the role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · the role of...

158
i The role of exercise in glycaemic control and inflammation in overweight/obese individuals with and without type 2 diabetes by Aaron Raman BSc (Hons), MSc Exercise and Sport Science Massey University, New Zealand A thesis submitted to Murdoch University to fulfil the requirements for the degree of Doctor of Philosophy in the discipline of Exercise Science and Exercise Immunology Perth, Western Australia 2018

Upload: others

Post on 02-Mar-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

i

The role of exercise in glycaemic control

and inflammation in overweight/obese

individuals with and without type 2

diabetes

by

Aaron Raman

BSc (Hons), MSc Exercise and Sport Science

Massey University, New Zealand

A thesis submitted to Murdoch University

to fulfil the requirements for the degree of

Doctor of Philosophy

in the discipline of

Exercise Science and Exercise Immunology

Perth, Western Australia

2018

Page 2: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

ii

Author’s Declaration

I declare that this thesis is my own account of my research and contains as its main content

work which has not previously been submitted for a degree at any tertiary education

institution.

Aaron Raman

Page 3: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

iii

Statement of Contributors

I declare that this thesis is composed of my own research and work. I was involved in the

conception of the work presented in each chapter; reviewing the literature for each chapter;

writing the first draft of each chapter; interpreting the data in each chapter; and, I was solely

responsible for all data collection in each chapter.

In addition, I claim the majority of authorship for each article presented in this thesis. In

doing so, I declare that the co-authors as recorded below contributed to the relevant article

by way of critically analysing and commenting on that article, as necessary, so as to

contribute to its interpretation. More specifically, Associate Professor Fairchild, Associate

Professor Peiffer, Professor Hoyne, Dr Currie and Mr Lawler contributed their expertise in

relation to the analysis and report of the quantitative data present in Chapters Four, Five and

Six. Each author provided their final approval of the relevant article prior to journal

submission.

Mr Aaron Raman

Associate Professor Timothy Fairchild

Associate Professor Jeremiah Peiffer

Professor Gerard Hoyne

Dr Andrew Currie

Mr Nathan Lawler

Page 4: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

iv

Publication and Award

Chapter Two

Raman, A., Peiffer, J. J., Hoyne, G. F., Fairchild, T. J. (2018). Exercise, glucose transport

and glucose regulation: A brief review. Prepared for submission to Sports Medicine

Chapter Four

Raman, A., Peiffer, J. J., Hoyne, G. F., Lawler, N. G., Currie, A. J., Fairchild, T. J. (2018).

Effect of exercise on acute postprandial glucose concentrations and interleukin-6 responses

in sedentary and overweight males. Applied Physiology, Nutrition and Metabolism. In Press.

Doi 10.1139/apnm-2018-0160.

Chapter Five

Raman, A., Peiffer, J. J., Hoyne, G. F., Lawler, N. G., Currie, A. J., Fairchild, T. J. (2018).

Biomarkers of cardiovascular disease and type 2 diabetes following acute exercise in

overweight/obese males. Does intensity matter? Under Review in the Journal of Applied

Physiology.

Chapter Six

Raman, A., Peiffer, J. J., Hoyne, G. F., Currie, A. J., Fairchild, T. J. (2018). Short-term

training effect on glycaemic control and the inflammatory response in type 2 diabetes.

Prepared for submission to Medicine and Science in Sports and Exercise

Conference publication:

Raman, A., Peiffer, J. J., Hoyne, G. F., Kanaley, J.A, Fairchild, T. J. (2016). Acute Post-

exercise Glucose Disposal: Exploring The Role For Exercise Intensity In Overweight/obese

Males. Paper presented at the 63rd ACSM Annual Meeting, Hynes Convention Center and

the Sheraton Boston Hotel, Boston, Massachusetts, USA.

Travel award:

Conference Travel Award (CTA) by Murdoch University 2016.

Page 5: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

v

Abstract

Evidence suggests the chronic low-grade inflammation associated with obesity increases the

risk of developing type 2 diabetes mellitus (T2DM). Increased rates of exercise participation

has been shown to reduce the risk of progression from overweight/obese to T2DM.

Paradoxically, exercise results in an acute inflammatory response. The purpose of the research

herein was to characterize the response in key inflammatory markers and glycaemia to acute

exercise and short-term training in sedentary, overweight individuals and individuals with

T2DM. Glucose tolerance and the interleukin-6 (IL-6) receptor pathway were assessed

following acute (1h and 25h post-exercise) continuous exercise of moderate intensity (CME),

and compared with a work- and duration-matched bout of high intensity intermittent exercise

(HIIE; Chapter 4). There was an improvement in glucose tolerance immediately but not 25h

post-exercise, concomitant with increased activity via the IL-6 classical signalling pathway.

However, there were no differences between exercise modes and no associations between

glucose tolerance and IL-6 signalling markers. The response in matrix metalloproteinase

(MMP)-2 and MMP-3, as well as osteopontin was assessed following acute exercise in chapter

5. Continuous glucose monitoring was performed the day prior to exercise (control), the day

of exercise, and the day after exercise and showed no between-day or condition (CME vs.

HIIE) differences. However, differences in MMP-3 and osteopontin were identified in a

direction consistent with protection against metabolic and cardiovascular disease. Effect of

short-term exercise training (12 consecutive days of treadmill exercise) on inflammation and

glucose control were assessed in previously diagnosed individuals with T2DM (Chapter 6).

There were improvements in glycaemic control and global inflammation status post-training.

Acute changes in pro- and anti-inflammatory cytokine responses following the first and last

exercise session were also observed. The significance of these findings and future direction of

research are discussed in chapter 7.

Page 6: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

vi

Acknowledgements

Completing a PhD in the field of exercise immunology and its relationship with Type 2

Diabetes was always the ‘next step’ I needed to take to contribute to making the world a

healthier place. To Associate Professor Timothy Fairchild, I am so thankful to have had this

opportunity to work with you and to learn from you. Your knowledge and passion in research

has shaped me to become the person I am today. Thank you for your time and mentoring ability

in decluttering new and complicated concepts which were foreign to me, now a clear picture

in my head.

Professor Gerard Hoyne and Dr Andrew Currie, I am grateful for sharing your level of

expertise in immunology and guiding me to learn new techniques which formed an essential

part of this thesis. The learning curve was steep at times, but the extra effort was worth it to

gaining better quality information within the research projects. Associate Professor Jeremiah

Peiffer, thank you for providing me with the additional support and for your clear vision and

direct approach to problem solving in many of our discussions.

To my postgraduate colleagues, Kieran, Mitch, Nikky and Shaun, it has been a pleasure to

have your support and entertainment throughout this journey. To Mr Nathan Lawler, thank

you for your friendship and support since day one when we started our PhD together. Your

attempt to convert this Kiwi to an Australian is still a work in progress, but your influence has

been much welcomed! To Nardine, thank you for your great friendship and always reminding

me to be myself. Mr Jack Burns, I must also thank you for your help and friendship in and

outside of the laboratory that made these research projects run as smoothly as it did.

Mum and Dad, your constant reminder on the importance of education has got me here. Thank

you for your encouragement and support over the last few years. I can finally say the thesis is

completed and I look forward to spending more time back in New Zealand with our family.

Finally, to my partner Linh, I cannot imagine how much harder this journey would have been

without your love and support. To put up with my ups and downs, listening to the moments I

have had and supporting me whenever I needed it most, I cannot thank you enough for being

there for me. The next adventure awaits us!

Page 7: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

vii

List of Abbreviations

APN Adiponectin

AUC Area Under the Curve

BMI Body Mass Index

CME Continuous Moderate Intensity Exercise

CGMS Continuous Glucose Monitoring System

CRP C-Reactive Protein

DXA Dual Energy X-Ray Absorptiometry

FBG Fasting Blasma Glucose

GLUT-4 Glucose Transporter 4

hsCRP High Sensitivity C-Reactive Protein

HbA1c Glycated Haemoglobin

HIIE High Intensity Intermittent Exercise

HRmax Heart Rate Max

IL-1β Interleukin 1 Beta

IL-1ra Interleukin 1 Receptor Antagonist

IL-4 Interleukin 4

IL-6 Interleukin 6

IL-8 Interleukin 8

IL-10 Interleukin 10

IIL-15 Interleukin 15

IP-10 Interferon Gamma-Induced Protein 10

Page 8: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

viii

M1 Classically Activated Macrophages

M2 Alternatively Activated Macrophages

MCP-1 Monocyte Chemoattractant Protein 1

MMP-2 Matrix Metalloproteinase 2

MMP-3 Matrix Metalloproteinase 3

OGTT Oral Glucose Tolerance Test

OPN Osteopontin

PPG Postprandial Glucose

RPE Rating of Perceived Exertion

sIL-6R Soluble Interleukin-6 receptor

sGP130 Soluble Glycoprotein 130

T2DM Type 2 Diabetes Mellitus

TGF-β Transforming Growth Factor-Beta

Tregs Regulatory T Cells

TNFα Tumour Necrosis Factor Alpha

VAT Visceral Adipose Tissue

VO2peak Peak oxygen uptake

WC Waist Circumference

Page 9: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

ix

Table of Contents

Author’s Declaration ..................................................................................................... ii

Statement of Contributors ............................................................................................. iii

Publication and Award ................................................................................................. iv

Abstract ................................................................................................................. v

Acknowledgements ...................................................................................................... vi

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

List of Figures ............................................................................................................. xiii

List of Tables ............................................................................................................... xv

Chapter 1 Introduction ........................................................................................... 17

Chapter 2 Exercise, glucose transport and glucose regulation: A brief review...... 21

2.1 Glucose Transport ................................................................................. 22

2.2 Insulin sensitivity and responsiveness ................................................... 26

2.3 Tissue specific glucose transport in humans ......................................... 27

2.4 Changes in systemic plasma glucose post-exercise: Looking beyond

skeletal muscle....................................................................................... 27

2.4.1 Glucoregulatory effects of key Hormones: Epinephrine, glucagon,

growth hormone and cortisol ................................................................. 28

2.4.2 The role of exercise intensity................................................................. 31

2.5 Short-term consecutive training............................................................. 32

2.6 Conclusion ............................................................................................. 34

Chapter 3 Low grade inflammation, exercise and type 2 diabetes: A brief review

............................................................................................................... 35

3.1 Inflammation, obesity and T2DM ......................................................... 38

3.1.1 Pro-inflammatory cytokine markers associated with T2DM ................. 38

3.1.2 Additional inflammatory markers associated with T2DM .................... 40

3.1.3 Anti-inflammatory markers associated with T2DM .............................. 41

3.2 Exercise and inflammation – intensity, mode and duration .................. 42

3.3 Inflammation in T2DM – exercise-induced improvements ................... 43

3.4 Conclusions ........................................................................................... 47

Chapter 4 Effect of exercise on acute postprandial glucose concentrations and

interleukin-6 responses in sedentary and overweight males .................. 51

4.1 Abstract ................................................................................................. 52

4.2 Introduction ........................................................................................... 53

4.3 Methods ................................................................................................. 55

4.3.1 Study Design ......................................................................................... 55

Page 10: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

x

4.3.2 Participants ............................................................................................ 55

4.3.3 Introductory Session .............................................................................. 56

4.3.4 Experimental Phases .............................................................................. 56

4.3.5 Exercise Conditions (Day-2) ................................................................. 57

4.3.6 Blood Analysis ...................................................................................... 58

4.3.7 Data Analyses ........................................................................................ 59

4.4 Results ................................................................................................... 61

4.4.1 Glucose Responses ................................................................................ 62

4.4.2 Insulin, C-peptide and Inflammatory Markers ...................................... 63

4.5 Discussion.............................................................................................. 67

Chapter 5 Glucose and inflammatory markers following acute exercise in

sedentary and overweight males ............................................................ 71

5.1 Abstract ................................................................................................. 72

5.2 New & Noteworthy ............................................................................... 73

5.3 Introduction ........................................................................................... 74

5.4 Methods ................................................................................................. 76

5.4.1 Study Design ......................................................................................... 76

5.4.2 Participants ............................................................................................ 76

5.4.3 Introductory Session .............................................................................. 77

5.4.4 Experimental Trials ............................................................................... 77

5.4.5 Exercise Conditions ............................................................................... 78

5.4.6 Blood Sampling ..................................................................................... 78

5.4.7 Physical Activity Monitoring ................................................................ 79

5.4.8 Data Analyses ........................................................................................ 79

5.5 Results ................................................................................................... 81

5.5.1 Inflammatory Cytokines ........................................................................ 82

5.5.2 Glucose: Continuous Glucose Monitoring ............................................ 83

5.6 Discussion.............................................................................................. 86

Chapter 6 Short-term training effect on glycaemic control and the inflammatory

response in type 2 diabetes. ................................................................... 90

6.1 Abstract ................................................................................................. 91

6.2 Introduction ........................................................................................... 92

6.3 Methods ................................................................................................. 94

6.3.1 Study Design ......................................................................................... 94

6.3.2 Participants ............................................................................................ 94

6.3.3 Introductory session ............................................................................... 95

6.3.4 Oral Glucose Tolerance Test (OGTT) ................................................... 96

6.3.5 Experimental Phases (Exercise intervention) ........................................ 96

Page 11: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xi

6.3.6 Biochemical analysis ............................................................................. 96

6.3.7 Data Analyses ........................................................................................ 98

6.4 Results ................................................................................................. 100

6.4.1 Fitness and Anthropometric data (Table 6.1) ...................................... 100

6.4.2 Glycaemic variables (Table 6.2) .......................................................... 100

6.4.3 Acute phase proteins and hsCRP ......................................................... 101

6.4.4 Cell populations (Table 6.3) ................................................................ 102

6.4.5 Functional inflammatory response ...................................................... 103

6.5 Discussion............................................................................................ 105

Chapter 7 Thesis summary ................................................................................... 110

7.1 Future research directions .................................................................... 113

Bibliography ............................................................................................................. 114

Appendix A Murdoch University Ethics Approval ................................................... 139

A.1 Murdoch University Ethics Approval (Chapters 4 and 5) ........................... 140

A.2 Murdoch University Ethics Approval (Chapters 6) .................................... 141

Appendix B Information Letters ................................................................................. 143

B.1 Information Letters (Chapter 4 and 5) ............................................................... 144

B.2 Information Letters (Chapter 6)......................................................................... 148

Appendix C Adult Pre-Exercise Screening Tool ........................................................ 152

Appendix D 24-h Food Diary ..................................................................................... 156

Appendix E Borg Scale .............................................................................................. 157

Appendix F CONSORT Diagram ............................................................................... 158

Page 12: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xii

Page 13: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xiii

List of Figures

Figure 2.1. Glucose Transporter number of subcellular membrane fractions from

skeletal muscle. Data derived from [76]. ............................................... 23

Figure 2.2. (A) Represents the glucose transporter number (ρmol/mg; based on

cytochalasin B binding sites) in non-exercise (Non-EX), immediately

after exercise (0h Post-EX), 0.5 h after exercise (0.5h Post-EX) and 2 h

post-exercise (2h Post-EX). The exercise consisted of treadmill running

at 20m/min up 10% gradient. Data based on [80]. (B) Represents the

effect of 1 and 2 days of swimming on the number of glucose

transporter protein (derived from muscle homogenates containing 50ug

of protein). Data based on [84]. ............................................................. 24

Figure 2.3. Glucose uptake and the effect of insulin. Control group was not exercised;

Postexercise group completed 45 minutes of treadmill running at

18m/min, with the last two minutes completed at 31m/min. Results are

means ± SE with n=2-13 observations per data point. p < 0.05 when

compared with control values [99]. ....................................................... 25

Figure 2.4. Rate of glucose appearance (Ra) and disappearance (Rd) during steady state

glucose infusion rate. A. Measures taken before the clamp (PLA),

during the clamp at low (Low Ins; 4mU/l) and high (High Ins; 51mU/l);

B. Measures taken before the clamp (PLA), during the clamp at low

(Low Ins; 4mU/l) and high (High Ins; 51mU/l) insulin infusion rates

with addition of epinephrine (Low Ins +A; High Ins + A); C. measures

taken during the Low insulin clamp (40 mU.(m2)-1.min-1; Ins),

epinephrine only (A), insulin with epinephrine and propranolol

(Ins+A+Pro), insulin with epinephrine and metoprolol (Ins+A+Met); D.

Measures taken during the High insulin clamp (1200 mU.(m2)-1.min-1;

Ins), epinephrine only (A), insulin with epinephrine and propranolol

(Ins+A+Pro), insulin with epinephrine and metoprolol (Ins+A+Met). . 30

Figure 3.1. A schematic of the relationship between exercise training, inflammation

and type 2 diabetes. ............................................................................... 45

Figure 4.1. Diagram of study design. ........................................................................... 57

Figure 4.2. (Top). Mean (± SD) Ratings of perceived exertion after completing high-

intensity intermittent exercise (HIIE, filled square) and continuous

moderate-intensity (CME, open circle) using Borg’s scale. a,

significantly different from CME (P < 0.01). (Bottom). Mean (± SD)

Heart rate response during high-intensity intermittent exercise (HIIE)

compared to continuous moderate-intensity (CME). No significant

differences between trials p=0.112. ....................................................... 61

Figure 4.3. Box and whiskers (95% CI) plot for plasma glucose area under the curve

(AUC) on Day-1 (baseline), Day-2 (exercise) and Day-3. A main effect

of time was present; p=0.000. ................................................................ 62

Figure 4.4. Mean differences (with 95% CI) in plasma glucose concentration between

A: HIIE and CME on Day-2; B: HIIE and CME on Day-3; C: Day-2

and Day-1; D: Day-3 and Day-2. Differences were calculated from

blood glucose concentrations for each participant at each time-point of

the OGTT from one study day and subtracted from the corresponding

time-point on the comparison study-day. Significant differences are

indicated where 95% confidence limits to not cross the zero band. ...... 63

Page 14: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xiv

Figure 4.5. Box and whiskers (95% CI) plot for insulin (Top) and c-peptide (Bottom)

measured pre-exercise (baseline), immediately post-exercise (IPE), 1h

post-exercise and 25h post-exercise. a) compared to pre-exercise,

p<0.05; b) compared to post-exercise, p<0.01; c) compared to post-

exercise, p<0.05. Open bars, CME; hatched bars, HIIE. ....................... 65

Figure 4.6. Box and whiskers (95% CI) plot for; A: IL-6; B: sIL-6R; C: IL-6/sIL-6R

complex, and D: sgp130; measured in 10 participants pre-exercise

(baseline), immediately post-exercise (IPE), 1h post-exercise and 25h

post-exercise. a) compared to IPE and 1h post-exercise, p<0.05; b)

compared to IPE and 1h post-exercise, p<0.01; c) compared to 1h post-

exercise and 24h post-exercise, p≤0.01. Open bars, CME; hatched bars,

HIIE. ...................................................................................................... 66

Figure 5.1. Mean (± SD) concentration of matrix metalloproteinase-2 (MMP-2),

matrix metalloproteinase-3 (MMP-3), osteopontin and adiponectin.

MMP-3; a, Significantly different than Pre (p=0.001) and 25 h

(p=0.001). Osteopontin; a, Significantly different than Pre (p = 0.006)

and 25 h (p = 0.001). No differences were found with adiponectin and

MMP-2. CME (open bars) vs HIIE (hatched bars)................................ 82

Figure 5.2. (Top) Mean (±SD) glucose concentration over each day calculated from

CGMS. a, significant difference between Day 1 and Day 3, p = 0.05.

(Bottom) Glucose concentration during OGTT on each day. A main

effect of time over the OGTT was present, p < 0.001 and a main effect

between days was present, p = 0.025. Mean glucose was lower on Day

1 vs Day 3, p = 0.043. CME (open bars) vs HIIE (hatched bars). ......... 84

Figure 5.3. Mean differences (with 95% CI) in 24h glucose concentrations between A:

HIIE on Day-2 minus Day-1; B: CME on Day-2 minus Day-1; C: HIIE

on Day-3 minus Day-1; D: CME on Day-3 minus Day-1. Subplot next

to each graph represents mean glucose difference concentration and

standard deviation over each respective 24 h period. Arrow indicates

when exercise was completed. ............................................................... 85

Figure 6.1. Identification of Tregs via flow cytometry. ............................................... 99

Figure 6.2. Mean (± SD) concentration of acute phase reactants; Alpha 1-acid

glycoprotein, Homocysteine, Fructosamine, high sensitivity C-reactive

Protein and Transforming Growth Factor-beta. *, post-exercise was

greater than pre-exercise in the 12th session, p < 0.001; ^, Post-training

was lower than pre-training values, p = 0.037. .................................... 102

Figure 6.3. Mean (± SD) concentration of unstimulated and LPS-stimulated

inflammatory cytokines pre- and post-exercise in the first and 12th

session of training. *, post-exercise was greater than pre-exercise in the

12th session, p < 0.05. Closed circles represent first session; open circles

represent 12th session. .......................................................................... 104

Page 15: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xv

List of Tables

Table 3.1. The relationship of pro-inflammatory and anti-inflammatory markers to

T2DM, exercise and glucose control ..................................................... 50

Table 4.1 Descriptive characteristics of the participants. All data are presented as

mean (SD). FBG, Fasting blood glucose; WC, waist circumference;

VO2, peak VO2 achieved. ...................................................................... 60

Table 5.1. Physical activity monitor measurement ....................................................... 81

Table 6.1. Descriptive characteristics of the participants at baseline (data presented as

mean (SD)) and changes in anthropometric measures in response to

exercise training; where effect of training is represented by Cohen’s d

effect size and p-values. BMI, body mass index; WC, waist

circumference; VAT, visceral adipose tissues; VO2peak, peak VO2

achieved. ................................................................................................ 95

Table 6.2. Measures of glycaemic control and insulin resistance pre- and post-training

(presented as mean (SD)) with associated effect (Cohen’s d) of the

exercise training intervention. FBG, Fasting blood glucose; AUC, Area

under the curve; HOMA2-IR, Homeostatic model assessement index 2;

HOMA2-B, beta cell function; HOMA2-S, insulin sensitivity; ISI,

Insulin sensitivity index ....................................................................... 101

Table 6.3. Absolute count and percentage of T cells in whole blood. Data displayed as

mean (SD). ........................................................................................... 103

Page 16: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

xvi

Page 17: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

17

Chapter 1 Introduction

Obesity and physical inactivity are risk factors associated with the development of type 2

diabetes mellitus (T2DM) [1, 2]. The adipose tissue accumulation characteristic of obesity is

linked with the onset of insulin resistance [3], both centrally (liver) and peripherally (skeletal

muscle, adipose tissue) [4]. The compensatory hyperinsulinemic response augments the

insulin resistance and in the longer-term leads to greater glycaemic impairment, consequently,

increasing the risk of T2DM [5].

