cns hormonal and nutritional regulation of glucose … · 2017. 12. 19. · title: cns hormonal and...

150
CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE AND ENERGY HOMEOSTASIS by Mona Anna Abraham A dissertation submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Physiology University of Toronto © Copyright by Mona Anna Abraham (2017)

Upload: others

Post on 07-Jun-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

CNS HORMONAL AND NUTRITIONAL REGULATION OF

GLUCOSE AND ENERGY HOMEOSTASIS

by

Mona Anna Abraham

A dissertation submitted in conformity with the requirements for the degree of Doctor of Philosophy

Department of Physiology University of Toronto

© Copyright by Mona Anna Abraham (2017)

Page 2: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

ii

Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

Degree: Doctor of Philosophy

Department: Physiology, University of Toronto

Year of Convocation: 2017

GENERAL ABSTRACT

The mediobasal hypothalamus (MBH) and the dorsal vagal complex (DVC) regions contain a

leaky blood-brain barrier and therefore act as critical sites of action for circulating hormones and

nutrients on glucose and energy homeostasis. For example, hormones such as insulin, leptin and

glucagon and nutrients such as lipids, glucose and amino acids act in the MBH and DVC to

regulate hepatic glucose production, glucose tolerance, food intake and body weight, but the

underlying CNS sensory mechanisms in rodents and humans remain elusive. The current

dissertation unveils novel mechanisms for glucagon action in the MBH and glycine sensing in

the DVC in rats and mice that maintain glucose and energy homeostasis. In Study 1, we

discovered that KATP channel is necessary for MBH glucagon action to exert glucose control. In

Study 2, we found that pharmacological and molecular manipulation of glycine transporter 1 in

the DVC enhances glycine sensing and potently regulates glucose and energy homeostasis. In

conclusion, I have discovered novel mechanisms for glucagon and glycine sensing in the brain

that regulate glucose and energy homeostasis, and have unveiled CNS therapeutic targets for

diabetes and obesity.

Page 3: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

iii

Acknowledgments

Completing my doctoral work at the University of Toronto has been one of the most rewarding

and challenging experiences of my life. I’m extremely thankful to all the intellectual mentors and

colleagues who have guided and supported me through the last 5 years.

To my PhD supervisor, Dr. Tony Lam. I have greatly benefitted from his work ethic, keen

scientific insight, and ability to simplify complex questions. Sincerely, I appreciate everything he

has done for me.

To my committee: Drs. Adria Giacca, Richard Bazinet, Daniel Drucker. They have been

extremely generous with their time and always provided helpful suggestions, valuable advice,

and support.

To my current and past colleagues in the lab. Every result in this dissertation was accomplished

with the assistance of fellow lab-mates. Dr. Jessica Yue, Dr. Beatrice Filippi, Mary, Dr. Frank

Duca, Paige and I worked together on several different projects, and without their efforts my

doctoral work would have been far more difficult.

I gratefully acknowledge the support of the Banting and Best Diabetes Centre Fellowship and

Canadian Institutes for Health Research for their financial support to make this work possible.

I’m extremely thankful for my grandparents and parents who instilled in me a love of learning,

and lastly, I’m grateful most of all to God, who opens doors of destiny.

Page 4: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

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

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

List of Publications that contributed to this Thesis ..........................................................................x

Chapter 1 Introduction .....................................................................................................................1

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

1.2 The role of the hypothalamus in the regulation of glucose and energy homeostasis ............3

1.1.1 Hormonal action .......................................................................................................4

1.1.2 Nutrient sensing .....................................................................................................13

1.2 The role of the dorsal vagal complex in the regulation of glucose and energy homeostasis ........................................................................................................................18

1.2.1 Hormonal action .....................................................................................................19

1.2.2 Nutrient sensing .....................................................................................................21

1.3 The role of glial cells in the regulation of glucose and energy homeostasis. ......................25

1. 4 Summary of Introduction/Rationale of Study 1 and Study 2 .............................................26

Chapter 2 ........................................................................................................................................28

Study 1 ...........................................................................................................................................28

2.1 Abstract ...............................................................................................................................29

2.2 Introduction .........................................................................................................................30

2.3 Materials and Methods ........................................................................................................32

2.3.1 Animal Preparation ...................................................................................................32

2.3.2 Adenovirus injection .................................................................................................32

2.3.3 Pancreatic-euglycemic clamp ...................................................................................32

2.3.4 Western blotting for phosphorylated ACC ...............................................................33

Page 5: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

v

2.3.5 PKC-δ activity assay .................................................................................................34

2.3.6 Biochemical Analysis ...............................................................................................35

2.3.7 Statistical Analysis ....................................................................................................35

2.4 Results .................................................................................................................................35

2.4.1 Role of MBH AMPK in glucagon action .................................................................35

2.4.2 Role of MBH PKC-δ in glucagon action ..................................................................37

2.4.3 Role of MBH KATP channels in glucagon action ......................................................37

2.5 Discussion ...........................................................................................................................38

Chapter 3 ........................................................................................................................................45

Study 2 ...........................................................................................................................................45

3.1 Abstract ...............................................................................................................................46

3.2 Introduction .........................................................................................................................47

3.3 Materials and Methods ........................................................................................................49

3.3.1 Animal preparation and surgical procedures ............................................................49

3.3.2 Intravenous glucose tolerance test ............................................................................50

3.3.3 DVC treatments ........................................................................................................51

3.3.4 Pancreatic basal insulin euglycemic clamp in rats ....................................................52

3.3.5 Hepatic branch vagotomy in rats ..............................................................................53

3.3.6 Microdialysis .............................................................................................................54

3.3.7 DVC virus injection ..................................................................................................54

3.3.8 Brain tissue sampling in rats .....................................................................................55

3.3.9 Microdialysate sample glycine analysis ....................................................................55

3.3.10 Acute (3-d) and chronic (28-d) high-fat feeding in rats ..........................................56

3.3.11 Intravenous ALX infusion clamps ..........................................................................56

3.3.12 Induction of experimental type 2 diabetes ..............................................................57

3.3.13 Fasting-refeeding experiments ................................................................................57

Page 6: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

vi

3.3.14 Pancreatic basal insulin euglycemic clamp in mice ................................................57

3.3.15 Western blot analyses .............................................................................................58

3.3.16 Biochemical analysis ..............................................................................................59

3.3.17 Calculations and statistics .......................................................................................59

3.4 Results .................................................................................................................................60

3.4.1 Gluco-regulation by DVC GlyT1 inhibition in healthy rodents ...............................60

3.4.2 Anti-diabetic effect of DVC GlyT1 inhibition ..........................................................63

3.4.3 Metabolic benefits of DVC GlyT1 inhibition in obesity ..........................................65

3.4.4 DVC GlyT1 inhibition regulates energy balance ......................................................66

3.5 Discussion ...........................................................................................................................68

Chapter 4 Summary, Discussion and Future Directions ................................................................86

4.1 Summary .............................................................................................................................86

4.2 Discussion ...........................................................................................................................89

4.3 Limitations and Future directions .......................................................................................98

References ....................................................................................................................................103

Page 7: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

vii

List of Tables

Table 2. 1 Plasma insulin, glucagon and glucose concentrations during basal and clamp

conditions. Data are means ±SEM. ............................................................................................... 44

Page 8: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

viii

List of Figures

Figure 2. 1 Schematic representation of the working hypothesis. ................................................ 40

Figure 2. 2 Role of MBH AMPK in glucagon action. .................................................................. 41

Figure 2. 3 Role of MBH PKC- δ in glucagon action. .................................................................. 42

Figure 2. 4 Role of MBH KATP channels in glucagon action. ....................................................... 43

Figure 3. 1 Schematic representation of the working hypothesis. ................................................ 72

Figure 3. 2 Chemical inhibition of DVC GlyT1 regulates glucose homeostasis in healthy rats. . 73

Figure 3. 3 Molecular inhibition of DVC GlyT1 regulates glucose homeostasis in healthy rats. 74

Figure 3. 4 DVC and iv infusion of ALX regulates glucose homeostasis in 3d-HFD rats. .......... 75

Figure 3. 5 Inhibition of DVC GlyT1 regulates glucose homeostasis in diabetic rats. ................ 76

Figure 3. 6 Chemical inhibition of DVC GlyT1 regulates glucose homeostasis in obese rats. .... 77

Figure 3. 7 Molecular inhibition of DVC GlyT1 regulates metabolic homeostasis in obese rats. 78

Figure 3. 8 Chemical and molecular inhibition of DVC GlyT1 regulate energy balance. ........... 79

Supplementary Figure 3. 1 Metabolic effects of chemical inhibition of DVC GlyT1 in healthy

rats. ................................................................................................................................................ 80

Supplementary Figure 3. 2 Metabolic effects of chemical inhibition of GlyT1 in the 4th ventricle

of mice and in the DVC of hepatic vagotomized rats. .................................................................. 81

Supplementary Figure 3. 3 Brain regions included in the GlyT1 protein analysis. ...................... 82

Page 9: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

ix

Supplementary Figure 3. 4 Metabolic effects of molecular inhibition of DVC GlyT1 in healthy

rats. ................................................................................................................................................ 83

Supplementary Figure 3. 5 Metabolic effects of DVC and iv infusion of ALX in 3d-HFD rats. 84

Supplementary Figure 3. 6 Metabolic effects of chemical and molecular inhibition of DVC

GlyT1 in obese rats. ...................................................................................................................... 85

Figure 4. 1 Summary of Study 1 and Study 2. .............................................................................. 88

Page 10: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

x

List of Publications that contributed to this Thesis

Study 1:

Abraham, M.A., Yue, J.T., LaPierre, M.P., Rutter, G.A., Light, P.E., Filippi, B.M., and Lam, T.K. (2014). Hypothalamic glucagon signals through the KATP channels to regulate glucose production. Mol Metab 3, 202-208.

Study 2:

Abraham, M.A.*, Yue, J.T.*, Bauer, P.V., LaPierre, M.P., Wang, P., Duca, F.A., Filippi, B.M., Chan, O., and Lam, T.K. (2016). Inhibition of glycine transporter-1 in the dorsal vagal complex improves metabolic homeostasis in diabetes and obesity. Nat Commun 7, 13501. *Equal contribution

Review Papers:

Abraham, M.A., and Lam, T.K. (2016). Glucagon action in the brain. Diabetologia 59, 1367-1371.

Abraham, M.A., Filippi, B.M., Kang, G.M., Kim, M.S., and Lam, T.K. (2014). Insulin action in the hypothalamus and dorsal vagal complex. Exp. Physiol. 99, 1104-1109.

LaPierre, M.P.*, Abraham, M.A.*, Filippi, B.M., Yue, J.T., and Lam, T.K. (2014). Glucagon and lipid signaling in the hypothalamus. Mamm. Genome 25, 434-441. *Equal contribution

Filippi, B.M.*, Abraham, M.A.*, Yue, J.T., and Lam, T.K. (2013). Insulin and glucagon signaling in the central nervous system. Rev. Endocr. Metab. Disord. 14, 365-375. *Equal contribution

Other studies contributing to the completion of this dissertation:

LaPierre, M.P., Abraham, M.A., Yue, J.T., Filippi, B.M., and Lam, T.K. (2015). 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.

Filippi, B.M., Bassiri, A., Abraham, M.A., Duca, F.A., Yue, J.T., and Lam, T.K. (2014). Insulin signals through the dorsal vagal complex to regulate energy balance. Diabetes 63, 892-899.

Page 11: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

1

Chapter 1 Introduction

1.1 Diabetes and Obesity In 1878, the French physiologist Claude Bernard described that ‘All the vital mechanisms,

however varied they may be, have only one object, that of preserving constant the conditions of

the internal environment which make a free and independent life possible’.1,2 This maintenance

of an internal state defended against changes is particularly true for the regulation of blood

glucose levels and body weight. Low levels of plasma glucose would deplete the brain of its only

energy source, leading to seizures, unconsciousness, and death. On the other hand, sustained

elevation of blood glucose can also be fatal as it causes diabetes and associated complications.

Therefore, it is vital for the body to maintain blood glucose at a fairly narrow range of 4-7

mmol/L. Glucose homeostasis is that process of maintaining blood glucose at a steady-state

level. Similarly, in a healthy body, body weight and body fat is also defended against acute

perturbations under the influence of a tightly regulated homeostatic process called energy

homeostasis. For instance, rats when subjected to caloric restriction display significant weight

loss but when returned to free access of food, quickly rebound regaining their initial body weight

within days3,4. Remarkably, this precise nature of body weight regulation is also true in humans,

both lean and obese. In fact, the literature documents the weight gain rate among obese men

(0.04 kg·BW/year) is slower than men with no history of obesity (0.18 kg·BW /year)5.

However, despite these robust homeostatic systems, Type 2 diabetes and obesity are the

two most challenging public health concerns of the 21st century. Where the global prevalence of

diabetes was 9.3% in 2015, this number is predicted to increase to 12.1 % by 20256. Coincident

with this diabetes epidemic, the prevalence rates of obesity has also been escalating, with about

Page 12: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

2

13% of the world’s adult population obese in 20147. People affected by diabetes and obesity are

predisposed to developing cardiovascular diseases, and cancer8, which in turn reduce their life

expectancy and cost heavily in health care expenses. In Canada alone, the economic burden of

diabetes, which was approximated at $12.2 billion in 2010, is projected to increase by another

$4.7 billion by 20209. Meanwhile, the annual economic burden of obesity in Canada ranges from

$ 4.6 billion to $7.1 billion10. Given the survival and financial crisis caused by these diseases and

the expected rise in the number of affected individuals, the need for therapeutic interventions

aimed at combating the diabetes and obesity epidemic is more than ever today.

In this regard, considerable advances have been made scientifically in understanding the

different mechanisms that provide effective feedback to regulate glucose and energy

homeostasis. It is now recognized that the central nervous system (CNS) plays a critical role in

coordinating and integrating various components of glucose and energy regulation. In particular,

hormonal and nutrient signals from the periphery, relaying the body’s energy status, are detected

by the brain and integrated in CNS pathways to maintain constant blood glucose levels and body

weight stability. It follows that Type 2 diabetes and obesity develops as a result of dysregulation

in the ability of the CNS hormone and sensing pathways to appropriately couple the body’s

energy needs with nutrient intake and endogenous nutrient output. As such, delineating the

mechanisms of CNS hormonal signaling and/or nutrient sensing is vital in understanding and

identifying potential molecular targets to therapeutically restore regulation of feeding behaviour

and glucose homeostasis. Till date, tremendous progress has been made to elucidate the

molecular and cellular pathways, primarily within the hypothalamus and hindbrain, comprising

hormonal action and nutrient sensing circuits (which will be reviewed as follows).

The goal of this dissertation is to characterize novel CNS mechanisms of hormonal

signaling and nutrient sensing involved in the control of glucose and energy balance, thereby

Page 13: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

3

unveiling potential new therapeutic targets and experimental approaches to improve metabolic

control in diabetes and obesity.

1.2 The role of the hypothalamus in the regulation of glucose and energy homeostasis Of the different anatomical regions in the brain, the hypothalamus in particular is a privileged

CNS site to sense and integrate peripheral signals to regulate metabolic homeostasis. The median

eminence located at the mediobasal hypothalamus (MBH) and adjacent to the arcuate nucleus

(ARC) is a circumventricular organ, which is lined by fenestrated brain endothelium. This

permits circulating molecules to traverse past the blood brain barrier (BBB) and access the

hypothalamic ARC. There are two sets of first order neurons in the ARC, on which peripheral

metabolic hormones such as insulin, leptin, and glucagon and nutrients such as fatty acids and

glucose act: 1) the neurons that produce the anorexigenic (appetite suppressing) neuropeptides,

pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART), and

2) the neurons that produce the orexigenic (appetite promoting) neuropeptides, agouti-related

peptide (AgRP) and neuropeptide Y (NPY)11. Projections from these first order neurons to

second order neurons in other hypothalamic areas such as the paraventricular hypothalamus,

lateral hypothalamus, and ventromedial hypothalamus or extrahypothalamic areas including the

nucleus tractus solitarius ultimately influence peripheral metabolism.

Page 14: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

4

1.1.1 Hormonal action

Insulin

Insulin is the principal metabolic hormone that controls blood glucose by promoting glucose

uptake into muscle and adipose tissues, and suppressing glucose production by the liver. It is

now well-established that the brain is an important insulin-sensitive organ that has a

physiological role in the regulation of glucose homeostasis as well as energy balance. Pioneering

experiments by Porte and group revealed that intracerebroventricular (icv) injection of insulin

stimulated insulin secretion from the pancreas of dogs12 and reduced body weight and intake of

food in baboons13. Similarly, intranasal administration of insulin also led to decreased plasma

glucose in circulation in dogs14 and rhesus monkeys15. Studies in the past decade have illustrated

that the hypothalamus specifically is an important CNS site of insulin action for metabolic

regulation. Obici et al. demonstrated that insulin receptors (IR) within the hypothalamus have a

physiological role in the regulation of food intake, fat mass and hepatic insulin action. They

showed that rats treated with injection of IR antagonists into the third cerebral ventricle (icv-3) to

block hypothalamic insulin signalling, failed to suppress hepatic glucose production during

hyperinsulinemic clamp studies as well as displayed hyperphagia and increased fat mass16.

Subsequent studies revealed that hypothalamic insulin signals via the insulin receptor substrate-

phosphatidylinositol 3-kinase (IRS-PI3K) pathway17. Hypothalamic overexpression of either

IRS-2 or protein kinase B (PKB, a main downstream signalling molecule of PI3K action

enhanced the ability of peripheral insulin treatment to lower glucose by approximately 2-fold in

rats with uncontrolled diabetes induced by streptozotocin (STZ) 17. Moreover, activation of ATP-

sensitive potassium (KATP channels) is also required for hypothalamic insulin signaling. Insulin

stimulates KATP channel activity in hypothalamic glucose-sensing neurons in lean rats, but not in

obese rats nor in the presence of the KATP channel inhibitor tolbutamide2. Likewise, in the

Page 15: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

5

presence of icv-3 infusion of the KATP channel inhibitor glibenclamide, central insulin failed to

lower glucose production and regulate peripheral glucose fluxes18. Indeed, the hepatic vagus

nerve transmit the brain-liver communication of central insulin action and, at the level of the

liver, the hepatic interleukin-6 (IL-6)/STAT3 signaling mediates the effect of brain insulin to

lower hepatic glucose production19. A recent study has proposed inhibition of the α7-nicotinic

acetylcholine receptor (nAchR) contributes to the effect of central insulin to activate hepatic IL-

6/STAT3 and regulate gluconeogenic responses20. The exact mechanism underlying this brain-

liver axis of insulin action remains unclear and merits investigation.

Regarding the neuronal circuitry, AgRP neurons play a major role in insulin’s ability to

lower glucose production21,22. In specifically targeted AgRP-neuron insulin receptor (IR)

knockout mice, insulin failed to suppress glucose production during hyperinsulinemic clamps21.

On the other hand, in L1 mice (mice that have ~90 % reduction of IR levels in the ARC and

which have hyperinsulinemia and impaired regulation of glucose production23), the restoration of

insulin signaling in AgRP neurons was sufficient for central insulin to lower glucose

production22.

Importantly, overnutrition appears to affect the ability of central insulin to lower food

intake and glucose production. Obese rats develop insulin resistance in the brain which prevents

an insulin-dependent decrease of food intake24. High-fat diet (HFD) feeding induces the

expression of several pro-inflammatory cytokines and inflammatory responsive proteins in the

hypothalamus; this together with the accumulation of pro-inflammatory lipids increases local

inflammation in the hypothalamus and impairs the anorectic effect of insulin25,26 inducing

hyperphagia and consequently, body weight gain. Recent evidence suggested that switching to a

low-fat diet reversed hypothalamic insulin resistance caused by diet-induced obesity and restored

the anorectic effect of hypothalamic insulin27. Similarly, HFD feeding for 3 days (3d HFD) also

Page 16: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

6

blunts the glucose production-lowering effect of hypothalamic insulin28. This is associated with

the activation of hypothalamic S6 kinase (S6K), which phosphorylates insulin receptor substrate

(IRS) adaptor proteins to create a negative feedback that shuts off insulin signaling28. Further,

overfeeding-induced endoplasmic reticulum (ER) stress could also prevent the ability of insulin

to lower glucose production, since icv-3 treatment with the ER stress- inducer, thapsigargin,

prevented the ability of central insulin to lower blood glucose levels29. Taken together, these

studies associate hypothalamic insulin resistance to diet-induced dysregulation of glucose and

energy balance.

Leptin

Like insulin, leptin is another hormone that has high relevance in diabetes and obesity. Leptin

circulates in the plasma in proportion to body fat stores, enters the brain and interacts with its

receptors to regulate food intake, body weight, and glucose homeostasis30. It is now evident that

the metabolic effects of leptin are likely mediated through the brain. In both rats31 and mice32, icv

administered leptin show similar metabolic effects as intravenously (iv) administered leptin. For

instance, low-dose icv leptin recapitulates the effects of iv leptin in reversing the insulin

resistance and diabetic phenotype of leptin deficient, ob/ob33 and lipodystrophic mice34.

Similarly, icv leptin normalizes hyperglycemia in STZ-induced Type 1 diabetic rats whereas iv

leptin at the same dose is unable to do so35,36. Notably, the hypothalamus per se can regulate

leptin-glucose control as evident by Coppari et al.’ and Morton et al.’s studies where selective

restoration of leptin receptors in the hypothalamus normalizes blood glucose levels of the obese,

hyperinsulinemic and severely diabetic leptin receptor (LepRb)-null mice37 and rats38,

respectively. Consistently, injection of leptin specifically into the ARC leads to reduced food

intake and body weight gain in rats39 and icv administration of leptin in ob/ob mice attenuates

Page 17: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

7

obesity37. The STAT3 pathway plays a critical role in central leptin’s regulation of energy

homeostasis while there is some controversy over its role in glucose regulation. Mice with a

whole-body disruption of LepRb à STAT3 signaling (s/s mice) are hyperphagic and obese with

reduced energy expenditure, but less hyperglycemic compared to LepRb deficient, db/db

mice40,41. In contrast, mice with neural-specific ablation of STAT3, in addition to being

hyperphagic and obese, show impaired glucose tolerance and hyperinsulinemia to the same

extent as db/db mice42. Mice with specific deletion of STAT3 from AgRP neurons display

obesity, increased NPY expression, but did not differ in glucose levels compared to control

mice43. STAT3 deletion from POMC neurons increases adiposity, but the effect is milder than

that observed for the AgRP-specific knockout, suggesting a greater role for STAT3 in leptin’s

regulation of energy homeostasis in AgRP neurons than in POMC cells44. It should be noted,

evidence also suggests most of the anorectic actions of leptin is dependent on the inhibition of

GABA neurons outside of the ARC causing a reduced reduced inhibitory tone to POMC

neurons45. Importantly, STAT3 activation is a functional determinant for hypothalamic leptin’s

regulation of glucose homeostasis, as pharmacological and molecular inhibition of STAT3 in the

third ventricle abolishes the ability of MBH leptin to lower glucose production in rodents fed a

high fat diet46. The extracellular signal–regulated kinase (ERK), a member of the mitogen-

activated protein kinase (MAPK) family, is another downstream pathway of the leptin receptor.

The ERK pathway appears to modulate brain leptin’s control on energy and glucose balance.

Chemical blockade of hypothalamic ERK1/2 negated the food intake- and weight-reducing

effects of icv leptin47, while genetic blockade of ERK signaling in POMC neurons negated the

glucose lowering effects of iv leptin with only a modest effect on food intake48. In addition to

STAT3 and ERK, the LepRb receptor also signals through the PI3K pathway. Pharmacological

blockade of PI3K activity in the hypothalamus abolishes the ability of leptin to lower food

Page 18: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

8

intake49. Interestingly, chemical blockade of PI3K prevents insulin-stimulated suppression of

glucose production otherwise seen upon restoration of LepRb in Koletsky fak/fak rats38. The PI3K

pathway is also implicated in leptin-induced depolarization of POMC neurons50, whose role in

regulating normoglycemia is also dependent on PI3K signaling51. Notably, like insulin, leptin

also activates KATP channels to hyperpolarize hypothalamic neurons obtained from lean rats, but

not in obese rats52.

Furthermore, AMP-activated kinase (AMPK) has also been identified as an important

player in leptin's hypothalamic intracellular signaling cascade. Exogenous administration of

leptin inhibits AMPK in the ARC, while constitutive activity of AMPK in the ARC abolishes

leptin's effect on anorexia, thereby indicating that inhibition of hypothalamic AMPK is necessary

for leptin's regulation on energy homeostasis53. Subsequent studies have demonstrated that

activation of acetyl-CoA carboxylase (ACC), the downstream target of AMPK inhibition, further

mediates hypothalamic leptin’s control on food intake and body weight54. Consistently, increased

levels of malonly-CoA, a direct product of ACC activation and an important mediator in the

lipid-sensing pathway, are also required for hypothalamic leptin-induced reduction in food

intake55.

Importantly, hypothalamic leptin resistance occurs in an obese state since icv leptin fails

to induce anorexia in diet-induced obese (DIO) rats56, contributing to the notion that

hypothalamic dysregulation of hormonal action leads to obesity. Surprisingly, the effect of icv

leptin to decrease glucose production is intact in both 3 d HFD-fed as well as STZ diabetic

rats35,57. It is possible, that these differences in CNS leptin resistance could be due to specific

signaling pathways activated by leptin. Where activation of hypothalamic STAT3 is required for

MBH leptin’s effect on glucose production46, HFD impaired the anorectic effects of leptin

despite intact STAT3 signaling58.

Page 19: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

9

Ghrelin

The peripherally derived-orexigenic hormone, ghrelin, is another critical regulator of metabolism

modulated through hypothalamic pathways. Ghrelin regulates feeding, and glucose homeostasis

via ghrelin receptors abundantly expressed in the ARC AgRP/NPY neurons 59,60. Indeed, direct

ghrelin administration into the ARC increases food intake, induces NPY and AgRP mRNA

expression in the ARC and its orexigenic effect on food intake was negated in the presence of icv

administration of anti-NPY or anti-AgRP antibodies 61. Further, icv ghrelin infusion has also

been reported to improve glucose tolerance in pair-fed mice, compared to icv ghrelin ad libitum

mice and icv vehicle treated mice 62. While the molecular players for the glucoregulatory action

of icv ghrelin remain largely unknown, the key pathway for the orexigenic effect of ghrelin

includes AMPK since icv ghrelin increases AMPK phosphorylation in the hypothalamus63 and

molecular inhibition of AMPK markedly blunted the feeding effect of ghrelin64. Consistently,

ghrelin-induced AMPK phosphorylation decreased ACC levels, accompanied by reduced

formation of malonyl-CoA levels and enhanced CPT-1 activity, which in turn leads to

mitochondrial fatty oxidation64. This hypothalamic fatty oxidation pathway increases reactive

oxygen species (ROS), activating uncoupling protein 2 (UCP2), which mediates sustained firing

of AgRP/NPY neurons, and increased GABAergic inhibitory tone to POMC neurons –

ultimately increasing feeding behaviour65. Whether an AMPK mediated pathway is involved in

the glucose regulating effects of icv ghrelin remains to be identified.

Glucagon

The second principal pancreatic hormone, glucagon, is well known for its catabolic effect on fuel

metabolism. Once released into the circulation, glucagon stimulates hepatic glucose output

through the breakdown of glycogen, inhibition of glycogen synthesis and stimulation of

Page 20: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

10

gluconeogenesis66. Glucagon exerts these biological effects by binding to G-protein coupled

glucagon receptors in the liver and activating the cyclic AMP-protein kinase A (cAMP-PKA)

signaling pathway67,68. In addition to the liver, glucagon receptors (GR) are also expressed in the

brain69,70, and the findings that other key components of the glucagon signaling pathway

including cAMP and PKA are present in the hypothalamus71, and that glucagon can be

transported from blood into the cerebrospinal fluid72 and into the brain to affect hypothalamic

neuronal activity73 suggests that circulating glucagon passes through the BBB and could exert

part of its biological effects through a hypothalamic pathway.

In fact, multiple studies have documented the ability of icv glucagon administration to

modulate peripheral glucose levels in different species of animals74-76. Most intriguing, however,

is the earliest study in dogs where a high dose icv injection of 10 ng of glucagon transiently

produced hypoglycemia followed by hyperglycemia77. The hypoglycemic effect was abolished in

vagotomised dogs, suggesting the involvement of a brain-liver axis in the glucose-lowering

effect of central glucagon; whereas pancreatectomy prevented the hyperglycemic effect,

attributing a pancreatic role to the rise of glucose from icv glucagon injections 77,78. Given the

non-specific administration of icv glucagon and the use of relatively high glucagon dosage in

these experiments, more recent studies have administered much lower doses of glucagon

specifically into the MBH and evaluated whether MBH glucagon action accounts for the

glucose-lowering effect or hyperglycemic effect of icv injections in the early dog studies.

Indeed, direct infusion of glucagon into the MBH actually lowered hepatic glucose

production in a pancreatic basal-insulin euglycemic clamp condition79. This effect of MBH

glucagon required the activation of MBH GRs, PKA signalling, and intact vagal nerves, since

their ablation negated the metabolic effects of MBH glucagon infusion. The inhibitory metabolic

effects of central glucagon were also confirmed in another rodent model; central glucagon, but

Page 21: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

11

not saline, administration in C57BL/6 normal mice during a pancreatic clamp increased the

glucose infusion rate required to maintain euglycemia, owing to a suppression in glucose

production79. Furthermore, a genetic knockout model involving mice that lack functional

glucagon receptors (Gcgr−/−) also confirmed the role of central glucagon receptors in mediating

the central glucagon effect since central infusion of glucagon in these mice failed to increase the

glucose infusion rate and lower glucose production compared to Gcgr+/+ control mice. Taken

together, these data illustrate that hypothalamic/central glucagon receptor signaling regulates

glucose production in both rats and mice and is possibly the mechanism responsible for the

initial hypoglycemic effect of central glucagon injections seen in dogs as previously reported77,78.

