

206   Lebovitz,  H.  E.  et  al.  Gastric  electrical  stimulation  treatment  of  type  2  diabetes:  effects  of  implantation  versus  meal-­‐mediated  stimulation.  A  randomized  blinded  cross-­‐over  trial.  Physiol  Rep  3,  doi:10.14814/phy2.12456  (2015).  

207   Kondoh,  T.  &  Torii,  K.  Brain  activation  by  umami  substances  via  gustatory  and  visceral  signaling  pathways,  and  physiological  significance.  Biological  &  Pharmaceutical  Bulletin  31,  1827-­‐1832  (2008).  

Page 87: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science

78

208   Steinert,  R.  E.,  Landrock,  M.  F.,  Horowitz,  M.  &  Feinle-­‐Bisset,  C.  Effects  of  Intraduodenal  Infusions  of  L-­‐phenylalanine  and  L-­‐glutamine  on  Antropyloroduodenal  Motility  and  Plasma  Cholecystokinin  in  Healthy  Men.  Journal  of  Neurogastroenterology  &  Motility  21,  404-­‐413,  doi:10.5056/jnm14143  (2015).  

209   Cote,  C.  D.  et  al.  Resveratrol  activates  duodenal  Sirt1  to  reverse  insulin  resistance  in  rats  through  a  neuronal  network.  Nat  Med  21,  498-­‐505,  doi:10.1038/nm.3821  (2015).  

210   Hu,  Y.  et  al.  Targeted  disruption  of  peptide  transporter  Pept1  gene  in  mice  significantly  reduces  dipeptide  absorption  in  intestine.  Mol  Pharm  5,  1122-­‐1130  (2008).  

211   Arrieta-­‐Cruz,  I.,  Su,  Y.  &  Gutierrez-­‐Juarez,  R.  Suppression  of  Endogenous  Glucose  Production  by  Isoleucine  and  Valine  and  Impact  of  Diet  Composition.  Nutrients  8,  doi:10.3390/nu8020079  (2016).  

212   Stengel,  A.  et  al.  High-­‐protein  diet  selectively  reduces  fat  mass  and  improves  glucose  tolerance  in  Western-­‐type  diet-­‐induced  obese  rats.  American  Journal  of  Physiology:  Regulatory,  Integrative  &  Comparative  Physiology  305,  R582-­‐591,  doi:10.1152/ajpregu.00598.2012  (2013).  

213   Zhang,  Y.  et  al.  Increasing  dietary  leucine  intake  reduces  diet-­‐induced  obesity  and  improves  glucose  and  cholesterol  metabolism  in  mice  via  multimechanisms.  Diabetes  56,  1647-­‐1654,  doi:10.2337/db07-­‐0123  (2007).  

214   Nuttall,  F.  Q.,  Mooradian,  A.  D.,  Gannon,  M.  C.,  Billington,  C.  &  Krezowski,  P.  Effect  of  protein  ingestion  on  the  glucose  and  insulin  response  to  a  standardized  oral  glucose  load.  Diabetes  Care  7,  465-­‐470  (1984).  

215   Gannon,  M.  C.,  Nuttall,  F.  Q.,  Neil,  B.  J.  &  Westphal,  S.  A.  The  insulin  and  glucose  responses  to  meals  of  glucose  plus  various  proteins  in  type  II  diabetic  subjects.  Metabolism:  Clinical  &  Experimental  37,  1081-­‐1088  (1988).  

216   Manders,  R.  J.  et  al.  Co-­‐ingestion  of  a  protein  hydrolysate  and  amino  acid  mixture  with  carbohydrate  improves  plasma  glucose  disposal  in  patients  with  type  2  diabetes.  Am  J  Clin  Nutr  82,  76-­‐83  (2005).  

Page 88: Upper Small Intestinal Protein Sensing in the Regulation ... · Upper small intestinal protein sensing in the regulation of glucose homeostasis Sophie Claire Hamr Master of Science