In lean heathy individuals, adipose tissue is predominantly infiltrated by alternatively activated

macrophages which results in a pervasive anti-inflammatory environment, supported by

regulatory T cells [6]. However, obesity facilitates a shift in the state of these infiltrated

macrophages towards a classically-activated state, increasing the secretion of pro-

inflammatory markers and reducing the suppressive capacity of regulatory T cells and anti-

inflammatory cytokines such as Interleukin 10 (IL-10) [7, 8], influencing insulin signalling

and glucose homeostasis [9]. Acute phase protein such as C-reactive protein (CRP) and pro-

inflammatory cytokines such as tumour necrosis-alpha (TNF-α) and interleukin-6 (IL-6) have

consistently been associated with obesity and T2DM [10-13]. The production of TNF-α in

adipose tissue is implicated with increased insulin resistance in mice [14]. Specifically,

neutralization of TNF-α with a soluble TNF-α receptor resulted in improvements in insulin-

resistance in mice [15], and that TNF-α mediated insulin resistance by impairing proteins in

the proximal insulin signalling pathway [15]. Given the importance of the insulin-stimulated

translocation of glucose transporter-4 (GLUT4) to the plasma membrane in tissues such as

liver, adipose tissue and skeletal muscle for glycaemic control, this impairment leads to

prolonged periods of hyperglycaemia [16]. The production of TNF-α is also of importance as

it induces the production of IL-6 which is associated with increased glucose intolerance and

reduced insulin sensitivity in pro-inflammatory environments [17, 18]. Furthermore, the

production of IL-6 induces the circulating concentration of CRP [10] which is also associated

with obesity [12], which suggests to not only be a traditional risk marker for cardiovascular

disease but also T2DM [17, 19]. In addition to these well-established inflammatory markers,

a growing list of inflammatory mediators such as adiponectin, matrix metalloproteinases and

osteopontin have been implicated in the adaptation to the pro-inflammatory state associated

with obesity-related diseases [20-26].

Regular exercise has proven to be an effective tool in managing glycaemic control when

compared to glucose lowering drugs in T2DM [27, 28]. In addition to insulin, exercise

promotes glucose uptake in the muscle through the translocation of GLUT4 transporters from

Page 18: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

18

its intracellular pool to the plasma membrane. Indeed, positive health outcomes are observed

following long-term exercise training including reduced adiposity, improved glucose tolerance

as well as reduced levels of pro inflammatory cytokines [29]. High-intensity intermittent

exercise, comprising several ‘bursts’ of intense efforts interspersed with periods of lower-

intensity exercise is an area of research that has received increasing attention due to it being

perceived as being more enjoyable than continuous moderate-intensity exercise [30] and

despite the reduced time-commitment relative to continuous moderate-intensity exercise, it

has been found to be at least equivalent in improving aerobic capacity [31]. Additionally,

various research groups have identified improvements in glycaemic outcomes following acute

high-intensity intermittent exercise in individuals with [32, 33] and without type 2 diabetes

[34], although not when compared to moderate-intensity exercise [35]. A recent review of

high-intensity intermittent exercise on glucose control has highlighted its effectiveness in

patients with and without type 2 diabetes [36]. In addition to acute exercise, improvements in

glycaemic control have been reported following high-intensity exercise training, even in the

short-term (≤ 2 weeks) [32, 33, 37, 38]. Aside from exercise intensity, previous meta-analysis

has also reported the importance of exercise volume (frequency and duration) to be a

significant factor in its effectiveness in improving glycaemic control [27]. With this in mind,

a number of short-term training studies have assessed the glycaemic responses to consecutive

exercise sessions for periods of 10 days [36, 39-41], and thereby increasing the frequency of

exercise in this short period, and reported improved glycaemic control. A review of exercise

and glycaemic control is covered in Chapter Two.

Whilst exercise training results in glycaemic improvements, exercise training similarly

demonstrates large immune-modulating effects, and specifically, improvements in reducing

chronic inflammatory markers such as CRP, IL-6 and TNFα [42]. Long term (≥ 3 months)

exercise training has been found to improve CRP [43], as well as reduce pro-inflammatory

and increase anti-inflammatory markers associated with the development of cardiovascular

disease and type 2 diabetes [29, 43, 44]. Additionally, the effects of exercise on changes in the

cellular immune compartments have been more recently assessed, with regulatory T-cells

being identified as important cells due to the suppressive anti-inflammatory nature of these

cells in down regulating the pro-inflammatory tissue environment [6, 45]. While a large body

of evidence exists which has examined the effects of exercise on glycaemic control and

immune responses independently, there is currently only limited research assessing the role

exercise on glucose control concomitant with detailed inflammatory responses. Chapter three

will therefore review the extant literature on exercise, inflammation and T2DM from an

immune perspective.

Page 19: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

19

Chapter Four investigated the role of acute exercise on glycaemic control and the subsequent

changes in the IL-6 related inflammatory markers. Although a positive association between

resting IL-6 and a risk for type 2 diabetes exists [46], an increase in IL-6 following exercise

has been implicated in controlling glycaemia where its response has been shown to associate

with increased exercise intensity [47]. The relationship between IL-6 and glycaemic control

may be a consequence of the IL-6 distinct pathways with its soluble receptors, IL-6 receptor

(IL-6R) and soluble glycoprotein (sgp130). This chapter assessed the role of exercise intensity

on glycaemic control as well as the acute response to IL-6 and its receptors. Specifically,

overweight sedentary adults performed either high-intensity intermittent exercise or

continuous moderate-intensity exercise (cross-over design) and glycaemia and IL-6 related

responses were assessed 24 h prior to exercise, immediately post-exercise, 1 h post-exercise,

and 25 h post-exercise. Glycaemia was assessed following an oral glucose tolerance test which

was completed to calculate glucose area under the curve (AUC) between each trial and the IL-

6 related pathways were measured to assess concurrent changes associated with glucose

tolerance. The exercise protocols in this study were matched for workload as well as exercise

duration.

Using the same design and participants from Chapter Four, the purpose of Chapter Five was

to assess the role of exercise intensity on diurnal glycaemic control and novel inflammatory

markers associated with the initial stages of adipose-tissue expansion and T2DM. In this vein,

the matrix metalloproteinases (MMPs) have now been shown to play potentially important

roles early in obesity-related disease progression [48-50] and appear to be differentially

expressed in clinical populations, with increased MMP-2 concentrations in obese patients [22];

increased MMP-2 concentrations [21] and decreased MMP-3 concentrations in patients with

type 2 diabetes mellitus (T2DM; [23]). Osteopontin (OPN) on the other hand is a potentially

important regulator of inflammation, obesity and diabetes [24], playing a particularly

important role in macrophage movement and accumulation [51] and stimulating expression of

MMP-2 [52]. The purpose of this chapter therefore, was to determine the acute (pre-,

immediately post-, 1 h post- and 25 h post-exercise) effects of exercise on the inflammatory

markers MMP-2, MMP-3 and OPN, and concomitantly assess changes in the 48 h glucose

response to two different types of exercise in overweight and sedentary individuals using

continuous glucose monitoring.

Chapter Six extends the findings from Chapters Four and Five, by way of recruiting

individuals previously diagnosed with T2DM. Additionally, this chapter sought to disentangle

the inflammatory responses to an acute exercise bout, in these sedentary individuals and the

training responses. To this end, this chapter adopted a short-term training program, consisting

Page 20: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

20

of 12 training sessions performed on successive days. To distinguish the acute and training

responses, inflammatory cytokine (unstimulated and LPS-stimulated cytokine) responses to

the first and last (12th) training session were assessed. The panels of cytokine included a range

of anti- and pro-inflammatory cytokines including IL-10, IL-6, and TNFα, amongst others. To

assess the impact of the training intervention, glycaemic control was assessed before and 48 h

after the last training session by completion of an oral glucose tolerance test. Additionally, an

alternative measure to glycated haemoglobin(HbA1c), fructosamine was also measured at

these time points which is more appropriate in capturing short term (around two weeks)

changes in glycaemic control [53]. With exercise training previously shown to be of anti-

inflammatory nature [29, 44], we also measured regulatory T cells to determine if production

of these cells were increased following exercise training which are responsible for promoting

the production of anti-inflammatory cytokines, in addition to maintaining immune

homeostasis [54] and its potential role in glucose control [55, 56].

Significance of the thesis

The contribution of the immune system in development of obesity-related disease, and

specifically T2DM, has long been acknowledged. In the past few years, the importance of

particular T-cell subsets such as the regulatory T-cells in obesity-induced inflammation have

become better understood and novel cytokines in the role of adipose tissue development been

identified. Concomitantly, the role of exercise in improving glycaemic control in individuals

with T2DM is now universally acknowledged, and the underlying mechanisms better

understood. The role of exercise in modulating components of the immune system have also

evolved rapidly in the last decade, with research shifting towards a better functional

understanding of the immune-changes in response to both acute and training responses. Much

work in these fields has occurred in isolation, focusing on the individual effects of exercise on

either glycaemic control or inflammation, but only limited research exists which has attempted

to bridge these fields with a view to gaining a deeper understanding on their interaction. The

work in this thesis aims to further our understanding of the inter-relationships between the

immune-modulating effects of exercise and the glycaemic responses to exercise, in sedentary

and overweight individuals with or without T2DM.

Page 21: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

21

Chapter 2 Exercise, glucose transport

and glucose regulation: A

brief review

Authors: Aaron Raman1, Gerard F. Hoyne2, Timothy J. Fairchild1

Affiliations:

1School of Psychology and Exercise Science, Murdoch University, Western Australia,

Australia;

2School of Health Sciences, University of Notre Dame Australia, Fremantle Campus,

Australia.

Correspondence: Aaron Raman

School of Psychology and Exercise Science

Murdoch University

90 South Street, Murdoch 6150, Perth, Western Australia

Email: [email protected]

The manuscript in this chapter is prepared for submission to the Sports Medicine.

Page 22: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

22

The maintenance of glycaemia within a physiological range is not trivial, with postprandial

glycaemic fluctuations occurring on the background of fasting glucose concentrations. Poor

glycaemic control is associated with increased risk of cardiovascular disease [57, 58], renal

disease [59], non-traumatic amputations [60] and all-cause mortality [61]. Unfortunately the

prevalence of poor glycaemic control, either as type 2 diabetes mellitus (T2DM), impaired

fasting glucose (IFG) or impaired glucose tolerance (IGT) is increasing globally; where an

estimated 347 million people have diabetes, 90% of whom have T2DM [62].

The benefit of longer-term exercise training on glyacemic control (assessed by

glycated haemoglobin [HbA1c]) is well established [63-65]. Improved glycaemic control is

primarily attributed to the improved insulin-mediated glucose disposal, either through

increased insulin sensitivity or insulin responsiveness [33, 34, 38, 66-68] rather than lower

fasting glucose concentration [69]. While the general consensus clearly outlines a beneficial

role of exercise with respect to glycaemic control, variability regarding the magnitude of

benefit on glycaemic control exists. This variability may be due to the study population, the

duration of the intervention, the type of exercise adopted or the outcome measure being used.

The purpose of this review is to synthesise the information garnered from classic in vitro and

in situ glucose uptake studies, and then apply this information to help explain the more recent

clinical findings in response to exercise. The role of exercise intensity and duration will be a

key focus of this review.

2.1 Glucose Transport

Cellular uptake of glucose occurs through facilitated diffusion using a carrier protein from the

glucose transporter (GLUT) family. Glucose transporter type 1 (GLUT-1) is ubiquitously

distributed and does not change in response to hormonal or other stimuli [70, 71]. For this

reason, GLUT-1 is primarily responsible for glucose transport under basal conditions. The

GLUT-4 on the other hand, are present primarily in adipose cells [72, 73] and striated (skeletal

and cardiac) muscle cells [16], and translocates to the cell membrane from intracellular storage

sites [74]. The translocation of GLUT4 from its intracellular compartments to the sarcolemma

and t-tubules may be initiated by insulin binding [75], or in the case of muscle cells, in response

to contraction (Figure 2.1;[76-80]).

Page 23: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

23

Increasing doses of insulin results in increased recruitment of GLUT4 transporters to the cell

membrane [81]. Accordingly, GLUT4 plasma membrane content increases with increasing

contraction rates [82]. Insulin has been shown to increase the GLUT4 plasma membrane

expression by up to a maximum ~7-fold (1mU/ml), whilst muscular contractions may

maximally increase (10 Hz for 5 min) the GLUT4 plasma membrane expression up to ~4-fold

[82]. When maximal insulin stimulation (1mU/ml) and muscle contractions (10 Hz for 5 min)

were combined, a 9.3-fold increase in the GLUT4 plasma membrane expression was observed

(versus the 7-fold and 4-fold increase by insulin and contraction alone, respectively) [82]. This

additive effect may be explained by the existence of discrete intracellular GLUT4 pools, which

are mobilized via discreet mechanisms [74, 77, 82]. Of relevance, GLUT4 protein content

varies between muscles comprising different muscle fibre compositions [83], and the

recruitment of GLUT4 to the cell membrane in response to either insulin or contraction has

also been shown to vary in Male Wistar rats. Specifically, contraction was shown to elicit

greater GLUT4 translocation in the flexor digitorum brevis (predominantly type 2a fibres)

muscle, while Soleus (predominantly type 1 fibres) demonstrated greater GLUT4 recruitment

with insulin (i.e., Soleus) at the same relative insulin concentration and contraction protocol

[83]. Of significance to the present review, is the observation that in individuals with T2DM,

exercise training has been shown to increase the total GLUT4 content in skeletal muscle [41]

and adipose tissue [73].

Figure 2.1. Glucose Transporter number of subcellular membrane fractions from skeletal

muscle. Data derived from [76].

Page 24: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

24

Figure 2.2. (A) Represents the glucose transporter number (ρmol/mg; based on cytochalasin

B binding sites) in non-exercise (Non-EX), immediately after exercise (0h Post-EX), 0.5 h

after exercise (0.5h Post-EX) and 2 h post-exercise (2h Post-EX). The exercise consisted of

treadmill running at 20m/min up 10% gradient. Data based on [80]. (B) Represents the effect

of 1 and 2 days of swimming on the number of glucose transporter protein (derived from

muscle homogenates containing 50ug of protein). Data based on [84].

Increased GLUT4 content in the cell membrane is associated with an increased rate of glucose

transport (Figure 2.2; [78, 82, 84-86]). Indeed, the increased glucose transport in response to

insulin [75] and contractions [87] as well as their additive effects (suggesting independent

mechanisms) [88] were established prior to the identification of GLUT4 (albeit a transporter

protein was known to be involved [75]), due to the relatively easier method of assessing

glucose transport. Using in vitro models, contraction-induced glucose transport was increased

1.5-fold, 1.6-fold, 2.7-fold, 3.1-fold and 4.7-fold over basal glucose transport at contraction

frequencies of 1 Hz, 2.5 Hz, 5 Hz, 7.5 Hz and 10 Hz, respectively; while GLUT4 content was

increased by 2.5-fold and 4.1-fold at contraction rates of 5Hz and 10Hz, respectively [82].

When the frequency of contraction was increased to 25 Hz, there was no additional glucose

uptake, indicating a plateau and that the maximum rate of glucose uptake was ~60% of a supra-

maximal (1mU/mL) concentration of insulin [82]. Since these earlier experiments, a number

of additional studies now support the increased glucose transport associated with contractions

and insulin, and the additive effect when compared to each stimulus independently [86, 88-

94].

Page 25: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

25

With respect to contractions, increases in glucose transport have not only been seen with

increases in the intensity of contractions, but also the duration of contractions [95-97]. These

increased rates of glucose transport persist into recovery following exhaustive exercise,

wherein 10-min post-exercise the rate of glucose transport is ~4.7-fold higher than pre-

exercise; 30 min post-exercise the rate is ~3.3-fold higher than pre-exercise; 60-min post-

exercise the rate is ~2.6-fold than pre-exercise; and 180-min post-exercise the rate is ~1.5-fold

higher than pre-exercise [98].

Figure 2.3. Glucose uptake and the effect of insulin. Control group was not exercised;

Postexercise group completed 45 minutes of treadmill running at 18m/min, with the last two

minutes completed at 31m/min. Results are means ± SE with n=2-13 observations per data

point. p < 0.05 when compared with control values [99].

Page 26: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

26

2.2 Insulin sensitivity and responsiveness

Insulin responsiveness refers to the rate of glucose transport associated with a maximally

effective insulin concentration (i.e., maximal glucose transport via insulin); while insulin

sensitivity refers to the concentration of insulin required to achieve 50% of this maximal effect

on glucose transport [100]. A classic example of improved insulin sensitivity following

exercise was demonstrated by Richter and colleagues (Figure 2.3 [99]). It is noteworthy that

no increase in glucose transport post-exercise was observed (as would be expected given the

GLUT4 translocation with contractions), however, animals were provided ad libitum food

access between exercise cessation and the perfusion experiments which were conducted in the

subsequent few hours [99].

The duration of increased insulin sensitivity following exercise may persist from 3 hours to 48

hours and is dependent on the dietary status [99, 101, 102] and likely affected by the exercise

performed (duration, intensity, type). In rats fed a carbohydrate-free diet post-exercise, the

exercise-stimulated increases in glucose uptake (7.8-fold above non-exercised levels) were

still largely present 18h later (3.5-fold increase above non-exercised levels), however in rats

fed a 60% carbohydrate diet the exercise-stimulated glucose uptake was no longer present

(1.1-fold increase above non-exercise levels) [103]. This indirectly aligns with the earlier

findings of Richter and colleagues [99], given the lack of difference in glucose transport post-

exercise when animals were provided food in this period.

It is important to note that despite the extent of evidence from in vitro, in vivo and in situ

studies demonstrating significant increases in glucose transport (measured by 3-

MethylGlucose [3-MG]) following exercise or contractions, albeit to varying rates of glucose

transport, some in vitro studies have reported no increase in glucose transport following

contractions [89, 100, 104]. This discrepancy led to the identification that serum, and

specifically a factor within serum, was critical for the increased glucose uptake following

muscle contractions [89, 100]. For instance, glucose transport (epitrochlearis muscle; 3-MG)

in the presence of insulin was i) increased 0.29±0.06 µmol.ml-1.10min-1 at rest; ii) increased

0.86±0.10 µmol.ml-1.10min-1 3.5h following swimming; iii) increased 0.19±0.05 µmol.ml-

1.10min-1 3.5h following in vitro contractions (performed 15-min post excision while

immersed in Krebs-Henseleit buffer; KHB) [89]. A follow-up experiment in the same study

showed insulin sensitivity was largely retained 3.5h following the in vitro contractions, when

performed immediately post-excision (0.43±0.04 µmol.ml-1.10min-1) versus a 15-min

incubation in KHB buffer prior to contractions (0.18±0.03 µmol.ml-1.10min-1), although

insulin sensitivity was not to the same degree as observed in situ. This rapid degradation in the

Page 27: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

27

contraction-stimulated increase in insulin sensitivity associated with incubation in KHB

buffer, suggested an important biological factor facilitating this increased sensitivity is rapidly

lost. However, the exact property or molecule of the serum factor remains unknown; other

than being a protein [100].

2.3 Tissue specific glucose transport in humans

The measurement of glucose transport in humans is complex and relies primarily on tracer-

labeled glucose (such as [13C] glucose, [2H]glucose) [105, 106], which when coupled with

tissue biopsies [107], measures of arterio-venous glucose (AVg) difference [108], or imaging

techniques (such as dynamic positron emission tomography [109] and magnetic resonance

spectroscopy [110]) allow specific tissue/site glucose uptake rates. The findings in humans

during 40 min of continuous exercise is consistent with animal work, demonstrating increased

glucose uptake with increasing (up to ~80% VO2peak) exercise intensity [111, 112]. The greater

recruitment of total muscle fibres and in particular, the greater recruitment of fast-twitch fibres,

likely underpin the higher rates of glucose uptake [111, 113], although there does appear to be

a high degree of heterogeneity in the glucose uptake rates in fast-twitch muscles of humans

[114]. Furthermore, the relationship between exercise intensity and glucose uptake is also of

importance when applied to disease (T2DM) such that a greater reduction in plasma glucose

was evident with increased intensity, reinforcing the significance of exercise for glucose

control [115]. In addition to exercise intensity, exercise duration is associated with an initial

(~30 min) rapid increase in glucose uptake [112] and a subsequent slower increase or plateau

in glucose uptake over the next 60-90min [87, 115]. Insulin secretion is suppressed during

exercise in a dose-dependent manner [116], wherein high intensity exercise is associated with

increased leg glucose uptake, strongly suppressing insulin secretion and its circulating

concentration [117-119].

2.4 Changes in systemic plasma glucose post-

exercise: Looking beyond skeletal muscle

The beneficial effect of acute exercise on insulin responsiveness and sensitivity in muscle

tissue has been confirmed during euglycaemic clamps performed prior to, and 1h and 24h

subsequent to a 60 min cycling bout [120]. However, despite this localised improvement in

glucose uptake, systemic postprandial glucose (PPG) tolerance remains either unchanged

Page 28: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

28

[121-124], increases [120, 125-129] or decreases [130-132] in the hours following an acute

bout of moderate- and high-intensity exercise. The disparity between peripheral and systemic

glucose concentrations may be due to an increased rate of hepatic glucose release, which

continues for approximately 30 min after cycling in overweight/obese individuals with NGT

[133] as well as a concomitant decrease in insulin sensitivity in tissues other than the

contracting musculature [134]. Although in healthy middle-aged individuals, the glucose-

AUC was shown to be highest (worst) immediately post-exercise, lowest (best) the day

following exercise, and then demonstrated a progressive increase in the subsequent days,

although even after 7 days, were still not as high as that experienced immediately post-exercise

[130]. Since systemic plasma glucose concentrations are critical to successful management of

individuals with impaired glucose tolerance, impaired fasting glucose and/or T2DM [64, 135],

the underlying mechanisms explaining this apparent discrepant finding between increased

localized glucose uptake and systemic glucose concentrations requires clarification.

Systemic plasma glucose concentration reflects the balance between the rate of

glucose appearance (Ra) and rate of glucose disappearance (Rd). This balance is complicated

by the multiple factors contributing to Ra and Rd respectively. For instance, glucose Ra in the

fasted period is principally governed by total hepatic glucose release, while in the postprandial

period is governed by total hepatic glucose release and the glucose in the portal vein (dietary

glucose absorbed from the small intestine) remaining following the first-pass; while Rd is the

sum total of glucose uptake by all cells of the body. The balance between Ra and Rd becomes

more complex with acute exercise, since both Ra and Rd may be affected, specifically i) acute

exercise may increase- in orders of magnitude- glucose uptake into the exercised/contracted

muscles, whilst concomitantly decreasing glucose uptake in other tissues of the body; ii) acute

exercise may increase net hepatic glucose release and rate of glucose absorption from the small

intestine [133]. The underlying mechanisms regarding these changes are discussed below, with

specific emphasis on free fatty acids (FFA), epinephrine, glucagon, growth hormone and

cortisol. A previous review has also proposed a model which discusses the potential

mechanisms of Akt Substrate (AS160) which may explain the increased post-exercise glucose

uptake by acting as a potential trigger, memory element and/or mediator of increasing insulin

sensitivity [136].