The next key question was whether the hypothalamic action of glucagon to lower glucose

production is physiologically relevant—does the MBH mediate the effect of circulating glucagon

to regulate glucose production? Despite the classical view of glucagon to be a gluco-stimulatory

hormone, continuous iv infusions of glucagon only transiently stimulate glucose production, with

a subsequent decline back to baseline after approximately 40 min in both dogs80 and humans81,82.

Interestingly, Mighiu et al. demonstrated blocking MBH glucagon signaling prolonged the

gluco-stimulatory effect of iv glucagon, thereby ascribing the short-lived elevation of glucose

production and glycemia induced by circulating glucagon to, and illustrates the physiological

relevance of, MBH glucagon action.

On the contrary, in diabetic83 and obese84 conditions chronic hyperglucagonemia is

associated with increased glucose production and blood glucose levels. For instance, in diabetic

rodents, antagonism of glucagon action by iv injection of either glucagon receptor antagonist

THG85 or monoclonal glucagon antibody86 alleviates hyperglycemia, suggesting that glucagon

leads to a sustained increase in blood glucose levels in diabetes. Therefore, given that glucagon’s

gluco-stimulatory effect is evanescent in normal physiology whereas it is sustained in diabetes, it

Page 22: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

12

is likely that the absence of glucagon’s transient effect in diabetes and obesity may be attributed

to hypothalamic glucagon resistance. In fact, 3 d HFD feeding has been shown to disrupt MBH

glucagon action to lower glucose production79. This indicates that hypothalamic glucagon

resistance indeed exists in a pathological state. However, this resistance could be reversed by

direct activation of hypothalamic PKA, which implies, at this preliminary stage, that the

hypothalamic glucagon signaling defect lies upstream of PKA. Investigating what lies

downstream of PKA in MBH glucagon signaling thus becomes of scientific interest, especially

from a therapeutic standpoint to identify targets that could potentially enhance or restore

hypothalamic glucagon action in diabetes and/or obesity. Interestingly, PKA decreases AMPK

activation in adipocytes87 and hypothalamic cell lines88. The aim of Study 1 in this dissertation is

to further uncover the molecular pathways that lie downstream of MBH glucagon-PKA signaling

in the regulation of glucose homeostasis.

Notably, the effects of MBH glucagon are not just limited to glucose regulation. The

earliest evidence that hypothalamic glucagon action regulates food intake comes from Inokouchi

et al.’s study which showed that icv-3 injection of glucagon significantly suppressed feeding in

rats more potently than the effect observed with peripheral administration89. Similar anorexigenic

effects of central glucagon were also demonstrated in chicks90 and sheep91. Notably, one study

showed that central glucagon administration did not alter food intake in baboons13; however, the

glucagon dose used in this study was 10 times lower than that what was used to achieve feeding

inhibition in rats89. Although iv infusion of glucagon reduces meal size and suppresses appetite

in humans92, whether this is due to an effect of glucagon in the hypothalamus is not clear.

In summary, peripherally derived hormones such as insulin, leptin and glucagon have

been documented to act on their respective receptors in the hypothalamus to regulate glucose and

energy homeostasis via seemingly distinct signaling pathways, but perhaps also converging at a

Page 23: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

13

common downstream effector (s). Notably, the activation of KATP channels presents as a likely

candidate, as not only is it necessary for the action of MBH insulin and leptin, but also in MBH

nutrient sensing mechanisms which be reviewed as follows.

1.1.2 Nutrient sensing

In addition to integrating hormonal signals, the hypothalamus is also a sensor of nutritional

inputs including those of fatty acids, glucose and amino acid leucine to modulate glucose

homeostasis and energy balance.

Fatty acids

Although fatty acids per se are not an energy source for the brain, lipid sensing mechanisms have

emerged as important indicators of nutrient availability to the CNS to regulate glucose and

energy homeostasis. In fact, icv-3 infusion of the long chain fatty acid (LCFA), oleic acid during

basal insulin clamping leads to a significant inhibition in the rate of hepatic glucose production,

and during an unclamped postprandial setting, effectively lowers plasma glucose levels93. The

hypothalamus can also sense circulating fatty acids to regulate glucose homeostasis94.

Circulating fatty acids can passively diffuse across the BBB95. At the same time, facilitated

transport of lipids can also occur via plasma membrane fatty acid transport proteins and fatty

acid translocases/CD36 96-98. Upon entry into cells, LCFAs are subsequently esterified to LCFA-

CoA by acyl-CoA synthetase (ACS) which prevents diffusion out of neurons. In fact, the

accumulation of intracellular LCFA-CoA is necessary to activate hypothalamic lipid sensing

pathway and lower glucose production as direct inhibition of MBH ACS negates the ability of

circulating LCFA to restrain glucose production94. Intracellular LCFA-CoA levels are also

dependent on carnitine palmitoyltransferase-1 (CPT1) activity, which regulates the uptake of

Page 24: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

14

LCFA-CoA into mitochondria for β-oxidation. In line with this, genetic inhibition of

hypothalamic CPT1 expression increased LCFA-CoA levels and suppressed glucose

production99. Therefore, factors regulating CPT1 activity are also key components of the lipid

sensing pathway. One such factor is malonyl-CoA which inhibits CPT1 to prevent lipid entry

into the mitochondria100. Thus, malonyl-CoA, which is a product of glucose metabolism and

therefore, abundant during nutrient availability, promotes the accumulation of LCFA-CoA to

lower glucose production and circumvent circulating nutrient excess101. AMPK is another

important negative regulator of hypothalamic lipid sensing. As described earlier, AMPK inhibits

ACC activity consequently preventing the formation of malonyl-CoA from acetyl-CoA and this

abolishes the glucose-lowering actions of MBH lipid sensing mechanisms102.

As with MBH insulin and leptin infusions, hypothalamic KATP channels are also activated

by hypothalamic lipid sensing, and are necessary for hypothalamic LCFA to suppress glucose

production103. Consistent with this, blockade of hypothalamic KATP channels by both

pharmacological and genetic approaches abolished the glucose production-lowering effect

achieved by physiological increases in circulating LCFAs94. The activation of KATP channels

subsequently signals through the hepatic vagus nerve to communicate with the liver as hepatic

vagotomy negates the hypothalamic lipid sensing mechanism to inhibit the hepatic expression of

gluconeogenic enzymes and glucose production104. Further, hypothalamic protein kinase C

(PKC) was later shown to be an important mediator in the hypothalamic lipid-KATP signaling to

regulate glucose homeostasis. While direct hypothalamic infusion of the PKC activator, OAG

lowered glucose production, this effect was eliminated by co-infusion with rottlerin, an inhibitor

specifically targeting the PKC-δ isoform as well as by blocking hypothalamic KATP channel105.

Lastly, inhibition of hypothalamic PKC negated the effect of hypothalamic lipids to lower

glucose production105.

Page 25: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

15

Hypothalamic fatty acid sensing also modulates energy balance. Rats that received a

bolus of icv-3 oleic acid 1 hour prior to the dark cycle showed a significant reduction in food

intake- an effect that lasted for 2 days103. Further, the reduced expression of hypothalamic NPY

mRNA levels observed in rats treated with icv-3 oleic acid vs vehicle-bolus, likely represents the

underlying mechanism behind the anorectic effect of icv oleic acid.

Similar to how various hypothalamic hormonal signaling mechanisms are disrupted in

models of high fat feeding, 3 d HFD feeding disrupts the ability of icv-3 oleic acid infusion to

lower glucose production93. Interestingly, genetic inhibition of hypothalamic CPT1 expression

by icv infusion of the ribozyme CPT1A-Ribo restored LCFA-CoA levels and rescued the gluco-

regulatory effect of hypothalamic lipid sensing in the 3d HFD fed rats106. Moreover, direct

activation of MBH PKC-δ was also able to restore the suppression of glucose production during

3d HFD105. These data are indicative that the HFD-induced defect in lipid sensing lies at the

level of CPT1 and LCFA-CoA accumulation, while the downstream signaling pathway of

LCFA-CoA remains intact after a 3d HFD. Notably, icv oleic acid also failed to suppress food

intake in rats fed 3d HFD103. This proves compelling evidence that hypothalamic lipid sensing

can become defective and is of therapeutic relevance in diabetes and obesity.

Because of the involvement of 1) AMPK and KATP channels in the hypothalamic action of

leptin and/or insulin 2) AMPK, PKC- δ, and KATP channels in hypothalamic lipid sensing

mechanism, and 3) that PKA inhibits AMPK, we tested in study 1, the role these respective

intracellular signaling effectors in MBH glucagon action.

Page 26: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

16

Glucose and lactate

Interestingly, the hypothalamic lipid sensing axis illustrated as above is positioned to mediate

hypothalamic glucose sensing. In addition to being the primary fuel for the brain, glucose

sensing in the hypothalamus plays an important role in the physiological control of blood glucose

and feeding regulation. It was Lam et al. who first provided evidence that during a pancreatic

clamp, a direct infusion of glucose or lactate, a byproduct of glucose metabolism, into the MBH

leads to an inhibition of hepatic glucose production, which, like MBH insulin and leptin action,

also requires activation of hypothalamic KATP channels107. Notably, when hypothalamic lipid

sensing axis is blocked via activation of MBH AMPK, it abolished the effect of MBH glucose

and lactate sensing to lower glucose production102. It is not surprising, increase in hypothalamic

glucose flux lowers food intake, and this anorectic effect is also mediated by inhibiting AMPK

and activating ACC activity, which in turn elevates hypothalamic malonyl–CoA108. Taken

together, AMPK-mediated lipid sensing appears to be a critical intermediary mechanism by

which hypothalamic glucose sensing works to regulate glucose production and feeding behavior.

The literature documents that defective hypothalamic glucose sensing maybe an

important pathogenic component of diabetes and obesity. This is particularly true for MBH

glucose sensing in uncontrolled diabetes. Chari et al. showed in rats with STZ-induced

uncontrolled diabetes, icv-3 administration of glucose was unable to suppress the rate of glucose

production, and they demonstrated this defect was due to a glucotoxic insult induced by

sustained hyperglycemia109. Intriguingly, MBH lactate administration, using the same dose as for

normal rats, retained its ability to inhibit glucose production in the STZ-induced diabetic rats, as

well as in experimentally induced hypoinsulinemic rats, and in rats fed a 3d HFD110. Thus, it is

likely that defective mechanisms upstream of lactate metabolism are at play in hindering

effective hypothalamic glucose sensing.

Page 27: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

17

Amino acid, leucine

Amino acid sensing in the hypothalamus also elicit potent metabolic influence on peripheral

glucose and energy regulation. Leucine is one of the three branched-chain amino acids that have

been documented to represent a physiological signal of hypothalamic amino acid availability.111

Dietary leucine escapes first-pass metabolism resulting in a rapid increase in plasma leucine

levels during postprandial conditions, and leucine reaches the brain sooner than other amino

acids112. A recent study shows that direct infusion of leucine in the MBH lowers blood glucose

through suppression of glucose production113. This gluco-regulatory effect requires the

metabolism of leucine to acetyl-CoA and the subsequent conversion to malonyl-CoA as well as

the activation of hypothalamic KATP channels. Similarly, leucine sensing also contributes to the

hypothalamus modeulated lowering of food intake and body weight, and this effect requires

leucine -induced activation of the hypothalamic mammalian target of rapamycin (mTOR)

signaling114. Studies investigating the downstream neural targets in hypothalamic leucine sensing

revealed that in addition to hypothalamic POMC neurons, MBH leucine also activates brainstem

DVC neurons to lower food intake111. This finding identified for the first time an MBH-DVC

neurocircuit engaged by MBH leucine to exert homeostatic control on energy homeostasis. As

with fatty acids and glucose, impaired hypothalamic leucine sensing may also have a role in

disease development. While molecular disruption of leucine sensing in the MBH resulted in

hyperglycemia in rats fed a high-protein diet113, 3d HFD feeding also attenuated the ability of

central leucine to modulate glucose metabolism115.

Page 28: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

18

1.2 The role of the dorsal vagal complex in the regulation of glucose and energy homeostasis

Although the historical emphasis on the CNS regulation of glucose and energy homeostasis has

been on the MBH, the DVC, in the brain stem, is another key brain site of hormonal action and

nutrient sensing that can regulate metabolic homeostasis. Like the MBH, the DVC contains a

circumventricular organ called the area postrema. In addition, the DVC contains the nucleus of

the solitary tract (NTS) and the dorsal motor nucleus of the vagus (DMX) with synaptic

connections to vagal afferent and efferent fibres, respectively. These features enable humoral and

neural signals to reach the DVC in order to modulate various autonomic functions including

glucose and energy regulation. As described previously, the DVC is a critical site for the

integration of lipid, glucose and leucine sensing inputs from the hypothalamus to affect glucose

and feeding control. This is because neuronal projections from the hypothalamus reach the

hindbrain forming a hypothalamic-DVC neuronal axis to exert homeostatic feedback on glucose

and feeding control. In this regard, studies have identified the N-Methyl-d-aspartate (NMDA)

receptors in the DVC to act as an important switchboard in integrating hypothalamic signals to

maintain glucose and energy homeostasis. Previously, neurotransmission by the DVC NMDA

receptors was shown to play an important role in relaying signals generated by intestinal nutrient

and hormonal signalling mechanisms to lower glucose production and feeding behavior116-120.

However, it was Lam et al. who directly tested and confirmed that pharmacological and

molecular blockade of DVC NMDA receptors indeed also negates the ability of hypothalamic

nutrient sensing mechanisms activated by glucose (or more specifically lactate) metabolism or

hypothalamic lipid sensing to lower glucose production121.

Page 29: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

19

In addition to the role of the DVC as a relay centre for hypothalamic-sensed signals,

multiple studies have also confirmed that the DVC is equally a direct sensor for different

hormones and nutrients to regulate metabolic homeostasis.

1.2.1 Hormonal action

Insulin

The earliest evidence that the DVC is insulin responsive came from Filippi et al122 who showed

that direct insulin infusion into the DVC (targeting the NTS) activated insulin receptor-mediated

signaling events in a dose-dependent manner. Indeed, DVC insulin signaling lowers glucose

production in rodents, while inhibition of DVC insulin receptors negated the glucose production-

lowering effect. Secondly, this study also addressed which of the DVC insulin receptor-mediated

signaling pathways regulate glucose production. When insulin was infused into the DVC at the

same dose as that which activated PI3K when administered into the MBH28,123, insulin failed to

activate DVC PI3K. Instead, DVC insulin activated Erk1/2, while inhibition of DVC Erk1/2

signaling by a chemical approach, as well as a molecular approach abolished the ability of

insulin to lower glucose production. In direct contrast, inhibition of DVC PI3K signaling did not

alter the ability of insulin to control glucose levels. Furthermore, directly activating DVC Erk1/2

lowered glucose production, strengthening the gluco-regulatory role of DVC Erk1/2 signaling.

Thus, insulin activates an Erk1/2-dependent signaling pathway in the DVC to lower glucose

production, as opposed to the MBH, where insulin lowers glucose production via PI3K signaling.

Finally, activation of KATP channels in the DVC was also found necessary for insulin–Erk1/2

signaling to inhibit glucose production.

Indeed, direct administration of insulin into the DVC also affected feeding behavior.

DVC insulin infusion lowered food intake as early as 90 min compared with DVC saline

Page 30: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

20

infusions124. The mechanism underlying DVC insulin-induced satiety involves the activation of

DVC Erk1/2, because molecular and chemical inhibition of Erk1/2 signaling negated the ability

of DVC insulin infusion to reduce feeding. In contrast, inhibition of the PI3K–Akt pathway did

not affect the action of insulin in the DVC, thereby suggesting that DVC insulin activates an

Erk1/2-dependent and a PI3K–Akt-independent pathway to regulate feeding.

High-fat feeding disrupts the action of insulin in the DVC and dysregulates feeding

control and glucose production in association with the inability of insulin to activate Erk1/2 in

the DVC122,124. However, future studies are warranted to characterize the underlying mechanisms

of insulin resistance in the DVC that concurrently dysregulate feeding and glucose control.

Leptin

As for the effects of insulin, the DVC is also a target for the inhibitory effect of leptin on food

intake. Rats treated with an injection of leptin into the 4th ventrice (icv-4), presumably targeting

the DVC, showed reduced food intake at 2, 4, and 24 h after injection and displayed significant

reduction in body weight gain125. Further, increased expression of the leptin transgene in the

DVC completely curbed the time-related gain in body weight in rats despite only a transient

suppression of food intake126.

Glucagon

In light of the fact that glucagon receptors are abundantly expressed in the brainstem70,127, and

peripherally administered glucagon alters neuronal activity in the brainstem128, it was plausible

that, analogous to the MBH, glucagon could initiate a signalling cascade in the DVC to regulate

peripheral metabolism. While it remains to be investigated whether glucagon acts in the DVC to

regulate food intake, a recent study has revealed that glucagon action in the DVC can impact

glucose homeostasis, and has postulated a novel physiological role of DVC glucagon action in

Page 31: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

21

postprandial conditions129. During pancreatic basal insulin clamp conditions, glucagon infusion

into the DVC reduced hepatic glucose production in healthy rodents. Using complementary

chemical and molecular loss-of-function approaches, blocking activation of the glucagon

receptor, PKA, ERK 1/2 or KATP channels in the DVC reversed the glucose production-lowering

effect of DVC glucagon infusion, thereby implicating each of these as the downstream signalling

mediators in the DVC glucagon signalling pathway. The physiological relevance of DVC

glucagon action in regulating glucose homeostasis was investigated in the context of the

postprandial period following high-protein meals. During fasting–refeeding experiments, high-

protein feeding acutely decreased plasma glucose levels compared with a low-protein diet; but

when DVC glucagon receptor signalling was blocked, the ability of high-vs low-protein feeding

to suppress the rise in plasma glucose was negated.

1.2.2 Nutrient sensing

Glucose

It has long been shown that there are glucose responding neurons in the DVC130. Specifically,

these neurons employ glucokinase and KATP channels to alter their action potential firing in

response to varying glucose concentrations131. Microinjections of glucose into the NTS or DMV

sites of the DVC lead to inhibition of gastric motility and increased intragastric pressure132.

Surprisingly, very little work has been done to investigate the functional effects of direct glucose

administration in the DVC neurons on the control of glucose and energy regulation, other than

the studies that have identified that glucose sensing neurons in the DVC participate in the

regulation of feeding and glycemic responses elicited by hypoglycemia133. For instance,

microinjection of glucose antimetabolites into various hindbrain sites has been shown to induce

at least 1.5 g more in food intake or at least more than 25 mg/dl in hyperglycemic response

Page 32: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

22

compared to vehicle-injected rats133. Further insights into the physiological potential of direct

DVC glucose sensing as a signal of energy availability remain unexplored.

Leucine

Similar to the MBH, the DVC possesses amino acid nutrient sensing capabilities modulating

metabolic homeostasis. Direct NTS administration of leucine, using a postprandial physiological

dose, significantly reduced meal size, 24 hour food intake and 24 hour body weight change, and

this effect is mediated by activation of the mTOR/S6K1 pathway134. The same study also

identified catecholaminergic and POMC neurons as the primary leucine- sensing neurons in the

DVC. Moreover, DVC leucine administration also elicits activation of DVC Erk1/2 signaling

pathway, which is also necessary for the anorectic effect of DVC leucine. Lastly, it was also

observed that co-administration of subthreshold doses of CCK admininstered ip or DVC

melanotan II (an analogue of the melanocortin peptide hormone) or DVC leptin enhanced the

acute anorectic effects of DVC leucine, thereby highlighting the integrating role DVC leucine

sensing plays with gut-derived signals, forebrain-descending melanocortinergic signals and

adiposity signals of energy availability to control energy homeostasis134.

As with DVC insulin action, HFD feeding leads to an impairment in DVC leucine

sensing in the regulation of feeding135. HFD negates the anorectic effects of DVC leucine

administration. This was accompanied by an increase in the baseline activity of the S6K1 and

impaired leucine-induced activation of this pathway in the DVC of HFD-fed mice, further

implicating impaired DVC mTOR sensing during HFD. Notably, the synergistic suppressive

effect of ip CCK and DVC leucine was also blunted in the DIO mice, thereby indicating that

HFD also impairs the gut brain integration in the DVC in the control of meal size.

Page 33: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

23

Glycine

Accumulating evidence shows that sensing of the amino acid glycine in the DVC has a

physiological role in the regulation of peripheral homeostasis. Data show there is a high

concentration of glycine terminals in the DVC and that glycine injections into the DVC elicit

changes in heart rate and blood pressure136. Glycine binds to the GluN1 subunit of the NMDA

receptor and acts a co-agonist along with glutamate to potentiate NMDA receptor activation137.

In line with this, direct administration of glycine in the DVC activates NMDA receptors to lower

hepatic glucose production, which was abolished in the presence of NMDA receptor antagonism

and hepatic vagotomy138. Subsequent studies have revealed that glycine sensing via DVC

NMDA receptors is sufficient to also lower hepatic-triglyceride secretion139. These data are in

agreement with other studies demonstrating that binding of glycine to NMDA receptor serves to

potentiate NMDA receptor-mediated neurotransmission 140-142.

Given that CNS hormonal and nutritional sensing mechanisms are impaired in models of

obesity and/or diabetes, the question arises whether the effectiveness of DVC glycine to lower

glucose production is intact in settings of insulin resistance, uncontrolled diabetes and DIO. We

are encouraged by the findings that DVC glycine infusion, at the same dose infused in normal

rodents, is still able to lower hepatic lipid production in an acute diet-induced insulin resistance

model139 and that other central nutrient sensing mechanisms, such as those of central lactate are

preserved in an early-onset model of STZ-diabetes110. Further, no studies till date have directly

tested whether DVC glycine sensing can regulate energy homeostasis. However, given that DVC

NMDA receptors play a role in the control of food intake (as described previously) elicited by

hormones such as cholecystokinin143, and that glycine binding acts as a modulator to NMDA

transmission in the DVC, it is plausible that glycine triggers a sensing mechanism via DVC

NMDA receptors to reduce appetite.

Page 34: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

24

However, glycine as a CNS-active drug suffers from the limitation of having a poor

pharmacokinetic profile144. For instance, following oral administration of glycine, although

elevated glycine levels in the cerebral cortex, there was a concomitant increase in the rate of

glycine uptake and rapid conversion of glycine into serine in brain tissue, thereby limiting brain

exposure to extracellular glycine145. Indeed, studies from the schizophrenia field have reported

the use of glycine administration as an approach to activate NMDA receptor-mediated

transmission in schizophrenic patients. Schizophrenia displays reduced NMDA receptor

function, and therefore increasing NMDA receptor function via pharmacological manipulation is

integral to the treatment for schizophrenia. While there are some encouraging studies reporting

that large doses of glycine can improve the negative symptoms in schizophrenic patients146,147,

there are studies that have failed to confirm the efficacy of glycine as a therapeutic agent148. It is

not clear whether the conflicting evidence is due to low CNS exposure to extracellular glycine.

In fact, the level of extracellular glycine in the CNS is primarily dependent on regulation

by the Na+/Cl−-dependent glycine transporters, GlyT1 and GlyT2149. In particular, GlyT1, which

is expressed on glial cells, is the primary transporter of glycine mediating the uptake of glycine

into cells near NMDA receptors. Thus, blockade of GlyT1 could increase synaptic glycine levels,

thereby potentiating activation of NMDA receptors. Indeed, GlyT1 inhibitors have been tested in

rodents and preliminary clinical studies and have proved to be beneficial in the treatment of

schizophrenia150-153.

Given the unfavourable properties of glycine administration, the question arises as to

whether GlyT1 inhibition in the DVC would be a favourable approach to trigger glycine sensing

in the brainstem and more importantly, whether there is a novel therapeutic potential for DVC

GlyT1 inhibition, in the treatment for diabetes and obesity to lower glucose levels and body

weight via the activation of NMDA receptors.

Page 35: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

25

1.3 The role of glial cells in the regulation of glucose and energy homeostasis. When glial cells were first discovered in the 1800s, they were viewed as merely “cellular glue”

for the brain, holding neurons together. However, in recent years, there has been accumulating

evidence pointing that far from being passive, glial cells play a critical role in the normal

functioning of the brain including synaptic plasticity, development, neurotransmission and

metabolism154. In fact, astrocytes, a type of glial cells, are emerging as important regulators of

nutrient and energy sensing mechanisms in the CNS, and they do so by expressing specific

hormonal receptors and nutrient transporters. For instance, in the hypothalamus, astrocytic

insulin receptors are indispensible for proper glucose and insulin entry into the brain, in turn

contributing to CNS regulation of systemic glucose and energy homeostasis155. Similarly, leptin

signaling in hypothalamic astrocytes has also been reported to play an important role in the CNS

control of feeding156. At the same time, studies also show that glial specific glucose transporter,

GLUT1 is vital for hypothalamic glucose sensing to regulate peripheral glucose levels109, and

that lipoprotein lipase (LPL) in astrocytes controls lipid uptake in the hypothalamus for central

regulation of body weight and glucose metabolism157. Notably, in addition to transporting

circulating nutrients and expressing hormonal receptors, glial cells can also contribute to

systemic metabolic control via uptake of neurotransmitters from the synaptic cleft. For instance,

glial cells exclusively expressing glutamate transporter (GLT)-1, mediates glutamate uptake into

astrocytes, which is critical for regulating synaptic transmission by this excitatory amino acid.

Further, glutamate uptake into astrocytes has also been reported to increase glycolysis and lactate

production, in turn modulating nutrient availability for neurons107,158. Therefore, CNS regulation

of nutrient sensing and hormonal signals is, atleast in part, directed by glial cells. However, what

remains to be shown and will be addressed in this dissertation, is whether the glial specific

Page 36: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

26

glycine transporter, GlyT1 like GLT1, can couple CNS nutrient mechanisms to control systemic

metabolic homeostasis.

1. 4 Summary of Introduction/Rationale of Study 1 and Study 2 Type 2 diabetes and obesity, the two largest public health concerns of today, are progressive

metabolic disorders of glucose and energy homeostasis. Over the last two decades, significant

progress has been made in support of the role the brain plays in regulating peripheral glucose and

energy homeostasis. The two key regions of the CNS: the MBH and the DVC can receive and

integrate information from hormones and nutrients to subsequently direct changes in hepatic

glucose production and feeding behaviour, and they do so by using distinct and sometimes

common receptors and intracellular signalling pathways. AMPK and KATP are two such crucial

intracellular signaling pathways responsible for the metabolic effects of MBH insulin, leptin,

glucose, fatty acids and amino acid leucine to regulate whole-body energy homeostasis and

glucose control. More recently, a novel gluco-regulatory role of MBH glucagon was discovered

to lower glucose production via the PKA pathway. Whether MBH glucagon action is

downstream mediated by AMPK and KATP signaling remains to be investigated. The focus of

Study 1 was to evaluate whether AMPK-mediated lipid sensing and KATP channels are

necessary for MBH glucagon for glucose regulation.

Moreover, the smallest amino acid glycine triggers neurotransmission in the DVC to

regulate metabolic homeostasis including glucose and lipid metabolism using NMDA receptors.

Importantly, DVC glycine sensing can normalize the hypersecretion of lipids induced by 3d

HFD. These findings highlight the therapeutic potential of glycine sensing to lower blood lipids

in individuals with obesity and diabetes. Could there be a novel therapeutic potential for DVC

glycine manipulation in lowering blood glucose and body weight in obese and diabetic

Page 37: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

27

individuals? Because glycine has poor pharmacokinetics, and because glycine levels in the

CNS are enhanced by GlyT1 inhibition, in Study 2, we investigated whether DVC GlyT1

inhibition could sufficiently trigger glycine sensing to improve glucose and energy

homeostasis in diabetes and obesity.

Page 38: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

28

Chapter 2 Study 1

Hypothalamic Glucagon Signals through the

KATP Channels to Regulate Glucose Production

Modified from:

Abraham, M.A., Yue, J.T., LaPierre, M.P., Rutter, G.A., Light, P.E., Filippi, B.M., and Lam,

T.K. (2014). Hypothalamic glucagon signals through the KATP channels to regulate glucose

production. Molecular Metabolism 3, 202-208.

Permission to reproduce portions of the above manuscript has been obtained from the copyright

owner: Elsevier Limited

Page 39: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

29

2.1 Abstract Background and Aims: Gluco-regulatory hormones such as insulin, leptin and GLP-1 signal in

the mediobasal hypothalamus (MBH) to lower hepatic glucose production glucose production.

MBH glucagon action also inhibits glucose production, but the downstream MBH signaling

mediators remain largely unknown. In parallel, a lipid-sensing pathway involving MBH AMPK -

> malonyl-CoA -> CPT-1 -> LCFA-CoA -> PKC-δ leading to the activation of MBH KATP

channels has been documented to lower glucose production. Given that glucagon signals through

the cAMP-PKA pathway in MBH to lower glucose production, and PKA inhibits AMPK in

hypothalamic cell lines, a possibility arises that glucagon-PKA action in the MBH would inhibit

AMPK, elevates LCFA-CoA levels to activate PKC-δ, and activates KATP channels to lower

glucose production. Methods: Using molecular and chemical approaches, we inhibited the MBH

lipid-sensing pathway in normal male Sprague-Dawley rats via (i) activation of MBH AMPK or

inhibition of MBH PKC-δ, and (ii) MBH KATP channels in the presence of MBH glucagon

stimulation and evaluated the changes in glucose kinetics during the pancreatic (basal insulin)

euglycemic clamps. Results: We found that neither molecular and chemical activation of MBH

AMPK nor inhibition of PKC-δ negated the gluco-regulatory effect of MBH glucagon. In

contrast, molecular and chemical inhibition of MBH KATP channels negated MBH glucagon’s

effect to lower glucose production. Conclusion: Our data collectively indicates that MBH

glucagon signals through a lipid-sensing independent but KATP channel-dependent pathway to

regulate glucose production.

Page 40: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

30

2.2 Introduction A role of glucagon action in the MBH was documented, in contrast to the hormone’s hepatic

stimulatory effect, to lower glucose production79. This glucose production-lowering effect

required the activation of the MBH glucagon receptor-cAMP-PKA signaling pathway. In an

experimental model of high-fat feeding, hypothalamic glucagon resistance disrupts the control on

glucose production. However, direct activation of MBH PKA bypasses this resistance to lower

glucose production79. Since MBH glucagon resistance lies upstream of PKA in response to a

high-fat diet, the potential downstream targets of PKA in MBH glucagon action warrants

investigation.

The activation of cAMP-PKA pathway has been documented to inhibit AMPK in

hypothalamic cell lines88 and adipocytes87. These findings are of interest as direct inhibition of

MBH AMPK is sufficient to lower glucose production102, while activating MBH AMPK negates

glucose sensing to inhibit glucose production102. It is believed that activation of MBH AMPK

negates the ability of glucose flux to increase malonyl-CoA levels and relieves the inhibition on

CPT-1, leading to a reduction of cytosolic LCFA-CoA levels101,118. An accumulation of MBH

LCFA-CoA levels is necessary to activate MBH PKC-δ105 and the KATP channels94,105 to lower

glucose production. Given that MBH PKA signaling is necessary for glucagon to inhibit glucose

production79 and that PKA inhibits AMPK in vitro as discussed above87,88, we here tested the

hypothesis that MBH lipid-sensing pathway involving AMPK -> LCFA-CoA -> PKC-δ and the

subsequent activation of the KATP channels are necessary for MBH glucagon to lower glucose

production (Figure 2.1).

With molecular and chemical approaches, we inhibited the (i) MBH lipid-sensing

pathway via activation of MBH AMPK or inhibition of MBH PKC-δ, and (ii) MBH KATP

Page 41: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

31

channels in the presence of MBH glucagon stimulation and evaluated the changes in the rate of

glucose production and glucose uptake in normal rats.

Page 42: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

32

2.3 Materials and Methods 2.3.1 Animal Preparation

Adult male Sprague Dawley rats aged 8 weeks (260-280g) from Charles River Laboratories

(Montreal, Quebec, Canada) were studied. Rats underwent stereotaxic implantation with a 26-

gauge stainless steel bilateral guide catheter (C235G, Plastics One Inc. Virgina, USA) placed

into the MBH using the coordinates 3.1 mm posterior to bregma, 0.4 mm lateral of midline and

9.6 mm below skull surface as described79. After six days of recovery, vascular catheters were

inserted into the internal jugular vein and carotid artery for infusion and blood sampling79,122. All

experiments in rats complied with the rules of the Institutional Animal Care and Use committee

of the University Health Network.

2.3.2 Adenovirus injection

Immediately following brain surgery, a group of rats received 3µl of adenovirus containing the

constitutively active (CA) form of AMPK (Ad-CA AMPK α1312 [T172D] (3.83x1010pfu/ml)102; or

the dominant negative (DN) form of PKC-δ or LacZ (4x108pfu/ml; gift from Dr. J Soh,

Biomedical Research Centre for Signal Transduction, Incheon, Korea)159 ; or the DN Kir6.2

AAA (3.1x1010pfu/ml) or green fluorescence protein (GFP) (3.0x1010 pfu/ml)105 through each

side of the MBH catheters, as described102,105,109.

2.3.3 Pancreatic-euglycemic clamp

Four days following vascular catheterization, conscious and unrestrained rats with at least 90%

recovery in their food intake and body weight were used in clamp studies. All rats were limited

to 15g of food the night before the clamp to ensure comparable nutritional status79,122. At the start

of the experiment (t=0), a primed continuous infusion of [3-3H]-glucose (Perkin-Elmer; 40µCi

Page 43: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

33

bolus, 0.4µCi min-1) was initiated and maintained until the end of the experiment (t=210) to

assess glucose kinetics using tracer dilution methodology. From t=90-210, a pancreatic basal

insulin clamp was performed during which somatostatin (3µg kg-1 body weight min-1) and

insulin (1.1mU kg-1 body weight min-1) were continuously infused to replace insulin to basal

levels, along with a variable infusion of 25% glucose to maintain euglycemia. At 10-min

intervals, plasma samples were taken for determination of [3-3H]-glucose concentrations, as well

as plasma insulin and glucagon concentrations. At the end of the experiment, rats were

anesthetized and injected with 3ul bromophenol blue through each side of the MBH catheter to

verify the correct placement of the catheter. The MBH wedges were then collected, frozen in

liquid nitrogen and stored at −80°C for subsequent analysis.

Treatments administered into the MBH at a rate of 0.006µl/min included: Saline (t=90-210);

5pg/µl glucagon (t=90-210); 25mmol/l AMPK activator AICAR (t=0-90) with 25mmol/l

AICAR+saline (t=90-210) or 25mmol/l AICAR+5pg/µl glucagon (t=90-210); 60µmol/l PKC-δ

inhibitor rottlerin (t=0-90) with 60µmol/l rottlerin+saline or 60µmol/l rottlerin+5pg/µl glucagon

(t=90-210); 100µmol/l KATP channel inhibitor glibenclamide (t=0-90) with 100µmol/l

glibenclamide+saline or 100µmol/l glibenclamide+5pg/µl glucagon (t=90-210 min).

2.3.4 Western blotting for phosphorylated ACC

MBH wedges were homogenized in a lysis buffer constituting 50mM Tris-HCl (pH 7.5), 1mM

EGTA, 1mM EDTA, 1% (w/v) Nonidet P40, 1mM sodium orthovanadate, 50mM sodium

fluoride, 5mM sodium pyrophosphate, 0.27M sucrose, 1µM Dithiotritolo (DTT), and protease

inhibitor cocktail (Roche). The Pierce 660nm protein assay (Thermo Scientific) was used to

measure protein concentrations of the homogenized tissues. Protein lysates (20ug) were

subjected to SDS-PAGE and transferred onto nitrocellulose membranes (Amersham). The

Page 44: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

34

membranes were first incubated for 1h at room temperature with blocking solution (5% BSA in

Tris-buffered saline and 0.2% Tween-20) and then overnight at 4°C with the primary antibody

(indicated below) diluted 1000-fold. Protein expression was detected using a horseradish

peroxidase (HRP)-linked rabbit-specific secondary antibody (diluted 1/4,000 in blocking

solution) and an enhanced chemoluminescence commercial kit (Pierce). The phosphorylation of

ACC was quantified by densitometry with the Quantity One software and normalized for the

total protein (ACC or β-tubulin). Primary antibodies include: anti-phospho ACC, anti-total ACC,

anti-β-tubulin (Cell signaling Technology).

2.3.5 PKC-δ activity assay

PKC-δ was immunoprecipitated from MBH wedges obtained after performing 10-minute MBH

infusions of saline, glucagon or OAG in rats. To do this, 3 MBH wedges from each treatment

group were pooled together to yield an n =1. MBH tissues were homogenized as described

above, following which 500ug of tissue lysate was incubated overnight with 8µg of PKC-δ

polyclonal antibody (sc-213; Santa Cruz Biotechnology) on a rotating wheel and then incubated

with 25µl of protein A/G sepharose beads for 2h at 4°C. The beads were then centrifuged at

8000rpm for 1min. After removal of the supernatant, the beads were then washed (2x with 1ml

lysis buffer (as above) with 0.5M NaCl, 1x with 1ml lysis buffer (as above) with 0.15M NaCl

and 2x with 1ml buffer A containing 50mM Tris-HCl pH 7.5, 0.1mM EGTA and 1µM DTT).

With the supernatant removed, 20µl of buffer A was further added giving a final sample volume

of 25µl. We then proceeded with the Biotrak protein Kinase C (PKC) enzyme assay system (GE

Healthcare). Additionally, to normalize for the amount of PKC-δ immunoprecipitated in each

sample, PKC-δ protein was separated from the beads using Laemmli sample buffer and subjected

Page 45: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

35

to SDS-PAGE and quantified (as described above). Results were then presented as

pmol/min/protein.

2.3.6 Biochemical Analysis

Plasma glucose levels were determined by the glucose oxidase method (Glucose analyzer GM9;

Analox Instruments, Lunenberg, MA). Radioimmunoassays (Linco Research, St. Charles, MO)

were used to determine plasma insulin and glucagon concentrations.

2.3.7 Statistical Analysis

Unpaired Student’s t tests and ANOVA as appropriate. Significance was accepted as P<0.05.

2.4 Results 2.4.1 Role of MBH AMPK in glucagon action

We activated MBH AMPK in the presence of MBH glucagon by infusing the AMPK activator

AICAR and examined whether it would negate the ability of MBH glucagon to lower glucose

production. This AICAR dose negated the ability of hypothalamic glucose infusion to lower

glucose production102. Firstly, consistent with previous finding79, MBH glucagon infusion led to

a significant increase in the glucose infusion rate (Figure 2.2A) to achieve euglycemia during

the pancreatic basal insulin clamp and significantly lowered glucose production (Figures 2.2 B,

C) compared to MBH saline treatment independent of changes in glucose uptake and plasma

levels of insulin, glucagon and glucose (Figure 2.2 D, Table 2.1). Interestingly, glucagon co-

infused with AICAR into the MBH still led to a significant increase in glucose infusion rate and

decrease in glucose production. Notably, rats that received MBH AICAR infusion alone showed

no significant difference in the glucose infusion rate and glucose production compared with the

MBH saline group, thereby showing that AICAR per se has minimal effect on glucose kinetics in

Page 46: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

36

these experimental conditions. These data indicate that chemical activation of hypothalamic

AMPK does not reverse the ability of MBH glucagon to lower glucose production.

Alternatively, we activated hypothalamic AMPK by injecting an adenovirus expressing

the CA form of AMPK, which has been previously shown to negate the metabolic effects of

hypothalamic nutrient-sensing mechanisms102. We examined whether MBH glucagon inhibition

on glucose production is then blocked in rats expressing MBH CA AMPK. Firstly, infusion of

MBH glucagon with prior Ad-GFP (control) injection significantly increased the glucose

infusion rate required to maintain euglycemia compared to MBH saline infusions (Figure 2.2E),

owing to a reduction in glucose production (Figures 2.2F, G) as opposed to a difference in

glucose uptake (Figure 2.2H). Similarly, in rats injected with MBH CA AMPK, MBH glucagon

also led to an increase in the glucose infusion rate and suppression of glucose production, while

glucose uptake was similar in all groups (Figures 2.2E-H). Similar to our chemical approach

data, these results show that molecular activation of hypothalamic AMPK does not negate the

ability of MBH glucagon to lower glucose production.

As added confirmation, we evaluated AMPK activity (i.e., protein content ratio of pACC

/ total ACC) in MBH wedges obtained from rats that received MBH saline or glucagon

treatments during clamps. Acetyl CoA carboxylase (ACC) is phosphorylated by AMPK; thus, a

lower ratio of phospho (P)-ACC/ total ACC is indicative of a lower degree of AMPK activation.

MBH glucagon infusion showed a similar degree of AMPK activation (0.6±0.1) as with MBH

saline infusion (0.5±0.1) (Figure 2.2I) (in contrast to our original hypothesis where MBH

glucagon action was postulated to inhibit AMPK). These results suggest that the MBH glucagon

infusion does not alter MBH AMPK activity in these experimental conditions and hence changes

in MBH AMPK activity is not necessary for glucagon to lower glucose production.

Page 47: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

37

2.4.2 Role of MBH PKC-δ in glucagon action

We next investigated whether inhibition of MBH PKC-δ will negate the glucose production-

regulating effect of MBH glucagon. MBH PKC-δ was inhibited by infusing PKC-δ inhibitor

rottlerin directly into the MBH at a dose previously shown to block MBH lipid or PKC activator

(OAG) infusion to lower glucose production and MBH OAG to activate PKC-δ105. We found

that MBH rottlerin did not attenuate the ability of MBH glucagon to increase the exogenous

glucose infusion rate (Figure 2.3A) and lower glucose production (Figures 2.3B, C) during the

clamp, while glucose uptake remained unchanged (Figure 2.3D). Alternatively, MBH glucagon

was also equally potent to increase the glucose infusion rate (Figure 2.3E) and lower glucose

production (Figures 2.3 F, G) in rats injected with MBH DN PKC-δ as compared with LacZ

injected rats, while glucose uptake remained similar between groups (Figure 2.3H). Of note,

injection of DN PKC-δ has been previously shown to reduce PKC-δ activity and negate the

metabolic effects of OAG in the duodenum159 . Collectively, these data suggest that both

chemical and molecular inhibition of hypothalamic PKC-δ do not block the ability of MBH

glucagon to lower glucose production. Consistent with these observations, no significant

difference in MBH PKC-δ activity between glucagon- and saline- treated MBH tissues was

detected, whereas MBH OAG (positive control) infusion markedly stimulated PKC-δ activity in

the same experimental conditions (Figure 2.3I).

2.4.3 Role of MBH KATP channels in glucagon action

We inhibited MBH KATP channels using glibenclamide in the presence of MBH glucagon to

examine whether the glucose production-lowering effect of MBH glucagon is abrogated. MBH

glibenclamide has been reported to negate MBH lipid105, OAG105 but insulin18 as well to lower

glucose production. Interestingly, co-infusion with glibenclamide attenuated the ability of MBH

Page 48: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

38

glucagon to increase the glucose infusion rate (Figure 2.4A) and lower glucose production

(Figures 2.4B, C), without any changes in glucose uptake (Figure 2.4D).

The Kir6.2-SUR1 KATP channels (which are blocked by glibenclamide) are expressed in

both the pancreatic β-cells and neurons18. The adenovirus expressing the DN form of Kir6.2

expresses an AAA mutant subunit of Kir6.2 that co-assembles with endogenous Kir6.2 and

prevents the KATP channels from conducting potassium current160. We here directly injected the

adenovirus DN Kir6.2 AAA into the MBH as described105 and tested the metabolic effect of

MBH glucagon. Consistent with our chemical approach data, MBH glucagon also failed to

increase the exogenous glucose infusion rate (Figure 2.4E) and lower glucose production

(Figures 2.4F, G) in rats expressing MBH DN Kir 6.2 AAA unlike GFP (control virus)-injected

rats, independent of changes in glucose uptake (Figure 2.4H). Together, these chemical and

molecular loss-of-function experiments indicate that MBH KATP channel is necessary for MBH

glucagon to lower glucose production.

2.5 Discussion We presently demonstrate that activation of MBH AMPK or inhibition of MBH PKC-δ did not

negate the glucose production-lowering effect of MBH glucagon. Activation of MBH AMPK

increases protein ratio of pACC / total ACC, inhibits ACC and prevents the formation of

malonyl-CoA (endogenous inhibitor of CPT-1) and LCFA-CoA54,101, while inhibiting MBH

PKC-δ negates LCFA-CoA to lower glucose production94. Since blocking the beginning or the

end of this malonyl-CoA -> CPT-1 -> LCFA-CoA -> PKC-δ, lipid sensing, pathway did not alter

the gluco-regulatory effect of MBH glucagon, MBH glucagon is demonstrated to signal through

a lipid-sensing independent pathway to lower glucose production.

Page 49: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

39

Like glucagon, leptin and insulin action in the MBH lowers glucose production18,46,161.

However, in contrast to the inability of MBH glucagon infusion to alter MBH pACC / total ACC

(i.e., AMPK activity) as currently reported, hypothalamic leptin and insulin administration lower

MBH AMPK activity, the protein ratio of pACC / total ACC and activate ACC activity53,54.

These findings raise the possibility that unlike glucagon, leptin and insulin in the MBH signal

through a lipid-sensing dependent pathway to lower glucose production. This working

hypothesis, however, remains to be investigated.

Interestingly, activation of the MBH KATP channels is necessary for MBH glucagon to

lower glucose production. Hypothalamic KATP channels, thus, become the common integrator of

hormonal and nutrient sensing to regulate glucose production as activation of hypothalamic KATP

channels is sufficient18 and necessary for insulin18, GLP-1162 and lipids94 as well to lower glucose

production in rodents. Of note, activating KATP channels in the whole brain of humans163 or the

dorsal vagal complex in rodents122 lowers glucose production, highlighting the gluco-regulatory

role of the KATP channels is not limited to the ones that are expressed in the hypothalamus.

Given that MBH glucagon receptor-PKA signaling79 and the activation of the MBH KATP

channels are required for glucagon to lower glucose production, and that PKA directly

phosphorylates and activates the Kir6.2/SUR1 subunits of the KATP channels164, our data

collectively indicate that glucagon action in the hypothalamus signals via a lipid-sensing

independent but KATP channel dependent pathway to regulate glucose production in vivo.

Page 50: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

40

Figure 2. 1 Schematic representation of the working hypothesis.

MBH glucagon may signal through the MBH lipid sensing- KATP channel pathway to lower

glucose production.

Page 51: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

41

Figure 2. 2 Role of MBH AMPK in glucagon action.

(A) Glucose infusion rate, (B) glucose production, (C) glucose production suppression expressed

as the percentage decrease from basal period (60-90 min) to the clamp period (180-210 min) and

(D) glucose uptake obtained during the clamps that received MBH saline (n=5), glucagon (n=5),

AICAR+saline (n=5) or AICAR+glucagon (n=7). (E) Glucose infusion rate, (F) glucose

production, (G) glucose production suppression expressed as the percentage decrease from basal

period (60-90 min) to the clamp period (180-210 min) and (H) glucose uptake obtained during

the clamps that received MBH GFP+saline (n=5), GFP+glucagon (n=5), CA AMPK+saline

(n=5) or CA AMPK+glucagon (n=6). (I): Phosphorylation of ACC. Shown above is the

representative western blot of pACC in saline (n=5) and glucagon (n=5) treated MBH wedges

normalized to total ACC and B-tubulin. Shown below is the quantification of pACC normalized

to total ACC. Data are shown as means+SE. *P<0.05.

Page 52: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

42

Figure 2. 3 Role of MBH PKC- δ in glucagon action.

(A) Glucose infusion rate, (B) glucose production, (C) glucose production suppression expressed

as the percentage decrease from basal period (60-90 min) to the clamp period (180-210 min) and

(D) glucose uptake obtained during the clamps that received MBH Saline (n=5); Glucagon

(n=5); Rot+saline (n=5); Rot+glucagon (n=5). Rot = Rottlerin. (E) Glucose infusion rate, (F)

glucose production, (G) glucose production suppression expressed as the percentage decrease

from basal period (60-90 min) to the clamp period (180-210 min) and (H) glucose uptake

obtained during the clamps that received MBH LacZ+saline (n=5); Lacz+glucagon (n=5); DN

PKC-δ+saline (n=5); DN PKC-δ+glucagon (n=5). I: PKC-δ activity in MBH wedges. Shown is a

representative quantification from three samples in each treatment group. Data are shown as

means+SE. *P<0.05.

Page 53: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

43

Figure 2. 4 Role of MBH KATP channels in glucagon action.

(A) Glucose infusion rate, (B) glucose production, (C) glucose production suppression expressed

as the percentage decrease from basal period (60-90 min) to the clamp period (180-210 min) and

(D) glucose uptake obtained during the clamps that received Saline (n=5); Glucagon (n=5);

Gli+saline (n=5); Gli+glucagon (n=4). Gli = Glibenclamide. (E) Glucose infusion rate, (F)

glucose production, (G) glucose production suppression expressed as the percentage decrease

from basal period (60-90 min) to the clamp period (180-210 min) and (H) glucose uptake

obtained during the clamps that received MBH GFP+saline (n=5); GFP+glucagon (n=5); DN

Kir6.2+saline (n=5); DN Kir6.2+glucagon (n=5). Values are shown as means+SE. *P<0.05

Page 54: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

44

Table 2. 1 Plasma insulin, glucagon and glucose concentrations during basal and clamp

conditions. Data are means ±SEM.

Page 55: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

45

Chapter 3 Study 2

Inhibition of Glycine Transporter-1 in the

Dorsal Vagal Complex improves Metabolic

Homeostasis in Diabetes and Obesity

Modified from:

Abraham, M.A.*, Yue, J.T.*, Bauer, P.V., LaPierre, M.P., Wang, P., Duca, F.A., Filippi, B.M.,

Chan, O., and Lam, T.K. (2016). Inhibition of glycine transporter-1 in the dorsal vagal complex

improves metabolic homeostasis in diabetes and obesity. Nat Commun 7, 13501. *Equal

contribution

Permission to reproduce portions of the above manuscript has been obtained from the copyright

owner: Elsevier Limited

Page 56: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

46

3.1 Abstract Background and Aims: The DVC located in the brainstem is a crucial site in sensing nutrients

and hormones to regulate peripheral glucose and energy homeostasis. Direct administration of

amino acid glycine into the DVC is sufficient to lower glucose production in healthy rats via

activation of NMDA receptors in the DVC. Further, NMDA receptors in the DVC can also relay

gut-hormonal dependent signals to regulate feeding. What remains to be shown is whether DVC

glycine- NMDA receptor axis regulates glucose production in diabetic and obese rodents, and

whether activating NMDA receptors with direct glycine infusion could potentially also regulate

feeding. However, given that glycine has poor pharmacokinetics in vivo, we here sought to

examine whether selectively increasing extracellular glycine levels in the DVC by inhibiting

DVC GlyT1 has a therapeutic potential in diabetes and obesity. Methods: We administered a

GlyT1 inhibitor and/or a molecular GlyT1 knockdown in the DVC of healthy, diabetic and obese

male Sprague-Dawley rats and evaluated changes in glucose regulation during ivGTT and basal-

insulin pancreatic clamps. Loss-of-function approaches targeting the NMDA receptors as well as

the vagus nerve were also utilized to delineate the mechanistic pathway relaying DVC GlyT1

inhibition and glucose regulation. Finally, to determine the effect of DVC GlyT1 inhibition on

food intake and body weight, we also performed a non-clamp fasting-refeeding protocol in

healthy rats. Results: We found that administration of a glycine transporter 1 (GlyT1) inhibitor,

or molecular GlyT1 knockdown, in the DVC suppresses glucose production, increases glucose

tolerance, and reduces food intake and body weight gain in healthy, obese, and diabetic rats.

Conclusions: These findings provide proof of concept that GlyT1 inhibition in the brain

improves glucose and energy homeostasis. We propose that GlyT1 inhibitors have potential as a

treatment of both obesity and diabetes.

Page 57: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

47

3.2 Introduction Obesity and diabetes have become a worldwide epidemic. Over 2.1 billion people worldwide are

overweight or obese8 and approximately 422 million are afflicted with diabetes165. Given the

combined economic burden of treating both these diseases and their complications, the

development of safe and effective therapeutic strategies is decidedly crucial. The dysregulation

of glucose and energy homeostasis in diabetes and obesity are caused in part by the aberrant

elevation of hepatic glucose production and energy intake166,167, pathologies which arise from the

collective failure of multiple homeostatic systems involving the liver, pancreas, adipose tissue,

brain, and gastrointestinal tract167-171. As such, the development of pharmacological approaches

to restore the impaired mechanisms within these systems is crucial to restore metabolic

homeostasis in diabetes and obesity. Neural circuits of the central nervous system (CNS) emerge

as a potential target for clinical intervention. The recently FDA-approved anti-obesity drug,

lorcaserin, activates hypothalamic 5-HT2C receptors to reduce food intake172. Moreover, the anti-

diabetic glucagon-like-peptide 1 receptor agonist drug, liraglutide, which is clinically

demonstrated to improve glycemia and reduce body weight173, requires activation of neuronal

glucagon-like-peptide 1 receptor to exert its anorectic effects174. The central actions of other

hormones such as insulin further demonstrate the potential of CNS-based therapies, as intranasal

insulin delivery in humans reduces food intake175 and glucose production176 and improves

whole-body insulin sensitivity177.

CNS nutrient sensing mechanisms also reduce food intake and body weight178 and lower

glucose levels in healthy107 and diabetic110 rodents. Specifically, hypothalamic nutrient sensing

activates a forebrain-hindbrain neuronal axis involving N-methyl-ᴅ-aspartate (NMDA) receptors

in the dorsal vagal complex (DVC) to suppress glucose production121, while these same DVC

NMDA receptors are required for intestinal sensing of nutrients, as well as metformin and

Page 58: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

48

resveratrol, to lower glucose production and food intake5,116,119,179,180. Furthermore, directly

targeting the DVC has metabolic benefits, as direct administration of glycine, an obligatory co-

agonist of the NMDA receptor, into the DVC of healthy rats lowers glucose production via

NMDA receptor activation138. Therefore, manipulating glycine levels in the DVC could present a

therapeutic target for the treatment of obesity and diabetes. However, administration of glycine

per se is not suitable as a therapy due to its poor pharmacokinetics in vivo.

On the other hand, regulating glycine concentration by manipulating glycine transporters

(GlyT) has demonstrated clinical feasibility. Since glycine uptake into cells is regulated by

glycine transporters, of which GlyT1 is the primary regulator of glycine levels in the vicinity of

NMDA receptors181, GlyT1 inhibition increases extracellular glycine levels to potentiate the

activation of NMDA receptors142. Modulation of NMDA receptor neurotransmission is currently

used as a therapy for schizophrenia, a disease that displays reduced NMDA receptor function. In

fact, clinical trials have shown that NMDA receptor augmentation via GlyT1 inhibitors improve

symptoms of schizophrenia182,183. However, no studies to date have investigated the therapeutic

potential of GlyT1 inhibition for the treatment of diabetes and obesity. Here, we examined

whether GlyT1 inhibition regulates glucose and energy homeostasis in healthy, obese, and

diabetic rodents (Figure 3.1). We demonstrate that direct inhibition of GlyT1 in the DVC

confers metabolic benefits including improved glucose tolerance, lowered glucose production,

reduced feeding, and lowered body weight gain in diabetic and obese rodents. We also report

that systemic infusion of GlyT1 inhibitor recapitulates the metabolic effects of DVC GlyT1

inhibition. Thus, inhibiting GlyT1 in the brain represents a potential novel therapeutic strategy to

lower plasma glucose levels and body weight in diabetes and obesity.

Page 59: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

49

3.3 Materials and Methods 3.3.1 Animal preparation and surgical procedures

Male Sprague-Dawley rats (Charles River Laboratories, Saint-Constant, QC, Canada) weighing

280–300 g (9-week old) were used. For chronic 28-d feeding studies (see below), a separate set

of rats initially weighing 200-220 g and fed with regular chow or a high-fat diet were used. Rats

individually housed, were subjected to a standard light–dark (0700 light, 1900 dark) cycle, and

had ad libitum access to drinking water and standard regular chow or a 10% lard-enriched chow

(high-fat diet, HFD) where indicated (see below). Rats were anesthetized during surgeries

(ketamine, 60 mg/kg; xylazine, 8 mg/kg). Bilateral, 26-gauge, stainless steel guide cannulae

(Plastics One Inc, Roanoke, VA, USA) were stereotaxically implanted into the DVC via

coordinates targeting the nucleus of the solitary tract within the DVC (NTS, 0 mm on the

occipital crest, 0.4 mm lateral to the midline, 7.9 mm below the cranial surface; Supplementary

Figure 3.1)139. Eight days following DVC surgery, indwelling catheters were surgically

implanted in the left carotid artery and right jugular vein for blood sampling and infusions,

respectively184. Post-surgical body weight and food intake were monitored daily. Rats attained a

minimum of 90% of their pre-vascular surgery body weight before undergoing experimentation 5

days following vascular surgery. Rats that did not fully recover were excluded from the study.

Rats were randomly allocated into groups prior to experiments but no blinding was done.

In parallel, microdialysis studies were performed on male Sprague-Dawley rats (Charles

River, Raleigh, NC) which started with a body weight of ~280-300g and were individually

housed in the Yale Animal Resources Center in temperature (22-23oC) and humidity controlled

rooms. The animals had free access to rat chow (Harlan Teklad, Indianapolis, IN, USA) and

water. Upon arrival at the Yale Animal Resources Center, the animals were acclimatized to

Page 60: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

50

handling and a 12-hour light cycle (lights on between 0700h and 1900h) for one week before

experimental manipulation. Principles of laboratory animal care were followed, and experimental

protocols were approved by the Institutional Animal Care & Use Committee at Yale University.

The rats were anesthetized with isoflurane and the heads were positioned into a stereotaxic frame

(David Kopf Instruments, Tujunga, CA). A single stainless steel guide cannula for

microinjection and microdialysis (Eicom Corporation, Japan) was implanted intracranially using

the following stereotaxic co-ordinates from Paxinos and Watson (0mm on the occipital crest,

5mm medial-lateral, and 7.4mm ventral at an angle of 35° for microdialysis). This targeted the

1mm microdialysis probe (Eicom Corporation, Japan) to the DVC within the NTS.

Male C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) at 18 weeks of age were

housed in a standard light-dark cycle with ad libitium access to drinking water and standard

chow. Mice were anesthetized during stereotaxic and vascular surgeries (Avertin, 0.6 mg/g). A

unilateral, 33-gauge, stainless steel guide cannula (Plastics One Inc) was stereotaxically

implanted into the 4th ventricle (ICV-4; 6.0 mm posterior to Bregma, 4.0 mm below the cranial

surface)79,122. One week following ICV-4 surgery, an indwelling catheter was surgically

implanted in the right jugular vein79,122. Post-surgical body weight and food intake were

monitored daily. Mice attained a minimum of 90% of their pre-vascular surgery body weight

before undergoing pancreatic clamp 3-5 days following vascular surgery.

3.3.2 Intravenous glucose tolerance test

Experiments were performed in overnight- (16 to 18 h) fasted male Sprague-Dawley rats 5 days

after vascular catheterization. Basal blood samples were obtained in conscious, unrestrained rats

immediately before the start of DVC infusions (0.33 µl/h, CMA 400 syringe pump, CMA

Microdialysis, Inc., North Chelmsford, MA) of 0.9% saline or ALX (ALX 5407, Tocris

Page 61: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

51

Bioscience, 40 nM), which were commenced at t = -240 min and maintained until the end of the

experiment at t = 60 min to ensure that rats received the same duration of DVC treatment as

clamp experiments (see below). After t = 0 min blood samples were obtained, an intravenous

bolus of glucose (20% glucose, 0.25 g/kg) was injected and flushed with saline. Injections were

administered via the jugular vein catheter, and blood was sampled from the carotid artery

catheter to measure plasma glucose and insulin levels for 60 min following glucose injection as

described79.