2.4.1 Glucoregulatory effects of key Hormones:

Epinephrine, glucagon, growth hormone and

cortisol

Increased hepatic glucose output is essential in sustaining exercise capacity and preventing

hypoglycaemia [137, 138], and the increased glucose output continues for up to 45-60 min

Page 29: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

29

into the recovery period [133]. This increased glucose output coincides with increases in

glucagon, catecholamine and cortisol concentrations, and decreases in insulin concentration

[138, 139]. Of these hormones, single infusions of epinephrine and glucagon are associated

with increases in plasma glucose during rest, with glucagon stimulating hepatic glucose output

to the greatest effect, while cortisol appears to have negligible effect in isolation [96, 140-

142]. In accordance, glucagon- in concert with the associated decrease in insulin

concentration- has been shown to play the greatest role in increasing the hepatic glucose

release observed during exercise [143, 144]. The role of the catecholamines, and specifically

epinephrine on increased hepatic glucose output have been shown to be minimal via studies

conducted with hepatic denervation [145], β- and α-adrenergic receptor blockade [146], and

adrenalectomy [147]. It is noteworthy that the hormones appear to act synergistically, with the

addition of epinephrine and glucagon being greater than either alone, and the addition of

cortisol appears to prolong the action of these hormones on the liver [140].

While the effects of glucagon appear to be largely limited to the liver, epinephrine has been

shown to play important roles in glucose clearance [148]. In perfused rat muscles, epinephrine

(25 nM) significantly increases in GLUT4 content and glucose transport (22%-48%), wherein

the greatest increase occurs in oxidative muscle (red gastrocnemius and soleus) when

compared to muscles perfused without epinephrine [149]. In contrast, infusion of epinephrine

(25 nM) together with insulin (20 mU/ml) partially inhibits the insulin stimulated glucose

transport when compared to insulin infusion (20 mU/ml) alone, despite the persistent increase

in GLUT4 plasma membrane content [149]. Oxidative muscle appears to be consistently more

sensitive to epinephrine which is likely a result of the greater number of β2-adrenergic

receptors [150]. Subsequent work found the inhibition by epinephrine (24 nM) persists at 50

mU/ml but not 100 mU/ml of insulin, even when the concentration of epinephrine was

increased to 500 nM [151], suggesting that very high levels of insulin overcome this inhibition

by epinephrine. To assess the mechanism associated with this downregulation, insulin receptor

substrate-1 associated phosphatidylinositol 3-kinase (PI3K) activity was assessed and found

to be suppressed with addition of epinephrine [151], although this inhibitory effect was ablated

at higher insulin (100mU/ml) concentration.

The effect of epinephrine in humans has been assessed during a euglycaemic clamp performed

with or without epinephrine, along with the β1-adrenergic receptor antagonist metoprolol, or

the β1- and β2-adrenergic receptor antagonist propranolol [152]. The major findings (Figure

2.4) were i) glucose Ra decreased substantially with the addition of insulin, while Rd

concomitantly increased; ii) Addition of epinephrine with the insulin, resulted in an increased

Ra relative to insulin infusion alone, while the Rd of glucose was reduced; iii) this effect was

Page 30: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

30

exerted through the β2-adrenergic receptors as evidenced by propranolol being able to ablate

the insulin-antagonizing effect of epinephrine while this inhibitory effect persisted in the

presence of metoprolol [152].

Figure 2.4. Rate of glucose appearance (Ra) and disappearance (Rd) during steady state

glucose infusion rate. A. Measures taken before the clamp (PLA), during the clamp at low

(Low Ins; 4mU/l) and high (High Ins; 51mU/l); B. Measures taken before the clamp (PLA),

during the clamp at low (Low Ins; 4mU/l) and high (High Ins; 51mU/l) insulin infusion rates

with addition of epinephrine (Low Ins +A; High Ins + A); C. measures taken during the Low

insulin clamp (40 mU.(m2)-1.min-1; Ins), epinephrine only (A), insulin with epinephrine and

propranolol (Ins+A+Pro), insulin with epinephrine and metoprolol (Ins+A+Met); D. Measures

taken during the High insulin clamp (1200 mU.(m2)-1.min-1; Ins), epinephrine only (A), insulin

with epinephrine and propranolol (Ins+A+Pro), insulin with epinephrine and metoprolol

(Ins+A+Met).

Page 31: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

31

An increase in growth hormone is also evident following exercise [143] concomitant with an

increase in exercise duration and intensity [139, 153]. Growth hormone has been implicated

in the development of insulin resistance, which was identified as being due to both increased

hepatic glucose output and reduced glucose clearance [154]. While growth hormone does

appear to promote gluconeogenesis and increased hepatic glucose production [155, 156], this

effect seems minimal [141]. In muscles, increased concentration of growth hormone has been

shown to down regulate glucose transporters (GLUT1 and GLUT4) and glucose uptake [157].

Increases in growth hormone and epinephrine are also accompanied with increased lipolysis,

and consequently, increased free fatty-acid (FFA) concentrations [139, 154, 155, 158-160].

The increase in circulating FFA interferes in glucose uptake via both insulin-dependent [161,

162] and independent effects via decreased cellular glucose utilisation [116, 163-165], which

can act synergistically [3]. During exercise in rats, increased concentration of FFA resulted in

significantly higher blood glucose [166], while in humans, elevated plasma FFA

concentrations following a high-fat diet resulted in increased blood glucose [167].

2.4.2 The role of exercise intensity

Increases in the concentration of catecholamines (epinephrine and norepinephrine), growth

hormone and glucagon are positively associated with increases in exercise intensity and

duration [168-171]. Likewise, utilisation of muscle glycogen increases with both intensity and

duration of exercise [172-176] with plasma glucose utilization demonstrating a similar trend

[172]. Accordingly, when matched for work, low-moderate intensity (40% VO2peak) exercise

has been shown to result in a similar GLUT4 recruitment to high-intensity (80% VO2peak)

exercise [177], and this translates to similar plasma glucose responses to an oral glucose

tolerance test performed 1h [126, 129] and 24h post-exercise of low- or high-intensity [126].

Reductions in muscle glycogen concentration, have been associated with increased glucose

uptake [115]. In longer-term training studies (6 months), total work has been shown to be more

important than either intensity or duration alone [178].

More recently, interval-based training in the form of high-intensity intermittent exercise

(acute) or high-intensity intermittent training (‘longer-term’) has gained increasing attention

[31, 179]. Indeed, HIIE has been shown to decrease post-meal glucose AUC and reduce

hyperglycaemia during the 24h following exercise as measured by a continuous glucose

monitoring system (CGMS) in overweight/obese subjects with T2DM [32, 33]. Despite the

improvement in 3h post-meal glucose AUC, the changes in postprandial glucose remains

Page 32: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

32

unclear as 24h average blood glucose was not different [32]. Additionally, very few studies

have adopted a comparator condition, with the initial studies adopting a single-arm, pre- and

post-assessment design only [33] or comparing glucose changes to a no-exercise control

condition [32]. Indeed, one study comparing the effect of interval-based training against

moderate-intensity (75% VO2peak) exercise, showed greater improvements in insulin sensitivity

with moderate-intensity exercise compared to sprint interval exercise in overweight/obese

individuals; although an improvement in glucose AUC was not evident [35]. Since the initial

landmark studies on high intensity interval exercise and high intensity intermittent training

[33, 180], questions remain on whether this form of training is superior to a bout of work-

matched continuous moderate-intensity exercise.

2.5 Short-term consecutive training

Meta-analyses have identified the benefits of chronic (>8 weeks) exercise-training on

improvements in HbA1c [28, 179]. There is also evidence of improved glycaemic control

following short-term (≤2 weeks; 3 to 10 sessions) exercise training [33, 34, 38, 40, 41, 67,

115, 181-185], despite large heterogeneity between studies.

Short-term high intensity training studies remain in conflict as mixed findings have been

reported in the improvement of glucose AUC in non-T2DM individuals following two weeks

of training. Although both training protocols comprised of Wingate exercise protocols (all-out

cycling efforts for 30s), a reduction of glucose AUC following an oral glucose tolerance test

was only evident in young non-obese individuals [67, 186], with no such improvement in

overweight/obese males [187]. Despite this difference, both studies found short-term training

to result in an improvement in insulin sensitivity, derived from insulin sensitivity indices.

Interestingly, the improvement of insulin sensitivity was evident two to three days following

exercise in the young individuals [67, 186], rather than 24 h following the last exercise bout

in the overweight/obese males [187]. Additionally, a comparable study did not find

improvements in insulin sensitivity, which may be explained by the lack of improvement in

GLUT4 recruitment measured post-training [185], contrary to previous work where increased

GLUT4 was reported after a week of exercise training [181]. The difference in training

stimulus may also be considered as a limiting factor as the protocol implemented an 8-12s

Page 33: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

33

active period followed by 80s of recovery which resulted in a lower training volume [185].

Similar short-term exercise training employing high intensity exercise has also been completed

in individuals with T2DM [33]. The exercise protocol was completed at 90% HRmax rather

than the exhaustive Wingate protocol, Little et al. demonstrated that six sessions of interval

training (10 x 60s @ 90% HRmax) reduced both 24 h blood glucose concentrations as well as

post-meal 3 h postprandial glucose [33]. Although insulin sensitivity was not measured, the

improvement in glycaemic control was accompanied by increased GLUT4 protein of ~360%

highlighting the importance of exercise intensity and volume [33]. With improvements seen

only after six sessions over a two-week period, further research has investigated the impact of

consecutive days of exercise training for up to seven days in individuals with T2DM [40, 41,

115, 182, 184, 188]. The consequence of seven days of consecutive training results in an

increase training volume although these protocols varied between 50 to 70 minutes of exercise

at 50% to 75% of VO2peak consisting of treadmill and cycling exercise, or both. The increase

in training volume resulted in improved insulin sensitivity as demonstrated by increase insulin-

stimulated glucose disposal [41, 188], as well as reduced insulin and glucose AUC [184].

Gorman et al. also demonstrated that the increased insulin sensitivity may be attributed to the

increase in GLUT4 content following 7 days of exercise [41]. These examples have shown

positive outcomes in insulin sensitivity and reduced glucose AUC, however, two studies [182,

183] failed to show improvements in glucose AUC, whilst Kang and colleagues did not find

improvements in either glucose AUC nor insulin sensitivity following an oral glucose

tolerance tests [115].

Currently, it is not known whether exercise intensity, exercise volume or a

combination of increased exercise intensity and exercise volume is important in driving the

improvements seen in insulin sensitivity and glucose tolerance.

Page 34: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

34

2.6 Conclusion

Despite existing evidence suggesting improvements in insulin sensitivity and glycaemic

control following exercise, there remains debate concerning the extent of improvement in

glucose tolerance. This debate remains due to significant heterogeneity in existing literature,

and methodological limitations such as a lack of a comparison group [32-34]; unmatched

groups in exercise duration and intensity [115]; volume [177, 178]; and non-standardised

glucose assessment via continuous glucose monitoring [33-35, 102] versus oral glucose

tolerance test [130, 131, 133]. Work by Knudsen [133] provided an updated insight into the

post-exercise meal effect on glucose tolerance which confirms the results of previous research

[130, 131] in healthy individuals such that plasma glucose and glucose AUC was significantly

increased post-exercise. This increase may be explained by the increased rate of glucose

appearance following exercise together with an increased rate of glucose disappearance.

Despite the increase in plasma glucose, insulin sensitivity was improved as measured by the

insulin sensitivity index which highlights an interesting point that reduced glucose AUC was

not evident together with this improvement. As suggested by the authors, the increase rate of

appearance may be due to reduced uptake in hepatic glucose following increased intestinal

absorption in splanchnic tissues. Further research is warranted to answer the limitations

discussed including time and work-matched exercise groups, a suitable comparison group

(moderate intensity exercise) over a training period, as well as the inclusion of relevant

comparison populations during high-intensity interval training. What can be confirmed

however is that plasma glucose is not consistently improved, and is instead increased following

exercise in subjects who have normal glucose tolerance [130, 132, 133] and with T2DM [96].

Page 35: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

35

Chapter 3 Low grade inflammation,

exercise and type 2

diabetes: A brief review

This chapter will not be submitted for publication.

Page 36: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

36

Obesity is characterised by the expansion of adipose tissue and is associated with an increased

risk of insulin resistance [3] and a chronic inflammatory response originating from adipose

tissue reaching systemic circulation [189]. As a consequence of obesity-induced insulin

resistance, chronic elevated blood glucose concentration leads to the development of Type 2

Diabetes (T2DM; defined by glycated haemoglobin (HbA1c) ≥ 6.5%). Evidence suggests that

increased adipose tissue contributes to a chronic state of low grade inflammation which is

more evident in obese individuals than lean healthy individuals, representing a link between

obesity, inflammation and T2DM [190, 191]. Although both subcutaneous and visceral

adipose tissue are associated with increased metabolic risk factors [192], studies have shown

greater importance of visceral adipose tissue on obesity related inflammation [193, 194].

Increased visceral adipose tissue leads to greater macrophage infiltration resulting in an

increase of several pro-inflammatory markers such as Interleukin (IL)-6 and tumour necrosis

factor alpha (TNFα) [7, 195], whilst elevated C-reactive protein (CRP) produced from

hepatocytes is also elevated [196]. These markers have been previously associated with T2DM

[46, 197]. The infiltration of macrophages can also be classed in at least two forms; anti-

inflammatory ‘alternatively-activated’ (M2) macrophages which may be polarised to

‘classically activated’ (M1) macrophages which produce pro-inflammatory cytokines

associated with insulin resistance [198].

An increase in obesity-induced pro-inflammatory marker, TNFα impairs insulin signalling is

associated with insulin resistance, affecting glucose homeostasis [14]. Neutralization of TNFα

in mice results in the attenuation of insulin resistance, further highlighting the direct

relationship between inflammation and insulin resistance [14]. Additionally, elevated IL-6 is

associated with an increased risk of T2DM through induction of CRP release which is also

associated with the development of T2DM [10]. In addition to the positive association of IL-

6 and CRP with T2DM, evidence has also identified a role of additional markers such as

Matrixmetalloproteinases (MMPs) which is related to the production of TNFα [199], whilst

IL-6 has been shown to mediate the expression of Osteopontin (OPN) which is elevated with

obesity and positively correlated with fasting blood glucose [200]. Obesity and T2DM are also

accompanied with a reduced anti-inflammatory profile, affecting factors such as adiponectin

[201], IL-10 [202] and regulatory T cells [6, 203] which are crucial in maintaining immune

homeostasis [204]. It is evident that inflammation is dysregulated in obesity and T2DM, thus

an appropriate intervention that targets both obesity and inflammation, such as increased

physical activity may be effective in the reducing the clinical effects of obesity-associated

chronic inflammation.

Regular exercise is beneficial in the management of blood glucose, reducing the risk of T2DM

[27]. Further to glycaemic control, acute exercise and exercise training also modulate the

Page 37: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

37

immune system, demonstrated by changes in the balance of both anti-inflammatory and pro-

inflammatory markers following exercise [29, 205, 206]. Although exercise stimulates an

immune response, physical inactivity leads to the impairment of glycaemic control [207]

which contributes to obesity and an increased risk of T2DM, consequently accompanied with

chronic inflammation. Therefore, regular exercise is not only important in managing

glycaemic control but may also contribute to exercise-induced improvements in chronic

inflammation, especially in individuals who are at risk, or have previously been diagnosed

with T2DM. Despite previous work highlighting the beneficial health effect of exercise, the

understanding of the inter-play between exercise and chronic low-grade inflammation and the

subsequent impact on T2DM remains limited. Since measurement of cytokines such as TNFα

or IL-6 does not directly cause T2DM in humans, but are rather associated with T2DM,

additional research is needed to understand the role of acute exercise and exercise training on

the regulation of classic inflammatory markers such as IL-6, CRP, TNFα and IL-10, as well

as related inflammatory markers including MMPs and OPN which have been reported to be

elevated and associated with obesity and/or T2DM [21, 208]. Combining the measurement of

inflammatory markers together with the assessment of glucose-related responses is crucial in

determining the role exercise may have in regulating both the inflammatory response and any

associated relationship with glucose homeostasis.

Although exercise and T2DM are accompanied with an inflammatory response which involve

a large number of inflammatory mediators, this review will focus on selected markers

including IL-1β, IL-6, IL-10, TNFα, CRP, adiponectin, OPN, MMPs and regulatory T cells.

Of the relevant markers, the review will then discuss the impact of acute exercise on the

inflammatory response and the beneficial role exercise training may have on regulating

chronic inflammation experienced in obesity and/or T2DM. A combination of these markers

were measured following acute exercise (Chapter Four and Five) in overweight/obese

individuals and following short-term training in individuals diagnosed with T2DM (Chapter

Six).

Page 38: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

38

3.1 Inflammation, obesity and T2DM

3.1.1 Pro-inflammatory cytokine markers associated

with T2DM

Studies have shown that a number of prominent inflammatory markers are elevated in T2DM

including CRP, IL-1 beta (IL-1β), IL-6 and TNFα [10, 11, 46, 209]. Of relevance, the release

of these markers are associated with increased risk towards the development of T2DM through

the disruption of insulin signaling where the insulin receptor is phosphorylated by serine

instead of tyrosine leading to the inhibition of downstream signal transduction, promoting

insulin resistance [15, 209-211].

Adipose tissue has been found to be a large source of IL-6 [212]. The production of IL-6 has

also been found to be affected by hyperglycaemia, both in vitro [213] and in vivo [214]. Since

hyperglycaemia is a consequence of obesity and insulin resistance, an elevated concentration

of IL-6 is correlated with increased BMI [215, 216], and is associated with the severity of

glucose intolerance and reduced insulin sensitivity [17, 18]. Thus, the increase in IL-6 in the

transition to obesity strongly supports the link between obesity and insulin resistance [217,

218], increasing the risk for development of T2DM [13, 219, 220]. Extending to the

association with insulin resistance, the release of IL-6 induces the production of CRP [217],

which is an important marker on vascular inflammation and atherosclerosis [221]. Plasma CRP

is correlated with body fat percentage, BMI, fasting plasma glucose and 2-hour plasma glucose

following a glucose tolerance test proving to be an important risk marker for not only

cardiovascular disease but also T2DM [10, 17, 19, 46]. Previous work has found CRP levels

to be inversely associated with insulin responsiveness [217], and significantly increased in

individuals with impaired glucose tolerance and T2DM, compared to control subjects [19, 46,

197, 217]. Additionally, a positive correlation between IL-6 and CRP with impaired glycaemic

control has been previously reported [17]. A progressive increase in the concentration of IL-

6 and CRP were also found together with increased severity of metabolic syndrome and T2DM

where they were lowest in non-diabetic individuals, intermediate levels in metabolic

syndrome-negative T2DM and highest concentration in metabolic syndrome-positive T2DM

subjects [219].

TNFα is a pro-inflammatory cytokine highly expressed in adipose tissue associated with

insulin resistance and T2DM. Seminal work by Hotamisligil and colleagues highlighted the

Page 39: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

39

relationship between TNFα and insulin resistance through neutralization of TNFα with a

soluble TNFα receptor in obese, insulin-resistant mice in vivo, which resulted in decreased

insulin resistance [14]. Further evidence have since supported the overproduction of TNFα in

obese human adipose tissue [222-224] particularly when compared to non-obese individuals

[225]. The role of TNFα was further demonstrated by an inverse association between TNFα

and insulin sensitivity demonstrated by reduced glucose disposal rate [224]. The main

mechanism by which TNFα promotes insulin resistance, occurs by limiting phosphorylation

of the insulin receptor and interfering with insulin action via TNFα-induced serine

phosphorylation of IRS-1 [15, 226]. The concentration of TNFα in obese individuals has been

demonstrated to reduce through weight loss following lifestyle modification with potential

positive effects on insulin sensitivity and insulin resistance evident from reduced insulin area

under the curve following a glucose challenge [227].

IL-1β, a major IL-1 family cytokine produced by macrophages [228], has previously been

implicated in type 1 diabetes through the destruction of pancreatic β cells [229]. In addition to

IL-6, TNFα and CRP, previous work has also reported elevated concentration of IL-1β in

T2DM [11, 13, 46]. This pro-inflammatory cytokine is a significant marker of the development

of T2DM, as the secretion of IL-1β has also been found to be promoted under high glucose

concentration during in vitro exposure of IL-1β supporting that the similar hyperglycaemic

environment experienced in T2DM may induce the production of IL-1β. Furthermore, in vivo

pancreatic sections of type 2 diabetic subjects were presented with elevated IL-1β but not in

nondiabetic control subjects [230], suggesting that the pancreas may be responsible for the

synthesis and release of IL-1β [231]. Additionally, the increase of IL-1β in T2DM patients

was also accompanied with reduced concentration of anti-inflammatory marker, IL-1 receptor

antagonist (IL-1ra) in vitro [232] highlighting a bias towards a pro-inflammatory environment

as a consequence of T2DM upsetting the balance of inflammatory cytokines. Thus, consistent

evidence has highlighted that T2DM is accompanied with elevated concentration of pro-

inflammatory markers including IL-6, CRP, TNFα and IL-1β. The release of these markers is

induced with increased levels of adipose tissue which is a main source of these markers in

addition to adipose-resident macrophages and crown like structures which reside in necrotic

adipocytes which infiltrate into adipose tissue as a consequence of obesity [233, 234]. These

inflammatory markers also extend further to associate with factors related to T2DM including

hyperglycaemia, insulin resistance, increased BMI, impaired glucose tolerance and decreased

insulin sensitivity.

Page 40: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

40

3.1.2 Additional inflammatory markers associated

with T2DM

In addition to the markers discussed above, three inflammatory markers associated with

vascular disease and coronary heart disease; Matrix Metalloproteinase (MMP) 2, MMP3 and

OPN have been previously linked with insulin resistance/T2DM [21, 208, 235, 236]. MMPs

are enzymes responsible for the breakdown activity in vascular extracellular matrix

remodeling during obesity-induced adipose tissue formation [48]. Their proteolytic activity

plays a role in plaque rupture and destabilization in atherosclerotic lesions [237]. Together as

a marker for vascular disease, the expression of increased MMP2, MMP3, MMP9 were

previously elevated in obese individuals with and without T2DM when compared to healthy

subjects [22, 23]. Goncalves et al. also highlighted that type 2 diabetic subjects presented

higher levels of MMPs when compared to those without T2DM, recognizing MMPs as

potential risk markers to consider for the development of T2DM [23]. Further work in cell

culture of adipocytes and high fat fed mice demonstrated enhanced MMP-3 release which

stimulated the release of TNFα and resulted in the inhibition of insulin-induced glucose uptake

in adipocytes. However, neutralization of MMP3 inhibited TNFα secretion, indicating that

increased MMP3 may promote insulin resistance through the release of TNFα [199]. Elevated

concentrations of MMP2 and MMP9 have also been found under high glucose conditions in

human umbilical vein endothelial cells in vitro, highlighting the significance of a

hyperglycemic environment as experienced in individuals at risk of T2DM, in addition to

vascular complications [238].

The formation of active MMP2 from pro-MMP2, can be induced by the release of OPN. OPN

is a pro-inflammatory glycoprotein associated with bone formation and is abundant in

atherosclerotic plaques, specifically in calcified plaques [239]. In addition to an increased risk

of atherosclerosis [240], OPN is associated with obesity, evident from increased concentration

in obese high-fat fed mice [241] and increased adipose tissue resulting from obesity in humans

[208]. Further work has also identified that an increase in IL-6 is a significant mediator to the

expression of OPN expression in macrophages [242]. Elevated OPN was found in obese

individuals with [20, 208] and without [208] T2DM when compared to healthy individuals

with normal glucose tolerance. This difference between populations was also highlighted from

a positive correlation of OPN with fasting blood glucose in obese individuals when compared

to a lean group [200]. Furthermore, increased OPN levels also correlated with an increased

percentage of body fat, as well as physical inactivity and physiological variables such as

reduced insulin, elevated QUICKI (insulin sensitivity index) and elevated CRP, highlighting

the relevance of OPN in the development of T2DM [243].

Page 41: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

41

3.1.3 Anti-inflammatory markers associated with

T2DM

The concentration of the anti-inflammatory cytokines IL-10 and adiponectin in blood are

inversely associated with the development of T2DM [244-247]. Adiponectin is exclusively

secreted and synthesized by adipose tissue macrophages and then released into blood [248].

Low adiponectin levels are also associated with decreased insulin sensitivity, with the lowest

levels seen in individuals with T2DM [249]. Furthermore, the production of adiponectin is

reduced by the release of TNFα, IL-6 and CRP [248, 250], suggesting that the obesity-induced

increase in pro-inflammatory markers is detrimental to the anti-inflammatory effect of

adiponectin. The protective effects of adiponectin is suggested to be glucose- and immune-

related, resulting in reduced blood glucose [251], increased insulin sensitivity [252] via the

induction of tyrosine phosphorylation of insulin receptor [247], inhibition of pro-inflammatory

cytokines TNFα and IL-6 [248], and stimulation of the production of anti-inflammatory

marker, IL-10 [253, 254].

IL-10 can be produced by a range of cells including monocytes, macrophages and regulatory

T cells, amongst other innate and adaptive immune cells [255]. A population based study

revealed that low concentration of IL-10 was associated with an increased risk of T2DM [203].

IL-10 is a potent anti-inflammatory cytokine that inhibits the production of pro-inflammatory

cytokines including IL-6, TNFα, and IL-1ẞ [256, 257]. Thus, the suppressive function of IL-

10 is crucial in maintaining and promoting an anti-inflammatory environment. The protective

effect of IL-10 in glucose control was also evident in IL-10-treated mice following a high fat-

fed diet, resulting in improved insulin signaling and sensitivity [198, 258] as well as the

attenuation of macrophage infiltration in insulin resistant mice [259]. In contrast, limited

production of IL-10 in murine models resulted in greater macrophage infiltration, contributing

to a greater pro-inflammatory response [260, 261]. The suppressive function of IL-10 is also

crucial to the function of regulatory T cells (Tregs) which produce IL-10 as a major

suppressive factor [262]. Tregs (CD4+CD25+CD127- T cells) play an important role in the

maintenance of self-tolerance and immune homeostasis [54]. Tregs can be generated in the

thymus and then migrate to peripheral tissues, or alternatively can be induced from peripheral

lymphocytes upon stimulation from cytokines [54]. Growing evidence have found that a

reduced number of circulating Tregs in obese individuals is associated with obesity and T2D

[202, 263-265] and is inversely correlated with BMI [6, 263] and HbA1c [202] which may

contribute to insulin resistance [6, 55, 266]. Additionally, the reduction of Tregs in visceral

adipose tissue has also been associated with decreased insulin sensitivity [55] and is also

Page 42: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

42

accompanied with reduced immune-suppressive capacity and a skewed pro-inflammatory

environment increasing the risk of insulin resistance [55, 263]. Tregs are important in the

suppression of a pro-inflammatory environment such as TNFα-induced insulin resistance via

secretion of IL-10 [198, 267]. Previous work has highlighted the importance Tregs such that

depletion of Tregs in obese mice has been demonstrated to lead to increased insulin resistance

in mice [55]. However, induction or the transfer of exogenous Tregs in mice resulted in

improved glucose tolerance and insulin sensitivity highlighting the therapeutic potential of

Tregs [55, 56].

3.2 Exercise and inflammation – intensity, mode

and duration

Regular physical activity has an important role in the prevention of T2DM [268],

cardiovascular disease [269] as well as all-cause mortality [270]. Exercise increases the rate

of glucose uptake through the translocation of GLUT4 transporters from intracellular storage

sites to the cell membrane to facilitate the transport of glucose into the muscle [271]. Increased

GLUT4 expression results in local muscles to be more sensitive to the action of insulin

improving insulin sensitivity [272, 273] and an additive effect in glucose uptake is evident if

both exercise and insulin are present when compared to each stimuli independently [86, 90].

Previous meta-analyses have suggested that the beneficial health effect of exercise was more

effective in reducing HbA1c in type 2 diabetics when compared to glucose-lowering drugs

[27, 28, 274]. Interestingly, consistent with the increased rate of glucose uptake accompanied

with increased exercise intensity, previous meta-analyses also support greater reduction in

weighted mean difference of HbA1c with exercise training at higher intensities [179, 275].

However, it must be noted that others have found the exercise-induced reduction in HbA1c

was associated with exercise volume, rather than intensity [179]. Overall this suggests that

although it is not clear whether exercise intensity or exercise volume is superior when

prescribing exercise, meta-analyses have identified similar or greater therapeutic benefit of

exercise when compared to glucose-lowering drugs.

Acute exercise stimulates the production of pro- and anti-inflammatory markers where it is

mediated by exercise duration, intensity and mode. Ostrowski et. al. demonstrated that acute

exercise induces the release of IL-6, which gradually increased over time during treadmill

running [276]. Further work following marathon running, highlighted that prolonged

strenuous exercise induced a significant pro-inflammatory response of up to 128-fold in IL-6,

Page 43: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

43

TNFα and IL-1β levels immediately following exercise, whereas anti-inflammatory cytokines

IL-10 and IL-1ra peaked to a lesser extent immediately post exercise and 1-hour post exercise,

respectively [204]. In addition to exercise duration, the inflammatory response is also driven

by exercise intensity. A number of studies found that the production of pro- and anti-

inflammatory cytokines including IL-6, IL-1β, IL-8, IL-10 and TNFα were increased together

with increased exercise intensity [47, 277-279]. However, previous work in healthy

individuals have found no differences in anti-inflammatory cytokines IL-4 and IL-10 [280].

Indeed, it may be plausible that the anti-inflammatory effect of exercise may be present at the

adipose tissue level, without any effect on plasma inflammatory cytokines [206].

Further to the variables of exercise intensity and duration, the mode of exercise may also

contribute to the degree of the inflammatory response. Nieman and colleagues have shown

that downhill running (eccentric contractions) induced significantly greater concentration of

inflammatory markers including IL-1β, IL-6, IL-10, TNFα and others when compared to

concentric cycling [281]. Their results also demonstrated that downhill running induced

greater level of muscle damage with elevated creatine kinase, myoglobin and delayed onset of

muscle soreness[281]. This confirms the findings of previous work, where eccentric cycling

induced greater creatine kinase and IL-6 when compared to concentric cycling [282].

However, although significant differences in cytokine response were found between modes of

exercise, evidence suggests that exercise intensity may have a stronger influence on cytokine

changes compared to muscle damage as the exercise-induced increase in stress hormones such

as cortisol and norepinephrine may have a greater influence on the systemic release of

cytokines rather than exercise-induced muscle damage [283]. Therefore, the inflammatory

response between mode of exercise should be interpreted with caution as multiple factors may

contribute to the magnitude of cytokine release.

3.3 Inflammation in T2DM – exercise-induced

improvements

In addition to the improvements seen with exercise training on glucose control as reviewed in

Chapter Two, a reduction in chronic inflammatory markers such as CRP [214, 284, 285] and

IL-6 [11, 42, 46] is also evident. Several studies showed that, following 3 to 12 months of

exercise training in T2DM patients, pro-inflammatory markers such as CRP, IL-1β, IL-18 and

Page 44: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

44

TNFα were reduced (up to ~40%), whilst anti-inflammatory cytokines IL-4 and IL-10

increased (up to ~84%). These changes in cytokine levels were associated with reduced insulin

resistance in T2DM patients [29, 44, 286]. In contrast, Dekker et.al found that while exercise

training in obese men with and without T2DM for 12 weeks did result in a reduction in IL-6,

there was no change in insulin sensitivity [287].

Whilst acute exercise results in an increased inflammatory response where most markers return

to resting concentrations within 24 hours, regular exercise promotes an inflammatory response

which reduces chronic inflammation in both chronic disease populations [288, 289] and in

healthy adults [290]. The exercise-induced reduction in chronic inflammation may be driven

by IL-6. Although IL-6 has been described as a pro-inflammatory marker primarily originating

from adipose tissue and has a strong association with obesity and insulin resistance, previous

work has also demonstrated its anti-inflammatory properties following exercise [291]. IL-6 is

one of the first cytokines released into circulation which increases up to 100-fold following

exercise, returning to baseline levels by 24h following exercise [204]. The anti-inflammatory

effects of IL-6 results from the induction of other anti-inflammatory cytokines including IL-

1ra and IL-10 [292, 293]. Further evidence of IL-6 on improved glucose homeostasis was

demonstrated by IL-6 infusion in T2DM patients, resulting in a decreased plasma insulin

response when compared to saline infusion [294]. IL-6 infusion has also been found to increase

the glucose infusion rate and glucose oxidation following assessment via a hyperinsulinemic

euglycaemic clamp [295]. These studies suggest that IL-6 has insulin sensitising properties

and enhances glucose clearance in both healthy and type 2 diabetic individuals. The fact that

IL-6 is released following exercise and is dependent upon exercise intensity and duration,

highlights the potential impact exercise has on mediating anti-inflammatory effects through

the IL-6-induced release of anti-inflammatory cytokine such as IL-10, which is also produced

from Tregs [296]. Thus, the pleiotropic role of IL-6 is dependent upon its source, where

obesity-induced IL-6 from adipose tissue has pro-inflammatory influence [225], whilst

muscle-derived IL-6 is of beneficial effect [297].

The impact of exercise on other inflammatory markers related to T2DM such as adiponectin,

MMPs, OPN and Tregs is not yet clear. Together with the obesity-induced reduction in anti-

inflammatory markers, current evidence suggests that adiponectin and Tregs are decreased

whilst MMPs and OPN are increased in association with obesity and T2DM. Limited evidence

is available on the role of exercise training on these markers and its association with glucose

control. This part of the review looks to summarize the recent findings of these relevant

markers (Table 3.1). A schematic of the relationship between exercise training, inflammation

and T2DM can be seen in Figure 1.

Page 45: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

45

Figure 3.1. A schematic of the relationship between exercise training, inflammation and type

2 diabetes.

Adiponectin

Whilst acute exercise has not been found to directly affect adiponectin [298], evidence on

exercise in regulating adiponectin levels was previously demonstrated in short- and long-term

training studies where as few as three days of exercise, and up to 12 weeks of training, resulted

in increased circulating adiponectin in overweight or obese individuals [299-302]. The

increase in adiponectin was associated with improved insulin sensitivity in obese individuals

further supporting the role adiponectin has on glucose homeostasis as per previous findings

[299, 301]. Racil and colleagues suggested that exercise intensity was associated with

induction of adiponectin production such that high-intensity intermittent exercise increased

adiponectin concentration to a greater degree than moderate intensity exercise when compared

to a control group. However, there were no differences of adiponectin between high-intensity

and moderate-intensity exercise [301].

Matrixmetalloproteinase

Matrix metalloproteinases were initially found to associate with cardiovascular disease and

atherosclerosis. However, growing evidence have revealed an association between increased

MMPs and T2DM [21-23]. Kadoglou et.al. previously investigated the effect of exercise

training for 16 weeks on plasma MMP2 and MMP9 in individuals with T2DM [235]. Although

no significant changes were found in MMP2 levels, exercise training significantly reduced

MMP9 concentrations, which was also associated with reductions in CRP and HbA1c [235].

These findings demonstrated a link between exercise-mediated improvements in MMP in type

Page 46: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

46

2 diabetics. Kadoglou et.al. also investigated the impact of exercise training for 12 weeks in

atherosclerotic mice with diabetes [303]. Exercise was beneficial as seen from reduced size of

atherosclerotic lesions in mice, which was also associated with reduced concentrations of

MMP2, MMP3, MMP9, IL-6 and reduced glucose AUC following an OGTT, further

strengthening the impact exercise training has on dampening the inflammatory milieu and

glucose tolerance seen with in diabetes and atherosclerosis.

Osteopontin

You et.al found that following 8 weeks of aerobic treadmill exercise in obese females, serum

OPN levels were significantly reducead in the morning following the last training session

[304]. Despite this positive finding, the effect of exercise remains inconclusive as a number of

previous studies did not find any changes in OPN levels [305-307]. A 12-week home-based

exercise program in colorectal cancer patients with normal BMI failed to report any significant

changes in OPN level [306]. A combination of aerobic and resistance exercise for 12 weeks in

overweight/obese middle-aged men with impaired glucose tolerance also failed to show any

improvements in OPN concentration [307]. Surprisingly, long term (12 months) training

completed in healthy postmenopausal women, consisting of regular (5 days per week)

moderate-intensity exercise, did not result in any changes in OPN either [305]. Despite the

lack of improvements here, it may be plausible that the reduction in OPN following 8 weeks

of treadmill exercise may be associated with weight loss [208], although weight loss through

bariatric surgery has been shown to increase serum OPN in morbidly obese individuals [308].

Altogether, these studies highlight mixed findings of OPN following exercise training and is

not consistently reduced following exercise training.

Regulatory T cells and IL-10

An understanding of the influence of Tregs on inflammation in T2DM following exercise is

still in its infancy as a limited number of studies have been conducted. It is evident in healthy

individuals that an acute swimming, increased Tregs number/proportions were found in the

blood of elite adolescent swimmers with and without rhinitis/asthma [309]. More relevantly,

12 weeks of Tai-Chi Chuan exercise (meditation type movement) for 60 minutes, increased

Tregs numbers in the blood of middle-aged volunteers, and this was associated with increases

in plasma levels of TGF-β and IL-10 [310]. This finding is of significance as the increase in

TGF-β and IL-10 has been found to drive further production of Tregs which leads to greater

suppressive function [267]. Further work found that 12 weeks of Tai Chi Chuan training also

increased the number of Tregs in type 2 diabetic patients when compared to a control group

[311]. The impact of exercise on Treg numbers in T2DM and its therapeutic potential was

further reinforced by a reduction in HbA1c post-training, although the significance and

Page 47: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

47

strength of this association was not assessed [311]. Despite the previous improvement in the

number of Tregs reported following Tai Chi Chuan, T2DM individuals who had completed 3

months of endurance exercise training (45 minutes cycling at lactate threshold) did not find

increased Treg numbers. Rather, exercise training resulted in a slight decrease which was close

to reaching statistical significance [312].

The production of Tregs may also be regulated by exercise intensity as demonstrated from

increased Treg cell proportion and production of IL-10 in mice following 6 weeks of high-

intensity exercise, but not moderate-intensity [26]. Recent work by Minuzzi et.al. has

suggested that the benefit of regular exercise is reflected in the anti-inflammatory status seen

following long-term training in masters athletes, who displayed greater levels of IL-10

compared to an age-matched controls, both before and after exercise [144]. Indeed, the anti-

inflammatory effect following six months of aerobic exercise training in T2DM patients was

effective as seen from increased IL-10 production, which was accompanied with a reduction

in CRP as well as improved glucose control and reduced insulin resistance [29]. Similar anti-

inflammatory changes were found following 12 months of exercise training where combined

aerobic and resistance exercise was demonstrated as the most effective training type resulting

in an increase in IL-10 together with reduced CRP, IL-1β and TNFα [44].

3.4 Conclusions

The evidence in this review have demonstrated that exercise has been shown to have a mixed

effect on the inflammatory response following acute exercise and exercise training. Although

acutely, an increased inflammatory response is seen following exercise, long term training

generally induces a beneficial adaptation to exercise leading to the reduction of a pro-

inflammatory markers together with an increase in anti-inflammatory markers associated with

T2DM. Indeed, further work is warranted to investigate the impact the mode, duration and

intensity of exercise has on modulating inflammation as well as its relationship with glycaemic

control. Although most of the exercise-induced interventions have found positive

Page 48: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

48

improvements, a number of studies have also showed no differences in outcome measures

related to inflammation and T2DM. This highlights the gap in the literature where the

mechanisms and the understanding in the relationship of chronic inflammation and T2DM is

still in its infancy.

In conclusion, chronic low-grade inflammation is a consequence of obesity-induced

infiltration of adipose tissue macrophages which increases the concentration of pro-

inflammatory markers associated with obesity and T2DM. Fortunately, regular exercise is not

only crucial in the management of blood glucose but emerging research has focused on the

role of exercise on reducing the risk of developing T2DM by assessing the relationship

between markers of chronic inflammation associated with T2DM. In addition to the reduction

in traditional inflammatory markers elevated in T2DM such as CRP and IL-6, evidence

suggests that regular exercise training is also beneficial in increasing regulatory T cells which

are crucial in the production of IL-10 as well as modulating other inflammatory components

of the immune system such as matrixmetalloproteinases and osteopontin which have been

previously implicated with cardiovascular disease, atherosclerosis and T2DM. This thesis

attempts to further investigate the role of exercise on glycaemic control and the subsequent

inflammatory response following acute exercise and a short-term training period.

Pro-inflammatory

Marker Roles in obesity/T2D Role in exercise Role in glucose control

IL-1β IL-1β = risk of T2DM [11, 13, 46].

IL-1β following acute exercise in highly trained and untrained post-exercise [313].

High glucose induces production of IL-1β in β-cells [230] Blocking of IL-1β reduces hyperglycaemia [209]

IL-6 IL-6 correlated with reduced IS [217] and insulin resistance [218].

Acute exercise IL-6 [47, 204, 276, 313].

IL-6 under high glucose conditions in vitro [213, 315]

Page 49: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

49

IL-6 increases risk of obesity/T2DM [13, 219].

between NGT and T2DM [20].

IL-6 following long term training in IGT [314] and T2DM [29].

IL-6 = insulin sensitivity in non-diabetic Pima Indians [316]

IL-6 in impaired glucose tolerance compared to normal glucose tolerance [214].

MMP2 MMP2 in T2DM patients vs healthy[21, 317].

MMP2 following 12 weeks exercise training [303].

MMP2 following 10 days of exercise [318].

High glucose exposure

in endotheliala cells MMP2 [238].

MMP3 MMP3 in T2DM patients [23, 319].

MMP3 following exercise training [303].

MMP3 inhibits glucose uptake and promotes TNFα [199].

OPN OPN = obesity in mice [241].

OPN in obese with [208] and without T2DM [20].

OPN after 8 weeks training in obese females [304].

OPN following 12 weeks training in cancer patients [306] and overweight/obese middle aged men [307].

OPN following 12mo training in healthy post-menopausal women [305].

Correlation between

OPN with fasting

blood glucose,

insulin, insulin resistance in obese [200].

TNFα TNFα associated with insulin resistance [14, 15, 222] and

IS [223]. TNFα higher in IGT compare to control subjects [320].

TNFα with acute exercise [313].

Exercise training TNFα and

insulin sensitivity [320].

TNFα in impaired glucose tolerance compared to normal glucose tolerance [214].

CRP CRP in individuals with impaired glucose tolerance and T2DM [19, 46, 197, 217].

Exercise training CRP [29, 57].

Positively correlated with insulin [10, 57]. Inversely associated with insulin responsiveness [217].

Anti-inflammatory markers

Marker Roles in obesity/T2DM Role in exercise Role in glucose control

Adiponectin Low concentration associated with obesity [19] and T2DM [246, 247]. Limits endogenous glucose production [251]. Negatively correlated with fasting plasma glucose [245]. Stimulates production of IL-10 [253, 254].

acute exercise [298].

with training [299-301].

Promotes insulin sensitivity in mice [252].

Page 50: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

50

Tregs

Tregs in obesity and T2DM [263-265].

Tregs = IL-10 production [262].

Tregs after acute exercise [309].

with training [310, 311].

IL-10 IL-10 = risk of T2DM [203, 302]. Impaired glucose tolerance =

IL-10 [17].

Treatment of IL-10 = insulin

signalling + glucose uptake +

insulin resistance [321].

IL-10 post-exercise, greater in high-intensity exercise compared to moderate-intensity [322].

IL-10 associated with high glucose & HbA1c [203].

Table 3.1. The relationship of pro-inflammatory and anti-inflammatory markers to T2DM,

exercise and glucose control

Page 51: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

51

Chapter 4 Effect of exercise on acute

postprandial glucose

concentrations and

interleukin-6 responses in

sedentary and overweight

males

Published in Applied Physiology, Nutrition and Metabolism. Doi: 10.1139/apnm-2018-0160.

Authors: Aaron Raman1, Jeremiah J. Peiffer1, Gerard F. Hoyne3, Nathan G. Lawler1,

Andrew J. Currie2 and Timothy J. Fairchild1

Affiliations:

1School of Psychology and Exercise Science, Murdoch University, Western Australia,

Australia;

2School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia;

3School of Health Sciences, University of Notre Dame Australia, Fremantle Campus,

Australia.

Co-authors

Aaron Raman: [email protected]

Jeremiah J. Peiffer: [email protected]

Gerard F. Hoyne: [email protected]

Nathan G. Lawler: [email protected]

Andrew J Currie: [email protected]

Corresponding author:

Timothy J Fairchild

School of Psychology and Exercise Science,

Murdoch University,

90 South Street,

Murdoch, 6150, Western Australia,

AUSTRALIA.

E-mail: [email protected]

Page 52: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

52

4.1 Abstract

This study examined the effect of two forms of exercise on glucose tolerance and concurrent

changes in markers associated with the interleukin-6 (IL-6) pathways. Fifteen sedentary,

overweight males (29.0±3.1 kg/m2) completed two separate, 3-day trials in randomised and

counterbalanced order. An oral glucose tolerance test (OGTT; 75g) was performed at the same

time on each successive day of the trial. Day two of each trial, consisted of a single 30-min

workload-matched bout of either high-intensity interval exercise (HIIE; alternating 100% and

50% of VO2peak) or continuous moderate intensity exercise (CME; 60% VO2peak)

completed 1h prior to the OGTT. Venous blood samples were collected pre-, immediately

post-, 1h post- and 25h post-exercise for measurement of insulin, C-peptide, IL-6 and the

soluble IL-6 receptors (sIL-6R; sgp130). Glucose area under the curve (AUC) were calculated

from capillary blood samples collected throughout the OGTT. Exercise resulted in a modest

(4.4%; p = 0.003) decrease in the glucose AUC when compared to the pre-exercise AUC;

however, no differences were observed between exercise conditions (p = 0.65). IL-6 was

elevated immediately and 1h post-exercise, whilst sgp130 and sIL-6R concentrations were

reduced immediately post-exercise. In summary, exercise was effective in reducing glucose

AUC which was attributed to improvements between 60 and 120 min of the OGTT, was in

parallel with an increased ratio of IL-6 to sIL-6R, which accords with an increased activation

via the ‘classical’ IL-6 signalling pathway. Our findings suggest acute HIIE did not improve

glycaemic response when compared to CME.