3.3.3 DVC treatments

(i) 0.9% saline

(ii) MK801 (NMDA receptor antagonist, 0.06 ng/min, dissolved in saline)

(iii) 7CKNA (7-chlorokynurenic acid, non-competitive antagonist of the glycine binding

site of the NMDA receptor, 30 µM, dissolved in saline),

(iv) ALX (ALX 5407, selective inhibitor of GlyT1, Tocris Bioscience, 40 nM, dissolved

in saline)

(v) glycine (10 µM, dissolved in saline).

Using the same DVC infusion protocol as the current study, glycine at 10 µM was validated to

elevate DVC glycine levels by ~1.2 fold and lower glucose production138 and secretion of

triglyceride rich very low density lipoproteins (VLDL-TG)139 in regular chow-fed healthy rats,

while MK-801 at 0.06 ng/min and 30 µM of 7CKNA blocked the effects of DVC glycine

infusion to lower glucose production138, and VLDL-TG secretion139. Thus, we have chosen to

use these same dose and concentrations for the inhibitors in this study examining the effect of

DVC GlyT1 inhibition (likely mediating glycine sensing). In fact, a total amount of ~20 ng of

MK-801 was delivered into the DVC over the course of 330 minutes in the current studies,

Page 62: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

52

which is comparable to the 50 ng of MK-801 delivered into the NTS (or DVC) that regulated

feeding behavior185. Similarly, our concentration of 30 µM 7CKNA was also well within the

range reported by other studies that have indicated that 10-50 uM of 7CKNA reduces 80-90% of

glycine binding to rat cerebral cortex synaptic plasma membrane186, while 30 uM of 7CKNA

inhibits NMDA-induced transmitter release from rat hippocampal slices187. More importantly,

DVC infusion of neither MK-801 at 0.06 ng/min nor 30 µM 7CKNA per se resulted in increased

glucose production but only blocked the effect of DVC glycine infusion to lower glucose

production138. Thus, any concern for the non-specific effects of these inhibitors in regulating

glucose homeostasis can be safely excluded. The concentration of 40 nM ALX was chosen based

on the IC50 of ALX for GlyT1 (4 nM)188 and factoring into a dilution factor when chemical

inhibitors are infused into the DVC.

3.3.4 Pancreatic basal insulin euglycemic clamp in rats

Experiments were performed in male Sprague-Dawley rats fasted for ~4-6 hrs before clamp

experiments to ensure comparable post-absorptive nutritional status. Basal blood samples were

obtained in conscious, unrestrained rats immediately before the start of DVC infusions (0.33

µl/h) of the following infusates: (i) saline, (ii) MK801, (iii) 7CKNA, (iii) ALX, (iv) ALX +

MK801, (v) ALX + 7CKNA, (vi) glycine. Infusions of MK801 or 7CKNA, when used, or saline

as a control, were commenced at t = -90 min; infusions of ALX ± MK801 or 7CKNA were

commenced at t = -60 min, and infusions of glycine ± 7CKNA were commenced at t = 0 min and

maintained for the duration of the experiment (Supplementary Figure 3.1 A). ALX, an

inhibitor of the GlyT1 transporter, inhibits the binding of glycine to its cellular transporter and

elevates extracellular levels of glycine189. ALX infusion was initiated earlier to allow for

extracellular levels of glycine in the DVC to accumulate. Clamp methodology was performed as

Page 63: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

53

follows79. A primed, continuous infusion (PHD2000 syringe pump, Harvard Apparatus, Saint

Laurent, QC) of [3-3H]-glucose (Perkin Elmer; 40 µCi bolus + 0.4 µCi infusion) was

commenced at t = 0 min and maintained until the end of the clamp experiment at t = 240 min to

measure glucose kinetics using tracer-dilution methodology. Glucose turnover was calculated

using steady-state formulae, in which the rate of appearance of glucose of glucose is calculated

using [3-3H]-glucose. The total rate of appearance of endogenous glucose production is

equivalent to the rate of glucose utilization during the basal period (t = 60-90 min). The

pancreatic basal insulin-euglycemic clamp was initiated at t = 90 min with the primed continuous

infusion of insulin (1.2 mU/kg/min, somatostatin (SST, 3 µg/kg/min), and a variable infusion of

25% glucose to maintain glycemia at a similar level to the basal period and was maintained until

t = 240 min. Plasma samples were obtained every 10 min for determination of [3-3H]-glucose

specific activity and glucose levels. Wedges containing the DVC, the left and right portions of

spinal trigeminal tr. (sp5), Spinal 5nu caudal part (Sp5C), Spinal 5nu, interpolar (Sp5I), and the

pyramidal tr. (py) (Supplementary Figure 3.3:i-v, see Brain tissue sampling section below)

were collected immediately after the experiments, frozen in liquid nitrogen, and stored at -80°C

for analysis.

3.3.5 Hepatic branch vagotomy in rats

A separate set of male Sprague-Dawley rats underwent hepatic branch vagotomy138 on the same

day as vascular catheterization surgeries. The hepatic branch of the ventral subdiaphragmatic

vagal trunk was transected, and the omentum between the liver and the esophagus was severed

such that any tissue connections between the liver and the esophagus were removed. The neural

communication between the central nervous system and the liver was disrupted upon transection

of the hepatic vagal nerve, and to a much lesser degree, the innervations to the gut were also

Page 64: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

54

disrupted. Sham-operated rats underwent similar procedure except for transection of the vagus.

After surgical recovery, rats underwent clamp experiments as described above.

3.3.6 Microdialysis

One week after surgery, the male Sprague-Dawley rats were fasted for 4-6 hrs before

experiments to ensure comparable post-absorptive nutritional status. On the day of the study, the

microdialysis-microinjection probe was inserted through the guide cannula and the animals were

allowed to recover for 2.5hrs prior to collection of the baseline sample (Supplementary Figure

3.2B). Artificial extracellular fluid was perfused through the probe at a rate of 0.5ul/min

throughout the study. Following the recovery period, we collected a baseline sample over the

course of 2 hours prior to the start of ALX (40nM in saline, Tocris Bioscience) infusion. The

ALX was infused into the DVC (via the microinjection needle) at a rate of 0.33ul/h for a total

duration of 300 minutes. Microdialysate samples were collected at 60, 180 and 300 minutes

following the start of ALX infusion. Control animals were infused with saline and sampled under

similar conditions. At the end of the study, the animals were euthanized with an overdose of

sodium pentobarbital.

3.3.7 DVC virus injection

Immediately after stereotaxic surgery while anesthetized, 3 µl of adenovirus or lentivirus were

injected over 30 s in each of the DVC cannulae with microsyringes. An adenovirus expressing

shRNA to the GluN1 subunit of the NMDA receptor (Ad- GluN1 shRNA, 4.0 x 1011 pfu/ml), or

a mismatch sequence as a control (Ad-MM, 4.0 x 1011 pfu/ml), was injected into the DVC for

one set of experiments using the same protocol that we have validated138. This adenoviral GluN1

shRNA knockdown procedure decreases GluN1 protein levels specifically in the region DVC138.

Page 65: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

55

In separate sets of experiments, a lentivirus expressing shRNA to GlyT1 (LV-GlyT1 shRNA, 1.0

x 106 infectious units) (sc-270432-V, Santa Cruz Biotechnology, Inc., Dallas, TX), or a mismatch

sequence as a control (LV-MM, 1.0 x 106 infectious units) (sc-108080, Santa Cruz

Biotechnology), was injected in the DVC. Eight days after DVC cannulation and virus injection,

vascular catheterization was performed as described above in rats that would undergo clamp

experiments. Thirteen days after DVC cannulation, virus-injected rats underwent clamp or

feeding experiments as described above.

3.3.8 Brain tissue sampling in rats

At the end of the experiments, rats were injected with 3 µl bromophenol blue through each side

of the bilateral DVC catheter to verify the correct placement of the catheter. Once the whole

brain is harvested from the anesthetized rat via decapitation, the cerebelleum is lifted to expose

the caudal part of the brain (Supplementary Figure 3.3:i-v). Only those data for rats that

showed injection of dye within the vagal triangle (Supplementary Figure 3.3: ii-v) were

included. A spatula was used to extract the section of vagal triangle overlaying the DVC (Blue;

Supplementary Figure 3.3:ii-v). Additionally, sections of tissues were also dissected out from

the left (Yellow) and right (Purple) lateral regions of the caudal brain containing sp5 (spinal

trigeminal tr.), Sp5C (spinal 5nu, caudal part), Sp5I (spinal 5nu, interpolar), as well from the

lower region containing py (pyramidal tr) of the distal caudal brain (Green; Supplementary

Figure 3.3:ii-v) to validate the specificity of the lentiviral injections into the DVC.

3.3.9 Microdialysate sample glycine analysis

Microdialysate samples collected at baseline, 180 and 300 minutes were analyzed using a

fluorometric assay kit (Glycine assay kit (Fluorometric), #K589-100, Biovision Incorporated,

Milpitas, CA) according to the manufacturer’s directions. A sample volume of 50ul was used in

Page 66: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

56

the assay and since glycine content was low, each microdialysate sample was spiked with

0.3nmol of the glycine standard to bring the values within a more reliable reading range of the

assay and the calculations were adjusted accordingly. Since glycine levels were relatively stable

during ALX infusion, the concentrations obtained from the last two microdialysate samples were

averaged together and compared to baseline levels. Similar calculations were performed for the

control animals.

3.3.10 Acute (3-d) and chronic (28-d) high-fat feeding in rats

Two separate sets of male Sprague-Dawley rats were fed a palatable, 10% lard-enriched high-fat

diet (HFD, TestDiet #571R, Purina Mills, Richmond, IN), either for 3-d (acute, 3-d HFD) or 28-

d (chronic, 28-d HFD) prior to clamp experiments. The composition of the HFD (3.9 kcal/g)

differs from regular chow (3.1 kcal/g): fat content (34 vs 18%); protein (22 vs 33%) and

carbohydrate (44 vs 49%) content. A 28-d regular chow-fed cohort of rats was used in parallel to

the 28-d HFD group. Both cohorts of HFD rats underwent the pancreatic clamp experiments as

described above. Rats did not overeat were excluded from the studies.

3.3.11 Intravenous ALX infusion clamps

In a separate group of 3-d HFD fed male Sprague-Dawley rats, the pancreatic clamp experiments

were performed as described above, with the exception that a continuous i.v. ALX (4.1

µg/kg/min, dissolved in 6% DMSO infused at 20 µl/min) or i.v. 6% DMSO (20 µl/min) as

vehicle was initiated at t = -90 min. At t = 0 min, a primed continuous infusion of 3[H3]- glucose

was commenced and maintained until the end of the experiment, t = 210 min. At t = 90 min, the

pancreatic basal insulin clamp was initiated with the primed continuous infusion of insulin (1.2

mU/kg/min), somatostatin (SST, 3 µg/kg/min), and a variable infusion of 25% glucose to achieve

euglycemia was administered until t = 210 min. Intravenous (i.v.) ALX was constantly infused at

Page 67: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

57

4.1 ug/kg/min to achieve a total amount of 1.23 mg/kg ALX delivered into the blood in 300 min.

This choice of dose is based on the fact that i.v. ALX injected at 1-2 mg/kg has been documented

to potentiate NMDA-evoked firing in PFC neurons of rats in vivo190.

3.3.12 Induction of experimental type 2 diabetes

Six days after DVC surgery, a separate set of male Sprague-Dawley rats was given an

intraperitoneal injection of nicotinamide (Nic, 170 mg/kg) followed by an intraperitoneal low-

dose injection of streptozotocin (STZ, 65 mg/kg) 15 min later and fed with a HFD for 7 days as

described179,180,191, prior to intravenous glucose tolerance tests as described above. Rats that did

not present with fed hyperglycemia (e.g. > 9 mM) were excluded from the study.

3.3.13 Fasting-refeeding experiments

Separate groups of male Sprague-Dawley rats were subjected to a 22-h fast (food removed at 7

pm) prior to undergoing the refeeding experiment. DVC injections (0.04 µl/min for 5 min using

CMA syringe pumps) of 0.9% saline, ALX (40 nM), or glycine (10 µM) were given at t = -60

min (ALX) or t = -10 min (glycine) (Fig 7a,e). To prevent backflow of the injected volume,

injection cannulae were left in guide cannulae for an additional 5 min with the pump off, and

dummy cannulae are re-inserted and secured with dust caps. Regular chow was returned to cages

at 5 pm, t = 0 min. Food intake was measured every 30 min for the first 4 h of the refeeding

experiment, and every 1 h until t = 360 min. Food intake and body weight were measured again

20 h (day 1) and 44 h (day 2) after rats were refed.

3.3.14 Pancreatic basal insulin euglycemic clamp in mice

Experiments were performed in male C57BL/6 mice fasted for ~4-6 hrs before clamp

experiments to ensure comparable post-absorptive nutritional status. Basal blood samples were

Page 68: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

58

obtained in conscious, unrestrained mice immediately before the start of ICV-4 infusions (1.02

µl/h) of saline or ALX (40 nM), which were commenced at t = -90 min and maintained for the

duration of the experiment. Clamp methodology in mice was performed as follows 79. A primed,

continuous infusion of [3-3H]-glucose (1 µCi bolus + 0.1 µCi infusion) was commenced at t = 0

min and maintained until the end of the clamp experiment at t = 180 min to measure glucose

kinetics. The basal period was defined as t = 50-60 min. The pancreatic basal insulin-

euglycemic clamp was initiated at t = 60 min with a primed continuous infusion of insulin (1.4

mU/kg/min, SST (8.3 µg/kg/min), and a variable infusion of 10% glucose to maintain glycemia

at a similar level to the basal period and was maintained until t = 180 min. Plasma samples were

obtained every 10 min for determination of [3-3H]-glucose specific activity and glucose levels.

3.3.15 Western blot analyses

GlyT1 protein levels were measured in purified plasma membrane fractions of brain tissue

wedges from rats that received LV GlyT1 shRNA or LV MM injections. Brain tissue wedges

were collected 13 d following lentivirus injection and immediately frozen in liquid nitrogen and

stored at -80°C until analysis. Purified plasma membrane protein fractions were isolated using a

commercial kit suitable for mammalian tissues (Plasma Membrane Protein Extraction Kit

#K268-50, BioVision Incorporated, Milpitas, CA)184. Purified plasma membrane fraction protein

concentrations were measured using a BCA Protein Assay kit (#K812-1000, BioVision

Incorporated, Milpitas, CA), and 6 µg of protein was subjected to electrophoresis on 8%

polyacrylamide gels and transferred to nitrocellulose membranes. Membranes were incubated

with blocking solution (5% BSA in Tris-buffered saline containing 0.2% Tween-20 (TBS-T)) for

1h at room temperature and overnight at 4°C in primary antibody solutions diluted 1/1000 in 5%

BSA in TBS-T of GlyT1 (ab113823 rabbit, Abcam, Cambridge, MA), or insulin receptor (IR) β

Page 69: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

59

(L55B10 mouse, #3020, Cell Signaling Technology, Danvers, MA) after 10 min shaking in

antibody stripping buffer (Gene Bio-Application Ltd, Yavne, Israel) and re-blocked as above.

Protein expression was detected using an HRP-linked secondary antibody (rabbit and mouse,

respectively, diluted 1/4000 in blocking solution) and an enhanced chemoluminescence reagent

(Pierce ECL Western Blotting Substrate, Thermo Scientific, Rockford, IL). Immunoblots were

detected using a MicroChemi 4.2 chemiluminescent imaging system and quantified with

GelQuant image analysis software (DNR Bio-Imaging Systems, Jerusalem, Israel). Plasma

membrane GlyT1 protein levels were normalized to the plasma membrane protein levels of IR.

3.3.16 Biochemical analysis

Plasma glucose concentrations were measured by the glucose oxidase method (Glucose Analyzer

GM9, Analox Instruments, Lunenburg, MA). Plasma insulin levels were determined by

radioimmunoassay (Millipore Canada Ltd, Etobicoke, ON).

3.3.17 Calculations and statistics

The sample size for each group was chosen based on study feasibility and prior knowledge of

statistical power form previously published experiments. For pancreatic clamp experiments in

rats, measurements during t = 60-90 min were averaged for the basal period, and t = 210-240

min, and for the intravenous ALX infusion clamps, t=180-210 min were averaged for the clamp

period. In mice, measurements during t = 50-60 min were averaged for the basal period, and t =

160-180 min were averaged for the clamp period. Integration of the area under the curve (AUC)

was calculated with GraphPad Prism 6 software (LaJolla, CA). Unpaired Student’s t-tests were

performed in the statistical analysis of two groups. Where comparisons were made across more

than two groups, ANOVA was performed, and if significant, was followed by Dunnett’s or

Tukey’s post-hoc tests when appropriate. Measurements that were taken repeatedly over time

Page 70: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

60

were compared using repeated measures ANOVA; if the time and treatment interaction between

groups was found to be significant, Sidak’s multiple comparisons test or t-tests were used to

determine the statistical significance at specific time points between groups. Differences in the

overall effects of HFD diet on body weight are indicated where significance was found following

repeated measures ANOVA. P value <0.05 was considered statistically significant.

3.4 Results 3.4.1 Gluco-regulation by DVC GlyT1 inhibition in healthy rodents

To first assess a gluco-regulatory function of GlyT1 inhibition in physiological conditions, we

infused the GlyT1 inhibitor, ALX188,192, into the DVC of conscious, unrestrained healthy rats and

monitored plasma glucose levels during an intravenous glucose tolerance test (ivGTT)

(Supplementary Figure 3.1A). DVC ALX infusion for 5 hrs improved glucose tolerance

(Figure 3.2A) independent of a rise in plasma insulin levels (Figure 3.2B) compared to DVC

saline infusions. To begin delineating the mechanism by which DVC GlyT1 inhibition improves

glucose tolerance independent of changes in insulin, we tested whether DVC GlyT1 inhibition

regulates glucose production or uptake during the pancreatic basal insulin euglycemic clamps in

both rats and mice (Supplementary Figure 3.1A), since DVC glycine infusion potentiates

NMDA receptors to inhibit hepatic glucose production138.

Infusion of ALX into the DVC of rats increases the requirement for exogenous glucose

infusion to maintain euglycemia (Figure 3.2C) and lowers the rate of glucose production

(Figure 3.2D) compared to infusions with saline, independent of differences in glucose uptake,

plasma glucose, plasma insulin or body weight (Supplementary Figure 3.1C-F). We also

performed pancreatic clamps in healthy mice that underwent stereotaxic and vascular

cannulation and demonstrated that ICV-4th ventricle ALX infusion correspondingly increases

Page 71: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

61

glucose infusion rates and suppresses glucose production without affecting glucose uptake or

plasma glucose levels (Supplementary Figure 3.2A-D).

Whereas inhibition of NMDA receptors with DVC infusion of NMDA receptor blocker

MK801 alone has no effect on glucose metabolism in rats, the ability of DVC ALX infusion to

increase glucose infusion rates and suppress glucose production is abolished with co-infusion of

MK801 (Figure 3.2C, D), without altering glucose uptake or plasma glucose (Supplementary

Figure 3.1C,D). The classical NMDA receptors, which are comprised of two glycine-binding

GluN1 subunits and two glutamate-binding GluN2 subunits, require co-agonism of their subunit

binding sites for activation137,141,142,193. Since GluN1 is an obligatory subunit193 and full agonism

at the glycine site is necessary for full NMDA receptor activation194, we tested the gluco-

regulatory ability of DVC ALX infusion when GluN1 is inactivated. Similar to that which was

observed with NMDA receptor inhibition, specific chemical antagonism of the GluN1 subunit of

NMDA receptors with 7-chlorokynurenic acid (7CKNA) into the DVC nullifies the ALX-

induced increase of the requirement for exogenous glucose and suppression of glucose

production without affecting glucose uptake or plasma glucose levels (Figure 3.2C,D,

Supplementary Figure 3.1C,D). Selective genetic inhibition of DVC GluN1 subunits with

injection of an adenoviral vector expressing GluN1 shRNA (Ad-GluN1 shRNA) likewise

reverses the ability of ALX infusion to increase glucose infusion rates and lower glucose

production compared to adenovirus injected mismatch sequence (Ad-MM) controls (Figure

3.2C,D, Supplementary Figure 3.1C,D).

To test whether hepatic vagal innervation mediates the gluco-regulatory effects of DVC

GlyT1 inhibition, we examined the effect of DVC ALX in rats with hepatic vagotomy vs. sham

surgery. While hepatic vagotomy or sham surgery per se do not affect glucose kinetics, the

Page 72: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

62

higher glucose infusion rate and lower glucose production observed in sham rats receiving DVC

ALX compared to DVC saline are negated in hepatic vagotomized rats, without any difference in

glucose uptake or plasma glucose (Figure 3.2E,F, Supplementary Figure 3.2E,F).

We next performed microdialysis to examine the effect ALX infused into the DVC would

have on the extracellular levels of glycine within the DVC in healthy rats in vivo

(Supplementary Figure 3.1B). When ALX vs. saline is infused into the DVC at a comparable

duration and dosage as the IVGTT and clamp infusion studies in healthy rats (Supplementary

Figure 3.1A), ALX results in a ~2.5-fold increase in extracellular glycine levels in the DVC

(Figure 3.2G). Taken together, DVC GlyT1 inhibition via ALX infusion increases glucose

tolerance and elevates extracellular glycine levels in the DVC to potentiate NMDA receptors and

activate a brain-liver axis to lower glucose production in healthy rodents in vivo.

We alternatively tested the gluco-regulatory role of hindbrain GlyT1 inhibition via the

targeted molecular knockdown of GlyT1 within the DVC. We first confirmed that lentiviral

injection of GlyT1 shRNA (LV-GlyT1 shRNA) into the DVC selectively reduces the expression

of both the 70- and 90-kDa isoforms of GlyT1 in plasma membrane fractions of only the DVC

tissue compared to lentiviral injection of mismatch sequence (LV-MM), but not in the 2 adjacent

left and right lateral regions of the DVC containing the Spinal trigeminal track (sp5), Spinal 5nu

caudal part (Sp5C) and Spinal 5nu interpolar (Sp5I), and the region inferior to the DVC

containing the pyramidal tract (py) of the same rats (Figure 3.3A-D, Supplementary Figure 3.3

i-v). The dominant band at the molecular weight of 70-75 kDa corresponds to GlyT1a and b

isoforms and the weaker band at 90-100 kDa corresponds to GlyT1c isoform found in the rat

brain as described195. The 90-100 kDa band is not detected in the sp5, Sp5C, Sp5I (right) and py

regions (Figure 3.3C,D). The immunoblot also reveals a strong band at 55 kDa (Figure 3.3A),

Page 73: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

63

which is consistent with the occurrence of partially glycosylated form of GlyT1 in the 55-60 kDa

range as indicated196-198. However, the 55 kDa GlyT1 band in the DVC of LV- GlyT1 shRNA vs.

- MM injected rats is not significantly different as compared to the effect on 70 kDa and 90 kDa

bands (Figure 3.3A). Nonetheless, the specific metabolic role of various forms of GlyT1 in the

brain warrants future investigation.

A 13-day chronic inhibition of GlyT1 robustly increases glucose infusion rates (Figure

3.3E) and diminishes rates of glucose production (Figure 3.3F) during the clamps, independent

of changes in glucose uptake and plasma glucose (Supplementary Figure 3.4A,B). The DVC

LV-GlyT1 shRNA and MM injected regular-chow fed rats received vascular surgery (for the

clamp studies) on day 8 post-DVC viral injection and the clamp studies were conducted on day

13 (Supplementary Figure 3.1A). The body weights of these viral injected rats remain

comparable on the morning of the clamps at which point rats were also fasted for 4-6 hrs

(Supplementary Figure 3.4C). Importantly, the gluco-suppressive effect of this chronic

molecular GlyT1 inhibition is also mediated through the activation of DVC NMDA receptors

since DVC infusion with MK801 abolishes the effect of LV-GlyT1 shRNA to increase glucose

infusion rates and lower glucose production, unaffected by differences in glucose uptake,

glycemia, or body weight (Figure 3.3E,F, Supplementary Figure 3.4A-C). These molecular

loss-of-function studies strengthen the role of DVC GlyT1 inhibition in elevating extracellular

glycine levels and activating NMDA receptors to lower glucose production in healthy rodents.

3.4.2 Anti-diabetic effect of DVC GlyT1 inhibition

We next sought to ascertain a therapeutic relevance for the glucose-lowering capacity of DVC

GlyT1 inhibition first in 3-day high fat diet (3-d HFD) fed rats (Figure 3.4A). Rats placed on a

3-d HFD were first confirmed to be hyperphagic (cumulative food intake: 258 ± 10 vs 178 ± 11

Page 74: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

64

kcal, P<0.01 3-d HFD (n=26) vs 3-d RC (n=11), t-tests) and hyperinsulinemic {3-d HFD (1.7 ±

0.2, n=5) vs. 3d RC rats (0.9 ± 0.1, n=8), P<0.05, t-tests}, consistent with the fact that 3d HFD

rats were validated in parallel under hyperinsulinemic-euglycemic clamp conditions in our

research facility to exhibit hepatic insulin resistance179. We here evaluated whether antagonism

of DVC GlyT1 modulates glucose homeostasis in these 3-d HFD rats to the same extent as direct

DVC glycine infusion during the pancreatic (basal insulin)-euglycemic clamp conditions, given

that DVC GlyT1 inhibition increases extracellular DVC glycine levels (Figure 3.2G). Indeed,

DVC GlyT1 inhibition with ALX increases the requirement of glucose (Figure 3.4B) and

suppresses the rate of glucose production (Figure 3.4C) independent of alterations in glucose

uptake (Supplementary Figure 3.5A) and plasma glucose levels (Supplementary Figure 3.5B)

during the pancreatic clamp in HFD rats to the same extent as DVC glycine infusion (Figure

3.4B,C). Further, this glucose production-lowering effect of DVC ALX or glycine in 3-d HFD

rats requires the activation of the NMDA receptor GluN1 subunits as co-infusion of 7CKNA

with ALX or glycine abates the glucose-suppressive ability of ALX and glycine (Figure

3.4B,C).

We next examined whether systemic administration of GlyT1 inhibitor ALX recapitulates

the glucose production-lowering effect of DVC ALX infused-dependent GlyT1 inhibition in 3-

day HFD-fed rats. Strikingly, constant intravenous (i.v.) infusion of ALX for 5 hrs leads to a

higher glucose infusion rate (Figure 3.4D) and lower glucose production (Figure 3.4E)

compared to i.v. 6% DMSO vehicle infusion during the pancreatic clamps, and these metabolic

changes occur independent of glucose uptake (Supplementary Figure 3.5C) and changes in

plasma glucose levels (Supplementary Figure 3.5D).

Page 75: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

65

Next, we evaluated the effects of DVC ALX infusion in a rat model of type 2 diabetes

(Figure 3.4A) that is considered a better representation of humans with type 2 diabetes191. Rats

were injected with nicotinamide (Nic) and low-dose streptozotocin (STZ) to prevent beta-cell

compensation for HFD-induced insulin resistance, and maintained on a HFD for 7 days (Figure

3.5A). We have confirmed these 7-d STZ/Nic/HFD rats have fasting hyperglycemia (Figure

3.5B) and validated in parallel in our research facility to exhibit elevated hepatic glucose

production179,180. In addition, diabetic 7-d STZ/Nic/HFD + DVC saline-infused rats are glucose

intolerant as they have markedly elevated total glucose excursions during ivGTT compared to

their non-diabetic regular chow fed DVC saline-infused counterparts (Figure 3.5C).

Interestingly, ALX infusion into the DVC markedly lowers total glucose excursions in diabetic

rats compared to DVC saline infusion (Figure 3.5C). Thus, these experiments indicate a gluco-

regulatory therapeutic potential for DVC GlyT1 inhibition in high-fat fed or diabetic rodents.

3.4.3 Metabolic benefits of DVC GlyT1 inhibition in obesity

We next assessed whether DVC GlyT1 inhibition improves glucose metabolism in 28-d

HFD-induced obese rats (Figure 3.6A). Rats fed a HFD for 28 days were first confirmed to be

obese (Figure 3.6B) and hyperinsulinemic {28d HFD rats (2.5 ± 0.2, n=10) vs. 28d RC rats (1.9

± 0.2, n=9), P<0.05, t-test}, consistent with the fact that this obese model was validated in

parallel under hyperinsulinemic-euglycemic clamp conditions in our research facility to exhibit

hepatic and peripheral insulin resistance179. Importantly, in both 28-d regular chow and HFD

cohorts, we here report that acute inhibition of DVC GlyT1 with ALX infusion into the DVC

increases glucose infusion rates (Figure 3.6C) and lowers glucose production (Figure 3.6D)

independent of changes in glucose uptake (Supplementary Figure 3.6A) and plasma glucose

levels (Supplementary Figure 3.6B) during the pancreatic (basal insulin)-euglycemic clamp

Page 76: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

66

conditions. Notably, the glucose production-lowering effect of acute DVC GlyT1 antagonism is

evident in spite of the weight gain incurred by chronic high-fat feeding (body weight on the

morning of clamp experiments: 419 ± 7 vs 390 ± 10 g, P<0.05 28-d HFD vs 28-d RC, t-test).

Given that acute inhibition of DVC GlyT1 improves glucose homeostasis in short-term

(Figure 3.4C) and long-term high-fat fed rats, we postulated that chronic inhibition of GlyT1 in

the DVC might confer a gluco-regulatory benefit during 28 days of HFD-induced obesity. We

tested this hypothesis by subjecting 28-d HFD-fed rats to targeted knockdown of GlyT1 in the

DVC (via DVC LV-GlyT1 shRNA injection on Day 16 after HFD; Figure 3.7A) to determine

whether this chronic (from Day 16-Day 29; Figure 3.7A) intervention modulates glucose

homeostasis. Indeed, chronic genetic inhibition of GlyT1 in the DVC robustly increases the

glucose infusion rate (Figure 3.7B) and suppresses glucose production (Figure 3.7C) as

compared with MM controls. This glucose-lowering effect occurs independent of changes in

glucose uptake (Supplementary Figure 3.6C) and plasma glucose levels (Supplementary

Figure 3.6D). Surprisingly, body weights on the morning of clamp experiments are markedly

lower in 28-d HFD-rats with chronic DVC GlyT1 inhibition (Figure 3.7D). In fact, this lowering

of body weight in 28-d HFD-induced obese rats is evident by 4 d post-viral (LV-GlyT1 shRNA

vs. LV-MM) injection (Figure 3.7E). However, it is unlikely that the gluco-regulatory

improvement results from a decrease in body weight since chronic DVC GlyT1 inhibition lowers

glucose production in healthy rats without affecting body weight on the morning of the clamp

studies (Figure 3.3F, Supplementary Figure 3.4C).