Trial registration: ACTRN12613001086752

Key words: Obesity, glucose regulation, interval training, area under the curve, high-intensity

Page 53: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

53

4.2 Introduction

Regular exercise is beneficial in the management of blood glucose and reducing the risk of

type 2 diabetes [27]. The exercising muscle stimulates glucose transporter-4 (GLUT4)

translocation and the associated increase in glucose uptake capacity during exercise is

dependent upon both the intensity [323-325] and duration of the activity [326, 327]. In

combination with increased blood glucose delivery and intramuscular metabolic flux [328],

the increased glucose transport capacity resulting from GLUT4 translocation can result in a 7-

fold (light exercise; ~63% max heart rate, HRmax), ~10-fold (moderate-heavy exercise;~80-

90% HRmax) and ~16-fold (~85% Wmax) increase in leg glucose uptake with increasing exercise

intensity [277, 325]. Increased (above baseline) glucose uptake can persist for up to 48 h after

exercise [329]. However, despite this localised improvement in glucose uptake, systemic

postprandial glucose (PPG) tolerance remains either unchanged [330-333], increases [334-

339] or decreases [340-342] in the hours following an acute bout of moderate- and high-

intensity exercise. The disparity between peripheral and systemic glucose concentrations may

be due to an increased rate of hepatic glucose release, which continues for approximately 30

min after cycling in overweight/obese individuals with normal glucose tolerance [343] as well

as a concomitant decrease in insulin sensitivity in tissues other than the contracting

musculature [328, 333, 343].

Interleukin-6 (IL-6) has been implicated in controlling glycaemia and the concentration of

circulating IL-6 in response to exercise has also been shown to increase in an intensity-

dependent manner [47, 204, 344]. Acutely, IL-6 increases glucose disposal [295, 345] in young

healthy males, although not in males with type 2 diabetes mellitus (T2DM) [346]. While

chronically elevated resting IL-6 concentration has been identified as a risk factor for

developing T2DM [46, 347] and is associated with poorer glycaemic control in individuals

with existing T2DM [348]. This equivocal relationship between IL-6 and glucose regulation

may arise as a consequence of the two distinct IL-6 signalling pathways. ‘Classical signalling’

occurs following binding of IL-6 to the membrane bound IL-6 receptor (IL-6R) and associated

glycoprotein 130 (gp130), forming an IL-6—IL-6R—gp130 complex [349]. ‘Trans-

signalling’ occurs when IL-6 binds to a soluble form of IL-6R (sIL-6R), and where this IL-

6—sIL-6R complex now directly binds gp130 at the plasma membrane to form the IL-6—sIL-

6R—gp130 complex [349, 350]. Since gp130 is ubiquitously expressed, cells which do not

express the membrane bound IL-6R may now become IL-6 responsive via this trans-signalling

pathway. It has been postulated that trans-signalling has detrimental effects on insulin

sensitivity by mediating the pro-inflammatory effects of IL-6, while the classical signalling

pathway promotes glucose uptake [350]. Trans-signalling may be inhibited via binding of

Page 54: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

54

soluble gp130 (sgp130) to the circulating IL-6—IL-6R complex; importantly, the sgp130 does

not bind either IL-6 or IL-6R alone and does not therefore affect the classical pathway [349].

Whether the inconsistent effect of acute exercise on PPG response [330-342] and more

specifically, the inconsistent findings regarding the role of exercise-intensity on PPG response

[186, 187, 351-353], is in part explained by the different responses in sIL-6R and sgp130 to

exercise, is less clear. Therefore, the purpose of the current study was to examine the effect of

exercise on IL-6, sIL-6R and sgp130 concentrations in sedentary and overweight males, and

assess glucose tolerance during a subsequent glucose challenge. We hypothesised that (i) acute

high-intensity intermittent exercise (HIIE) would improve PPG (glucose area under the curve)

to a greater degree than a work- and duration-matched continuous exercise bout, and this will

persist for up to 24 hours following exercise; (ii) Acute exercise will increase IL-6, and

decrease sIL-6R and sgp130 concentrations, wherein these effects will be amplified following

HIIE versus a work- and duration-matched continuous exercise bout.

Page 55: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

55

4.3 Methods

4.3.1 Study Design

The study was completed using a randomized cross-over design. All participants attended the

Exercise Physiology labs at Murdoch University for a total of seven visits (introductory

session; two experimental sessions comprising three visits each). The introductory session

(visit 1) involved the consent process, a familiarization session and assessment of fitness (peak

oxygen uptake, VO2peak; maximal aerobic power assessment). The two experimental phases

comprised three study days each (Day-1, Day-2, Day-3), and were completed using a

randomized cross-over design, with order randomization and allocation conducted by a

researcher not involved in the delivery of the intervention (TJF). Participants were informed

of the possible risks associated with the study before giving written informed consent. Ethics

approval was granted by Human Research Ethics Committee at Murdoch University (Project

number: 2013/121). This trial was prospectively registered with Australian New Zealand

Clinical Trials Registry: ACTRN12615000613505.

4.3.2 Participants

Fifteen sedentary (≤2 bouts of low-to-moderate intensity exercise per week) and overweight

(BMI >25 kg/m2) males were recruited from the local community via advertisements posted

on public noticeboards. Participants were deemed eligible if they were aged between 18 and

44 years old, had not been previously diagnosed with T2DM (fasting plasma glucose<7

mmol/L) and had not smoked within the previous 6 months. Participants were excluded if they

were taking anti-inflammatory medication, glucose controlling drugs (i.e. metformin), had

uncontrolled hypertension, and had either an acute or chronic inflammatory disease/infection.

Participants were pre-screened for exercise using the Exercise Science and Sport Science

Australia (ESSA) medical screening tool.

Page 56: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

56

4.3.3 Introductory Session

On arrival, participants were fully informed of all study procedures and written informed

consent obtained. Body height, body mass and waist circumference were determined. Body

composition was measured using the Dual Energy X-ray Absorptiometry (DXA; Hologic

Discovery QDR series, Hologic Inc., Bedford, MA, USA). Thereafter, participants completed

an incremental cycling test (50W; with 25W increments every 2 min) until volitional

exhaustion on an electronically braked Velotron cycle ergometer (RacerMate, Seattle, WA,

USA) for measures of VO2peak. During the incremental cycling test, expired ventilation was

collected as 15-second mean values using Parvomedics TrueOne metabolic cart (Parvomedics,

Sandy, UT, USA) and heart rate (Polar T31, Kempele, Finland) was collected at 1 Hz. VO2peak

was determined as the highest 30s average VO2 from the final stage of the incremental test.

Heart rate and Borg’s [354] rating of perceived exertion (RPE) was collected throughout the

maximal graded exercise test. This session was completed at least three days prior to the first

experimental phase.

4.3.4 Experimental Phases

On Day-1 of the experimental phase, participants reported to the laboratory in a fasted state at

~8am during which an oral glucose tolerance test (OGTT; 75g Carbotest, ThermoFisher,

Australia) was completed. Capillary blood samples were obtained via finger stick prior to the

OGTT, and at 15, 30, 60, 90 and 120 min following ingestion of the solution for measurement

of glucose (Hemocue Glucose 201 RT glucose; Hemocue AB, Ängelholm, Sweden). The

Hemocue glucose analyser uses a dual wavelength photometer that measures glucose

following a modified glucose dehydrogenase reaction and has been validated against the

Yellow Springs Instrument glucose oxidase analyser [355]. Participants were then provided

with a standardized breakfast (Up and Go, Sanitarium, Australia; two muesli bars, Kellogg’s,

Australia) before leaving the laboratory. Food intake for the remaining portion of the day was

at the participants’ discretion and was recorded in a food diary and repeated in the subsequent

trial.

Page 57: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

57

Twenty-four hours after the initial OGTT, and following an overnight fast, participants arrived

at the laboratory for their second visit in which they provided a venous blood sample (8ml)

from the antecubital vein collected into EDTA and SST tubes (Becton Dickinson, NJ, USA).

Participants then, completed a warm up at 75W before completing the assigned exercise

condition. Immediately upon the cessation of exercise, a second venous blood sample was

collected, followed by an hour of recovery where participants were advised to relax and remain

seated throughout the recovery period prior. A third venous blood sample was collected after

the recovery period prior to the completion of an OGTT using the same methodology as the

initial OGTT (Day-1) and at the same time of day. Participants were then provided with the

standardized breakfast before leaving the laboratory.

Day-3 comprised the same design as Day-1, with participants reporting to the laboratory in a

fasted state with the final venous blood sample collected prior to the final OGTT at the same

time as the previous days. A schematic of the timeline can be seen in Figure 4.1.

Figure 4.1. Diagram of study design.

4.3.5 Exercise Conditions (Day-2)

Each of the two experimental phases followed an identical three-day design, with the exception

being the exercise condition performed on Day-2. Day-2 comprised either a continuous

moderate intensity exercise bout (CME; 30 min at 60% of VO2peak), or a high intensity

intermittent exercise (HIIE) bout. Participants completed the desired workload based on power

output derived from the VO2peak test. The Velotron bike is electromagnetically braked and the

wattage was pre-set according to the protocol for each condition. The HIIE bout comprised six

efforts at high- (1 min at 100% VO2peak) and low- (4 min at 50% VO2peak) intensity which lasted

30 min in duration. Participants were advised to maintain a cadence between 80-110

revolutions per min (rpm) during the intense bouts and to maintain a comfortable cadence

during the recovery period. This exercise protocol was based on a previously published

protocol [356]. Both exercise conditions preceded with a standardized warm-up at 75W for

five min. Heart rate and RPE were recorded throughout each exercise trial.

Page 58: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

58

4.3.6 Blood Analysis

Serum samples (SST; 5mL) were allowed to clot for at least 30 min at room temperature,

before subsequently being centrifuged at 1,300g for 15-min. Plasma samples (EDTA; 6ml)

were spun immediately at 1,300g for 15-min. The supernatant was then immediately aliquoted

into triplicates and stored at -80°C. Samples were batch analysed for plasma glucose using a

commercial kit (GAHK20, Sigma-Aldrich, MO, USA); serum insulin and C-peptide (Bio-Plex

Pro Human Diabetes Assay, CA, USA; Inter-assay coefficient of variation, insulin: 5.0%, C-

peptide: 4.0%; Limit of detection, insulin: 1.0pg/ml, C-peptide: 14.5pg/ml); interleukin-6

receptor (sIL-6R; Inter-assay coefficient of variation: 3.2%; Limit of detection: 1.5pg/ml) and

glycoprotein 130 (sgp130; Bio-Plex Pro Human TH17 Assay, CA, USA; Inter-assay

coefficient of variation: 5.9%; Limit of detection: 16.9pg/ml) using the Bio-plex MAGPIX

multiplex reader (Bio-Rad, CA, USA). IL-6 was measured in serum using a high-sensitivity

enzyme-linked immunosorbent assay (ELISA; R&D Systems Human IL-6 Quantikine HS-

ELISA; MN, USA; Inter-assay coefficient of variation: 7.8%; Limit of detection: 1.77pg/ml).

Analyses of the standard curve for sgp130 and IL-6R used 5PL (Five Parameter Logistic)

equation which optimises the fit of the standard curve by default. The output provides an

assessment of the ‘goodness’ of the fit providing statistical parameters such as ‘fit probability’

(where 1 is a perfect fit and 0 has no fit) and ‘residual variance’ (where a small residual

variance indicates a good fit). The fit probability and residual variance for sgp130 and IL-6R

were: 0.829, 0.295 and 0.889, 0.210, respectively, which is similar to the manufacturer’s data.

These statistical parameters should also be interpreted with slight caution as it is a probability

outcome measuring the quality of fit, rather than R-squared which is not a sensitive metric for

assessing the quality of a curve fit.

Page 59: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

59

4.3.7 Data Analyses

Data are presented as mean ± SD, unless otherwise stated. Glucose concentrations from the

oral glucose tolerance test are presented using the total glucose area under the curve (AUC)

using the trapezoidal rule (Graphpad, PRISM), and as comparisons in raw glucose values using

95% confidence limits between corresponding days [357]. Separate bivariate regression

analysis was conducted to explore the associations between (i) glucose AUC on Day-2, with

the AUC (time-points: pre-exercise, immediately post-exercise, 1h post-exercise) of IL-6, sIL-

6R, sgp130; and (ii) glucose AUC on Day-3 with the concentrations of IL-6, sIL-6R, sgp130

on Day-3 (25h concentration). Data management and statistical analyses were performed using

IBM SPSS version 22.0 software (IBM Corp, Armonk, NY). Treatment effects were estimated

using separate, random-intercept linear mixed models (LMM) for each outcome variable

(blood analytes; heart rate; RPE; glucose AUC). Condition (CME, HIIE) and time (i.e., pre-

exercise, post-exercise, 1h post-exercise and 25h post-exercise; or, within exercise time-

points) were modelled as fixed effects. The primary hypothesis of interest was the time by

group interaction with main effects of condition and time examined with pairwise comparisons

of the estimated marginal means and reported accordingly. To assess magnitude of change,

Cohen’s d effect sizes were calculated and classified as: small effect (>0.2); moderate effect

(>0.5), and; large effect (>0.8) [358]. Significance was accepted at p 0.05.

Participant Characteristics

Page 60: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

60

(n = 15)

Age (y) 28.2 (6.9)

Height (m) 1.80 (0.05)

Weight (kg) 94.1 (10.3)

BMI (kg/m2) 29.0 (3.1)

FBG (mmol/L) 5.2 (0.5)

WC (cm) 96.6 (7.1)

Total fat mass (kg) 25.8 (6.6)

Fat (%) 27.5 (4.2)

VO2 (ml/kg/min) 35.7 (5.1)

Table 4.1 Descriptive characteristics of the participants. All data are presented as mean (SD).

FBG, Fasting blood glucose; WC, waist circumference; VO2, peak VO2 achieved.

Page 61: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

61

4.4 Results

Descriptive data of participants are displayed in Table 4.1. All participants completed the two

experimental phases. One participant completed only 5 sprints during the HIIE bout, whilst

one other participant completed all six sprints in the HIIE bout, but did not complete the last

four min (low-intensity portion of the protocol) of the bout. All data were included in the

analyses.

Mean RPE (mean±SD) was higher (p<0.01) during exercise for HIIE (16 ± 2) compared to

CME (14 ± 2; Figure 4.2). Accordingly, the interaction of trial by time revealed greater

perceived effort during HIIE than CME from at the time point corresponding from bout-3 to

bout-6 (Figure 4,2). Mean heart rate during exercise was higher during HIIE when compared

to CME; 147 ± 8 beats per min and 139 ± 11 beats per minute, respectively. There was no

main effect for HR between exercise conditions. Figure 4.2; p=0.080). Energy expenditure

(mean±SD) following CME (268 ± 31 Cal) was not different to HIIE (260 ± 10 Cal; p=0.167).

Figure 4.2. (Top). Mean (± SD) Ratings of perceived exertion after completing high-intensity

intermittent exercise (HIIE, filled square) and continuous moderate-intensity (CME, open

circle) using Borg’s scale. a, significantly different from CME (P < 0.01). (Bottom). Mean (±

SD) Heart rate response during high-intensity intermittent exercise (HIIE) compared to

continuous moderate-intensity (CME). No significant differences between trials p=0.112.

Page 62: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

62

4.4.1 Glucose Responses

The plasma glucose AUC are presented in Figure 4.3. A main effect of time (p<0.001) such

that plasma glucose AUC was significantly lower on Day-2 following exercise when compared

to Day-1 (mean difference [95% CI] in glucose: -0.36 [-0.65, -0.08] mmol/L; ES: 0.755) and

Day-3 (-0.56 [-0.79, -0.34] mmol/L; ES: 1.078) was observed. However, no other differences,

interaction (condition x day; p=0.302) or main effect of condition (i.e. between exercise

conditions; p=0.912) were observed.

In accordance with the AUC data, there were no differences between conditions on Day-2

(OGTT conducted 1 h post exercise) or Day-3 (25h post exercise) (Figure 4.3). When the

glycaemic response was explored between Day-2 and Day-1, exercise (HIIE and CME

combined; Figure 4.4) was shown to reduce the plasma glucose concentrations at 60 min, 90

min and 120 min of the OGTT. The AUC on Day-3 showed a significant increase at 15, 30,

60 and 90 min of the OGTT, while no differences between Day-3 and Day-1 were identified

(data not shown).

D a y O n e

(B a s e lin e )

D a y T w o D a y T h re e

0

4

5

6

7

8

9

AU

C-g

luc

os

e

(mm

ol.

min

)

C M E

H IIE

p = .0 0 2

p < .0 0 1

Figure 4.3. Box and whiskers (95% CI) plot for plasma glucose area under the curve (AUC)

on Day-1 (baseline), Day-2 (exercise) and Day-3. A main effect of time was present; p=0.000.

Page 63: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

63

Figure 4.4. Mean differences (with 95% CI) in plasma glucose concentration between A: HIIE

and CME on Day-2; B: HIIE and CME on Day-3; C: Day-2 and Day-1; D: Day-3 and Day-2.

Differences were calculated from blood glucose concentrations for each participant at each

time-point of the OGTT from one study day and subtracted from the corresponding time-point

on the comparison study-day. Significant differences are indicated where 95% confidence

limits to not cross the zero band.

4.4.2 Insulin, C-peptide and Inflammatory Markers

Data from all participants were available for analysis insulin and C-peptide, whilst data from

only ten participants were available for the analysis of the inflammatory marker panel. A main

effect time (C-peptide: p=0.001; insulin: p=0.007) was observed. However, no other

differences, interaction (condition x day; C-peptide: p=0.296; insulin: p=0.154) or main effect

of condition (C-peptide: p=0.283; insulin: p=0.443) were observed. When compared to pre-

exercise concentrations, both insulin and C-peptide concentrations demonstrated similar

trends with concentrations significantly elevated immediately post-exercise and lower 1 h and

25 h after exercise (Figure 4.5).

Page 64: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

64

There was a main effect of time on IL-6 (p=0.001) concentrations. When compared to

immediately post-exercise and 1 h post-exercise concentrations (Figure 4.6), IL-6 was lower

at pre-exercise (p=0.033, ES:-0.461; p=0.011, ES:-0.607, respectively) and 25 h post-exercise

(p=0.002, ES:-0.806; p<0.001, ES:-1.053, respectively). A main effect for condition was not

present, nor was a time by condition interaction. A main effect of time was present for both

sgp130 (p=0.006) and sIL-6R (p=0.010), with significantly lower concentrations in both

sgp130 and sIL-6R immediately post-exercise when compared to 1 h post-exercise (p=0.007,

ES:-0.558; p=0.019, ES:-1.012, respectively) and 25 h post-exercise (p=0.001, ES:-0.696;

p<0.001,ES:-0.905, respectively). No significant differences were observed in the IL-6/sIL-

6R complex concentration between the two trials (p=0.287), although a main effect of time

was present (p<0.001, Figure 4.6). When compared to immediately post-exercise and 1 h post-

exercise, IL-6/sIL-6R complex concentration was lower at pre-exercise (p=0.005, ES:-0.721;

p<0.001, ES:-0.805, respectively) and 25 h post-exercise (p<0.001, ES:-0.806; p<0.001, ES:-

1.242, respectively). There were no significant correlations obtained during either the HIIE or

CME conditions between the glucose-AUC and sGP130 (All p≥0.329) or sIL-6R (All

p≥0.222). However, the Day-2 (averaged across pre-exercise, immediately post-exercise and

1 hr post exercise) IL-6 concentration demonstrated a positive association with glucose AUC

(CME: r=0.627, p=0.053; HIIE: r=0.658, p=0.039), and this trend (non-significant) was also

observed between glucose-AUC and the ratio of IL-6/sIL-6R on Day-2 in the CME condition

(r=0.622, p=0.055) but not the HIIE condition (r=0.400, p=0.252).

Page 65: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

65

Figure 4.5. Box and whiskers (95% CI) plot for insulin (Top) and c-peptide (Bottom)

measured pre-exercise (baseline), immediately post-exercise (IPE), 1h post-exercise and 25h

post-exercise. a) compared to pre-exercise, p<0.05; b) compared to post-exercise, p<0.01; c)

compared to post-exercise, p<0.05. Open bars, CME; hatched bars, HIIE.

Page 66: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

66

Figure 4.6. Box and whiskers (95% CI) plot for; A: IL-6; B: sIL-6R; C: IL-6/sIL-6R complex,

and D: sgp130; measured in 10 participants pre-exercise (baseline), immediately post-exercise

(IPE), 1h post-exercise and 25h post-exercise. a) compared to IPE and 1h post-exercise,

p<0.05; b) compared to IPE and 1h post-exercise, p<0.01; c) compared to 1h post-exercise and

24h post-exercise, p≤0.01. Open bars, CME; hatched bars, HIIE.

Page 67: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

67

4.5 Discussion

This study sought to determine the effects of acute HIIE and CME on blood glucose responses

during an OGTT performed 1 h and 25 h post exercise in sedentary, overweight/obese men.

The main findings were: i) glucose AUC was significantly lower during the OGTT performed

1 h post exercise (Day-2) than compared to baseline (Day-1) or 25 hours after exercise

cessation (Day-3); ii) This reduction in glucose AUC appeared to result from a significant

reduction in glucose concentrations between 60-120 min of the OGTT when compared to

baseline (Day-1); iii) There were no differences in the glucose responses between HIIE or

CME conditions. To assess the role of IL-6 in moderating the effects of exercise on glucose

tolerance, IL-6, sIL-6R and sgp130 concentrations were also examined. The concentration of

IL-6 increased immediately post-exercise and 1 h post exercise, while the concentration of

both sIL-6R and sgp130 decreased immediately post-exercise. Consequently, the ratio of IL-

6 to sIL-6R was elevated immediately post-exercise and 1 h post-exercise which is expected

to result in a proportionate increased activation of the classical IL-6 signalling pathway; while

the concurrent changes in sIL-6R and sgp130 concentrations expected to result in the net

activation of the IL-6 trans-signalling pathway remained largely unaffected.

The finding that exercise reduced the subsequent glucose AUC during an OGTT in this study

is in accordance with our stated hypothesis and findings of some [334-339] but not all [330-

333, 340-342] previous studies. These differences are likely attributed to the timing of the

OGTT relative to the cessation of exercise. Indeed, higher glucose AUCs (i.e., worse) have

been observed when the OGTT is performed within 30 min of exercise cessation [340-343]

compared to; i) 60 min later [339]; ii) over the subsequent 24 h period (continuous glucose

monitoring; [334]); iii) or 24 h after cessation of exercise [340, 342]. The underlying

mechanism explaining this disparity may reside in the increased net hepatic glucose release

occurring during exercise and which continues for approximately 30 min after cycling in

overweight/obese individuals with normal glucose tolerance [343]. Since exercise in this study

was initiated 60 min post-exercise, it is unlikely that hepatic glucose release was still elevated.