3.4.4 DVC GlyT1 inhibition regulates energy balance

Given that acute DVC GlyT1 inhibition lowers glucose production in 28-d HFD obese rats

(Figure 3.6D), it was important to next investigate whether the local elevation of glycine in the

Page 77: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

67

DVC associated with DVC GlyT1 inhibition can also regulate energy balance. First, we tested

the direct effect of DVC glycine sensing on appetite and body weight regulation in healthy rats

that received DVC surgery 13 days prior (Figure 3.8A). Following a 22-h fast, injection of

glycine into the DVC begins to lower food intake compared to saline infused controls by ~120

min post-injection and refeeding (Figure 3.8A,B), an effect that becomes significant by 180 min

and persisted for 1 day after refeeding (Figure 3.8C). DVC glycine has no significant effect on 1

or 2 day post-refeeding percent body weight gain (Figure 3.8D). Secondly, we assessed whether

chemical inhibition of GlyT1 in the DVC could recapitulate these glycine-induced satiation

effects. Indeed, DVC ALX injection reduces food intake by 60 min after refeeding (or 120 min

post-ALX injection) (Figure 3.8E,F) with the effect still present 1 day post-refeeding (Figure

3.8G). DVC ALX also reduces the percent body weight gain after 1 and 2 day but not 3 day of

refeeding (Figure 3.8H). Finally, we evaluated whether molecular inhibition of GlyT1 in the

DVC could regulate energy balance. LV-GlyT1 shRNA or LV-MM was injected into the DVC

of healthy rats to knockdown DVC GlyT1, resulting in reduced body weight of LV-GlyT1

shRNA rats 4 day post-viral injection compared to LV-MM (Figure 3.8I), similar to the effect

observed in obese rats (Figure 3.7E). The viral-injected regular chow-fed rats were then

subjected to a 22-h fast in the evening of day 4, and genetic knockdown of DVC GlyT1 lowers

cumulative food intake as early as 120 min following refeeding (Figure 3.8J) and up to 1 day

post-refeeding (Figure 3.8K). In parallel, LV-GlyT1 shRNA vs LV-MM injection lowers the

percent body weight gain following 1 and 2 day but not 3 day post-refeeding (Figure 3.8L).

Taken together, we provide evidence that DVC GlyT1 inhibition and subsequent glycine

elevation triggers a sensing mechanism in the DVC to lower feeding and body weight in rats.

Page 78: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

68

3.5 Discussion We have shown that targeted inhibition of DVC GlyT1 through either administration of a GlyT1

inhibitor or a chronic molecular knockdown improves glucose homeostasis and lowers body

weight gain in diabetic and obese rodents.

The effect of DVC GlyT1 inhibition on glucose production regulation requires a hepatic

vagal-dependent communication between the brain and the liver. Although the neurocircuitry

involved in food intake and body weight regulation by DVC GlytT1 inhibition (or glycine

sensing) remains unclear, the underlying neuronal relay is likely different than glucose

production regulation (since glucose production is altered by DVC GlyT1 inhibition independent

of changes in food intake and body weight (Figure 3.2D, Figure 3.3F, Figure 3.6D) as well as

blood pressure and heart rate regulation (since DVC injection of glycine or glutamate induce

changes of blood pressure and heart rate at a much faster rate199,200 than changes in feeding

induced by DVC glycine injection (Figure 3.8B)). It would be important to follow-up on the

potential long term control of food intake and body weight regulation via repeated injections of

glycine or ALX, particularly knowing that a knock-down of DVC GlyT1 for 13 days exerts an

anti-obesity effect.

Although the individual cells in the DVC involved in the metabolic control of DVC

GlyT1 inhibition remain to be identified, the potentiation and activation of the GluN1/GluN2-

containing NMDA receptor in the DVC is necessary for the gluco-regulatory effect of DVC

GlyT1 inhibition (Figure 3.2-3.4). Given that the NMDA receptors are expressed in the plasma

membrane and are necessary for the metabolic effect of DVC GlyT1 inhibition, DVC ALX

infusion increases extracellular glycine levels within the DVC as assessed by microdialysis

(Figure 3.2G), and that DVC glycine infusion (like GlyT1 inhibition) potentiates DVC NMDA

receptors to lower glucose production in healthy138 and 3-d HFD (Figure 3.4C) rats, glycine is

Page 79: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

69

proposed to be the endogenous agonist that mediate the metabolic control of DVC GlyT1

inhibition. D-serine, like glycine, is also a co-agonist of the NMDA receptors. However, it is

unlikely that D-serine is the endogenous agonist that mediates the effects of GlyT1 inhibition as

injection of GlyT1 inhibitor elevates extracellular glycine but not serine and glutamate levels in

the brain of rats189. Future studies are necessary to dissect the specific role of glycine vs. serine

per se as well as in the presence of GlyT1 inhibition in regulating glucose and energy

homeostasis.

Although ketamine (a partial NMDA receptor antagonist) was used to anaesthetize the

animals for brain and vascular surgeries, any potential confounding effects of ketamine on the

gluco-regulatory studies should be absent by the time we carry out the infusion experiments as

body weight and food intake of the rodents have fully recovered. In addition, MK-801 inhibits

the GluN1/GluN2 but not the GluN1/GluN3 NMDA receptors201. Given that in our current study,

DVC MK-801 fully reverses the ability of both DVC ALX infusion and DVC LV-GlyT1 shRNA

viral injection to inhibit glucose production (Figures 3.2, 3.3), it is likely that activation of the

GluN1/GluN2 receptors, and not GluN1/GluN3, is essential for the metabolic effects of DVC

GlyT1 inhibition and glycine sensing. Consistent with this hypothesis, bi-directional changes of

NMDA receptors in the DVC via DVC infusion of NMDA or NMDA receptor antagonist AP5

alter glucose production138, while strychnine-sensitive glycine receptors do not appear to mediate

DVC glycine sensing to regulate glucose production138, altogether strengthening the claim that

GluN1/GluN2-containing NMDA receptors mediate the glucose-lowering effect of DVC GlyT1

inhibition. Nonetheless, a role for DVC GluN1/GluN3 NMDA receptor in glucose regulation

remains to be directly assessed.

DVC GlyT1 inhibition improves glucose tolerance independent of a rise in plasma insulin

levels and lowers glucose production when insulin levels are maintained at basal during the

Page 80: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

70

pancreatic clamps. Thus, it is tempting to speculate that DVC GlyT1 inhibition may improve

glucose homeostasis in type 1 diabetic insulin-deficient conditions, particularly knowing that

leptin action in the brain and the gut, as well as nutrient sensing in the gut, have been

documented to improve glucose homeostasis in insulin-deficient type 1 diabetic rodents119,202-204.

This working hypothesis warrants future investigation. On the other hand, it would also be of

future interest to assess whether DVC GlyT1 inhibition reverses insulin resistance in type 2

diabetic and obese rodents using the hyperinsulinemic-euglycemic clamp technique to achieve

insulin-stimulated conditions.

The finding that systemic administration of ALX can recapitulate the glucose production-

lowering effect of GlyT1 inhibition in the DVC during the pancreatic (basal insulin)-euglycemic

clamp settings further substantiates the potential therapeutic relevance of GlyT1 inhibitors in

diabetes and obesity. However, given that NMDA receptors are also expressed in the islets and

alter glucose-stimulated insulin secretion205, future studies are warranted to investigate the short

and long-term metabolic benefits of ALX administration in non-clamp conditions.

Our current set of findings serve as proof of concept for potential of GlyT1 inhibition as a

singular therapeutic target for the concurrent treatment of both diabetes and obesity, in addition

to its current use in the treatment of schizophrenia. Interestingly, patients with schizophrenia

have over four times the risk for abdominal obesity and twice the risk for diabetes compared to

general population controls206, highlighting the possibility that common pathologies may

contribute the development of these diseases.

Among several GlyT1 inhibitors that have undergone clinical trials, bitopertin has seen

the most success by advancing to phase III trials for the treatment of schizophrenia182,183. ALX,

on the other hand, has demonstrated relatively poorer tolerance in vivo207. However, although

ALX never entered clinical trials, it is extensively used as a pharmacological tool for the study of

Page 81: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

71

glycine transporter function207. Interestingly, DVC administration of ALX in the present study

increases DVC extracellular glycine levels which mimics a comparable effect of a low oral dose

of bitopertin on CSF glycine levels in rats208. These two drugs may therefore trigger a similar

degree of NMDA receptor-mediated neurotransmissions to elicit comparable metabolic effects.

Given that several GlyT1 inhibitors have successfully demonstrated safety and efficacy in

humans and that systemic ALX infusion recapitulates the ability of ALX infusion into the DVC

to lower glucose production in HFD rats, we propose that GlyT1 inhibitors be considered as

pharmacological agents for the restoration of glucose and energy homeostasis in obesity and

diabetes.

Page 82: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

72

Figure 3. 1 Schematic representation of the working hypothesis.

Glycine transporter-1 (GlyT1) facilitates the cellular uptake of glycine in the dorsal vagal

complex (DVC). Chemical (via DVC ALX infusion) or genetic (via DVC lentiviral injection of

GlyT1 shRNA) inhibition of GlyT1 increases extracellular glycine levels in the DVC, which

potentiates the activation of DVC N-methyl-D-aspartate (NMDA) receptors to regulate glucose

production and glucose tolerance, and food intake and body weight gain. MK-801, NMDA

receptor ion channel blocker. 7-chlorokynurenic acid, 7CKNA-antagonist to the GluN1 subunit

of NMDA receptors.

Page 83: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

73

Figure 3. 2 Chemical inhibition of DVC GlyT1 regulates glucose homeostasis in healthy

rats.

(A) Plasma glucose levels (inset: integrated area under the curve (AUC)) and (B) plasma insulin

levels during ivGTT with DVC infusion of ALX (n=8, black squares) or saline (n=7, white

squares). †P<0.04, ††P<0.0008 determined by Sidak’s multiple comparisons test following

repeated measures ANOVA. *P<0.05 determined by t-test. (C) Glucose infusion rates and (D)

glucose production during clamps with DVC infusion of saline (n=11), ALX (n=9), MK801

(n=9), ALX+MK801 (n=5), ALX+7CKNA (n=5), Ad-MM+ALX (n=5), or Ad-GluN1

shRNA+ALX (n=5). (C *P<0.002 vs saline, MK801, ALX+MK801, and ALX+7CKNA

determined by ANOVA and Dunnett’s post hoc test; †P<0.002 vs Ad-GluN1 shRNA+ALX

determined by t-test; D: *P<0.02 vs saline, MK801, ALX+MK801, and ALX+7CKNA

determined by ANOVA and Dunnett’s post hoc test; †P<0.0008 vs Ad-GluN1 shRNA+ALX

determined by t-test.) (E) Glucose infusion rates and (F) glucose production during clamps with

DVC ALX infusion in vagotomized (n=7) or sham-operated (n=5) rats or DVC saline infusion in

vagotomized (n=7) or sham-operated rats (n=5) rats. (E,F*P<0.01 compared to all other groups

determined by ANOVA and Dunnet’s post hoc test.). (G) Extracellular glycine levels within the

DVC following DVC infusion of ALX (n=7) or saline (n=7) in microdialysis studies. *P<0.03 vs

saline determined by t-test. Data are shown as the mean + SEM.

Page 84: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

74

Figure 3. 3 Molecular inhibition of DVC GlyT1 regulates glucose homeostasis in healthy

rats.

(A) Representative Western blots and protein levels of plasma membrane GlyT1 (55, 70 and 90

kDa isoforms) normalized to insulin receptor (IR) in DVC wedges of rats 13 day-post DVC

lentiviral (LV) injection of GlyT1 shRNA (black bars, n=14) or a mismatch sequence (MM;

white bars, n=11) as a control. *P<0.01, **P<0.001 determined by t-test. (B-D) Representative

Western blots and protein levels of plasma membrane GlyT1 (70 and/or 90 kDa isoforms)

normalized to IR in sp5, Sp5C, Sp5I (L), sp5, Sp5C, Sp5I (R) and py wedges of rats 13 day-post

DVC LV injection of GlyT1 shRNA (black bars, n=5) or MM (white bars, n=5). (E) Glucose

infusion rates and (F) glucose production during clamps in rats injected with LV-MM (n=7), LV-

GlyT1 shRNA (n=7), or LV-GlyT1 shRNA with DVC MK801 infusion (n=6). (E: *P<0.006; F:

*P<0.003 vs LV-MM control and LV-GlyT1 shRNA+MK801 determined by ANOVA and

Dunnett’s post hoc test.). Data are shown as the mean + SEM.

Page 85: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

75

Figure 3. 4 DVC and iv infusion of ALX regulates glucose homeostasis in 3d-HFD rats.

(A) Experimental protocol for panels B-C. (B) Glucose infusion rates and (C) glucose

production during clamps with DVC infusion of saline (n=5), glycine (n=6), ALX (n=5),

glycine+7CKNA (n=5), and ALX+7CKNA (n=5). (B: *P<0.0003 vs saline and

Glycine+7CKNA; †P<0.001 vs saline and ALX+7CKNA; determined by ANOVA and

Dunnett’s post hoc test; C: *P<0.006 vs saline and Glycine+7CKNA; †P<0.002 vs saline and

ALX+7CKNA; determined by ANOVA and Dunnett’s post hoc test.) (D) Glucose infusion rates

and (E) glucose production during clamps with iv infusion of 6% DMSO (n=7) or ALX (n=7) in

3-d HFD rats. (D,E: *P<0.001 vs iv DMSO determined by t-test.)

Page 86: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

76

Figure 3. 5 Inhibition of DVC GlyT1 regulates glucose homeostasis in diabetic rats.

(A) Experimental protocol for panels B-C. (B) Plasma levels of glucose in overnight-fasted 7d

STZ/Nic/HFD diabetic rats (black bars, n=17) compared to non-diabetic, regular chow-fed

counterparts (white bars, n=13); *P<0.01 determined by t-test. (C) Plasma glucose levels (inset:

integrated area under the curve (AUC)) during ivGTT with DVC infusion of ALX (n=9, grey

triangles) or saline (n=8, black triangles) in 7d STZ/Nic/HFD rats or DVC saline in regular chow

rats (n=7, white squares). †P<0.05, ††P<0.01, ††† P<0.001 vs DVC saline + regular chow rats; ‡

P<0.05, ‡‡ P<0.01 vs DVC ALX + 7d STZ/Nic/HFD rats determined by ANOVA and Dunnett’s

post hoc test; AUC: *P<0.05, **P<0.01 vs. DVC saline + 7d STZ/Nic/HFD rats determined by

ANOVA and Dunnet’s post-hoc test. Data are shown as the mean + SEM.

Page 87: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

77

Figure 3. 6 Chemical inhibition of DVC GlyT1 regulates glucose homeostasis in obese rats.

(A) Experimental protocol for panels B-D (B) Body weight gain in rats that were fed with HFD

(white circles, n=18) or regular chow (RC, white squares, n=6). Inflections of the body weight

curves at d 16 and d 24 represent DVC cannulation and vascular catheterization surgery days,

respectively. *P<0.02 main effect of diet, F(1,22)=6.964 determined by repeated measures

ANOVA. (C) Glucose infusion rates and (D) glucose production during clamps in 28d RC-fed

rats with DVC infusion of saline (n=5) or ALX (n=5) and in 28d HFD-fed rats with DVC

infusion of saline (n=7) or ALX (n=7) (C,D: *P<0.01 vs. the respective DVC saline determined

by ANOVA and Tukey’s post-hoc test).

Page 88: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

78

Figure 3. 7 Molecular inhibition of DVC GlyT1 regulates metabolic homeostasis in obese

rats.

(A) Experimental protocol for panels B-E. (B) Glucose infusion rates and (C) glucose

production during clamps in 28d HFD-fed rats with DVC lentivirus (LV) injection of GlyT1

shRNA (n=10) or a mismatch sequence (MM, n=9) as a control. (B, C: *P<0.001 vs 28d-

HFD+MM determined by t-test.) (D) Body weights on the morning of clamp experiments in 28d

HFD-fed rats with DVC LV MM or GlyT1 shRNA. *P<0.04 determined by t-test. (E) Percent

body weight change on days 4 and 5 following DVC injection of LV-MM (white circles, n=9) or

GlyT1 shRNA (black circles, n=9) in 28d HFD-fed rats and of MM fed with regular chow (white

squares, n=5). *P<0.05 compared to all other groups determined by ANOVA and Dunnett’s post

hoc test. Data are shown as the mean + SEM.

Page 89: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

79

Figure 3. 8 Chemical and molecular inhibition of DVC GlyT1 regulate energy balance.

(A) Experimental protocol for feeding experiments in rats that received DVC injection of

glycine (black squares, n=11) or saline (white squares, n=11). (B) Cumulative food intake during

the feeding experiment. (C) Daily food intake on day 1 and day 2 after food was returned during

the feeding experiment. (D) Percent body weight gain on day 1 or day 2 after the feeding

experiment. (E) Experimental protocol for feeding experiments in rats that received DVC

injection of ALX (black squares, n=8) or saline (white squares, n=8). (F) Cumulative food intake

during the feeding experiment. (G) Daily food intake on day 1 and day 2 after food was returned

during the feeding experiment. (H) Percent body weight gain on day 1, day 2 or day 3 after the

feeding experiment. (I) Percent body weight change on day 4 following DVC injection of LV-

MM (n=8) or GlyT1 shRNA (n=10) in regular chow-fed rats. (J) Cumulative food intake during

the feeding experiment in rats injected with LV-MM (n=6) or GlyT1 shRNA (n=7). (K) Daily

food intake on day 1 and day 2 after food was returned during the feeding experiment. (L)

Percent body weight gain on day 1, day 2 or day 3 after the feeding experiment. †P<0.05,

††P<0.01, †††P<0.001 determined by t-test at each time point, ‡P<0.05 determined by t-test at

each time. Data are shown as the mean + SEM.

Page 90: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

80

Supplementary Figure 3. 1 Metabolic effects of chemical inhibition of DVC GlyT1 in

healthy rats.

(A) Experimental protocols for experiments shown in Figure 2. In intravenous glucose tolerance

tests (ivGTT), infusions of saline or ALX into the DVC commenced at t=-240min and were

maintained for the duration of the experiment. In clamps experiments, pre-infusions of saline,

MK801, or 7CKNA into the DVC commenced at t=-90min. Infusions of saline, MK801, ALX,

ALX+MK801, or ALX+7CKNA into the DVC commenced at t=-60min and were maintained for

the duration of the clamps. (B) Experimental protocol for microdialysis studies shown in Fig. 1i.

(C) Glucose uptake, (D) basal and clamp plasma glucose levels during clamps with DVC

infusion of saline (n=11), ALX (n=9), MK801 (n=9), ALX+MK801 (n=5), ALX+7CKNA (n=5),

Ad-MM+ALX (n=5), or Ad-GluN1 shRNA+ALX (n=5), (E) basal and clamp plasma insulin

levels during DVC infusion of saline (n=6) or ALX (n=6), and (F) body weights on the morning

of clamp experiments before DVC infusion of saline (n=11) or ALX (n=9). Data are shown as

the mean + SEM.

Page 91: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

81

Supplementary Figure 3. 2 Metabolic effects of chemical inhibition of GlyT1 in the 4th

ventricle of mice and in the DVC of hepatic vagotomized rats.

(A) Glucose infusion rates, (B) rates of clamp glucose production, (C) glucose uptake, and (D)

clamp plasma glucose levels during clamp experiments with ICV-4th ventricle infusion of ALX

(n=5) or saline (n=6) in C57BL/6 mice; *P<0.02 vs saline determined by t-test. (E) Glucose

uptake, and (F) basal and clamp plasma glucose levels during clamp experiments with DVC

infusion of ALX in vagotomized (n=7) or sham-operated (n=5) rats or DVC infusion of saline in

vagotomized (n=7) or sham-operated rats (n=5) rats. Data are shown as the mean + SEM.

Page 92: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

82

Supplementary Figure 3. 3 Brain regions included in the GlyT1 protein analysis.

This include the DVC region (blue), the left (L) lateral region (yellow) containing spinal

trigeminal tr. (sp5), spinal 5nu, caudal part (Sp5C), spinal 5nu, interpolar (Sp5I), the right (R)

lateral region (purple) containing sp5, Sp5C, Sp5I and the bottom region (green) containing

pyramidal tr. (py). i: Saggital image representing the rat brain. ii: Vagal triangle overlaying the

DVC located in the caudal part of the brain. iii-v: three coronal images representing the

proximal, medial and distal regions of the caudal brain indicating all the regions included in the

analysis of the GlyT1 protein.

Page 93: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

83

Supplementary Figure 3. 4 Metabolic effects of molecular inhibition of DVC GlyT1 in

healthy rats.

(A) Glucose uptake, (B) basal and clamp plasma glucose levels during clamps, and (C) body

weights on the morning of clamp experiments in rats injected with DVC LV-MM (n=7), LV-

GlyT1 shRNA (n=7), or LV-GlyT1 shRNA with DVC MK801 infusion (n=6). Data are shown

as the mean + SEM.

Page 94: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

84

Supplementary Figure 3. 5 Metabolic effects of DVC and iv infusion of ALX in 3d-HFD

rats.

(A) Glucose uptake and (B) basal and clamp plasma glucose levels during clamps with DVC

infusion of saline (n=5), glycine (n=6), ALX (n=5), glycine+7CKNA (n=5), or ALX+7CKNA

(n=5) in 3-d HFD rats, (C) Glucose uptake and (D) basal and clamp plasma glucose levels during

clamps with iv infusion of 6% DMSO (n=7) or ALX (n=7) in 3-d HFD rats. Data are shown as

the mean + SEM.

Page 95: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

85

Supplementary Figure 3. 6 Metabolic effects of chemical and molecular inhibition of DVC

GlyT1 in obese rats.

(A) Glucose uptake and (B) basal and clamp plasma glucose levels during clamps in 28d RC-fed

rats with DVC infusion of saline (n=5) or ALX (n=5) and in 28d HFD-fed rats with DVC

infusion of saline (n=7) or ALX (n=7), (C) Glucose uptake and (D) basal and clamp plasma

glucose levels during clamps in 28d HFD-fed rats injected with DVC LV-GlyT1 shRNA (n=10)

or LV-MM (n=9). Data are shown as the mean + SEM.

Page 96: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

86

Chapter 4 Summary, Discussion and Future

Directions 4.1 Summary The ability of the CNS to detect and integrate peripheral humoral signals is an important

requirement for the regulation of glucose and energy homeostasis in a mammalian organism. The

current set of studies were aimed at further characterizing novel molecular mechanisms of CNS

hormone and nutrient sensing that control glucose and energy homeostasis.

Previously, our lab demonstrated that glucagon action in the MBH, via glucagon

receptor-PKA signaling pathway leads to an inhibition of hepatic glucose production, which is

disrupted in rats fed a high-fat diet for three days. This indicates hypothalamic glucagon

resistance exists in a pathological state. However, this resistance could be reversed by direct

activation of hypothalamic PKA, which implies, that the hypothalamic glucagon signaling defect

lies upstream of PKA. Investigating what lies downstream of PKA in MBH glucagon signaling

thus became of therapeutic interest, to identify targets that could potentially enhance or restore

hypothalamic glucagon action in diabetes and/or obesity. In hypothalamic cell lines, PKA has

been demonstrated to inhibit AMPK88 while MBH AMPK-mediated lipid sensing mechanisms

has been shown to lower glucose production102. It was unknown whether lipid sensing was a

downstream mechanism of MBH glucagon action to regulate glucose homeostasis. In Study 1,

we have shown that MBH glucagon does not signal through the lipid sensing axis involving

AMPK and PKC-δ rather activates KATP channels that lie downstream of MBH glucagon-PKA

signaling to lower glucose production (Figure 4.1).

Page 97: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

87

Similar to the MBH, the DVC is anatomically poised to detect hormones and nutrients to

regulate metabolic homeostasis. Direct administration of glycine into the DVC activates NMDA

receptors to lower glucose production in healthy rodents138. What remains to be shown is

whether DVC glycine-NMDA receptor axis works in diabetic and obese rodents to regulate

glucose homeostasis, as well as control energy balance. Given the poor pharmacokinetics of

glycine, Study 2 aimed to address whether inhibiting DVC GlyT1, the main regulator of glycine

levels for NMDA receptors, would sufficiently trigger endogenous glycine sensing to regulate

glucose and energy homeostasis. The major findings of these studies were that DVC GlyT1

inhibition elevates extracellular glycine availability to activate NMDA receptors in the DVC,

leading to improved glucose tolerance, lowered glucose production, reduced body weight gain

and food intake in healthy, diabetic and obese rodents (Figure 4.1). We have also reported that

intravenous infusion of GlyT1 inhibitors resulted in similar metabolic effects to those of DVC

GlyT1 inhibition. The significance of these major findings as well as potential avenues of further

research are discussed below.

Page 98: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

88

Figure 4. 1 Summary of Study 1 and Study 2.

Summarized representation illustrating that glucagon action in the MBH exerts an AMPK-

independent but a KATP channel- dependent pathway to exert glucose control, and that GlyT1

inhibition in the DVC sufficiently triggers glycine sensing in the DVC to modulate energy and

glucose homeostasis via activation of NMDA receptors.

Page 99: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

89

4.2 Discussion Ion channels in the CNS regulation of glucose and energy homeostasis.

The findings of this dissertation highlight the importance ion channels play in the CNS pathways

regulating glucose balance and food intake. KATP channels are one of the most ubiquitously

expressed ion channels in the brain209, where opening (or activation) of KATP channels induces

efflux of K+ ions and hyperpolarization of the cell membrane leading to suppressed neuronal

activity and excitability. In study 1, the loss of MBH glucagon’s effect to lower glucose

production by MBH injection of the dominant negative Kir6.2 mutant virus as well as by MBH

infusion of the pharmacological inhibitor glibenclamide is indicative that KATP channels mediate

the glucose regulating effects of MBH glucagon action. More specifically, our data suggests that

glucagon induces KATP channel-dependent hyperpolarization in MBH neurons to lower glucose

production. Consistent with this idea, our lab previously documented the co-localization of MBH

glucagon receptors with AgRP neurons79, which makes it conceivable that MBH glucagon

infusion could be causing hyperpolarization and inhibited firing of the orixegenic AgRP neurons

(mediated via KATP channel activation) to lower glucose production. Indeed, activated KATP

channels have been previously reported to mediate the hyperpolarization and inhibitory effects of

insulin on AgRP neurons, which in turn leads to suppressed glucose production21,210. Further,

Study 1 also corroborates those reports that showed pharmacologically depolarizing

hypothalamic KATP channels (via application of KATP channel inhibitors into the MBH) blocks

the glucose production lowering effects that are induced by MBH insulin, glucose, fatty acids or

leucine. Taken together, hypothalamic KATP channels represent a critical and common CNS ion

channel through which different types of hormones and nutrients regulate glucose homeostasis.

Although the role of KATP channels in the regulation of energy homeostasis remains elusive, it

has been shown that activation of hypothalamic KATP channels contributes to age-dependent

Page 100: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

90

obesity. For instance, in old mice, there is an increase of KATP channel activity, which causes

silencing of hypothalamic POMC neurons to reduce their leptin-induced release of the

anorexigenic α-MSH, displaying a hyperphagic and obese phenotype211. Investigation of KATP

channel activity within specific hypothalamic neurons (AgRP vs POMC) merits future

investigation for our thorough understanding of the role these channels play for energy

homeostasis.

Study 2 demonstrated that NMDA receptors, which are ligand-gated ion (Ca2+) channels,

in the DVC or more specifically the NTS, play a critical role in mediating the metabolic effects

of GlyT1 inhibition. Earlier studies have shown that NMDA receptor signaling relay ascending

signals from the gut to regulate glucose and energy homeostasis116,117,120, and that direct

activation of NTS NMDA receptors is sufficient138 and necessary121 for hypothalamic nutrient

sensing to lower hepatic glucose production. Recent studies further place emphasis on the role

NMDA receptor activation in the hindbrain play in the reduction of food intake and body

weight212. Upon co-activation by glutamate and glycine, NMDA receptors elicit membrane

depolarization of glutamatergic neurons and Ca2+-dependent signaling cascades via increased

conductance of Na+ and Ca2+ ions. Depolarization of the glutamatergic neurons in the NTS in

turn leads to excitation of DMV neurons, whose axons form the efferent limb of the vagus

nerve213. Consistently, in Study 2, we did show that DVC ALX treatments completely failed to

lower glucose production in hepatic-vagotomozied rats, confirming that glucose-regulation by

DVC GlyT1 inhibition indeed involves a specific neuronal relay via the vagus nerve to the liver.

Interestingly, the gluco-regulatory effects of MBH glucagon also required an intact

hepatic vagal signaling79. This together with the fact that NMDA receptors in the DVC can

integrate information from hypothalamic nutrient signals such as MBH lactate to regulate

Page 101: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

91

glucose homeostasis, it opens up the question as to whether MBH glucagon action (from Study1)

are also downstream mediated by NMDA receptors in the DVC. While future experiments are

warranted to test this hypothesis, there is some evidence showing that activation of KATP channel

and its neuronal hyperpolarization influence in fact play a protective role against over-

stimulation of NMDA receptors (or glutamate excitotoxicity), typically known to cause neuronal

cell death214. Interestingly, it is also suggested that NMDA receptor stimulation can lead to KATP

channel activation. For instance, NMDA receptor activation stimulates nitric oxide (NO)

production by Ca2+ -dependent activation of nitric oxide synthase (NOS)215-217, while NO-

mediated activation of cGMP-dependent protein kinase in turn can activate KATP channel

opening218. Whether this coupling of NMDA receptors to KATP channels or vice versa play a role

in CNS regulation of metabolic homeostasis remain to be investigated.

Indeed, NMDA receptors are also present in the hypothalamus but interestingly these

hypothalamic channels are shown to influence energy homeostasis in the opposite direction as in

the DVC. For instance, in the LH, it is the injection of NMDA receptor antagonists that lowers

feeding219 while intrahypothalamic injection of glutamate analogs, that are specific to NMDA

receptors, increases food intake220. Further, deletion of GluN1 or GluN2 subunits from

hypothalamic AgRP neurons cause lowering of food intake and body weight221,222. Nonetheless,

glutamatergic action and NMDA receptors in both the hypothalamus and the brainstem are

important for energy balance control. Of note, there is little evidence on the role of

hypothalamic NMDA receptors in regulating glucose homeostasis.

Page 102: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

92

The relevance of MBH glucagon action and DVC GlyT1 inhibition in health and disease

Indeed, the prime objective of this dissertation was to unveil novel molecular mechanisms in the

CNS that would serve as therapeutic targets to lower blood glucose, food intake and body weight

gain in diabetes and obesity. As our previous study has demonstrated that hypothalamic glucagon

resistance in the context of high fat feeding is manifested by the inability of glucagon receptor

signaling to activate PKA79, the findings of Study 1 indicate that activating hypothalamic KATP

channels may be therapeutically advantageous. In fact, activation of hypothalamic KATP channels

by oral administration of the KATP channel activator, diazoxide has already been implicated in

lowering glucose production in humans163. This study performed the same euglycemic –

somatostatin clamping technique as in our dissertation work, fixing the gluco-regulatory

hormones in circulation and showed that in healthy individuals, oral diazoxide treatment led to a

30% suppression of glucose production. Additional studies in healthy rats confirmed that

diazoxide’s inhibitory effects on glucose production are negated in the presence of the KATP

channel blocker glibenclamide, suggesting these effects are likely mediated by KATP channels in

the brain163. Furthermore, intranasal administration of insulin at doses that increases the insulin

concentration in the cerebrospinal fluid (CSF) led to a suppression of glucose production during

pancreatic clamps in humans, likely through activation of CNS KATP channels176.

Additionally, our study contributes to the evolving association between increased

glucagon action and a metabolically healthier phenotype- a theme that has been garnering

scientific attention in the recent years. Multiple studies have reported that activation of glucagon

receptors in conjunction with other G protein-coupled receptors is metabolically advantageous in

diabetes and obesity. For example, a triagonist aimed at simultaneous activation of glucagon,

GLP-1 and glucose-dependent insulinotropic (GIP) receptors improved metabolic and glycemic

profiles in obese and diabetic rodents223. In addition, dual activation of glucagon and GLP-1

Page 103: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

93

receptors normalized glucose tolerance and reduced food intake in mice with diet-induced

obesity224,225. These pre-clinical studies have attributed the beneficial effects of glucagon

primarily to increased energy expenditure and decreased food intake, and its hyperglycemic

effects to be countered by the actions of GLP-1 and/or GIP. Whether these polyagonists reach

the brain and activate central glucagon signalling to improve the diabetogenic effects in these

polyagonist therapeutic strategies remain to be investigated. In the same light, a recent

investigation reports that reduced glucagon suppression 2 hours after an intravenous glucose

challenge is associated with a healthier metabolic phenotype including lower BMI, higher insulin

sensitivity and reduced risk of impaired glucose tolerance226. These findings together with our

study reshape our understanding of glucagon’s physiological role in health and disease.

In Study 2, we took advantage of multiple disease models to test the therapeutic potential

of DVC GlyT1 inhibition/glycine sensing in diabetes and obesity. The fasting hyperglycemic

(type 2 diabetic) model used in our dissertation, arguably is the closest rodent model

recapitulating the pathogenesis of type 2 diabetes in humans. Elegantly described by Samuel et

al.191, a low dose of STZ protected by nicotinamide injection induces partial destruction of beta

cells, thereby preventing beta cell compensation for 7 d HFD-induced insulin resistance but still

maintaining basal insulin levels, consequently leading to fasting hyperglycemia secondary to an

elevation of hepatic glucose production179,180. Other rodent models of diabetes including the

Zucker diabetic fatty and Goto-Kakisaki diabetic rats as well as the db/db mice have their

diabetic characteristics confounded by a rise in glucocorticoid levels, which are not represented

in the hormonal profile of type 2 diabetic patients, and therefore are not accurate models of

clinical diabetes. In comparison, the plasma corticosterone levels are not increased in the

STZ/Nic/HFD model of hyperglycemic rats191. Importantly, we showed that 7d STZ/Nic/HFD

Page 104: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

94

diabetic rats are glucose intolerant and that infusion of GlyT1 inhibitor into the DVC leads to

improvement of glucose tolerance.

Further, our study indicates that DVC GlyT1 inhibition could also be effective as an early

intervention for insulin resistance and pre-diabetic conditions. While the 3d HFD-model we used

in Study 2 is not a diabetic model, it is a well-established model for hepatic insulin resistance

validated under hyperinsulinemic pancreatic clamp conditions28,227-229. In fact, a recent study

from our own lab testing the insulin sensitizing effects of resveratrol had validated 3d HFD rats

to be insulin resistant in regulating glucose production under hyperinsulinemic clamp conditions,

which could be reversed upon resveratrol infusion into the duodenum179. Whether GlyT1

inhibitors, like resveratrol, sensitizes insulin to regulate glucose production warrants

investigation; but under basal-insulin clamp conditions, our findings conclusively show that

DVC GlyT1 inhibition directly lowers glucose production independent of changes in basal

insulin levels. Further, based on the observation that 3d HFD rats display hyperphagia, a key

driving force of obesity, our data also implicate that DVC GlyT1 inhibition treatments are

effective in regulating glucose production in conditions of early onset diet-induced obesity as

well. However, instead of solely relying on an early onset obesity model (which is not in fact

obese), we also used a 28-d HFD obese model to show that DVC GlyT1 inhibition is effective in

regulating glucose production even at a later stage in diet-induced obesity. Previously, these 28d

HFD obese rats failed to show suppression of glucose production and stimulation of glucose

uptake compared to regular chow-fed rats under insulin-stimulated conditions (i.e.,

hyperinsulinemic clamps)179, thereby indicating hepatic and peripheral insulin resistance.

The therapeutic relevance of GlyT1 inhibition was further corroborated by our data

showing that systemic administration of GlyT1 inhibitors result in similar desirable metabolic

Page 105: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

95

effects to those of direct DVC GlyT1 inhibition. However, given that GlyT1 and NMDA

receptors are also present in non-neural tissues such as the pancreas, the possible side effects of

using GlyT1 inhibitors and/ or increasing glycine systemically will have to be assessed

cautiously. A previous study reports that inhibition of NMDA receptor transmission in the islets

enhances glucose-stimulated insulin secretion205. It is possible then, because glycine is a co-

agonist of the NMDA receptor, that systemic infusion of GlyT1 inhibitors and thus, elevated

systemic glycine levels would have the undesirable side effect of enhancing NMDA receptor

transmission in the islets and consequently lead to reduced insulin secretion- an effect especially

detrimental for Type 2 diabetic patients. Interestingly though, multiple studies have documented

circulating glycine levels to be inversely associated with Type 2 diabetes risk230-232. A recent

study has in fact validated that glycine treatment results in increased insulin secretion from intact

human islets, and a disruption in this glycine-insulin action contributes to impaired insulin

secretion in Type 2 diabetes233. Of particular note, the effect of glycine to stimulate insulin

secretion was mediated via activation of the glycine receptors (GlyR) in the pancreatic islets

since antagonism of GlyR with strynchnine-prevented glycine induced insulin secretion.

Whether elevation of endogenous glycine levels in the pancreas during systemic GlyT1

inhibition could potentiate NMDA receptors to alter insulin secretion merits future investigation.

The use of divergent and sometimes common signaling pathways by circulating hormones

and nutrients in the CNS to regulate glucose and energy homeostasis.

Recent studies have highlighted this theme that a commonly derived pathway for CNS

hormonal action could regulate both glucose and energy homeostasis. As described in Chapter 1,

insulin action in the MBH and the DVC regulates both energy and glucose homeostasis234. In the

MBH, it does via a PI3K-dependent pathway to regulate glucose and feeding control, whereas in

Page 106: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

96

the DVC, insulin signals through an ERK-dependent pathway to regulate feeding, body weight

and glucose production122. In regards to CNS glucagon action, a recent study by Quiñones et al.

reported that hypothalamic glucagon requires glucagon receptor and activation of downstream

PKA in the hypothalamic ARC to lower food intake, since icv. co-infusion of the glucagon

receptor antagonist des-His1-[Glu9] glucagon amide or the PKA inhibitor H-89 negated the

ability of central glucagon to decrease feeding235. Of note, these were the same chemical

inhibitors employed to confirm the role of glucagon receptor and PKA in mediating MBH

glucagon action on glucose control79, thereby suggesting that any associated molecular players in

the glucagon receptor–PKA branch are likely to be a part of a common pathway for both feeding

and glucose regulation by CNS glucagon. Moreover, Quiñones et al. also reported changes in

AgRP expression associated with the anorectic action of central glucagon, again consistent with

the observation that MBH glucagon receptors co-localised with AgRP neurons, thereby

demonstrating that in brain glucagon action, AgRP neurons mediate both glucose and feeding

regulation.

However, it appears that not the entire signaling pathway converges for the glycaemic

and satiety effects of hypothalamic glucagon action. Contrary to how MBH glucagon signals to

exert glucose control independent of MBH AMPK as shown in Study 1, the suppressive effect

central glucagon exerts on feeding involved inhibition of AMPK and activation of the

downstream target ACC. Specifically, molecular activation of AMPK in the ARC via injection

of a constitutively active AMPK virus blunted the anorectic effects of central glucagon

injections, whereas we showed that activation of MBH AMPK via the same viral approach had

no effect on the glucose production-lowering effect of MBH glucagon. Consistent with this,

there were decreased AMPK and increased ACC in the ARC associated with the satiety effect of

central glucagon injections, whereas we reported that the glucose-lowering effect of MBH

Page 107: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

97

glucagon infusions was associated with no differences in pACC/total ACC levels in the MBH. It

is likely these distinct mechanisms by which brain glucagon acts potentially allows for selective

and independent control of glucose and feeding regulation. It is also possible that there are

different populations of glucagon-responsive neurons within the MBH in which glucagon

signaling pathways might be distinct for glucose and energy regulation, and this warrants future

investigation. However, it cannot be overlooked that perhaps the differences in glucagon dose

and administration—a single bolus i.c.v. glucagon injection at a dose of 480 ng for feeding vs 2 h

of constant MBH glucagon infusion with a much lower dose of 3.6 pg for glucose control —

could explain some of the differences in the regulation of molecular targets for feeding and

glucose regulation by brain glucagon. Alternatively, these findings could be due to differences in

the times at which the glucagon-treated tissues were obtained for molecular analysis: changes in

AMPK and ACC, which mediate the anorectic action of hypothalamic glucagon, were measured

1 h after the i.c.v. glucagon single bolus injection vs after 2 h of constant infusion of MBH

glucagon.

Study 2 describes that DVC GlyT1 inhibition plays a role in glucose tolerance and

glucose production in response to elevated extracellular glycine levels and activated NMDA

receptors. Although the involvement of NMDA receptors in the food intake and body weight

regulation by DVC GlyT1 inhibition was not directly shown in our study, we are encouraged by

other reports that show that blockade of NMDA receptors in the DVC enhances feeding236, and

that activation of DVC NMDA receptors is also required for CCK’s vagally mediated

suppression of food intake143,237. Thus, by postulation, DVC NMDA receptors act as the common

downstream mediator for DVC GlyT1 inhibitory control of glucose and energy homeostasis.

Notably, though, other studies have shown the existence of differential mechanisms of CNS

nutrient sensing regulating glucose and energy balance. For instance, activation of the mTOR

Page 108: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

98

pathway is required for the inhibitory effect of MBH leucine on food intake111, while the

suppressive effect of MBH leucine on glucose production is independent of mTOR113. Whether

mechanisms of DVC GlyT1 inhibition (and DVC glycine sensing) diverge downstream of

NMDA receptors to allow for selective and autonomous control of feeding and glucose

regulation remain to be investigated.

In the pursuit of new pharmacological targets that would treat diabetes and obesity, in

order to design molecules where the main goal is to curb both hyperglycemia and hyperphagia it

becomes important to identify the point of convergence in these CNS fuel-sensing mechanisms

that control both glucose and energy homeostasis. However, it becomes equally important to

distinguish the point of divergence when the goal is to target one homeostatic regulation but not

the other. For instance, a lean diabetic individual would not require lowering body weight, as

opposed to reducing his glucose levels. In the same line, an obese non-diabetic individual would

only want to improve his energy balance as opposed to glucose regulation. Nonetheless, our

studies along with others’ point to the finding that hormonal action and nutrient sensing in the

CNS act via common as well as distinct signaling mechanisms to mediate glucose and energy

metabolism.

4.3 Limitations and Future directions Lack of a direct electrophysiological assessment confirming that MBH KATP channels were

inhibited by our molecular and pharmacological approaches is a major limitation in Study 1 that

concludes MBH glucagon activates KATP channels to lower glucose production. Given that

blockade of CNS KATP channels leads to membrane depolarization and increased electrical

activity, we acknowledge performing complementary patch-clamp studies to record changes in

membrane potential and firing activity, presumably, in AgRP neurons of MBH slices in response

Page 109: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

99

to DN Kir6.2 (vs GFP) or glibenclamide (vs saline) with glucagon treatment would have greatly

strengthened our findings. An important future direction of Study 1 would also be to show

evidence for a direct PKA-mediated phosphorylation of KATP channels in the hypothalamus. It

has been shown that Kir6.2 can be phosphorylated by PKA at S372 and at S1571 for SUR1 in

pancreatic beta cells164. We could transfect hypothalamic cells lines (e.g. GT1-7, known to

express AgRP) with either wild type Kir6.2 or mutant Kir6.2 cDNA (mutated at S372) together

with either wild type SUR1 or mutant SUR1 cDNA (mutated at S1571), and test their ability to

be phosphorylated after PKA stimulation or glucagon treatment using in vitro phosphorylation

assays.

Further, we also did not measure malonyl- and LCFA-CoA levels in Study 1 that

concludes MBH glucagon works through a lipid-sensing independent mechanism. Interestingly

though, studies show activating or inhibiting the AMPK -> malonly-CoA sensing pathway does

not always translate in changes in LCFA-CoA levels in the hypothalamus55. Exogenous leptin

administration has been shown to increase the levels of malonyl-CoA level without subsequently

affecting the LCFA-CoA levels in the ARC54 whereas ghrelin signaling in the hypothalamus is

known to increase LCFA-CoA levels while inhibiting hypothalamic ACC, which reduces the

malonyl-CoA level65. These studies cast doubt on the reliability of malonyl-CoA and LCFA-

CoA levels as readout for lipid sensing activation. Perhaps then, the strength of Study 1 ought to

be the fact that we did not solely focus on blocking the beginning of the lipid sensing pathway,

we also showed neither inhibition of MBH PKC-δ (blocking the end of the lipid sensing

pathway) affected MBH glucagon, thereby ruling out a lipid sensing dependent mechanism for

MBH glucagon’s effect of glucose homeostasis.

The principal findings of Study 1 were generated using a single manipulation of glucose

homeostasis, the pancreatic basal-insulin clamp. Though closer to physiological conditions than

Page 110: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

100

the hyperinsulinemic-euglycemic clamp, our findings would be even more convincing in light of

additional methods. Specifically, what is the relevance of this system in the post-prandial state,

where multiple systems are acting on glucose homeostasis in concert? Performing iv GTTs and

mixed-meal tolerance tests would provide further insight and confirmation into the MBH

glucagon signaling axis.

Given the work of polyagonist therapeutic studies targeting glucagon in conjunction with

GLP-1 and GIP receptors in the management of obesity and diabetes223, another important

question is whether concurrent infusions of glucagon, GLP-1 and GIP into the MBH would

result in redundant, additive or synergistic effects in lowering glucose production. This will

begin addressing whether the CNS penetrance of these polyagonists and whether activation of

MBH glucagon signalling plays a role in counteracting the hyperglycemic effects of peripheral

glucagon in these polyagonist therapeutic strategies.

We have identified, for the first time, that DVC GlyT1 inhibition and thus, DVC glycine

sensing plays a critical role in the regulation of energy balance as defined by changes in food

intake and body weight. However, components of energy expenditure (i.e. physical activity and

thermogenesis) are just as important on the energy balance equation. Given that obesity develops

when energy intake exceeds energy expenditure, the goal of any anti-obesity treatment should be

to reduce energy intake, promote energy expenditure, or both. Therefore, to truly repurpose

GlyT1 inhibitors as an effective obesity therapy, it becomes critical to determine the effect of

DVC GlyT1 inhibition on energy expenditure. Interestingly, the DVC as well as the NMDA

receptors in the DVC has directly been implicated in promoting the thermogenic activity of the

brown adipose tissue238,239. For instance, activation of DVC NMDA receptors is shown to

mediate the effect of increasing brown adipose thermogenesis in response to lipid infusion into

the duodenum239. In light of this, we would expect treatment with DVC GlyT1 inhibitors would

Page 111: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

101

sufficiently activate NMDA receptors in the DVC to increase energy expenditure. Indeed, to

confirm this, our next step would be to monitor rats treated with DVC ALX (vs. saline) and/or

injected with LV-GlyT1 shRNA (vs. LV-MM) using metabolic chambers, as described

previously240.

It should be noted that most of the viral vectors we used to modulate various molecular

targets in our studies including AMPK activity or GlyT1 expression were under the ubiquitous

promoter, cytomegalovirus (CMV), thereby altering gene expression in non-target cells which is

a limitation in our work. In the future, we could use cell-specific promoters such as the neuron

specific enolase (NSE) or glial fibrillary acidic protein (GFAP) to alter our molecular targets

specifically in neuronal cells versus glial cells, respectively241.

With state-of the-art technologies that allow real-time manipulation of genetically defined

neuronal populations, an important future direction would be to map out the hypothalamic and

neuronal circuitry in the DVC involved in the regulation of glucose and energy homeostasis. As

an example, NTS neurons are known to excite brain regions such as the lateral parabrachial

nucleus (PBN) to modulate feeding behaviour. Recently, distinct population of neurons in the

PBN expressing the neuropeptide calcitonin gene-related protein (CGRP) (CGRPPBN) were

identified to lower feeding242,243. It is likely that lateral PBN neurons or CGRPPBN neurons would

be a downstream target mediating the effects of DVC GlyT1 inhibition to lower feeding and

body weight gain, especially given that DVC GlyT1 inhibition activates NMDA receptors and

that glutamatergic signalling activates CGRPPBN neurons244. Interesting future direction would be

to check for c-fos labeling in CGRPPBN neurons following DVC ALX treatments as well as to

check whether optogenetically inactivating CGRPPBN neurons (using the inhibitory

channelrhodopsin protein construct) would abolish the anorectic effects of DVC GlyT1

inhibition.

Page 112: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

102

By contrast, our understanding of the glucoregulatory regulatory neurocircuits is far less

defined. However, a recent study employing optogenetic circuitry mapping approaches, revealed

that activating AgRP → LHA projections as well as activation of AgRP → anterior bed nucleus

of the stria terminalis (aBNST)vl projections impair systemic insulin sensitivity245. Clearly, future

studies are needed to know whether these neuronal circuits underlie MBH glucagon or DVC

GlyT1’s ability to alter peripheral glucose homeostasis.

Page 113: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

103

References

1 Bernard, C. Leçons sur les Phenonemes de la Vie Commune aux Animaux et aux Vegetaux. (Bailliere et Fils, 1879).

2 Spanswick, D., Smith, M. A., Mirshamsi, S., Routh, V. H. & Ashford, M. L. Insulin activates ATP-sensitive K+ channels in hypothalamic neurons of lean, but not obese rats. Nat. Neurosci. 3, 757-758, doi:10.1038/77660 (2000).

3 Levin, B. E. & Keesey, R. E. Defense of differing body weight set points in diet-induced obese and resistant rats. Am. J. Physiol. 274, R412-419 (1998).

4 Woods, S. C., Seeley, R. J., Rushing, P. A., D'Alessio, D. & Tso, P. A controlled high-fat diet induces an obese syndrome in rats. J. Nutr. 133, 1081-1087 (2003).

5 Van Wye, G., Dubin, J. A., Blair, S. N. & Dipietro, L. Adult obesity does not predict 6-year weight gain in men: the Aerobics Center Longitudinal Study. Obesity (Silver Spring) 15, 1571-1577, doi:10.1038/oby.2007.186 (2007).

6 Beagley, J., Guariguata, L., Weil, C. & Motala, A. A. Global estimates of undiagnosed diabetes in adults. Diabetes Res. Clin. Pract. 103, 150-160, doi:10.1016/j.diabres.2013.11.001 (2014).

7 Mendis, S., Davis, S. & Norrving, B. Organizational update: the world health organization global status report on noncommunicable diseases 2014; one more landmark step in the combat against stroke and vascular disease. Stroke 46, e121-122, doi:10.1161/STROKEAHA.115.008097 (2015).

8 Ng, M. et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766-781, doi:10.1016/S0140-6736(14)60460-8 (2014).

9 Association, C. D. An economic tsunami: the cose of diabetes in Canada. (2009).

10 Anis, A. H. et al. Obesity and overweight in Canada: an updated cost-of-illness study. Obes. Rev. 11, 31-40, doi:10.1111/j.1467-789X.2009.00579.x (2010).

11 Roh, E., Song do, K. & Kim, M. S. Emerging role of the brain in the homeostatic regulation of energy and glucose metabolism. Exp. Mol. Med. 48, e216, doi:10.1038/emm.2016.4 (2016).

12 Chen, M., Woods, S. C. & Porte, D., Jr. Effect of cerebral intraventricular insulin on pancreatic insulin secretion in the dog. Diabetes 24, 910-914 (1975).

13 Woods, S. C., Lotter, E. C., McKay, L. D. & Porte, D., Jr. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 282, 503-505 (1979).

Page 114: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

104

14 Harai, S., Ikenaga, T. & Matsuzawa, T. Nasal absorption of insulin in dogs. Diabetes 27, 296-299 (1978).

15 Anand Kumar, T. C., Sehgal A. & Bajaj, J. S. Intranasal administration of insulin reduces circulating levels of glucose in the Rhesus monkey. Vol. 49 (Current Science, 1980).

16 Obici, S., Feng, Z., Karkanias, G., Baskin, D. G. & Rossetti, L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat. Neurosci. 5, 566-572, doi:10.1038/nn861 (2002).

17 Gelling, R. W. et al. Insulin action in the brain contributes to glucose lowering during insulin treatment of diabetes. Cell Metab. 3, 67-73, doi:10.1016/j.cmet.2005.11.013 (2006).

18 Pocai, A. et al. Hypothalamic K(ATP) channels control hepatic glucose production. Nature 434, 1026-1031, doi:10.1038/nature03439 (2005).

19 Inoue, H. et al. Role of hepatic STAT3 in brain-insulin action on hepatic glucose production. Cell Metab. 3, 267-275, doi:10.1016/j.cmet.2006.02.009 (2006).

20 Kimura, K. et al. Central Insulin Action Activates Kupffer Cells by Suppressing Hepatic Vagal Activation via the Nicotinic Alpha 7 Acetylcholine Receptor. Cell Rep 14, 2362-2374, doi:10.1016/j.celrep.2016.02.032 (2016).

21 Konner, A. C. et al. Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production. Cell Metab. 5, 438-449, doi:10.1016/j.cmet.2007.05.004 (2007).

22 Lin, H. V. et al. Divergent regulation of energy expenditure and hepatic glucose production by insulin receptor in agouti-related protein and POMC neurons. Diabetes 59, 337-346, doi:10.2337/db09-1303 (2010).

23 Okamoto, H. et al. Transgenic rescue of insulin receptor-deficient mice. J. Clin. Invest. 114, 214-223, doi:10.1172/JCI21645 (2004).

24 Carvalheira, J. B. et al. Selective impairment of insulin signalling in the hypothalamus of obese Zucker rats. Diabetologia 46, 1629-1640, doi:10.1007/s00125-003-1246-x (2003).

25 De Souza, C. T. et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 146, 4192-4199, doi:10.1210/en.2004-1520 (2005).

26 Posey, K. A. et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. Am. J. Physiol. Endocrinol. Metab. 296, E1003-1012, doi:10.1152/ajpendo.90377.2008 (2009).

Page 115: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

105

27 Begg, D. P. et al. Reversal of diet-induced obesity increases insulin transport into cerebrospinal fluid and restores sensitivity to the anorexic action of central insulin in male rats. Endocrinology 154, 1047-1054, doi:10.1210/en.2012-1929 (2013).

28 Ono, H. et al. Activation of hypothalamic S6 kinase mediates diet-induced hepatic insulin resistance in rats. J. Clin. Invest. 118, 2959-2968, doi:10.1172/JCI34277 (2008).

29 Purkayastha, S. et al. Neural dysregulation of peripheral insulin action and blood pressure by brain endoplasmic reticulum stress. Proc. Natl. Acad. Sci. U. S. A. 108, 2939-2944, doi:10.1073/pnas.1006875108 (2011).

30 Meek, T. H. & Morton, G. J. Leptin, diabetes, and the brain. Indian J. Endocrinol. Metab. 16, S534-542, doi:10.4103/2230-8210.105568 (2012).

31 Liu, L. et al. Intracerebroventricular leptin regulates hepatic but not peripheral glucose fluxes. J. Biol. Chem. 273, 31160-31167 (1998).

32 Kamohara, S., Burcelin, R., Halaas, J. L., Friedman, J. M. & Charron, M. J. Acute stimulation of glucose metabolism in mice by leptin treatment. Nature 389, 374-377, doi:10.1038/38717 (1997).

33 Campfield, L. A., Smith, F. J., Guisez, Y., Devos, R. & Burn, P. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269, 546-549 (1995).

34 Asilmaz, E. et al. Site and mechanism of leptin action in a rodent form of congenital lipodystrophy. J. Clin. Invest. 113, 414-424, doi:10.1172/JCI19511 (2004).

35 Lin, C. Y., Higginbotham, D. A., Judd, R. L. & White, B. D. Central leptin increases insulin sensitivity in streptozotocin-induced diabetic rats. Am. J. Physiol. Endocrinol. Metab. 282, E1084-1091, doi:10.1152/ajpendo.00489.2001 (2002).

36 Hidaka, S. et al. Chronic central leptin infusion restores hyperglycemia independent of food intake and insulin level in streptozotocin-induced diabetic rats. FASEB J. 16, 509-518 (2002).

37 Coppari, R. et al. The hypothalamic arcuate nucleus: a key site for mediating leptin's effects on glucose homeostasis and locomotor activity. Cell Metab. 1, 63-72, doi:10.1016/j.cmet.2004.12.004 (2005).

38 Morton, G. J. et al. Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons. Cell Metab. 2, 411-420, doi:10.1016/j.cmet.2005.10.009 (2005).

39 Satoh, N. et al. The arcuate nucleus as a primary site of satiety effect of leptin in rats. Neurosci. Lett. 224, 149-152, doi:10.1016/S0304-3940(97)00163-8 (1997).

40 Bates, S. H. et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 421, 856-859, doi:10.1038/nature01388 (2003).

Page 116: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

106

41 Bates, S. H. et al. LRb-STAT3 signaling is required for the neuroendocrine regulation of energy expenditure by leptin. Diabetes 53, 3067-3073 (2004).

42 Gao, Q. et al. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc. Natl. Acad. Sci. U. S. A. 101, 4661-4666, doi:10.1073/pnas.0303992101 (2004).

43 Gong, L. et al. Signal transducer and activator of transcription-3 is required in hypothalamic agouti-related protein/neuropeptide Y neurons for normal energy homeostasis. Endocrinology 149, 3346-3354, doi:10.1210/en.2007-0945 (2008).

44 Xu, A. W., Ste-Marie, L., Kaelin, C. B. & Barsh, G. S. Inactivation of signal transducer and activator of transcription 3 in proopiomelanocortin (Pomc) neurons causes decreased pomc expression, mild obesity, and defects in compensatory refeeding. Endocrinology 148, 72-80, doi:10.1210/en.2006-1119 (2007).

45 Vong, L. et al. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 71, 142-154, doi:10.1016/j.neuron.2011.05.028 (2011).

46 Buettner, C. et al. Critical role of STAT3 in leptin's metabolic actions. Cell Metab. 4, 49-60, doi:10.1016/j.cmet.2006.04.014 (2006).

47 Rahmouni, K., Sigmund, C. D., Haynes, W. G. & Mark, A. L. Hypothalamic ERK mediates the anorectic and thermogenic sympathetic effects of leptin. Diabetes 58, 536-542, doi:10.2337/db08-0822 (2009).

48 do Carmo, J. M. et al. Shp2 signaling in POMC neurons is important for leptin's actions on blood pressure, energy balance, and glucose regulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 307, R1438-1447, doi:10.1152/ajpregu.00131.2014 (2014).

49 Niswender, K. D. et al. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature 413, 794-795, doi:10.1038/35101657 (2001).

50 Hill, J. W. et al. Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice. J. Clin. Invest. 118, 1796-1805, doi:10.1172/JCI32964 (2008).

51 Hill, J. W. et al. Phosphatidyl inositol 3-kinase signaling in hypothalamic proopiomelanocortin neurons contributes to the regulation of glucose homeostasis. Endocrinology 150, 4874-4882, doi:10.1210/en.2009-0454 (2009).

52 Spanswick, D., Smith, M. A., Groppi, V. E., Logan, S. D. & Ashford, M. L. Leptin inhibits hypothalamic neurons by activation of ATP-sensitive potassium channels. Nature 390, 521-525, doi:10.1038/37379 (1997).