The observed reduction in glucose AUC between 60 to 120 min in this study is consistent with

findings of Rynders et al. [339]. Of relevance to our findings, Knudsen et al. [343] observed

peak glucose disappearance ~90-100 min into the OGTT of individuals who were

overweight/obese but of normal glucose tolerance, both at rest and post-exercise. Although

glucose appearance/disappearance was not measured in the present study, the improved

glycaemic response is likely the result of the exercise-stimulated GLUT4 translocation to the

sarcolemmal membrane [323-325, 359].

Page 68: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

68

In the current study, the glucose AUC during the OGTT on Day-3 was similar to Day-1 and

significantly higher than the glucose AUC obtained 1 h post-exercise; indicating that the

beneficial effect of exercise on glucose tolerance had disappeared 24 h post exercise. This

finding is in contrast to the stated hypothesis and the findings of Bonen, Ball-Burnett [337]

and King, Baldus [340] whom observed significantly improved glucose AUCs 24 h post

exercise. However, differences in findings may partly be explained by the age of participants

and differences in the exclusion criteria between studies. The current study excluded

individuals who were physically active or had a BMI 25 kg.m-2 and had a mean participant-

age of 28 years old. King, Baldus [340] recruited moderately trained and lean individuals,

whilst Bonen et al. (1988) recruited middle-aged (40 to 55 years old) inactive individuals but

without exclusion based on BMI (men: 27.3 ± 0.7; women: 23.6 ± 1.5).

The responses in insulin and C-peptide following the exercise protocols are in agreement with

previous work [360-362]. Specifically, low-moderate intensity exercise results in a well

characterized decrease in insulin concentration, however supra-maximal (or very high-

intensity) exercise has been associated with a transient (min-hours) increase in insulin and C-

peptide which may be explained by a 14- to 18-fold increase in catecholamines [339, 361].

Indeed, we hypothesized that the increase in inflammatory markers in individuals

unaccustomed to exercise would partly negate the benefits of exercise on the glucose AUC.

However, this appeared not to be the case given i) the 1 h post-exercise AUC was improved;

ii) there were no significant associations between any of the measured inflammatory markers

and glucose AUCs on Day-2 or Day-3. It is important to note however, that the exercise-

induced decrease in the glucose AUC (Day-2) in the current study, while significant, was

smaller than anticipated. It is possible, that a concomitant decrease in insulin sensitivity in

tissues other than the contracting musculature [328, 333, 343] contributed to the relatively

small improvement in the systemic glucose AUC post-exercise.

Although resting IL-6 is associated with increased adiposity [215] and an increase in risk of

T2DM [46], the transient release of IL-6 from muscle during acute exercise can promote

muscle glucose uptake and insulin action [277, 295, 363]. The increase in circulating IL-6

concentration following exercise is dependent on the mode, duration and intensity of exercise

[47, 276, 364-366]. The anti-inflammatory effect of IL-6 is believed to be associated with the

improved glucose control [346, 349, 350], and is associated with the release of IL-10 [291]. In

accordance with the findings from this study, acute HIIE has been shown to increase IL-6

concentration [364, 367]. Although IL-10 was not measured in the current study, previous

work reported an increase in both IL-6 and IL-10 post HIIE [368], with the increase in IL-6

occurring 45 min prior to the increase in IL-10. It is important to note that the current study

Page 69: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

69

did not see a difference between exercise conditions, which is in contrast to the findings of

Leggate, Nowell [364], who observed significantly greater IL-6 concentrations following HIIE

compared to work-matched CME in lean and physically active participants. While the

participants in the study of Leggate et al. (2010) were different, the exercise protocol of

Leggate et al. (2010) was 60 min in duration (HIIE: 10 x 4 min, recovery 2 min) while the

current study was only 30 min in duration. Considering IL-6 has previously been postulated

to act as a signalling molecule indicating muscle glycogen levels (greater glycogen depletion

leading to greater IL-6 release) [365], it is possible the 60 min exercise duration adopted by

Leggate et al. (2010) resulted in a greater total depletion of muscle glycogen which was

sufficient to see a difference between the two exercise protocols.

Interestingly, both sgp130 and sIL-6R were lowest immediately post-exercise. When IL-6 is

expressed relative to sIL-6R (IL-6/sIL-6R; Figure 4.5), the post-exercise increase is enhanced,

which is in accordance with previous findings [364]. This finding is important since a

concomitant increase in IL-6 and sIL-6R is expected to increase activation of the ‘trans-

signalling’ pathway, while a reduction in sIL-6R is expected to reduce signalling via this

pathway due to reduced extracellular binding capacity of IL-6—IL-6R. The trans-signalling

pathway has been implicated in decreasing glucose tolerance in the longer-term and is

associated with infiltration of adipose tissue macrophages [350]; correspondingly, sIL-6R is

elevated in T2DM [348]. The purported deleterious effects of trans-signalling on glucose

regulation, may be ameliorated via the binding of sgp130 to the circulating IL-6—IL-6R

complex [349], since this reduces the availability of the complex to bind membrane-associated

gp130. It is interesting to note the corresponding time-course of sgp130 and IL-6R in the

current study, which indicates the trans-signalling pathway is unlikely to have been altered to

any great degree. In contrast, the increase in IL-6 with the concurrent decrease in sIL-6R is

consistent with an increased activation of the classical IL-6 signalling pathway which is linked

with stimulating increased skeletal muscle glucose uptake [369]. Multiple independent

correlations were conducted to explore associations between IL-6 and IL-6 related proteins,

and whilst caution is required in the interpretation of multiple repeated correlations on a small

sample, it is of note that positive relationships between glucose-AUC and both IL-6 and ratio

of IL-6/IL-6R were observed, which would be in accordance with an increased activation of

the classical signalling pathway as proposed. Further work in this field would benefit from the

Page 70: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

70

adoption of co-immunoprecipitation methods to explore these interactions in greater detail.

The study herein had a number of strengths including i) matching the workload and duration

of the HIIE and the CME conditions; ii) recruiting a clinically relevant population, namely

individuals who are inactive and overweight- which has, to our knowledge not previously been

conducted in a study of this kind; iii) adopting a randomized cross-over design; iv) each trial

included a specific baseline AUC which was conducted 24 hour prior to the testing sessions

and included a 25 h post AUC (with all AUCs being conducted at the same time of day).

However, there were also some limitations associated with this study. Specifically, the number

of blood samples collected by venepuncture were limited, and therefore restricted the

information on IL-6 and IL-6 receptors, as well as insulin and glucagon during the OGTT

which would have enhanced our interpretation of findings. Additionally, despite IL-6 and its

receptors being measured in serum, the study did not measure direct activation of each

signalling pathway, and can therefore only postulate on the changes based on the serum

markers. While an obese and sedentary population was recruited, inclusion of a broader group

of individuals spanning healthy to T2DM would have allowed assessment across clinical and

pre-clinical stages. Finally, although glucose tolerance was assessed, it must be stressed that

our findings cannot confirm increased glucose disposal/clearance as an isotope tracer

technique was not employed to measure hepatic glucose production in the present study.

In conclusion, 30-min of acute exercise reduced the glucose AUC during the OGTT performed

one-hour post exercise, although the improvements in glucose AUC were ameliorated 25 hours

later. The changes in IL-6, sIL-6R and sgp130 are consistent with increased activation of the

‘classical’ signalling pathway immediately post-exercise and 1 h post-exercise, which is

postulated to have contributed to the improvement in post-exercise glucose AUC. Further

work is warranted to directly assess the activation of the IL-6 pathways in response to both

acute exercise and exercise training in pre-clinical and clinical populations. Additional

methods such as isotope-labelled tracer technique should also be employed in order to

accurately determine the direct impact of these pathways on glucose disposal across tissues.

The understanding of these pathways has clinical implications, particularly with respect to

exercise prescription, wherein decisions on frequency, mode, intensity and duration of exercise

are made. Finally, the acute HIIE protocol did not confer additional benefits on glucose

concentration, or alter either the endocrine or IL-6 and IL-6 related soluble receptors when

compared to a work- and duration-matched CME protocol.

Conflicts of interest: The authors have no conflicts of interests to declare.

Page 71: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

71

Chapter 5 Glucose and inflammatory

markers following acute

exercise in sedentary and

overweight males

Authors: Aaron Raman1, Jeremiah J. Peiffer1, Gerard F. Hoyne2, Nathan G. Lawler1 and

Timothy J. Fairchild1

Affiliations:

1School of Psychology and Exercise Science, Murdoch University, Western Australia,

Australia;

2School of Health Sciences, University of Notre Dame Australia, Fremantle Campus,

Australia.

Corresponding Author:

Aaron Raman

School of Psychology and Exercise Science,

Murdoch University,

90 South Street,

Murdoch, 6150, Western Australia,

AUSTRALIA.

E-mail: [email protected]

The manuscript in this chapter is currently under review, submitted to the Journal of Applied

Physiology.

Page 72: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

72

5.1 Abstract

Inflammatory markers associated with adipose-tissue expansion have been implicated in the

early progression of metabolic disease. Consequently, Matrix metalloproteinase-2 (MMP-2)

and -3 (MMP-3), and osteopontin have garnered recent attention. The purpose of the current

study was to assess the circulating concentration of these markers, along with adiponectin and

glucose concentrations in response to two different modes of exercise. Fourteen sedentary,

overweight males (29.0 ± 3.1 kg/m2) completed two separate, 3-day trials in randomised and

counterbalanced order. Oral glucose tolerance tests (OGTT) were performed on each day of

the trial. Day two of each trial, consisted of a single 30-min workload-matched bout of either

high-intensity interval exercise (HIIE; alternating 100% and 50% of VO2peak) or continuous

moderate intensity exercise (CME; 60% VO2peak) completed 1h prior to the OGTT.

Continuous glucose and physical activity monitoring occurred throughout each trial, and

venous blood samples were collected pre-, 0h post-, 1h post- and 25h post-exercise for

measurement of MMP-2, MMP-3, osteopontin and adiponectin. No significant trial-by-time

interaction effects were noted for MMP-2, MMP-3, osteopontin or adiponectin. There was a

main effect of time (both p<0.01) with exercise transiently increasing MMP-3 and decreasing

osteopontin, but both returned to baseline values by 1h post-exercise. Exercise did not improve

glucose values over the subsequent 48h. Mean day glucose and area under the curve from the

OGTT was higher on Day 2 and 3 compared to day 1 (main effect of time; both p<0.05). Acute

exercise improved risk markers of metabolic disease without improvements in glucose control.

Trial Registration: ACTRN12613001086752

Key Words: Obesity, cardiovascular disease, glucose regulation, interval training, high-

intensity

Page 73: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

73

5.2 New & Noteworthy

This study demonstrated that regardless of exercise intensity, exercise was effective in

inducing changes to MMP-3 and OPN immediately post-exercise. We also assessed

continuous glucose concentrations for 72h to assess glycemic control under free-living

conditions. Interestingly, no improvements in glucose tolerance were evident over the three-

day period. Whereas previous study has shown an improvement in glucose response within 24

h following exercise.

Page 74: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

74

5.3 Introduction

The adipose tissue expansion associated with obesity results in the infiltration of macrophages

expressing pro-inflammatory cytokines [241, 370, 371]. The resultant nett increased release

of pro-inflammatory cytokines from adipose tissue can impair insulin signalling [9] and lead

to microvascular and macrovascular complications [220, 372-374]. These complications

underpin the increased rates of morbidity and mortality from cardiovascular disease in

individuals with diabetes mellitus [375, 376].

Attention has now focussed on identifying the initiating markers implicated in the shift to the

predominantly pro-inflammatory state associated with obesity and obesity-related disease

states [25, 377]. In this vein, matrix metalloproteinases (MMPs) appear to play potentially

important roles early in obesity-related disease progression. In particular, adipose tissue

releases MMP-2 during adipocyte differentiation, which is understood to regulate the vascular

extracellular matrix (ECM) remodelling during obesity-induced adipose tissue formation [48,

49]. In contrast, the release of MMP-3 appears to have an inhibitory effect on adipocyte

differentiation and therefore hyperplastic adipose expansion [50]. In an in vitro cell culture

model utilising human umbilical vein endothelial cells (HUVEC), hyperglycaemia (25 mM)

significantly increased concentrations of MMP-2 and reduced concentrations of MMP-3 [238].

This altered pattern in MMPs is further observed in clinical populations, with increased MMP-

2 concentrations in obese patients [22]; increased MMP-2 concentrations [21] and decreased

MMP-3 concentrations in patients with type 2 diabetes mellitus (T2DM; [23]).

Osteopontin (OPN) has also emerged as a potentially important regulator of inflammation,

obesity and diabetes [24]. Whilst OPN appears to play multiple immune-modulating roles, the

roles of OPN including the regulation of macrophage movement and accumulation [51] and

stimulating expression of MMP-2 [52] are of particular interest herein. Accumulation of

macrophages, particularly classically activated macrophages in adipose tissue acts as a source

for pro-inflammatory cytokines. These cytokines are associated with chronic inflammatory

diseases such as T2DM amongst others [241]. Clinically, increased concentration of OPN was

previously observed in individuals with T2DM [20]. Whilst the generation of pro-

inflammatory cytokines are associated with T2DM, anti-inflammatory cytokine, adiponectin

(APN) is inversely associated with insulin resistance [17] and its immunosuppressive effect is

mediated through the release of IL-10 [254]. Interestingly, adiponectin has been found to

increase MMP-3 expression [378], suggesting a role in the interplay of inflammatory cytokines

between OPN and MMPs.

Page 75: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

75

The longer-term benefits of exercise in controlling weight, specifically decreasing visceral

adipose tissue [379] and controlling blood glucose [64, 65] are well established. However, the

response of APN, OPN and MMPs to exercise are largely unexplored in humans and existing

research equivocal [298, 300, 380]. Moreover, despite the benefits of exercise, the role of

exercise intensity on postprandial glucose is unclear for individuals at risk of developing

T2DM. The purpose of this study therefore, was to determine the acute effects of exercise on

the inflammatory markers MMP-2, MMP-3 and OPN along with APN, and concomitantly

assess changes in the 48 h glucose response to two different types of exercise in overweight

and sedentary individuals.

Page 76: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

76

5.4 Methods

5.4.1 Study Design

The study followed a randomized cross-over design. All participants attended the Exercise

Physiology labs at Murdoch University for a total of seven visits (introductory session; two

experimental phases comprising three visits each). The introductory session (visit 1) involved

the consent process, a familiarization session and assessment of fitness (peak oxygen

consumption, VO2peak; maximal aerobic power assessment). The two experimental phases

comprised three study days each, with each phase completed in randomized order. The

randomization sequence was generated using a computer generated random number list

comprising 1’s and 2’s. The allocation of participants was conducted by a researcher not

involved in the delivery of the intervention (TJF), by placing pre-numbered cards into opaque,

sealed envelopes and recording the study ID’s. Participants were informed of the possible risks

associated with the study before giving written informed consent. Ethics approval was granted

by Human Research Ethics Committee at Murdoch University (Project number: 2013/121).

5.4.2 Participants

Fourteen sedentary (≤ 2 bouts of low-to-moderate intensity exercise per week) and overweight

(BMI > 25 kg/m2) males were recruited from the local community via advertisements posted

on public noticeboards. Participants were deemed eligible if they were aged between 18 and

44 years old, had not been previously diagnosed with T2DM (fasting plasma glucose < 7

mmol/L) and had not smoked within the previous six months. Participants were excluded if

they were taking anti-inflammatory medication, glucose controlling drugs (i.e. metformin),

had uncontrolled hypertension, and had either an acute or chronic inflammatory

disease/infection. Participants were pre-screened for exercise using the Exercise Science and

Sport Science Australia (ESSA) medical screening tool.

Page 77: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

77

5.4.3 Introductory Session

On arrival, participants were fully informed of all study procedures and written informed

consent obtained. Body height, body mass and waist circumference measures were taken, and

participants then shown the finger-stick procedure for collecting capillary blood for

determination of blood glucose using a glucometer (Accu-Chek Go, Roche, Mannheim,

Germany) required for the calibration of the continuous glucose monitor. Body composition

was measured using the Dual Energy X-ray Absorptiometry (DXA; Hologic Discovery QDR

series, Hologic Inc., Bedford, MA, USA). Thereafter, participants completed an incremental

cycling test (50W; with 25W increments every 2 minutes) until volitional exhaustion on an

electronically braked Velotron cycle ergometer (RacerMate, Seattle, WA, USA) for measures

of VO2peak. During the incremental cycling test, expired ventilation was collected as 15 s mean

values using the Parvomedics TrueOne metabolic cart (Parvomedics, Sandy, UT, USA) and

heart rate (Polar T31, Kempele, Finland) was collected at 1 Hz. Peak VO2 was determined

from the highest 30 s average VO2 during the final stages of the incremental test. Heart rate

and Borg’s [354] rating of perceived exertion (RPE) was collected throughout the maximal

graded exercise test. This session was completed at least three days prior to the first

experimental phase.

5.4.4 Experimental Trials

On day one of the experimental phase, participants reported to the laboratory (~0700 h) in a

fasted state for the insertion of the continuous glucose monitoring system (CGMS; iPro2,

Medtronic, Northridge, CA, USA). The sensor was inserted into an area of subcutaneous fat

in the abdomen as recommended by the manufacturer. Following insertion of the sensor,

participants were provided with a glucose meter (Accuchek Go, USA) in order to self-obtain

finger-prick blood samples required to calibrate the glucose monitor over the three-day trial.

In addition, participants were provided with an Activity Monitor (ActiGraph LLC, FL, USA)

to assess physical activity patterns over the three-day period. An hour following insertion of

the CGMS, an oral glucose tolerance test (OGTT; Carbotest, ThermoFisher, Australia) was

completed (Day 1; baseline). Participants were then provided with a standardized breakfast

(Up and Go, Sanitarium, Australia; two muesli bars, Kellogg’s, Australia) before leaving the

laboratory. Food intake for the remaining portion of the day was recorded in a food diary and

repeated during the second phase.

Page 78: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

78

On day two, participants arrived at the laboratory at 0600 h fasted (overnight fast; water

permitted). Venous blood sample was collected into EDTA and SST tubes (Becton Dickinson,

NJ, USA) from the antecubital vein. Participants then, completed a warm up at 75W before

completing the assigned exercise condition. Immediately upon the cessation of exercise, a

second venous blood sample was collected, followed by an hour of recovery where participants

were advised to relax and remain seated throughout the recovery period. A third venous blood

sample was collected after the recovery period prior to the completion of an OGTT using the

same methodology as the initial OGTT (day one) and at the same time of day (~0800 h).

Thereafter, participants were provided with the standardized breakfast before leaving the

laboratory.

On day three, participants reported to the laboratory in a fasted state prior to having the final

venous blood sample collected and the final OGTT, which was performed at the same time as

the previous days.

5.4.5 Exercise Conditions

Each of the two experimental phases followed an identical three-day design, with the exception

being the type of exercise performed on day two. Day two comprised either a continuous

moderate intensity exercise bout (CME; 30 minutes at 60% of VO2peak), or a high intensity

intermittent exercise (HIIE) bouts. The HIIE bout comprised six efforts at high- (1 minute at

100% VO2peak) and low- (4 minutes at 50% VO2peak) intensity which lasted 30 minute in

duration. Participants were advised to maintain a cadence between 80-110 revolutions per

minute (rpm) during the intense bouts and to maintain a comfortable cadence during the

recovery period. This exercise protocol was based on a previously published protocol from

our group [356]. Both exercise conditions proceeded with a standardized warm-up at 75W for

five minutes. Heart rate and RPE were recorded throughout each exercise trial.

5.4.6 Blood Sampling

Serum samples (SST; 5mL) were allowed to clot for at least 30 minutes at room temperature,

before being centrifuged at 1,300g for 15 minutes. Plasma samples (EDTA; 6ml) were spun

immediately after collection at 1,300g for 15 minutes. The supernatant was then aliquoted in

triplicate and stored at -80°C for later analysis. Samples were batch analysed using a multiplex

cytokine assay for serum adiponectin (APN; Bio-Plex Pro Human Diabetes Assay); matrix

metalloproteinase-2 (MMP2), MMP-3 and osteopontin (OPN; Bio-Plex Pro Human TH17

Assay) using the MAGPIX multiplex reader (Bio-Rad, Richmond, CA, USA).

Page 79: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

79

5.4.7 Physical Activity Monitoring

A physical activity monitor (ActiGraph GT3X Activity Monitor; ActiGraph LLC, FL, USA)

was provided to participants in order to record daily physical activity. Physical activity data

were then assessed using the ActiLife software (version 5.6.1; ActiGraph LLC, FL, USA).

Energy expenditure (kCal) over the three-day period was estimated by the algorithm

incorporated within the software, specifically set for adults.

5.4.8 Data Analyses

Data are presented as mean ± SD, unless otherwise stated. Sample size was calculated with an

effect size of F = 0.3112 with 80% power at a significance level of 5% based on IL-6 from

Chapter 4. If sphericity was violated during the analysis, the Huynh-Feldt correction was used.

Raw data from the CGMS and Actigraph were inspected for missing values and outliers prior

to time synchronization and analysis. The raw values from the CGMS was evaluated as

described below. In addition, the integrated area under the glucose curve (AUC-glucose)

during the OGTT was calculated using the Trapezoidal rule (Graphpad PRISM) for each day

and each condition, wherein the pre-OGTT value (time 0) was used as the baseline.

Differences in blood analytes (MMP-2, MMP-3, OPN, APN), AUC-glucose, heart rate, RPE,

mean-daily physical activity levels were analysed using a linear mixed model (LMM) with

repeated measures (time), with one between factor (trial: CME vs HIIE) and one within factor

(time: pre-exercise, post-exercise,1h post-exercise and 25h post-exercise) modelled as fixed

effects (intercept modelled as random effects). The primary outcome of interest was the time

by condition interaction, while significant main effects of time were explored.

To avoid the problem of multiple point-wise comparisons over time, a bootstrapping analysis

was used to assess the glucose values obtained from the CGMS. To this end, the blood glucose

concentration at each time point on the comparator day (i.e., day 1 of each phase), was

subtracted from the blood glucose concentration at each time point on the study day (i.e., Day

2 or Day 3 of each phase). This provided a difference or change score for each participant

across the two days being compared, and enabled the calculation of a group-mean and 95%

confidence interval (CI) bands over the course of the day. The change in glucose concentration

was plotted against time (as mean ± 95% CI), wherein a significant response was defined as

occurring when the lower 95% confidence limit for the curve was greater than zero, and a

significant reduction in blood glucose was defined as occurring when the upper 95%

confidence limit was less than zero [357]. All statistical analyses were completed using

commercially available software (SPSS 24 Windows; SPSS, Chicago, IL, USA). Significance

was accepted at p 0.05.