53 Minokoshi, Y. et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 428, 569-574, doi:10.1038/nature02440 (2004).

Page 117: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

107

54 Gao, S. et al. Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit food intake. Proc. Natl. Acad. Sci. U. S. A. 104, 17358-17363, doi:10.1073/pnas.0708385104 (2007).

55 Gao, S. et al. Malonyl-CoA mediates leptin hypothalamic control of feeding independent of inhibition of CPT-1a. Am. J. Physiol. Regul. Integr. Comp. Physiol. 301, R209-217, doi:10.1152/ajpregu.00092.2011 (2011).

56 Widdowson, P. S., Upton, R., Buckingham, R., Arch, J. & Williams, G. Inhibition of food response to intracerebroventricular injection of leptin is attenuated in rats with diet-induced obesity. Diabetes 46, 1782-1785 (1997).

57 Pocai, A. et al. Central leptin acutely reverses diet-induced hepatic insulin resistance. Diabetes 54, 3182-3189 (2005).

58 Metlakunta, A. S., Sahu, M. & Sahu, A. Hypothalamic phosphatidylinositol 3-kinase pathway of leptin signaling is impaired during the development of diet-induced obesity in FVB/N mice. Endocrinology 149, 1121-1128, doi:10.1210/en.2007-1307 (2008).

59 Zigman, J. M., Jones, J. E., Lee, C. E., Saper, C. B. & Elmquist, J. K. Expression of ghrelin receptor mRNA in the rat and the mouse brain. J. Comp. Neurol. 494, 528-548, doi:10.1002/cne.20823 (2006).

60 Wang, Q. et al. Arcuate AgRP neurons mediate orexigenic and glucoregulatory actions of ghrelin. Mol Metab 3, 64-72, doi:10.1016/j.molmet.2013.10.001 (2014).

61 Lawrence, C. B., Snape, A. C., Baudoin, F. M. & Luckman, S. M. Acute central ghrelin and GH secretagogues induce feeding and activate brain appetite centers. Endocrinology 143, 155-162, doi:10.1210/endo.143.1.8561 (2002).

62 Stark, R. et al. Acyl ghrelin acts in the brain to control liver function and peripheral glucose homeostasis in male mice. Endocrinology 156, 858-868, doi:10.1210/en.2014-1733 (2015).

63 Kola, B. et al. Cannabinoids and ghrelin have both central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. J. Biol. Chem. 280, 25196-25201, doi:10.1074/jbc.C500175200 (2005).

64 Lopez, M. et al. Hypothalamic fatty acid metabolism mediates the orexigenic action of ghrelin. Cell Metab. 7, 389-399, doi:10.1016/j.cmet.2008.03.006 (2008).

65 Andrews, Z. B. et al. UCP2 mediates ghrelin's action on NPY/AgRP neurons by lowering free radicals. Nature 454, 846-851, doi:10.1038/nature07181 (2008).

66 Jiang, G. & Zhang, B. B. Glucagon and regulation of glucose metabolism. Am. J. Physiol. Endocrinol. Metab. 284, E671-678, doi:10.1152/ajpendo.00492.2002 (2003).

Page 118: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

108

67 Beale, E., Andreone, T., Koch, S., Granner, M. & Granner, D. Insulin and glucagon regulate cytosolic phosphoenolpyruvate carboxykinase (GTP) mRNA in rat liver. Diabetes 33, 328-332 (1984).

68 Lok, S. et al. The human glucagon receptor encoding gene: structure, cDNA sequence and chromosomal localization. Gene 140, 203-209 (1994).

69 Dunphy, J. L., Taylor, R. G. & Fuller, P. J. Tissue distribution of rat glucagon receptor and GLP-1 receptor gene expression. Mol. Cell. Endocrinol. 141, 179-186 (1998).

70 Hoosein, N. M. & Gurd, R. S. Identification of glucagon receptors in rat brain. Proc. Natl. Acad. Sci. U. S. A. 81, 4368-4372 (1984).

71 Wetsel, W. C., Eraly, S. A., Whyte, D. B. & Mellon, P. L. Regulation of gonadotropin-releasing hormone by protein kinase-A and -C in immortalized hypothalamic neurons. Endocrinology 132, 2360-2370, doi:10.1210/endo.132.6.8504741 (1993).

72 Graner, J. L. & Abraira, C. Glucagon in the cerebrospinal fluid. N. Engl. J. Med. 312, 994-995 (1985).

73 Inokuchi, A., Oomura, Y., Shimizu, N. & Yamamoto, T. Central action of glucagon in rat hypothalamus. Am. J. Physiol. 250, R120-126 (1986).

74 Marubashi, S. et al. Hyperglycaemic effect of glucagon administered intracerebroventricularly in the rat. Acta Endocrinol. (Copenh.) 108, 6-10 (1985).

75 Honda, K. et al. The mechanism underlying the central glucagon-induced hyperglycemia and anorexia in chicks. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 163, 260-264, doi:10.1016/j.cbpa.2012.08.005 (2012).

76 Amir, S. Central glucagon-induced hyperglycemia is mediated by combined activation of the adrenal medulla and sympathetic nerve endings. Physiol. Behav. 37, 563-566 (1986).

77 Agarwala, G. C. & Bapat, S. K. Effect of centrally administered glucagon on blood glucose levels in dogs. Indian J. Med. Res. 66, 323-330 (1977).

78 Agarwala, G. C., Mishra, R., Jaiswal, G. & Bapat, V. Effect of centrally administered glucagon on liver glycogen & enzymes in anaesthetised dogs. Indian J. Med. Res. 90, 372-378 (1989).

79 Mighiu, P. I. et al. Hypothalamic glucagon signaling inhibits hepatic glucose production. Nat. Med. 19, 766-772, doi:10.1038/nm.3115 (2013).

80 Eigler, N., Sacca, L. & Sherwin, R. S. Synergistic interactions of physiologic increments of glucagon, epinephrine, and cortisol in the dog: a model for stress-induced hyperglycemia. J. Clin. Invest. 63, 114-123, doi:10.1172/JCI109264 (1979).

Page 119: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

109

81 Felig, P., Wahren, J. & Hendler, R. Influence of physiologic hyperglucagonemia on basal and insulin-inhibited splanchnic glucose output in normal man. J. Clin. Invest. 58, 761-765, doi:10.1172/JCI108523 (1976).

82 Bomboy, J. D., Jr., Lewis, S. B., Lacy, W. W., Sinclair-Smith, B. C. & Liljenquist, J. E. Transient stimulatory effect of sustained hyperglucagonemia on splanchnic glucose production in normal and diabetic man. Diabetes 26, 177-174 (1977).

83 Muller, W. A., Faloona, G. R., Aguilar-Parada, E. & Unger, R. H. Abnormal alpha-cell function in diabetes. Response to carbohydrate and protein ingestion. N. Engl. J. Med. 283, 109-115, doi:10.1056/NEJM197007162830301 (1970).

84 Reaven, G. M., Chen, Y. D., Golay, A., Swislocki, A. L. & Jaspan, J. B. Documentation of hyperglucagonemia throughout the day in nonobese and obese patients with noninsulin-dependent diabetes mellitus. J. Clin. Endocrinol. Metab. 64, 106-110, doi:10.1210/jcem-64-1-106 (1987).

85 Johnson, D. G., Goebel, C. U., Hruby, V. J., Bregman, M. D. & Trivedi, D. Hyperglycemia of diabetic rats decreased by a glucagon receptor antagonist. Science 215, 1115-1116 (1982).

86 Brand, C. L. et al. Immunoneutralization of endogenous glucagon with monoclonal glucagon antibody normalizes hyperglycaemia in moderately streptozotocin-diabetic rats. Diabetologia 37, 985-993 (1994).

87 Djouder, N. et al. PKA phosphorylates and inactivates AMPKalpha to promote efficient lipolysis. EMBO J. 29, 469-481, doi:10.1038/emboj.2009.339 (2010).

88 Damm, E., Buech, T. R., Gudermann, T. & Breit, A. Melanocortin-induced PKA activation inhibits AMPK activity via ERK-1/2 and LKB-1 in hypothalamic GT1-7 cells. Mol. Endocrinol. 26, 643-654, doi:10.1210/me.2011-1218 (2012).

89 Inokuchi, A., Oomura, Y. & Nishimura, H. Effect of intracerebroventricularly infused glucagon on feeding behavior. Physiol. Behav. 33, 397-400 (1984).

90 Honda, K. et al. Central administration of glucagon suppresses food intake in chicks. Neurosci. Lett. 416, 198-201, doi:10.1016/j.neulet.2007.02.011 (2007).

91 Kurose, Y. et al. Effects of central administration of glucagon on feed intake and endocrine responses in sheep. Anim Sci J 80, 686-690, doi:10.1111/j.1740-0929.2009.00685.x (2009).

92 Geary, N., Kissileff, H. R., Pi-Sunyer, F. X. & Hinton, V. Individual, but not simultaneous, glucagon and cholecystokinin infusions inhibit feeding in men. Am. J. Physiol. 262, R975-980 (1992).

93 Morgan, K., Obici, S. & Rossetti, L. Hypothalamic responses to long-chain fatty acids are nutritionally regulated. J. Biol. Chem. 279, 31139-31148, doi:10.1074/jbc.M400458200 (2004).

Page 120: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

110

94 Lam, T. K. et al. Hypothalamic sensing of circulating fatty acids is required for glucose homeostasis. Nat. Med. 11, 320-327, doi:10.1038/nm1201 (2005).

95 Rapoport, S. I. In vivo labeling of brain phospholipids by long-chain fatty acids: relation to turnover and function. Lipids 31 Suppl, S97-101 (1996).

96 Mitchell, R. W., Edmundson, C. L., Miller, D. W. & Hatch, G. M. On the mechanism of oleate transport across human brain microvessel endothelial cells. J. Neurochem. 110, 1049-1057, doi:10.1111/j.1471-4159.2009.06199.x (2009).

97 Le Foll, C., Irani, B. G., Magnan, C., Dunn-Meynell, A. A. & Levin, B. E. Characteristics and mechanisms of hypothalamic neuronal fatty acid sensing. Am. J. Physiol. Regul. Integr. Comp. Physiol. 297, R655-664, doi:10.1152/ajpregu.00223.2009 (2009).

98 Le Foll, C., Dunn-Meynell, A., Musatov, S., Magnan, C. & Levin, B. E. FAT/CD36: a major regulator of neuronal fatty acid sensing and energy homeostasis in rats and mice. Diabetes 62, 2709-2716, doi:10.2337/db12-1689 (2013).

99 Obici, S., Feng, Z., Arduini, A., Conti, R. & Rossetti, L. Inhibition of hypothalamic carnitine palmitoyltransferase-1 decreases food intake and glucose production. Nat. Med. 9, 756-761, doi:10.1038/nm873 (2003).

100 McGarry, J. D. What if Minkowski had been ageusic? An alternative angle on diabetes. Science 258, 766-770 (1992).

101 Yue, J. T. & Lam, T. K. Lipid sensing and insulin resistance in the brain. Cell Metab. 15, 646-655, doi:10.1016/j.cmet.2012.01.013 (2012).

102 Yang, C. S. et al. Hypothalamic AMP-activated protein kinase regulates glucose production. Diabetes 59, 2435-2443, doi:10.2337/db10-0221 (2010).

103 Obici, S. et al. Central administration of oleic acid inhibits glucose production and food intake. Diabetes 51, 271-275 (2002).

104 Pocai, A., Obici, S., Schwartz, G. J. & Rossetti, L. A brain-liver circuit regulates glucose homeostasis. Cell Metab. 1, 53-61, doi:10.1016/j.cmet.2004.11.001 (2005).

105 Ross, R. et al. Hypothalamic protein kinase C regulates glucose production. Diabetes 57, 2061-2065, doi:10.2337/db08-0206 (2008).

106 Pocai, A. et al. Restoration of hypothalamic lipid sensing normalizes energy and glucose homeostasis in overfed rats. J. Clin. Invest. 116, 1081-1091, doi:10.1172/JCI26640 (2006).

107 Lam, T. K., Gutierrez-Juarez, R., Pocai, A. & Rossetti, L. Regulation of blood glucose by hypothalamic pyruvate metabolism. Science 309, 943-947, doi:10.1126/science.1112085 (2005).

Page 121: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

111

108 Wolfgang, M. J. et al. Regulation of hypothalamic malonyl-CoA by central glucose and leptin. Proc. Natl. Acad. Sci. U. S. A. 104, 19285-19290, doi:10.1073/pnas.0709778104 (2007).

109 Chari, M. et al. Glucose transporter-1 in the hypothalamic glial cells mediates glucose sensing to regulate glucose production in vivo. Diabetes 60, 1901-1906, doi:10.2337/db11-0120 (2011).

110 Chari, M., Lam, C. K., Wang, P. Y. & Lam, T. K. Activation of central lactate metabolism lowers glucose production in uncontrolled diabetes and diet-induced insulin resistance. Diabetes 57, 836-840, doi:10.2337/db07-1464 (2008).

111 Blouet, C., Jo, Y. H., Li, X. & Schwartz, G. J. Mediobasal hypothalamic leucine sensing regulates food intake through activation of a hypothalamus-brainstem circuit. J. Neurosci. 29, 8302-8311, doi:10.1523/JNEUROSCI.1668-09.2009 (2009).

112 Kanamori, K., Ross, B. D. & Kondrat, R. W. Rate of glutamate synthesis from leucine in rat brain measured in vivo by 15N NMR. J. Neurochem. 70, 1304-1315 (1998).

113 Su, Y. et al. Hypothalamic leucine metabolism regulates liver glucose production. Diabetes 61, 85-93, doi:10.2337/db11-0857 (2012).

114 Cota, D. et al. Hypothalamic mTOR signaling regulates food intake. Science 312, 927-930, doi:10.1126/science.1124147 (2006).

115 Arrieta-Cruz, I., Su, Y. & Gutierrez-Juarez, R. Suppression of Endogenous Glucose Production by Isoleucine and Valine and Impact of Diet Composition. Nutrients 8, 79, doi:10.3390/nu8020079 (2016).

116 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).

117 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).

118 Lam, T. K. Neuronal regulation of homeostasis by nutrient sensing. Nat. Med. 16, 392-395, doi:10.1038/nm0410-392 (2010).

119 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).

120 Cummings, D. E. & Overduin, J. Gastrointestinal regulation of food intake. J. Clin. Invest. 117, 13-23, doi:10.1172/JCI30227 (2007).

121 Lam, C. K., Chari, M., Rutter, G. A. & Lam, T. K. Hypothalamic nutrient sensing activates a forebrain-hindbrain neuronal circuit to regulate glucose production in vivo. Diabetes 60, 107-113, doi:10.2337/db10-0994 (2011).

Page 122: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

112

122 Filippi, B. M., Yang, C. S., Tang, C. & Lam, T. K. Insulin activates Erk1/2 signaling in the dorsal vagal complex to inhibit glucose production. Cell Metab. 16, 500-510, doi:10.1016/j.cmet.2012.09.005 (2012).

123 Obici, S., Zhang, B. B., Karkanias, G. & Rossetti, L. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat. Med. 8, 1376-1382, doi:10.1038/nm798 (2002).

124 Filippi, B. M. et al. Insulin signals through the dorsal vagal complex to regulate energy balance. Diabetes 63, 892-899, doi:10.2337/db13-1044 (2014).

125 Grill, H. J. et al. Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology 143, 239-246, doi:10.1210/endo.143.1.8589 (2002).

126 Boghossian, S., Lecklin, A., Dube, M. G., Kalra, P. S. & Kalra, S. P. Increased leptin expression in the dorsal vagal complex suppresses adiposity without affecting energy intake and metabolic hormones. Obesity (Silver Spring) 14, 1003-1009, doi:10.1038/oby.2006.115 (2006).

127 Hansen, L. H., Abrahamsen, N. & Nishimura, E. Glucagon receptor mRNA distribution in rat tissues. Peptides 16, 1163-1166 (1995).

128 Parker, J. A. et al. Glucagon and GLP-1 inhibit food intake and increase c-fos expression in similar appetite regulating centres in the brainstem and amygdala. Int. J. Obes. (Lond.) 37, 1391-1398, doi:10.1038/ijo.2012.227 (2013).

129 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).

130 Mizuno, Y. & Oomura, Y. Glucose responding neurons in the nucleus tractus solitarius of the rat: in vitro study. Brain Res. 307, 109-116 (1984).

131 Balfour, R. H., Hansen, A. M. & Trapp, S. Neuronal responses to transient hypoglycaemia in the dorsal vagal complex of the rat brainstem. J. Physiol. 570, 469-484, doi:10.1113/jphysiol.2005.098822 (2006).

132 Ferreira, M., Jr. et al. Glucose effects on gastric motility and tone evoked from the rat dorsal vagal complex. J. Physiol. 536, 141-152 (2001).

133 Ritter, S., Dinh, T. T. & Zhang, Y. Localization of hindbrain glucoreceptive sites controlling food intake and blood glucose. Brain Res. 856, 37-47 (2000).

134 Blouet, C. & Schwartz, G. J. Brainstem nutrient sensing in the nucleus of the solitary tract inhibits feeding. Cell Metab. 16, 579-587, doi:10.1016/j.cmet.2012.10.003 (2012).

Page 123: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

113

135 Cavanaugh, A. R., Schwartz, G. J. & Blouet, C. High-fat feeding impairs nutrient sensing and gut brain integration in the caudomedial nucleus of the solitary tract in mice. PLoS One 10, e0118888, doi:10.1371/journal.pone.0118888 (2015).

136 Cassell, M. D., Roberts, L. & Talman, W. T. Glycine-containing terminals in the rat dorsal vagal complex. Neuroscience 50, 907-920 (1992).

137 Paoletti, P. & Neyton, J. NMDA receptor subunits: function and pharmacology. Curr. Opin. Pharmacol. 7, 39-47, doi:10.1016/j.coph.2006.08.011 (2007).

138 Lam, C. K. et al. Activation of N-methyl-D-aspartate (NMDA) receptors in the dorsal vagal complex lowers glucose production. J. Biol. Chem. 285, 21913-21921, doi:10.1074/jbc.M109.087338 (2010).

139 Yue, J. T., Mighiu, P. I., Naples, M., Adeli, K. & Lam, T. K. Glycine normalizes hepatic triglyceride-rich VLDL secretion by triggering the CNS in high-fat fed rats. Circ. Res. 110, 1345-1354, doi:10.1161/CIRCRESAHA.112.268276 (2012).

140 Kleckner, N. W. & Dingledine, R. Requirement for glycine in activation of NMDA-receptors expressed in Xenopus oocytes. Science 241, 835-837 (1988).

141 Baptista, V. & Varanda, W. A. Glycine binding site of the synaptic NMDA receptor in subpostremal NTS neurons. J. Neurophysiol. 94, 147-152, doi:10.1152/jn.00927.2004 (2005).

142 Bergeron, R., Meyer, T. M., Coyle, J. T. & Greene, R. W. Modulation of N-methyl-D-aspartate receptor function by glycine transport. Proc. Natl. Acad. Sci. U. S. A. 95, 15730-15734 (1998).

143 Wright, J. et al. Reduction of food intake by cholecystokinin requires activation of hindbrain NMDA-type glutamate receptors. Am. J. Physiol. Regul. Integr. Comp. Physiol. 301, R448-455, doi:10.1152/ajpregu.00026.2011 (2011).

144 Noetzel, M. J., Jones, C. K. & Conn, P. J. Emerging approaches for treatment of schizophrenia: modulation of glutamatergic signaling. Discov. Med. 14, 335-343 (2012).

145 Kawai, N. et al. Pharmacokinetics and cerebral distribution of glycine administered to rats. Amino Acids 42, 2129-2137, doi:10.1007/s00726-011-0950-y (2012).

146 Heresco-Levy, U. et al. Efficacy of high-dose glycine in the treatment of enduring negative symptoms of schizophrenia. Arch. Gen. Psychiatry 56, 29-36 (1999).

147 Heresco-Levy, U., Ermilov, M., Lichtenberg, P., Bar, G. & Javitt, D. C. High-dose glycine added to olanzapine and risperidone for the treatment of schizophrenia. Biol. Psychiatry 55, 165-171 (2004).

148 Buchanan, R. W. et al. The Cognitive and Negative Symptoms in Schizophrenia Trial (CONSIST): the efficacy of glutamatergic agents for negative symptoms and cognitive

Page 124: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

114

impairments. Am. J. Psychiatry 164, 1593-1602, doi:10.1176/appi.ajp.2007.06081358 (2007).

149 Betz, H., Gomeza, J., Armsen, W., Scholze, P. & Eulenburg, V. Glycine transporters: essential regulators of synaptic transmission. Biochem. Soc. Trans. 34, 55-58, doi:10.1042/BST0340055 (2006).

150 Black, M. D. et al. Procognitive and antipsychotic efficacy of glycine transport 1 inhibitors (GlyT1) in acute and neurodevelopmental models of schizophrenia: latent inhibition studies in the rat. Psychopharmacology (Berl.) 202, 385-396, doi:10.1007/s00213-008-1289-2 (2009).

151 Pinard, E. et al. Selective GlyT1 inhibitors: discovery of [4-(3-fluoro-5-trifluoromethylpyridin-2-yl)piperazin-1-yl][5-methanesulfonyl-2-(( S)-2,2,2-trifluoro-1-methylethoxy)phenyl]methanone (RG1678), a promising novel medicine to treat schizophrenia. J. Med. Chem. 53, 4603-4614, doi:10.1021/jm100210p (2010).

152 Boulay, D., Bergis, O., Avenet, P. & Griebel, G. The glycine transporter-1 inhibitor SSR103800 displays a selective and specific antipsychotic-like profile in normal and transgenic mice. Neuropsychopharmacology 35, 416-427, doi:10.1038/npp.2009.144 (2010).

153 D'Souza, D. C. et al. Glycine transporter inhibitor attenuates the psychotomimetic effects of ketamine in healthy males: preliminary evidence. Neuropsychopharmacology 37, 1036-1046, doi:10.1038/npp.2011.295 (2012).

154 Garcia-Caceres, C., Fuente-Martin, E., Argente, J. & Chowen, J. A. Emerging role of glial cells in the control of body weight. Mol Metab 1, 37-46, doi:10.1016/j.molmet.2012.07.001 (2012).

155 Garcia-Caceres, C. et al. Astrocytic Insulin Signaling Couples Brain Glucose Uptake with Nutrient Availability. Cell 166, 867-880, doi:10.1016/j.cell.2016.07.028 (2016).

156 Kim, J. G. et al. Leptin signaling in astrocytes regulates hypothalamic neuronal circuits and feeding. Nat. Neurosci. 17, 908-910, doi:10.1038/nn.3725 (2014).

157 Gao, Y. et al. Disruption of Lipid Uptake in Astroglia Exacerbates Diet Induced Obesity. Diabetes, doi:10.2337/db16-1278 (2017).

158 Pellerin, L. & Magistretti, P. J. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc. Natl. Acad. Sci. U. S. A. 91, 10625-10629 (1994).

159 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).

Page 125: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

115

160 Baczko, I. et al. Plasma membrane KATP channel-mediated cardioprotection involves posthypoxic reductions in calcium overload and contractile dysfunction: mechanistic insights into cardioplegia. FASEB J. 19, 980-982, doi:10.1096/fj.04-3008fje (2005).

161 German, J. et al. Hypothalamic leptin signaling regulates hepatic insulin sensitivity via a neurocircuit involving the vagus nerve. Endocrinology 150, 4502-4511, doi:10.1210/en.2009-0445 (2009).

162 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).

163 Kishore, P. et al. Activation of K(ATP) channels suppresses glucose production in humans. J. Clin. Invest. 121, 4916-4920, doi:10.1172/JCI58035 (2011).

164 Beguin, P., Nagashima, K., Nishimura, M., Gonoi, T. & Seino, S. PKA-mediated phosphorylation of the human K(ATP) channel: separate roles of Kir6.2 and SUR1 subunit phosphorylation. EMBO J. 18, 4722-4732, doi:10.1093/emboj/18.17.4722 (1999).

165 Global report on diabetes. World Health Organization. Geneva (2016).

166 Taylor, S. I. Deconstructing type 2 diabetes. Cell 97, 9-12 (1999).

167 Morton, G. J., Cummings, D. E., Baskin, D. G., Barsh, G. S. & Schwartz, M. W. Central nervous system control of food intake and body weight. Nature 443, 289-295, doi:10.1038/nature05026 (2006).

168 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).

169 Schwartz, M. W. et al. Cooperation between brain and islet in glucose homeostasis and diabetes. Nature 503, 59-66, doi:10.1038/nature12709 (2013).

170 Samuel, V. T. & Shulman, G. I. Mechanisms for insulin resistance: common threads and missing links. Cell 148, 852-871, doi:10.1016/j.cell.2012.02.017 (2012).

171 Sun, K., Kusminski, C. M. & Scherer, P. E. Adipose tissue remodeling and obesity. J. Clin. Invest. 121, 2094-2101, doi:10.1172/JCI45887 (2011).

172 Brashier, D. B., Sharma, A. K., Dahiya, N., Singh, S. K. & Khadka, A. Lorcaserin: A novel antiobesity drug. J. Pharmacol. Pharmacother. 5, 175-178, doi:10.4103/0976-500X.130158 (2014).

173 Buse, J. B. et al. Exenatide once weekly versus liraglutide once daily in patients with type 2 diabetes (DURATION-6): a randomised, open-label study. Lancet 381, 117-124, doi:10.1016/S0140-6736(12)61267-7 (2013).

Page 126: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

116

174 Sisley, S. et al. Neuronal GLP1R mediates liraglutide's anorectic but not glucose-lowering effect. J. Clin. Invest. 124, 2456-2463, doi:10.1172/JCI72434 (2014).

175 Hallschmid, M., Higgs, S., Thienel, M., Ott, V. & Lehnert, H. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Diabetes 61, 782-789, doi:10.2337/db11-1390 (2012).

176 Dash, S., Xiao, C., Morgantini, C., Koulajian, K. & Lewis, G. F. Intranasal insulin suppresses endogenous glucose production in humans compared with placebo in the presence of similar venous insulin concentrations. Diabetes 64, 766-774, doi:10.2337/db14-0685 (2015).

177 Heni, M. et al. Central insulin administration improves whole-body insulin sensitivity via hypothalamus and parasympathetic outputs in men. Diabetes 63, 4083-4088, doi:10.2337/db14-0477 (2014).

178 Lam, T. K., Schwartz, G. J. & Rossetti, L. Hypothalamic sensing of fatty acids. Nat. Neurosci. 8, 579-584, doi:10.1038/nn1456 (2005).

179 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).

180 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).

181 Cubelos, B., Gimenez, C. & Zafra, F. Localization of the GLYT1 glycine transporter at glutamatergic synapses in the rat brain. Cereb. Cortex 15, 448-459, doi:10.1093/cercor/bhh147 (2005).

182 Harvey, R. J. & Yee, B. K. Glycine transporters as novel therapeutic targets in schizophrenia, alcohol dependence and pain. Nat Rev Drug Discov 12, 866-885, doi:10.1038/nrd3893 (2013).

183 Porter, R. A., and Dawson, L.A. GlyT-1 Inhibitors: From Hits to Clinical Candidates. In Small Molecule Therapeutics for Schizophrenia, S. Celanire, and S. Poli, eds., 51-99 (Cham: Springer International Publishing, 2015).

184 Yue, J. T. et al. A fatty acid-dependent hypothalamic-DVC neurocircuitry that regulates hepatic secretion of triglyceride-rich lipoproteins. Nat Commun 6, 5970, doi:10.1038/ncomms6970 (2015).

185 Gillespie, B. R., Burns, G. A. & Ritter, R. C. NMDA channels control meal size via central vagal afferent terminals. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289, R1504-1511, doi:10.1152/ajpregu.00169.2005 (2005).

186 Kemp, J. A. et al. 7-Chlorokynurenic acid is a selective antagonist at the glycine modulatory site of the N-methyl-D-aspartate receptor complex. Proc. Natl. Acad. Sci. U. S. A. 85, 6547-6550 (1988).

Page 127: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

117

187 Woodward, J. J. A comparison of the effects of ethanol and the competitive glycine antagonist 7-chlorokynurenic acid on N-methyl-D-aspartic acid-induced neurotransmitter release from rat hippocampal slices. J. Neurochem. 62, 987-991 (1994).

188 Zhang, J. et al. Pharmacological characterization of [(3)H]CHIBA-3007 binding to glycine transporter 1 in the rat brain. PLoS One 6, e21322, doi:10.1371/journal.pone.0021322 (2011).

189 Nagy, K. et al. Alterations in brain extracellular dopamine and glycine levels following combined administration of the glycine transporter type-1 inhibitor Org-24461 and risperidone. Neurochem. Res. 35, 2096-2106, doi:10.1007/s11064-010-0241-0 (2010).

190 Chen, L., Muhlhauser, M. & Yang, C. R. Glycine tranporter-1 blockade potentiates NMDA-mediated responses in rat prefrontal cortical neurons in vitro and in vivo. J. Neurophysiol. 89, 691-703, doi:10.1152/jn.00680.2002 (2003).

191 Samuel, V. T. et al. Fasting hyperglycemia is not associated with increased expression of PEPCK or G6Pc in patients with Type 2 Diabetes. Proc. Natl. Acad. Sci. U. S. A. 106, 12121-12126, doi:10.1073/pnas.0812547106 (2009).

192 Atkinson, B. N. et al. ALX 5407: a potent, selective inhibitor of the hGlyT1 glycine transporter. Mol. Pharmacol. 60, 1414-1420 (2001).

193 Benarroch, E. E. NMDA receptors: recent insights and clinical correlations. Neurology 76, 1750-1757, doi:10.1212/WNL.0b013e31821b7cc9 (2011).

194 Kemp, J. A. & McKernan, R. M. NMDA receptor pathways as drug targets. Nat. Neurosci. 5 Suppl, 1039-1042, doi:10.1038/nn936 (2002).

195 Aroeira, R. I., Sebastiao, A. M. & Valente, C. A. GlyT1 and GlyT2 in brain astrocytes: expression, distribution and function. Brain Struct Funct 219, 817-830, doi:10.1007/s00429-013-0537-3 (2014).

196 Olivares, L., Aragon, C., Gimenez, C. & Zafra, F. The role of N-glycosylation in the targeting and activity of the GLYT1 glycine transporter. J. Biol. Chem. 270, 9437-9442 (1995).

197 Zafra, F., Gomeza, J., Olivares, L., Aragon, C. & Gimenez, C. Regional distribution and developmental variation of the glycine transporters GLYT1 and GLYT2 in the rat CNS. Eur. J. Neurosci. 7, 1342-1352 (1995).

198 Barrera, S. P. et al. PKC-Dependent GlyT1 Ubiquitination Occurs Independent of Phosphorylation: Inespecificity in Lysine Selection for Ubiquitination. PLoS One 10, e0138897, doi:10.1371/journal.pone.0138897 (2015).

199 Talman, W. T., Perrone, M. H. & Reis, D. J. Evidence for L-glutamate as the neurotransmitter of baroreceptor afferent nerve fibers. Science 209, 813-815 (1980).

Page 128: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

118

200 Talman, W. T. & Robertson, S. C. Glycine, like glutamate, microinjected into the nucleus tractus solitarii of rat decreases arterial pressure and heart rate. Brain Res. 477, 7-13 (1989).

201 Chatterton, J. E. et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 415, 793-798, doi:10.1038/nature715 (2002).

202 Fujikawa, T., Chuang, J. C., Sakata, I., Ramadori, G. & Coppari, R. Leptin therapy improves insulin-deficient type 1 diabetes by CNS-dependent mechanisms in mice. Proc. Natl. Acad. Sci. U. S. A. 107, 17391-17396, doi:10.1073/pnas.1008025107 (2010).

203 German, J. P. et al. Leptin activates a novel CNS mechanism for insulin-independent normalization of severe diabetic hyperglycemia. Endocrinology 152, 394-404, doi:10.1210/en.2010-0890 (2011).

204 Rasmussen, B. A. et al. Jejunal leptin-PI3K signaling lowers glucose production. Cell Metab. 19, 155-161, doi:10.1016/j.cmet.2013.11.014 (2014).

205 Marquard, J. et al. Characterization of pancreatic NMDA receptors as possible drug targets for diabetes treatment. Nat. Med. 21, 363-372, doi:10.1038/nm.3822 (2015).

206 Vancampfort, D. et al. A meta-analysis of cardio-metabolic abnormalities in drug naive, first-episode and multi-episode patients with schizophrenia versus general population controls. World Psychiatry 12, 240-250, doi:10.1002/wps.20069 (2013).

207 Harsing, L. G., Jr. et al. Glycine transporter type-1 and its inhibitors. Curr. Med. Chem. 13, 1017-1044 (2006).

208 Alberati, D. et al. Glycine reuptake inhibitor RG1678: a pharmacologic characterization of an investigational agent for the treatment of schizophrenia. Neuropharmacology 62, 1152-1161, doi:10.1016/j.neuropharm.2011.11.008 (2012).

209 Sun, H. S. & Feng, Z. P. Neuroprotective role of ATP-sensitive potassium channels in cerebral ischemia. Acta Pharmacol. Sin. 34, 24-32, doi:10.1038/aps.2012.138 (2013).

210 van den Top, M., Lee, K., Whyment, A. D., Blanks, A. M. & Spanswick, D. Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate nucleus. Nat. Neurosci. 7, 493-494, doi:10.1038/nn1226 (2004).

211 Yang, S. B. et al. Rapamycin ameliorates age-dependent obesity associated with increased mTOR signaling in hypothalamic POMC neurons. Neuron 75, 425-436, doi:10.1016/j.neuron.2012.03.043 (2012).

212 Campos, C. A. & Ritter, R. C. NMDA-type glutamate receptors participate in reduction of food intake following hindbrain melanocortin receptor activation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R1-9, doi:10.1152/ajpregu.00388.2014 (2015).

213 Bach, E. C., Halmos, K. C. & Smith, B. N. Enhanced NMDA receptor-mediated modulation of excitatory neurotransmission in the dorsal vagal complex of

Page 129: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

119

streptozotocin-treated, chronically hyperglycemic mice. PLoS One 10, e0121022, doi:10.1371/journal.pone.0121022 (2015).

214 Soundarapandian, M. M., Zhong, X., Peng, L., Wu, D. & Lu, Y. Role of K(ATP) channels in protection against neuronal excitatory insults. J. Neurochem. 103, 1721-1729, doi:10.1111/j.1471-4159.2007.04963.x (2007).

215 Hopper, R. A. & Garthwaite, J. Tonic and phasic nitric oxide signals in hippocampal long-term potentiation. J. Neurosci. 26, 11513-11521, doi:10.1523/JNEUROSCI.2259-06.2006 (2006).

216 Sattler, R. et al. Specific coupling of NMDA receptor activation to nitric oxide neurotoxicity by PSD-95 protein. Science 284, 1845-1848 (1999).

217 Snyder, S. H. Nitric oxide: first in a new class of neurotransmitters. Science 257, 494-496 (1992).

218 Shen, K. Z. & Johnson, S. W. Ca2+ influx through NMDA-gated channels activates ATP-sensitive K+ currents through a nitric oxide-cGMP pathway in subthalamic neurons. J. Neurosci. 30, 1882-1893, doi:10.1523/JNEUROSCI.3200-09.2010 (2010).

219 Stanley, B. G., Willett, V. L., 3rd, Donias, H. W., Dee, M. G., 2nd & Duva, M. A. Lateral hypothalamic NMDA receptors and glutamate as physiological mediators of eating and weight control. Am. J. Physiol. 270, R443-449 (1996).

220 Stanley, B. G., Ha, L. H., Spears, L. C. & Dee, M. G., 2nd. Lateral hypothalamic injections of glutamate, kainic acid, D,L-alpha-amino-3-hydroxy-5-methyl-isoxazole propionic acid or N-methyl-D-aspartic acid rapidly elicit intense transient eating in rats. Brain Res. 613, 88-95 (1993).

221 Liu, T. et al. Fasting activation of AgRP neurons requires NMDA receptors and involves spinogenesis and increased excitatory tone. Neuron 73, 511-522, doi:10.1016/j.neuron.2011.11.027 (2012).

222 Uner, A. et al. The role of GluN2A and GluN2B NMDA receptor subunits in AgRP and POMC neurons on body weight and glucose homeostasis. Mol Metab 4, 678-691, doi:10.1016/j.molmet.2015.06.010 (2015).

223 Finan, B. et al. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat. Med. 21, 27-36, doi:10.1038/nm.3761 (2015).

224 Pocai, A. et al. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes 58, 2258-2266, doi:10.2337/db09-0278 (2009).

225 Day, J. W. et al. A new glucagon and GLP-1 co-agonist eliminates obesity in rodents. Nat. Chem. Biol. 5, 749-757, doi:10.1038/nchembio.209 (2009).

226 Wagner, R. et al. Non-Suppressed Glucagon after Glucose Challenge as a Potential Predictor for Glucose Tolerance. Diabetes, doi:10.2337/db16-0354 (2016).

Page 130: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

120

227 Kraegen, E. W. et al. Development of muscle insulin resistance after liver insulin resistance in high-fat-fed rats. Diabetes 40, 1397-1403 (1991).

228 Wang, J. et al. Overfeeding rapidly induces leptin and insulin resistance. Diabetes 50, 2786-2791 (2001).

229 Samuel, V. T. et al. Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J. Biol. Chem. 279, 32345-32353, doi:10.1074/jbc.M313478200 (2004).

230 Thalacker-Mercer, A. E. et al. BMI, RQ, diabetes, and sex affect the relationships between amino acids and clamp measures of insulin action in humans. Diabetes 63, 791-800, doi:10.2337/db13-0396 (2014).

231 Wang-Sattler, R. et al. Novel biomarkers for pre-diabetes identified by metabolomics. Mol. Syst. Biol. 8, 615, doi:10.1038/msb.2012.43 (2012).

232 Floegel, A. et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes 62, 639-648, doi:10.2337/db12-0495 (2013).

233 Yan-Do, R. et al. A glycine-insulin autocrine feedback loop enhances insulin secretion from human beta-cells and is impaired in type 2 diabetes. Diabetes, doi:10.2337/db15-1272 (2016).

234 Abraham, M. A. & Lam, T. K. Glucagon action in the brain. Diabetologia 59, 1367-1371, doi:10.1007/s00125-016-3950-3 (2016).

235 Quinones, M. et al. Hypothalamic CaMKKbeta mediates glucagon anorectic effect and its diet-induced resistance. Mol Metab 4, 961-970, doi:10.1016/j.molmet.2015.09.014 (2015).

236 Hung, C. Y., Covasa, M., Ritter, R. C. & Burns, G. A. Hindbrain administration of NMDA receptor antagonist AP-5 increases food intake in the rat. Am. J. Physiol. Regul. Integr. Comp. Physiol. 290, R642-651, doi:10.1152/ajpregu.00641.2005 (2006).

237 Ritter, R. C., Covasa, M. & Matson, C. A. Cholecystokinin: proofs and prospects for involvement in control of food intake and body weight. Neuropeptides 33, 387-399, doi:10.1054/npep.1999.0051 (1999).

238 Skibicka, K. P. & Grill, H. J. Hypothalamic and hindbrain melanocortin receptors contribute to the feeding, thermogenic, and cardiovascular action of melanocortins. Endocrinology 150, 5351-5361, doi:10.1210/en.2009-0804 (2009).

239 Blouet, C. & Schwartz, G. J. Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One 7, e51898, doi:10.1371/journal.pone.0051898 (2012).

Page 131: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

121

240 Xu, Y. et al. PI3K signaling in the ventromedial hypothalamic nucleus is required for normal energy homeostasis. Cell Metab. 12, 88-95, doi:10.1016/j.cmet.2010.05.002 (2010).

241 Li, M. et al. Optimal promoter usage for lentiviral vector-mediated transduction of cultured central nervous system cells. J. Neurosci. Methods 189, 56-64, doi:10.1016/j.jneumeth.2010.03.019 (2010).

242 Wu, Q., Clark, M. S. & Palmiter, R. D. Deciphering a neuronal circuit that mediates appetite. Nature 483, 594-597, doi:10.1038/nature10899 (2012).

243 Roman, C. W., Derkach, V. A. & Palmiter, R. D. Genetically and functionally defined NTS to PBN brain circuits mediating anorexia. Nat Commun 7, 11905, doi:10.1038/ncomms11905 (2016).

244 Carter, M. E., Soden, M. E., Zweifel, L. S. & Palmiter, R. D. Genetic identification of a neural circuit that suppresses appetite. Nature 503, 111-114, doi:10.1038/nature12596 (2013).

245 Steculorum, S. M. et al. AgRP Neurons Control Systemic Insulin Sensitivity via Myostatin Expression in Brown Adipose Tissue. Cell 165, 125-138, doi:10.1016/j.cell.2016.02.044 (2016).

Page 132: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

Malonyl-CoACPT-1

GlucoseProduc5on

Ro7lerin/DNPKC-δ

Glibenclamide/Kir6.2

LCFA-CoA PKC-δ? Sur1-Kir6.2KATPchannel?

AICAR/CAAMPKACC

Acetyl-CoA ?

MediobasalHypothalamusGlucagon

Glucagon Receptor

PKA

AMPK

Figure2.1

Page 133: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

-90

-40

0

5

10

15

GFP+salineGFP+glucagonCAAMPK+salineCAAMPK+glucagon

0

2

4

6

8

024681012

0

5

10

0

2

4

6

8

-90

-40

A

B

C

D

E

F£basal

£basal nclampnclamp

H

MBHsaline

MBHglucagon

pACC(265kDa)

TotalACC(265kDa)

β-tubulin(55kDa)

00.10.20.30.40.50.60.7

MBHSaline MBHGlucagon

pAC

C/TotalACC

I* * * *

* * **

* * **

GlucoseIn

fusion

Rate

(mgkg

-1m

in-1)

GlucoseProdu

c5on

(m

gkg-1m

in-1)

GlucoseProdu

c5on

Supp

ression(%

)GlucoseUptake

(mgkg-1

min

-1)

GlucoseIn

fusion

Rate

(mgkg

-1m

in-1)

GlucoseProdu

c5on

(m

gkg-1m

in-1)

GlucoseProdu

c5on

Supp

ression(%

)GlucoseUptake

(mgkg-1

min

-1)

G

0

5

10

SalineGlucagonAICAR+salineAICAR+glucagon

+ – – –– + – –– – + –– – – +

SalineGlucagonAICAR+salineAICAR+glucagon

+ – – –– + – –– – + –– – – +

SalineGlucagonAICAR+salineAICAR+glucagon

+ – – –– + – –– – + –– – – +

SalineGlucagonAICAR+salineAICAR+glucagon

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

GFP+salineGFP+glucagonCAAMPK+salineCAAMPK+glucagon

GFP+salineGFP+glucagonCAAMPK+salineCAAMPK+glucagon

+ – – –– + – –– – + –– – – +

GFP+salineGFP+glucagonCAAMPK+salineCAAMPK+glucagon

+ – – –– + – –– – + –– – – +

250kDa

250kDa

50kDa

Figure2.2

Page 134: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

0.2

0.4

0.6

0.8

Saline Glucagon OAG

*

LacZ+salineLacZ+glucagonDNPKCδ+salineDNPKCδ+glucagon

0

5

10

0

5

10

15

-100

-50

0

0

5

10

15

0

5

10

0

5

10

15

-100

-50

0

0

5

10

15

A

B

E

C

D

F

G

H

* * * *

* * * *

**

* *

GlucoseIn

fusion

Rate

(mgkg

-1m

in-1)

GlucoseProdu

c5on

(m

gkg-1m

in-1)

GlucoseProdu

c5on

Supp

ression(%

)GlucoseUptake

(mgkg-1

min

-1)

GlucoseIn

fusion

Rate

(mgkg

-1m

in-1)

GlucoseProdu

c5on

Supp

ression(%

)GlucoseUptake

(mgkg-1

min

-1)

SalineGlucagonRot+salineRot+glucagon

SalineGlucagonRot+salineRot+glucagon

SalineGlucagonRot+salineRot+glucagon

SalineGlucagonRot+salineRot+glucagon

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

GlucoseProdu

c5on

(m

gkg-1m

in-1)

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

LacZ+salineLacZ+glucagonDNPKCδ+salineDNPKCδ+glucagon

LacZ+salineLacZ+glucagonDNPKCδ+salineDNPKCδ+glucagon

LacZ+salineLacZ+glucagonDNPKCδ+salineDNPKCδ+glucagon

PKC-δac5v

ity

(pmol/m

in/protein)

I

Figure2.3

Page 135: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

-100

-50

0

0

5

10

0

5

10

15

0

5

10

15

0

5

10

0

5

10

15

-100-80-60-40-200

A

B

C

D

E

F£basal

£basalnclamp

nclamp

0

5

10

G

H

**

**

* *

GlucoseInfusio

nRa

te

(mgkg

-1m

in-1)

GlucoseProd

ucMo

n(m

gkg

-1 min

-1)

GlucoseProdu

c5on

Supp

ression(%

)GlucoseUptake

(mgkg

-1m

in-1)

GlucoseInfusio

nRa

te

(mgkg

-1m

in-1)

GlucoseProd

ucMo

n(m

gkg

-1 min

-1)

GlucoseProdu

c5on

Supp

ression(%

)

GlucoseUptake

(mgkg

-1m

in-1)

SalineGlucagonGli+salineGli+glucagon

SalineGlucagonGli+salineGli+glucagon

SalineGlucagonGli+salineGli+glucagon

SalineGlucagonGli+salineGli+glucagon

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

+ – – –– + – –– – + –– – – +

GFP+salineGFP+glucagonDNKir6.2+salineDNKir6.2+glucagon

GFP+salineGFP+glucagonDNKir6.2+salineDNKir6.2+glucagon

GFP+salineGFP+glucagonDNKir6.2+salineDNKir6.2+glucagon

GFP+salineGFP+glucagonDNKir6.2+salineDNKir6.2+glucagon

+ – – –– + – –– – + –– – – +

Figure2.4

Page 136: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

Figure3.1

Page 137: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

2

4

6

8

10

12

14

0

1

2

3

4

5

6

0

0.001

0.002

0.003

0.004

0.005

Saline ALX0

2

4

6

8

10

12

0

1

2

3

4

5

6

100

120

140

160

180

200

220

240

260

basal 0 2 5 10

15

20

25

30

40

50

60

8100

8600

9100

£Basal nClamp

Glucoseprodu

c5on

(mg/kg/min)

D

*†

Plasmaglucose(m

g/dl)

0

1

2

3

4

basal 0 2 5 10

15

20

25

30

40

50

60

Plasmainsulin

(ng/ml)

DVCsalineDVCALX

DVCsalineDVCALX

Time(min)

AUC(0-60min)

*

††

††

Glucoseinfusion

rate(m

g/kg/min)

SalineSHAMALX

HVAG

+ – + –+ + – –– + – +– – + +

£Basal nClamp

Glucoseprodu

c5on

(mg/kg/min)

A B C

Glucoseinfusion

rate(m

g/kg/min)

+ – + – – – –– + – + + + +– – + + – – –– – – – + – –– – – – – + –– – – – – – +

+ – + – – – –– + – + + + +– – + + – – –– – – – + – –– – – – – + –– – – – – – +

E F

SalineALX

MK8017CKNAAd-MM

Ad-GluN1shRNA

SalineALX

MK8017CKNAAd-MM

Ad-GluN1shRNA

+ – + –+ + – –– + – +– – + +

* †

Time(min)

*

G

DVCextracellular

glycineconcen

tra5

on(n

mol/ul) £Baseline

nInfusion*

*

Figure3.2

Page 138: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

0.4

0.8

1.2

GlyT1(5

5):IR

A B

0

1

2

―IR

70―100― ―GlyT190

―GlyT170

MW(kDa)

0

0.5

1

1.5

GlyT1(9

0):IR

GlyT1(7

0):IR

GlyT1(9

0):IR

DVCLV-GlyT1shRNA

100―

MW(kDa)

70―

100―

―GlyT170

―IR

―GlyT190

GlyT1(7

0):IR

DVCLV-MM

0

0.4

0.8

1.2

**

0

0.4

0.8

1.2

*

DVC sp5,Sp5C,Sp5I(L)

D

70―

―IR100―

―GlyT170

0

0.5

1

1.5

2

C

70― ―GlyT170

100― ―IR

0

0.5

1

1.5

2

sp5,Sp5C,Sp5I(R)MW(kDa)

MW(kDa)

0

1

2

3

4

5

6

7

8

Glucoseinfusion

rate(m

g/kg/min)

Glucoseprodu

c5on

(mg/kg/min)

+ – –– + +– – +

£Basal nClamp*

LV-MM + – –LV-GlyT1shRNA – + +MK801 – – +

E Fpy

0

2

4

6

8

10

12

*

100―

100―

DVCLV-MM + –DVCLV-GlyT1shRNA – +

+ –– +

+ –– +

+ –– +

100―

DVCLV-MM

DVCLV-GlyT1shRNA

DVCLV-GlyT1shRNA

DVCLV-MM

GlyT1(7

0):IR

+ –– +

DVCLV-MM

DVCLV-GlyT1shRNA

GlyT1(7

0):IR

―GlyT155

+ –– +

55―

DVCLV-MM + –DVCLV-GlyT1shRNA – +

Figure3.3

Page 139: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

2

4

6

8

10

12

0

1

2

3

4

5

6

7

0

2

4

6

8

10

12

14

13

Glucoseinfusion

rate(m

g/kg/min)

Glucoseprodu

c5on

(mg/kg/min)

3dHFDSaline

GlycineALX

7CKNA

+ + + + ++ – – – –– + – + –– – + – +– – – + +

+ + + + ++ – – – –– + – + –– – + – +– – – + +

B C

£Basal nClamp

A

DVCinfusion:saline/glycine/ALX/glycine+7CKNA/ALX+7CKNA

SST(3µgkg-1min-1)

Insulin(1.2mUkg-1min-1)

-90min

Glucose(asneeded)

[3-3H]-glucose(0.4µCimin-1)

0 240

day0

DVCcannula5on Vascularcatheteriza5on Clamp

8

90

DVCinfusion:saline/7CKNA

10 HFD

-60

*

* †

Glucoseinfusion

rate(m

g/kg/min)

+ ++ –– +

3dHFDIvDMSO

IvALX

+ ++ –– +

£Basal nClamp

*

*

D

Glucoseprodu

c5on

(mg/kg/min)

E

0

1

2

3

4

5

Figure3.4

Page 140: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

‡‡‡

100

120

140

160

180

200

220

240

260

280

300

basal 0 2 5 10

15

20

25

30

40

50

60

‡‡

‡‡

8000

10000

12000

***

0

20

40

60

80

100

120

140

160

A

day0

DVCcannula5on Vascularcatheteriza5on ivGTT

8 136 HFD

STZ/Nicinjec5on

DVCinfusion:saline/ALX

-240min 600

i.v.glucose(0.25g/kg)

Fasted

plasm

aglucose(m

g/dl)

B

Non-diabeMc + –7dSTZ/Nic/HFD – +

*

Plasmaglucose(m

g/dl)

CAUC

(0-60min)

†††

†† ††

‡‡

ࠠ

†††

RC+DVCsaline7-dSTZ/Nic/HFD+DVCsaline7-dSTZ/Nic/HFD+DVCALX

Figure3.5

Page 141: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

A

Glucoseinfusion

rate(m

g/kg/min)

Glucoseprodu

c5on

(mg/kg/min)

28dRC28dHFDSalineALX

+ + – –– – + ++ – + –– + – +

+ + – –– – + ++ – + –– + – +

C D

0

1

2

3

4

5

6

0

1

2

3

4

5

6

7

8

9

10

200

250

300

350

400

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28

Bodyweight(g)

B

Time(days)

28dHFD28dRC

£Basal nClamp

Clamp

DVCinfusion:saline/ALX

SST(3µgkg-1min-1)

Insulin(1.2mUkg-1min-1)

Glucose(asneeded)

[3-3H]-glucose(0.4µCimin-1)

day0

DVCcannula5on

Vascularcatheteriza5on

16 29

0 24090

24

RCorHFD

-60min

*

*

*

*

*

Figure3.6

Page 142: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

SST(3µgkg-1min-1)

Insulin(1.2mUkg-1min-1)

Glucose(asneeded)

[3-3H]-glucose(0.4µCimin-1)

day0

DVCcannula5on±LV-MM/

LV-GlyT1shRNA

Vascularcatheteriza5on

16

0min 24090

24

HFD

29

A

+ +

+ –

– +

28dHFD + +

DVCLV-MM + –

DVCLV-GlyT1shRNA – +

+ +

+ –

– +

Clamp

0

1

2

3

4

5

6

0

1

2

3

4

5

6

7

8

9

10

Glucoseinfusion

rate(m

g/kg/min)

Glucoseprodu

c5on

(mg/kg/min)

B C£Basal nClamp

E

300

320

340

360

380

400

420

Bodyweight(g)

D

*

*

*

-6

-4

-2

0

2

4

6

%Bod

yweightcha

nge

28dHFD+DVCLV-MM28dHFD+DVCLV-GlyT1shRNA

RC+DVCLV-MM

-6

-4

-2

0

2

4

6

Day4postDVCviralinjecMon

Day5postDVCviralinjecMon

**

Figure3.7

Page 143: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

2

4

6

8

10

12

14

16

18

Day1 Day2 Day3

20

25

30

35

40

45

50

Day1

15

20

25

30

35

Day1

20

25

30

35

40

Day220

25

30

35

40

45

50

Day10

2

4

6

8

10

12

14

Day1 Day2 Day3

0

5

10

15

20

25

30

0 60 120 180 240 300 360

Cumula5

vefo

odintake(g)

0

5

10

15

20

25

30

0 60 120 180 240 300 360

Cumula5

vefo

odintake(g)

Time(min)

B

F

DVCsalineDVCglycine

DVCsalineDVCALX

† †† ††††

††

† † †††

†††

††††

††††††

Foodreturned

day0

FeedingExperiment

13

DVCinjec5on:saline/ALX

-60min 3600

Foodreturned

day0

DVCcannula5on FeedingExperiment

13

DVCinjec5on:saline/glycine

-10min 3600

A

E

%Bod

yweightg

ain

from

re-fe

eding

C D

G H

Dailyfo

odintake(g)

Dailyfo

odintake(g)

I

-8

-6

-4

-2

0

%Bod

yweightcha

nge DVCLV-MM

DVCLV-GlyT1shRNA

Day4postDVCviralinjec5on

0

5

10

15

20

25

0 60 120 180 240 300 360

Cumula5

vefo

odintake(g)

DVCLV-MMDVCLV-GlyT1shRNA

J

Dailyfo

odintake(g)

Daysagerre-feeding

K

L

Daysagerre-feeding

0

2

4

6

8

10

Day1 Day2

‡‡

%Bod

yweightg

ain

from

re-fe

eding

%Bod

yweightg

ain

from

re-fe

eding

20

25

30

35

Day2

20

25

30

35

40

Day2

††

††

††

Figure3.8

Page 144: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

Glucose(asneeded)

[3-3H]-glucose(0.4µCimin-1)

SST(3µgkg-1min-1)

Insulin(1.2mUkg-1min-1)

0

2

4

6

8

10

12

60

Glucoseuptake(m

g/kg/min)

SalineALX

MK8017CKNAAd-MM

Ad-GluN1shRNA

+ – – – – – –– + – + + + +– – + + – – –– – – – + – –– – – – – + –– – – – – – +

C

0

20

40

60

80

100

120

140

160

Plasmaglucose(m

g/dl)

+ – – – – – –– + – + + + +– – + + – – –– – – – + – –– – – – – + –– – – – – – +

£Basal nClamp

Plasmainsulin

(ng/ml)

SalineALX

+ –– +

0.0

0.1

0.2

0.3

0.4

Bodyweight(kg)

+ –– +

A

E

DVCinfusion:saline/MK801/ALX/ALX+MK801/ALX

+7CKNA-90min 0 240

day0

DVCcannula5on±Ad-GluN1shRNA/Ad-MM±LV-GlyT1shRNA/LV-MM

Vascularcatheteriza5on

Experiment

8 13

Clamp

ivGTT

90

DVCinfusion:saline/

MK801/7CKNADVCinfusion:saline/ALX-240min 0

i.v.glucose(0.25g/kg)

-60

D F£BasalnClamp

Bday0

DVCcannula5on MicrodialysisExperiment

8

-120 0 300

DVCinfusion:saline/ALXBaseline

-270min

Recovery

Microdialysisinfusion:aECF

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

SupplementaryFigure3.1

Page 145: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

5

10

15

20

25

0

2

4

6

8

10

12

0

6

12

18

24

30

0

30

60

90

120

150

180Glucoseinfusion

rate(m

g/kg/min)

ICV-4saline + –ICV-4ALX – +

Glucoseprodu

c5on

(mg/kg/min)

+ –– +

Glucoseuptake(m

g/kg/min)

+ –– +

Plasmaglucose(m

g/dl)

+ –– +

*

Glucoseuptake(m

g/kg/min)

Plasmaglucose(m

g/ml)

E£Basal nClamp

*

A B C D

F

SalineSHAMALX

HVAG

+ – + –+ + – –– + – +– – + +

+ – + –+ + – –– + – +– – + +

0

2

4

6

8

10

12

14

0

20

40

60

80

100

120

140

160

SupplementaryFigure3.2

Page 146: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

A i

ii

iii

iv

v

SupplementaryFigure3.3

Page 147: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

2

4

6

8

10

12

0

20

40

60

80

100

120

140

Glucoseuptake(m

g/kg/min)

Plasmaglucose(m

g/dl)

LV-MM + – –LV-GlyT1shRNA – + +

MK801 – – +

+ – –– + +– – +

0.0

0.1

0.2

0.3

0.4

Bodyweight(kg)

+ – –– + +– – +

A B C£BasalnClamp

SupplementaryFigure3.4

Page 148: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

20

40

60

80

100

120

140

160

0

2

4

6

8

10

12

0

20

40

60

80

100

120

140

160

180

3dHFDSaline

GlycineALX

7CKNA

+ + + + ++ – – – –– + – + –– – + – +– – – + +

AGlucoseuptake(m

g/kg/min)

+ + + + ++ – – – –– + – + –– – + – +– – – + +

B£Basal nClamp

0

2

4

6

8

10

12

14

Glucoseuptake(m

g/kg/min)

D

Plasmaglucose(m

g/dl)

£BasalnClamp

3dHFDIvDMSO

IvALX

+ ++ –– +

+ ++ –– +

C

Plasmaglucose(m

g/dl)

SupplementaryFigure3.5

Page 149: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

0

1

2

3

4

5

6

7

8

9

10

A

Glucoseuptake(m

g/kg/min)

B

Plasmaglucose(m

g/dl)

0

1

2

3

4

5

6

7

8

9

10

0

20

40

60

80

100

120

140

160

28dRC28dHFD

SalineALX

+ + – –– – + ++ – + –– + – +

+ + – –– – + ++ – + –– + – +

C

Glucoseuptake(m

g/kg/min)

D

Plasmaglucose(m

g/dl)

0

20

40

60

80

100

120

140

160£Basal nClamp £Basal nClamp

28dHFD + +DVCLV-MM + –

DVCLV-GlyT1shRNA – +

+ ++ –– +

SupplementaryFigure3.6

Page 150: CNS HORMONAL AND NUTRITIONAL REGULATION OF GLUCOSE … · 2017. 12. 19. · Title: CNS Hormonal and Nutritional regulation of Glucose and Energy homeostasis Name: Mona Anna Abraham

!MBH

KATP channel!

Glucagon!

GR!

PKA!↓

↓↓

AMPK!Acetyl-CoA!

Malonyl-CoA!↓ACC!

LCFA!

LCFA!LCFA-CoA!

PKC-𝛿!

CPT-1!

Glucagon!

DVC

Glycine!

GlyT1!NMDAr!

Glucose!homeostasis!

Glucose!homeostasis!

Figure4.1