Page 80: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

80

Page 81: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

81

5.5 Results

The participants in the study were: 27.4 ± 6.3 years old; classified as overweight or obese

(BMI: 29.0 ± 3.2 kg/m2) with excessive adiposity (total body fat %: 27.2 ± 4.2%; visceral body

fat: 613.2 ± 122.7 cm3); normoglycemic (fasting glucose: 5.2 ± 0.5 mmol/L); and had a VO2

peak of 35.7 ± 5.1 ml/kg/min. All participants completed the two experimental phases. One

participant completed only five sprints during the HIIE bout, whilst one other participant

completed all six sprints in the HIIE bout, but did not complete the last 4 minutes (low-

intensity portion of the protocol) of the bout. CME elicited a mean-sessional heart rate and

RPE (mean ± SD) of 137 ± 25 beats per minute (bpm) and 14 ± 2, respectively; whilst HIIE

elicited a significantly higher mean-sessional heart rate and RPE (151 ± 33 bpm and 16 ± 2,

respectively).

All subjects wore the activity monitor over the three-day period. Energy expenditure (kCal),

total time of sedentary (Sed Time) and the vector magnitude (VM) was no different between

days (p > 0.178 for all variables) or condition (p>0.196 for all variables). Data displayed in

Table 5.1.

CME HIIE

kCal Sed.

Time(min)

VM(cpm) kCal Sed.

Time(mins)

VM(cpm)

D1 401(181) 881(83) 7566(1688) 504(370) 813(146) 9162(5602)

D2 498(312) 881(192) 8697(3402) 493(485) 875(179) 9037(6683)

D3 546(450) 840(174) 9393(6247) 629(533) 821(169) 10137(6646)

Data as mean ± SD. There were no significant differences. Energy expenditure expressed as

kilocalories (kCal); Time spent in sedentary expressed as Sed. Time in minutes; total vector

magnitude expressed as VM. Counts per minute, expressed as cpm; time expressed as minutes

(min).

Table 5.1. Physical activity monitor measurement

Page 82: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

82

5.5.1 Inflammatory Cytokines

Data from ten participants were available for analysis of the inflammatory cytokine panel

(Figure 5.1). There were no time by condition interactions observed for the markers of interest

(MMP-2: p = 0.876; MMP-3: p = 0.886; OPN: p = 0.663; APN: p = 0.778). A main effect of

time for MMP-3 (p = 0.003) and OPN (p = 0.006) were observed (Figure 5.1). Exercise

resulted in an acute increase in MMP-3, which was ablated by 1h post-exercise. In contrast,

OPN decreased significantly in response to exercise, returning to pre-exercise levels by 25h

post-exercise (Figure 5.1).

Figure 5.1. Mean (± SD) concentration of matrix metalloproteinase-2 (MMP-2), matrix

metalloproteinase-3 (MMP-3), osteopontin and adiponectin. MMP-3; a, Significantly different

than Pre (p=0.001) and 25 h (p=0.001). Osteopontin; a, Significantly different than Pre (p =

0.006) and 25 h (p = 0.001). No differences were found with adiponectin and MMP-2. CME

(open bars) vs HIIE (hatched bars).

Page 83: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

83

5.5.2 Glucose: Continuous Glucose Monitoring

Following the evaluation of data from the CGMS, data from 3 participants were excluded due

to significant (>4 hours) lapses in the measurements. There was a main effect of time (p =

0.037) on mean-daily glucose values, with the average glucose concentration on Day 3 being

higher than Day 1 (Glucose: 5.6 ± 0.2 vs Glucose: 5.2 ± 0.1mmol/L; p = 0.050), but there was

no significant interaction effect (p = 0.755; Figure 5.2). There were only minor periods

throughout the day, where significant differences between study days were observed as

indicated by the upper- or lower-95% CI bands crossing zero (Figure 5.3).

The glucose profile from the CGMS during the OGTT revealed a main effect of time

(p <0.001), but no condition by time interaction effects (p = 0.840; Figure 5.2). The AUC-

glucose revealed a significant effect of time (day; p=0.027) but no interaction effect (p =

0.078). The AUC for glucose on Day 1 was significantly lower than day 2 (mean difference:

0.28 mmol/L/min; value: p = 0.043) and day 3 (mean difference: 0.46 mmol/L/min; p = 0.008).

Page 84: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

84

Figure 5.2. (Top) Mean (±SD) glucose concentration over each day calculated from CGMS.

a, significant difference between Day 1 and Day 3, p = 0.05. (Bottom) Glucose concentration

during OGTT on each day. A main effect of time over the OGTT was present, p < 0.001 and

a main effect between days was present, p = 0.025. Mean glucose was lower on Day 1 vs Day

3, p = 0.043. CME (open bars) vs HIIE (hatched bars).

Page 85: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

85

Figure 5.3. Mean differences (with 95% CI) in 24h glucose concentrations between A: HIIE

on Day-2 minus Day-1; B: CME on Day-2 minus Day-1; C: HIIE on Day-3 minus Day-1; D:

CME on Day-3 minus Day-1. Subplot next to each graph represents mean glucose difference

concentration and standard deviation over each respective 24 h period. Arrow indicates when

exercise was completed.

Page 86: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

86

5.6 Discussion

Emerging roles for MMP-2, MMP-3 and OPN in the development of adipose tissue and more

broadly T2DM are now being elucidated [21, 23, 24, 48-50]. The response of OPN and MMPs

to exercise are largely unexplored in humans and existing research equivocal [380]. We sought

to determine the blood glucose response and changes in circulating MMP-2, MMP-3 and OPN,

to two different exercise types in overweight and sedentary individuals. The main findings

were i) exercise did not improve diurnal glucose profiles as demonstrated by similar glucose

profiles across the day; and indeed appeared to inflate the glucose response; (ii) the glucose

profile during the OGTT supported the diurnal pattern, with the AUC performed 1 h after

exercise (Day 2) and 25 h after exercise (Day 3) being significantly higher than Day 1; iii)

MMP-3 was acutely increased in response to exercise while MMP-2 did not demonstrate

significant changes in response to exercise; iv) OPN was acutely decreased following exercise,

and returned to baseline levels by 25h.

Osteopontin appears to play an important initiating role in adipose-tissue expansion, inducing

the expression of MMP-2 via pro-MMP-2, and inflammation [20, 52]; while MMP-3 appears

to have an inhibitory effect on adipocyte differentiation [50]. The decrease in OPN

concentration immediately post-exercise however, did not correspond with a significant

decrease in MMP-2 concentration. Although previous findings of reduced OPN in mice was

beneficial in reducing adipose tissue inflammation and improving glucose tolerance [241,

381], the reduction in OPN in the current study was only transient and appeared not to have

an effect on glucose tolerance. Indeed, while the respective response of these markers to

exercise are consistent with potentially longer-term health benefits; in the current study, these

changes were not associated with changes in diurnal glucose profiles as evidenced by a lack

of significance over time and the higher glucose responses 1 h and 25 h post-exercise (Figure

5.2 and 5.3).

To our knowledge, this is the first study that has investigated the effect of exercise intensity

on both MMP-2 and MMP-3 in overweight/obese sedentary individuals. The increase in

MMP-3 immediately post-exercise was similar to Urso et al. [382] who also found a transient

increase in MMP-3 immediately following acute exercise. While decreased MMP-3 is

observed over the longer term with exercise training in diabetic mice [303], evidence in

humans remain limited. Previous cell culture work [378] found that APN induced the

expression of MMP-3 which may explain the increase in MMP-3 observed immediately post-

exercise. However, the present findings did not see changes in APN regardless of condition or

day. The acute effect of exercise on APN remain equivocal with increases observed following

Page 87: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

87

acute moderate-to-high intensity (55% to 70% VO2max) aerobic exercise [300, 383], whilst

others have not seen changes following moderate-intensity (60-65% VO2max) aerobic exercise

[298, 384].

Physical inactivity has been shown to impair glycemic control [207]. As a consequence, the

assessment of postprandial glucose is crucial where if elevated, can lead to the development

of type 2 diabetes [385]. Elevated postprandial glucose is also an independent risk factor for

cardiovascular events in individuals with or without type 2 diabetes [386]. Fortunately, poor

glycemic control can be ameliorated following increased physical activity [207]. However,

despite the improvements seen in blood glucose following acute exercise [207, 387], neither a

single bout of CME or HIIE in the present study was sufficient in improving glycemic control

as seen from a lack of improvement in 24 h and 48 h blood glucose upon cessation of exercise

(Figure 5.3). Our findings were in contrast to the findings of Oberlin et al whom had found

improved glucose response in the 24 h following acute exercise, but not in the subsequent 24

h in type 2 diabetic individuals [334]. Interestingly, despite a ~10% reduction in glucose

response during the first 24 h, significantly lowered postprandial glucose AUC response was

only found following one of three meals [334]. In addition to improvements seen 2.5 h post-

breakfast glucose on the first day of exercise, van Dijk and colleagues found that a single bout

of exercise was consistently reduce post-breakfast glucose on the second day following

exercise [388]. Some differences of the current study may be attributed to our population of

interest which did not present with T2DM, resulting in less room for improvement of glycemic

control. Secondly, the present study utilized OGTTs for the glucose challenge, as opposed to

mixed meals used in both studies where its macronutrient distribution may have affected

glycemic responses. The timing of exercise and the subsequent meal may have also affected

glucose responses such that in the present study the OGTT was consumed in a fasted state 1 h

following exercise rather than 2 h following exercise [334]. In the study by van Dijk et al.

exercise was performed following breakfast, which may have further altered the glucose

response following exercise [388]. Physical activity throughout both trials were monitored

using a tri-axial activity monitor over each three-day period. Physical activity monitoring

found that energy expenditure, sedentary time and total vector counts were no different

between exercise conditions and days indicating that neither conditions promoted an increase

Page 88: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

88

in activity throughout each day regardless of exercise intensity.

This study had a number of strengths including i) duration- and workload-matched CME and

HIIE conditions; ii) recruiting a clinically relevant population, namely individuals who were

inactive and overweight- which has, to our knowledge not previously been conducted in a

study of this kind; iii) adopting a randomized cross-over design; iv) each trial included a

specific baseline compromising of 24h continuous glucose monitoring which were used to

compare 24h differences following exercise for two days post exercise. However, there were

also some limitations associated with this study. Although the use of the CGMS provides

valuable information on the rapid short-term changes in glycaemic fluctuations, it does not

directly measure glucose concentrations. Rather, it measures a signal that is proportional to

the glucose level in the interstitial fluid. Another limiting factor previously experienced

included the time lag between the variation of blood glucose and those from the subcutaneous

interstitial fluid [389-391]. Errors were minimized by only using two continuous glucose

monitors throughout the study, with each subject using the same unit for both phases. Despite

the measurement of MMP-2 and MMP-3 in this study, we did not measure activity of tissue

inhibitors of MMPs which would have accounted for the regulatory activity of these MMPs

which may have provided further insight into the acute effect of MMPs following exercise.

Additionally, although exploring the changes of inflammatory markers between exercise-

intensity provides an insight on the acute effect of exercise, implementing this study in a type

2 diabetic population would be a strength for future studies that would have allowed

assessment across clinical and pre-clinical stages. Although the strict timing on completion of

the OGTT allowed for consistency over each trial, providing all participants with standardized

meal over the three days at strict meal times would have allowed for greater control of dietary

intake. Finally, conducting multiple regression analysis to assess the effect of respective

markers on glucose profile would provide more substantial and concrete answers, however a

greater number of participants would be required to run such an analysis. Due to the lack of

significant change in the glucose profile following exercise, we were not able to determine the

impact of the changes in these inflammatory markers on the glucose response.

Page 89: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

89

The present study demonstrated that a single bout of moderate- and high-intensity intermittent

exercise was effective in inducing changes to MMP-3 and OPN immediately following

exercise which is consistent previous work, although this did not persist and returned to

baseline levels by the next morning. An improvement in glucose tolerance was not evident

following completion of OGTTs over the three days which is in contrast to previous findings,

which was also reflected in continuous glucose concentrations in the 48 h period following

exercise. Further research is warranted to address limiting factors such as glycemic status,

exercise and meal timing (preprandial vs postprandial) across an at-risk population as well as

clinically diagnosed type 2 diabetes individuals.

Funding: AR was supported by a Murdoch University Research Scholarship during

completion of this work. This research did not receive any specific grant from funding agencies

in the public, commercial, or not-for-profit sectors.

Disclosures: No conflicts of interests to declare.

Author Contributions

A.R, T.J.F, J.J.P conceptualized and designed the study; A.R. and N.G.L performed

experiments; A.R and N.G.L analysed data; A.R, G.F.H and T.J.F interpreted results of

experiments; AR and T.J.F drafted manuscript, A.C, A.R, G.F.H, J.J.P, N.G.L and T.J.F edited

and revised manuscript. All authors approved the final version of manuscript.

Page 90: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

90

Chapter 6 Short-term training effect on

glycaemic control and the

inflammatory response in

type 2 diabetes.

Authors: Aaron Raman1, Jeremiah J. Peiffer1, Gerard F. Hoyne2 and Timothy J. Fairchild1

Affiliations:

1School of Psychology and Exercise Science, Murdoch University, Western Australia,

Australia;

2School of Health Sciences, University of Notre Dame Australia, Fremantle Campus,

Australia.

Corresponding Author:

Aaron Raman

School of Psychology and Exercise Science,

Murdoch University,

90 South Street,

Murdoch, 6150, Western Australia,

AUSTRALIA.

E-mail: [email protected]

The manuscript in this chapter is prepared for submission to the Medicine and Science in

Sport and Exercise.

Page 91: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

91

6.1 Abstract

This study examined the effect of short-term training on glycaemic control and the

corresponding inflammatory response associated with type 2 diabetes mellitus (T2DM).

Thirteen (5m/8f) obese (32.0±4.7kg/m2) individuals previously diagnosed with T2DM

completed 12 exercise training (45 minutes of treadmill exercise; 65% VO2peak) sessions in

successive days. An oral glucose tolerance test (OGTT; 75g) was completed before and 48 h

after the training period, along with fasting plasma measurements of fructosamine, glucose,

insulin, and C-Reactive Protein. To distinguish the acute and training responses, inflammatory

cytokine (unstimulated and LPS-stimulated) responses immediately following the first and last

exercise session were assessed in addition to the measurement of regulatory T cells which are

of anti-inflammatory nature. The acute response of exercise resulted in increased pro- (IL-1β,

TNFα and IP-10) and anti-inflammatory (IL-10) cytokine response following the last training

session (p≤0.045); however, no concurrent differences were observed in regulatory T cells at

any time point (p>0.650). Although an improvement in glucose and insulin area under the

curve was not evident (p>0.135), exercise training was accompanied with a 5% reduction in

glycaemic marker, fructosamine (p=0.023) which was concomitant with reduced (~27%)

global inflammation measured from CRP (p=0.037). Although exercise training was effective

in improving glycaemic control and reducing global inflammation as measured from CRP, the

acute increase in cytokine response indicates that longer term training may be of benefit to

demonstrate a reduced inflammatory response as a result of the training adaptation which

would be expected with a greater proportion of regulatory T cells.

Trial Registration: ACTRN12615000613505

Page 92: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

92

6.2 Introduction

Maintaining an appropriate balance between anti-inflammatory and pro-inflammatory

cytokines in the systemic circulation appears critical to avoiding inflammatory-related disease,

including type 2 diabetes (T2DM) [263]. To this end, adipose tissue in lean individuals is

largely comprised of alternatively-activated macrophages (M2) which promote a Th2-type

response producing anti-inflammatory cytokines such as Interleukin (IL)-10 and IL-1 receptor

antagonist (IL-1ra) [198]; balancing the pro-inflammatory cytokines produced by the

classically-activated (M1) macrophages [198, 392]. The anti-inflammatory environment of the

adipose tissue is further supported by regulatory T cells (Tregs) through the production of IL-

10 and Transforming Growth Factor-beta (TGF-β) [392, 393]. The increased accumulation of

adipose tissue associated with obesity facilitates an increased shift in the polarization of these

macrophages towards the M1-like phenotype [198]. This increased M1 infiltration and

subsequent increase in pro-inflammatory cytokines is associated with the characteristic insulin

resistance in obesity-linked T2DM [12, 14, 197]. Concomitantly, there are a reduced number

of adipose tissue resident Tregs associated with obesity and T2DM, and the functional

suppressive capacity of these cells are reduced [263]. Accordingly, increased circulating

concentrations of acute phase proteins such as C-reactive protein (CRP) and alpha-l acid

glycoprotein (AAGP), as well as classic pro-inflammatory cytokines such as IL-1β, TNFα,

and IL-6 are characteristic in individuals with obesity and T2DM [394-396]. Furthermore,

evidence suggest that Monocyte Chemoattractant Protein (MCP-1) and IL-8, IL-15 and

Interferon Gamma-Induced Protein 10 (IP-10) may also contribute to insulin resistance. MCP-

1 is a pro-inflammatory chemokine that has been shown to promote macrophage infiltration

and insulin resistance in mice [397]. MCP-1 may also play a significant role in insulin

resistance by affecting insulin sensitivity in adipocytes which reduced insulin-stimulated

glucose uptake following incubation of a low-dose of MCP-1 in vitro [398]. IL-8 and IP-10

are also chemokines which have been previously found to positively associate with insulin

resistance and T2DM [399, 400]. Importantly, both these markers are also reported to associate

with hsCRP and fasting glucose which may prove to be a significant risk marker of T2DM

[399, 400]. On the other hand, IL-15 is a growth factor produced in skeletal muscle which has

been found to stimulate adiponectin, an adipose-derived anti-inflammatory cytokine known to

promote insulin sensitivity [401]. Indeed, previous work has supported this theory as seen in

obese mice following IL-15 treatment which resulted in improved insulin sensitivity [402]

Therefore, systemic low grade chronic inflammation is not only believed to be characteristic

of T2DM, but is attributed to play an important role in the development of obesity-related

insulin resistance and T2DM [195, 403].

Page 93: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

93

Exercise is recommended as a key component in the management of individuals with T2DM

[404]. Exercise increases the translocation of glucose transporters (GLUT4) to the plasma

membrane promoting increased glucose uptake acutely [32, 272, 273, 387, 405-407]. In the

longer-term, exercise training in individuals with T2DM results in increased GLUT4 content

and improved glucose tolerance [184], and is associated with significantly reduced glycated

haemoglobin (HbA1c) [27, 408], a marker of glycaemic control. This improvement in HbA1c

is comparable to the use of non-insulin lowering medication [28, 408]. Of relevance, training

studies of shorter duration (i.e., <12 weeks) observed a 0.8% decrease in HbA1c which was

more pronounced than studies of longer duration (i.e., >12 weeks; 0.7%) [409]. These

improvements may be partially attributed to increased exercise volume and the specific

progressive nature of the exercise training adopted [409]. Subsequent short-term training

studies from as few as six sessions [33] to seven days of consecutive training [40, 182] have

reported improvements in glycaemic control, which is likely explained by improved insulin

action [41, 188].

Acute exercise elicits an exaggerated inflammatory response [410, 411] while exercise training

attenuates this response, although large heterogeneity in responses are seen between

populations and training variables [410]. Exercise training in obese non-T2DM [205, 206]

and T2DM appears to have beneficial effects on the inflammatory status such as decreased

concentration of global marker of inflammation, hsCRP (high-sensitivity CRP), as well as

decreased inflammatory cytokines including IL-1β, TNFα and increased IL-10 [29, 44].

Whether changes in inflammatory markers are observed concurrent with improvements in

glycaemic control in individuals with T2DM in response to short-term training, is not known.

Therefore, this study sought to determine the impact of 12 days of continuous exercise on

glycaemic control, changes in inflammatory cytokines and the population of regulatory T cells.

Glycaemic control was assessed using fructosamine, which is a short-term (~2 weeks) marker

of glycaemic control [412], and glucose response during an oral glucose tolerance test. We

hypothesised that exercise training would be associated with i) decrease in fructosamine and

improved glucose tolerance; ii) decrease in pro-inflammatory markers and an increase in anti-

inflammatory markers. A secondary aim was to assess the acute inflammatory response to

exercise, by assessing inflammatory markers pre- and post-exercise on the first and last (12th)

exercise session. To aid the interpretation of changes in the circulating inflammatory markers,

we also assessed the direct functional ability of immune cells to produce an inflammatory

response in vitro. We hypothesised that the inflammatory response would be attenuated in the

12th exercise session relative to the first session, but the inflammatory response to in vitro

stimulation (using a toll-like receptor 4 ligand) would be unaffected.

Page 94: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

94

6.3 Methods

6.3.1 Study Design

This study adopted a single-arm, repeated measures design. All participants attended the

Exercise Physiology laboratory at Murdoch University for a total of sixteen visits over a three-

week period. The first visit involved the consent process, a familiarisation session and

assessment of fitness (peak oxygen consumption, VO2peak assessment). At least two days

following familiarisation, participants visited the laboratory in the morning for the completion

of a baseline oral glucose tolerance test (OGTT) before commencing the first of twelve training

session the next day. Following the two-week training period, an OGTT was repeated 16-24h

following the last training session. In addition to the blood sampling during the OGTT, venous

blood samples were collected via venepuncture prior to and immediately following the first

and last exercise sessions during the training period. Participants were informed of the possible

risks associated with the study before providing written informed consent. Ethics approval was

granted by Human Research Ethics Committee at Murdoch University. This trial was

prospectively registered with Australian New Zealand Clinical Trials Registry:

ACTRN12617000286347.

6.3.2 Participants

Thirteen (5m / 8f; aged 51 ± 7 y) sedentary, overweight/obese (BMI > 27 kg/m2) volunteers

previously diagnosed with T2DM (HbA1c range; 6.2% to 10.2%) were recruited from the local

community (Table 6.1). Participants were deemed eligible if they were between 18 and 65

years old, and completed less than or equal to two bouts of low-to-moderate intensity exercise

per week). Exclusion criteria included the use of exogenous insulin, contraindications to

exercise training and smoking. Participants were pre-screened for exercise using the Exercise

Science and Sport Science Australia (ESSA) medical screening tool. 12 participants were on

anti-diabetic medication, 4 were on anti-hypertensive medication and 2 were on anti-

depressive medication.

Page 95: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

95

Pre-training Post-training Effect of training

(p-value)

Age (y) 51 (7)

HbA1c (mmol/mol) 53.8 (21.5)

Height (m) 1.66 (0.07)

Weight (kg) 90.6 (14.8) 90.7 (14.6) ES: 0.007; p = 0.809

BMI (kg/m2) 32.7 (4.9) 32.7 (4.6) ES: 0.000; p = 0.931

WC (cm) 103.9 (11.9) 101.1 (10.3) ES: 0.244; p = 0.134

Fat (%) 37.1 (6.4) 36.2 (7.1) ES: 0.129; p = 0.051

VAT (g) 901.6 (340.3) 925.8 (366.9) ES: -0.034; p = 0.450

VAT (cm3) 974.7 (367.9) 1000.9 (396.6) ES: -0.066; p = 0.491

VO2peak (ml/kg/min) 23.1 (4.9) 24.1 (4.7) ES: -0.202; p = 0.011

Table 6.1. Descriptive characteristics of the participants at baseline (data presented as mean

(SD)) and changes in anthropometric measures in response to exercise training; where effect

of training is represented by Cohen’s d effect size and p-values. BMI, body mass index; WC,

waist circumference; VAT, visceral adipose tissues; VO2peak, peak VO2 achieved.

6.3.3 Introductory session

On arrival, participants were fully informed of all study procedures and written informed

consent obtained. Body height, body mass, waist circumference was measured, whilst body

composition was assessed using Dual Energy X-ray Absorptiometry (DXA; Hologic

Discovery QDR series, Hologic Inc., Bedford, MA, USA). Thereafter, participants completed

a cardiorespiratory fitness test (start at 2.5km/h, 1km/h ramp every 2 minutes until 5.5km/h

followed by 2% increase in gradient thereafter) until volitional exhaustion on a treadmill

(Trackmaster, Full Vision, Kansas, USA) for measure of VO2peak. During the cardiorespiratory

fitness test, expired ventilation was collected in 15-second averages using a metabolic cart

(Parvomedics TrueOne, Sandy, UT, USA) and heart rate (Polar T31, Kempele, Finland) was

collected at 1 Hz. VO2peak was determined as the highest 30s average VO2 from the final stage

of the ramp test. Heart rate and Borg’s (6) rating of perceived exertion (RPE) was collected

throughout the maximal graded exercise test. This session was completed at least three days

prior to the first training session. These tests were repeated upon completion of the training

period.

Page 96: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

96

6.3.4 Oral Glucose Tolerance Test (OGTT)

OGTTs (75 g glucose) were performed after an overnight (10–12 h) fast at the same time of

day before and following the training period. Participants refrained from exercise and

medication use on the morning of testing. Post-intervention OGTTs were performed within

36-48 h of the final bout of exercise. Prior to the ingestion of the glucose-containing drink, a

catheter was placed in an antecubital vein for venous blood samples collected at 30 min

intervals for measurement of glucose and insulin. Food intake prior to each OGTT was at the

participants’ discretion and were advised to refrain from dense sugar containing foods in the

evening prior to the OGTT. Participants recorded their food intake in a 24h food recall and

were advised to replicate their meal to their best ability prior to the OGTT post training.

6.3.5 Experimental Phases (Exercise intervention)

On 12 consecutive days, all volunteers performed 45 min of supervised exercise for 12

consecutive days at ~65% VO2peak. A heart rate monitor was worn during each session to

monitor heart rate corresponding to the desired workload from the cardiorespiratory fitness

test. All sessions were completed from participants with 100% compliance.

6.3.6 Biochemical analysis

6.3.6.1 Glycated haemoglobin (HbA1c) and high

sensitivity CRP (hsCRP)

Fasting venous blood samples were collected into EDTA and lithium heparin tubes for the

analysis of HbA1c and hsCRP a local commercial laboratory (Western Diagnostics Pathology,

Myaree, Western Australia). HbA1c was measured pre-training whilst hsCRP was collected

pre- and post-training (within 36-48 h of the final bout of exercise).

6.3.6.2 Glucose, Insulin, inflammatory markers

Blood samples from the OGTT were collected into EDTA tubes containing aprotinin

(50KIU/ml blood; Becton Dickinson, Plymouth, United Kingdom) and serum clot activator

tubes (Becton Dickinson, Plymouth, United Kingdom). The EDTA tube was immediately

centrifuged at 1300g for 10 minutes whilst serum tubes were allowed to clot for 60 minutes

before centrifuging at the same speed. Plasma and serum samples were aliquoted into

triplicates and stored at -80˚C for later analysis. Alpha-1 acid glycoprotein, blood glucose and

fructosamine were assessed via chemical analyser (COBAS Integra 400 plus, Roche

Page 97: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

97

Diagnostics Ltd, Switzerland) and insulin by enzyme linked immunoassay (Mercodia;

Uppsala, Sweden). The area under the curve (AUC) for glucose and insulin during the OGTT

were calculated using the trapezoidal method. Insulin resistance was calculated using the

homeostasis model assessment (HOMA) of insulin resistance (HOMA2-IR), β-cell function

(HOMA2%beta) and insulin sensitivity (HOMA2%S) from fasting glucose and insulin

concentrations (HOMA2 calculator (v2.2); available online at

http://www.dtu.ox.ac.uk/homacalculator/), the insulin sensitivity index (ISI) composite was

calculated using glucose and insulin values at the 0 and 120 min from an OGTT [413].

Additionally, the hepatic insulin resistance index was calculated using the 0 and 30min glucose

and insulin values; although fasting indices are thought to reflect hepatic insulin resistance,

this measure is believed to be more specific to hepatic insulin resistance [414]. To assess

peripheral insulin sensitivity, the Cederholm index was calculated as previously described

[415].

6.3.6.3 Toll-like receptor stimulation and blood culture

Venous blood samples were collected via venepuncture prior to and immediately following

the first and last (12th) exercise session during the training period. The in vitro inflammatory

response to toll-like receptor (TLR) stimulation was assessed by adding 180 μL of diluted

blood to each well in a 96-well plate containing the TLR4 ligand, prepared as previously

described at the University of British Columbia in Vancouver, Canada [416]. The plates

contained the TLR4 ligand, lipopolysaccharide (LPS; TLR-4, InvivoGen) at 10 ng/mL. The

control condition consisted of a 96-well plate which did not contain LPS. Following addition

of blood, plates were incubated at 37°C in 5% CO2 for 24 hours after which the plates were

centrifuged at 600g and 50 μL of supernatant was removed and stored at -80°C for later

analysis.

6.3.6.4 Regulatory T cells (Tregs)

Venous blood collected via venepuncture prior to and immediately following the first and last

exercise session were stained for Tregs using fluorescent-conjugated monoclonal antibodies

to identify cell surface (CD3-BV510, CD4-APCH7, CD8-PerCpCy5.5, CD25-PeCy7, CD127-

BV421, CD120B-PE and live/dead stain-FITC) and intracellular (Foxp3-AF647) markers via

eight-colour flow cytometer (BD FACS CantoII, Becton Dickinson, UK). Antibodies were

mixed in 150L of whole blood containing EDTA into a TruCount tube (Becton Dickinson,

UK) and set to incubate for 20 minutes on ice. Red blood cells were then lysed with lysing

solution (BD Pharmlyse, Becton Dickinson, UK) following further incubation for 15 minutes

Page 98: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

98

before being spun at 600g at 4°C for 5 minutes and washed twice using phosphate-buffered

saline (PBS). The supernatant was then aspirated and cells were resuspended in PBS followed

by incubation with a live/dead stain (Zombie Fixable Viability kit, Biolegend, CA, USA) for

15 minutes. Cells were then washed twice and resuspended in PBS before fixation and

permeabilisation (Foxp3 fix/perm buffer set, Biolegend, CA, USA) as per manufacturer’s

recommendations for the intracellular staining of foxp3. 150uL of stabilizing fixative (Becton

Dickinson) was then added prior to flow cytometry analysis within 24h. Forward-scatter

versus side-scatter plot was used to gate lymphocytes based on size and density. Live cells

were included from the live/dead stain followed by the inclusion of single cells by gating SSC-

W by SSC-H. Viable single cells were then gated for CD3+CD4+CD8- T cells of which Tregs

were identified by further gating of CD127-CD25+ cells (Figure 6.1). The FoxP3, CD25 and

CD120B gates were set using FMO controls in order to correctly identify a positive population.

From this population, confirmation of Tregs were gated by foxp3. Absolute cell counts were

calculated from TruCount tubes by dividing the number of positive cell events (X) by the

number of bead events (Y), and then multiplying by the BD Trucount bead concentration (N/V,

where N = number of beads per test* and V = test volume); A = X/Y × N/V.

6.3.6.5 Cytokine measurement

Supernatants were thawed at room temperature and assayed by multiplex assay. The high-

biotin protocol was used to measure the levels of the following cytokines: IL-10, IL-15, IL-

1β, IL-6, IL-8, IP-10, MCP-1, TNF-α (eBioscience custom multiplex, Jomar Life Research,

Australia) and TGF- were measured from a ProcartaPlex simplex bead set (eBioscience

catalog, EPX01A-10249-901, Jomar Life Research, Australia) which was combined with the

Procarta Human Basic Kit (eBioscience, catalog EPX010-10420-901, Jomar Life Research,

Australia) All assays were measured using the Bioplex system (BioRad, Hercules, CA, USA)

running the Bioplex analysis software.

6.3.7 Data Analyses

Data is presented as mean ± SD, unless otherwise stated. Sample size was calculated with an

effect size of F = 0.4326 with 80% power at a significance level of 5% based on Foxp3. If

sphericity was violated during the analysis, the Huynh-Feldt correction was used. Glucose and

insulin concentrations from the oral glucose tolerance test were used to calculate the area under

the curve (AUC) using Graphpad software (Graphpad, PRISM). The inflammatory cytokine

panel, cell populations and acute phase reactants were analysed using a two-way analysis of

variance (ANOVA) with repeated measures; with one between factor (Training; 1st session vs

Page 99: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

99

12th session) and one within factor (acute exercise; pre-exercise, post-exercise). The

hypothesis of interest was the interaction effect (training x acute exercise), however, main

effects were explored with post Hoc analysis using least squared differences). Paired student’s

t-test were calculated for pre- and post-training time points for anthropometric variables,

VO2peak, HOMA2-related outcomes, glucose-AUC, insulin-AUC, hsCRP and fructosamine.

Pearson’s correlation was calculated to determine relationships between variables of interest

(hsCRP, body fat percentage and VO2peak). All statistical analyses were completed using

commercially available software (SPSS 21 Windows; SPSS, Chicago, IL, USA). Significance

was accepted at p.05.

Figure 6.1. Identification of Tregs via flow cytometry.

Page 100: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

100

6.4 Results

All participants completed twelve training sessions with 100% compliance. Due to difficulties

with blood samples before and after acute exercise on the 1st and 12th session, data was only

available from 9 of 13 participants (4m/5f) at these time points. Participants were on average,

well controlled (HbA1c: 53.8 mmol/mol; 7.1%), although ten participants had HbA1c over

6.5% and four had HbA1c over 8%.

6.4.1 Fitness and Anthropometric data (Table 6.1)

All participants completed the VO2peak test with a respiratory exchange ratio of greater than

1.10 and VO2 reaching a plateau at the end of the cardiorespiratory fitness test. Following 12

consecutive days of treadmill exercise, participants increased VO2peak (p = .011; d = 0.202) and

reduced body fat percentage (p = .051; d = 0.129). There were no significant changes in body

weight or BMI (both p ≥ .809), and there were no significant changes in waist circumference

or visceral adipose tissue (volume and weight) (all p ≥ .134; d ≥ 0.244; Table 1).

6.4.2 Glycaemic variables (Table 6.2)

Fructosamine was significantly (p = .023; d = 0.202) decreased following the 12 days of

exercise training (Table 2). However, exercise training did not result in significant differences

in fasting blood glucose, 2 h glucose AUC or 2 h insulin AUC (All p ≥ .135; All d ≤ 0.18).

Indices of insulin sensitivity, indicative of total, hepatic and muscle insulin sensitivity, were

also not significantly altered after the short-term training period (All p ≥ .086; All d ≤ 0.5).

Page 101: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

101

Pre-training Post-training

Effect of training

(p-value)

Glycaemic control

Fructosamine (µmol/L) 307 (76.6) 291.7 (67.4) ES: 0.202; p = 0.023)

FBG (mmol/L) 10.3 (3.9) 9.8 (4.0) ES: 0.123; p = 0.301

2h Glucose AUC (mmol/L) 1931 (514) 1830 (590) ES: 0.177; p = 0.135

2h Insulin AUC (µU/mL) 4820 (3909) 4464 (3522) ES: 0.007; p = 0.225

Insulin resistance

HOMA2-IR 2.48 (1.44) 2.26 (1.24) ES: 0.159; p = 0.134

HOMA2-B (%) 58.5 (51.8) 60.2 (49.2) ES: -0.017; p = 0.554

HOMA2-S (%) 49.0 (18.0) 54.1 (21.6) ES: -0.248; p = 0.113

ISI (composite) 2.24 (1.01) 2.38 (1.02) ES: -0.069; p = 0.331

Peripheral insulin resistance

Matsuda muscle -0.113 (0.103) -0.079 (0.090) ES: -0.174; p = 0.301

Cederholm 21.4 (10.7) 24.4 (12.6) ES: 0.499; p = 0.086

Table 6.2. Measures of glycaemic control and insulin resistance pre- and post-training

(presented as mean (SD)) with associated effect (Cohen’s d) of the exercise training

intervention. FBG, Fasting blood glucose; AUC, Area under the curve; HOMA2-IR,

Homeostatic model assessement index 2; HOMA2-B, beta cell function; HOMA2-S, insulin

sensitivity; ISI, Insulin sensitivity index

6.4.3 Acute phase proteins and hsCRP

Exercise training was associated with a significant decrease in hsCRP (27.2%; p = 0.037;

Figure 6.2). Although the change in hsCRP did not correlate with changes in body fat

percentage (r = -0.259; p= 0.202) or VO2peak (r = 0.156; p = 0.446). A significant interaction

effect in the acute phase protein alpha-l acid glycoprotein (AGP; p = 0.001) was identified,

wherein the acute response (pre- and post- exercise session) was altered following the 12

consecutive training bouts. Specifically, the pre-exercise AAGP concentration was ~5.2%

lower at the 12th exercise session compared to the 1st exercise session; while the concentration

of AAGP post-exercise were comparable between sessions (Figure 6.2). No significant

differences were found for homocysteine over the training period (p = 0.233).

Page 102: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

102

Figure 6.2. Mean (± SD) concentration of acute phase reactants; Alpha 1-acid glycoprotein,

Homocysteine, Fructosamine, high sensitivity C-reactive Protein and Transforming Growth

Factor-beta. *, post-exercise was greater than pre-exercise in the 12th session, p < 0.001; ^,

Post-training was lower than pre-training values, p = 0.037.

6.4.4 Cell populations (Table 6.3)

Exercise was not associated with changes in either absolute cell count or relative percentage

of CD3+CD4+ T cells or CD3+CD8+ T-cell populations, either in response to acute exercise

(pre- versus post-exercise session) or training (1st session to 12th session; all main effects p ≥

0.483, Table 3). This highlighted that exercise did not induce further immune activation

following exercise as demonstrated by no changes in proportionality in either CD3+CD4+ or

CD3+CD8+ T cell production. Additionally, the ratio of CD4+ to CD8+ was not altered by

acute exercise (p = 0.842) or exercise training (p = 0.462). Unsurprisingly, the lack of changes

in the T-cell population was also reflected in the percentage of CD4+CD127-CD25+ Tregs

and CD4+CD127-Foxp3+ Tregs which were not affected by neither acute exercise nor

training (p > .650; Table 6.3). Further function of the Tregs assessed by mean fluorescence

intensity for CD120B did not reveal a main effect of time (p = 0.910) nor a training effect (p

= 0.811).

1st session 12th Session

Page 103: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

103

Pre-training Post-training Pre-training Post-training

CD3+CD4+ (cells/ul) 293 (157) 300 (152) 320 (105) 289 (111)

CD3+CD8+ (cells/ul) 128 (102) 110 (140) 121 (41) 119 (45)

CD4+ (% cells) 58.4 (11.7) 58.0 (9.9) 55.7 (10.4) 54.7 (11.3)

CD8+ (% cells) 22.1 (9.7) 21.5 (10.1) 21.6 (8.1) 22.8 (8.7)

CD4+/CD8+ ratio 3.2 (1.7) 3.3 (1.8) 3.0 (1.5) 2.8 (1.3)

CD127-CD25+ (% cells) 3.3 (1.2) 2.8 (1.4) 2.7 (0.6) 3.4 (1.2)

CD127-foxP3+ (% cells) 1.2 (0.8) 1.2 (1.0) 1.0 (0.8) 1.0 (0.7)

Table 6.3. Absolute count and percentage of T cells in whole blood. Data displayed as mean (SD).

6.4.5 Functional inflammatory response

Half the lowest concentration of the standard curve was used for time points which presented

with data that were out of range [417]. Following acute exercise during the 12th session, a

significant effect of acute exercise was only present for IP-10 (p = 0.012) in the unstimulated

condition, which demonstrated a 37.3% increase post-exercise when compared to pre-exercise

(Figure 6.3). No significant interactions (p > 0.182) or main effects of time (pre- versus post-

acute; (p > 0.113) or training session (Session 1 vs Session 12; p > 0.255) were found for any

other cytokines in the unstimulated condition.

When whole blood was stimulated with LPS to determine the response of cytokine production,

no significant interaction effects (acute exercise response x training session; p > 0.147) or main

effects of training (session 1 vs session 12; p > 0.274) were found. However, there were

significant main effects in the acute exercise response (Figure 6.3), which were predominantly

driven by the acute-exercise response in the 12th session. Of the pro-inflammatory cytokines,

IL-1β, TNFα and IP-10 demonstrated an increase of 25% (p = .014), 55% (p = 0.045) and

140% (p = 0.022), respectively. The anti-inflammatory cytokine IL-10 also demonstrated an

increase by 32% (p = 0.023) (Figure 6.3). However, despite the potential role of IL-8, IL-15

and MCP-1 on insulin resistance, none of these markers were affected following acute exercise

nor exercise training (p > 0.165). Surprisingly, the increased production of IL-6 immediately

post exercise in the 1st and 12th training session did not result in a significant difference (p =

0.063).

Page 104: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

104

Figure 6.3. Mean (± SD) concentration of unstimulated and LPS-stimulated inflammatory

cytokines pre- and post-exercise in the first and 12th session of training. *, post-exercise was

greater than pre-exercise in the 12th session, p < 0.05. Closed circles represent first session;

open circles represent 12th session.

Page 105: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

105

6.5 Discussion

T2DM is characterized by an increased pro-inflammatory status [46], while exercise training

is associated with an increase in anti-inflammatory status [418]. Acute exercise has also been

shown to have profound effects on the inflammatory status [204], although paradoxically,

acute exercise acutely increases pro-inflammatory cytokines. The purpose of this study was to

determine whether an intensified training program, comprising 12 successive exercise training

days can improve glucose tolerance in individuals with T2DM, and determine the associated

changes in T-cell populations and their functional capacity in response to stimulation via a

TLR4 ligand. The main findings were i) exercise training improved fructosamine

concentrations, a short-term (2-3 week) marker of glycaemic control, although this was not

associated with significant improvements in other markers of glucose tolerance or insulin

sensitivity; ii) exercise training significantly reduced hsCRP, which was associated with a

significant decrease in the pre-exercise concentration of AGP; iii) exercise training did not

significantly alter T-cell populations, and resulted in a variable cytokine response. Given the

profound immune-modulating effect of acute exercise, this study also sought to determine

whether the acute response to exercise would be altered in response to the short-term training.

The main findings were an increase in a number of the pro-inflammatory cytokines during the

12th session when compared to the first session, which appeared to be larger in magnitude than

the rise in anti-inflammatory cytokines, which was contrary to our hypothesis.

Considering the short-term nature of the exercise protocol, the primary measure of glycaemic

control adopted in the present study was a change in fructosamine concentration [412].

Fructosamine is an alternative measure of glycaemic control to HbA1c [53, 419], and provides

a measure of glycosylated proteins (primarily albumin) which have shorter lifespans (1-3

weeks) than red blood cells (~17 weeks). In agreement with the stated hypothesis, there was a

significant improvement in fructosamine concentration after the exercise training period.

However, there were no significant improvements in fasting blood glucose, glucose AUC and

insulin AUC, indices of insulin sensitivity (HOMA2-IR, HOMA2%beta, HOMA2%sensitivity

& ISI composite); although all indices demonstrated favourable shifts in response to the

exercise intervention. A number of studies have previously employed short-term training

studies (≥7 consecutive days of exercise) in T2DM individuals and demonstrated

improvements in measures of glycaemic control [40, 41, 188]. Similarly, participants with

metabolic syndrome, but not T2DM improved glucose tolerance following 10 days of

treadmill walking [420]. Fasting blood glucose was not improved which is consistent with

previous work [40, 188] but not all studies, with O’Gorman and colleagues demonstrating

significant improvements in fasting blood glucose and insulin sensitivity following 7 days of

exercise, which was attributed to an increase in muscle GLUT4 content [41]. Of note, the

Page 106: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

106

Cederholm index, a measure of peripheral insulin sensitivity [421], was associated with a

moderate improvement (d = 0.5) consistent with data indicating that exercise-based

interventions are more likely to improve muscle sensitivity as opposed to hepatic insulin

sensitivity [89].

The short-term exercise training was also effective in reducing the concentration of the acute

phase reactant hsCRP, which has been well documented to be associated with T2DM [11, 12,

19] in addition to the development of cardiovascular disease [221]. This training-induced

reduction highlights the lowered global level of inflammation and reduced cardiovascular risk

despite the average concentration within this type 2 diabetic population remaining in the high-

risk classification [10]. The reduction in CRP is also consistent with the improvements seen

in longer term training studies lasting up to 10 months in individuals with [422] and without

[43, 423] T2DM, further highlighting the effectiveness of just 12 days of exercise. Although

IL-6 is correlated and a well-known precursor to the production of CRP [43], the reduction in

CRP was not accompanied with a reduction in IL-6. This may be attributed to the lack of

significant weight loss as previous work has reported an association between weight loss and

a reduction in CRP and IL-6 [289].

AAGP is an acute phase glycoprotein that is primarily synthesised by hepatocytes in the liver

and released as part of the acute phase response (peaking at a later time to CRP) which has

been shown to positively associate with T2DM [396, 424-426]. Similarly to CRP, IL-6 in

addition to IL-1β and TNFα have previously been reported to induce the expression of AAGP

[425]. Hence, it is not surprising that AAGP was previously found to correlate with IL-6 and

CRP in addition to all three markers predicting all-cause mortality [427]. Despite the lack of

differences in resting concentration between the 1st and 12th exercise session, pre-exercise

AAGP was lower in the 12th exercise session when compared to the 1st exercise session.

Despite the immediate increase of AAGP after exercise on the 12th session, the concentration

was similar to both pre- and post-exercise concentration of the 1st training session. As an acute

phase reactant, the increase of AAGP is consistent with previous work which increased in

concentration following a marathon [428]. However, training studies lasting between two to

six months have previously found a reduction in this acute phase protein, which was associated

with reduced visceral fat mass [429], a significant risk factor for the development of T2DM.

Although body fat percentage and visceral adipose tissue did not change with exercise training,

the lowered baseline AAGP concentration may be related to the outcome of CRP post training

since both markers have previously been found to correlate with each other [427] and are

associated with the risk of developing T2DM [46, 426]. It is recognized that AAGP was not

measured at the same time as CRP which will limit interpretation of the relationship between

Page 107: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

107

these two markers. However, the significant reduction in CRP demonstrates the beneficial

impact exercise has on reducing systemic inflammation. which is also evident following long-

term training of up to 10 months [43, 422, 423].

Unlike long-term training studies lasting between 3 and 12 months [44, 205, 430], we did not

find any improvements in the inflammatory cytokines measured pre-exercise between the first

and last exercise session. Together with hsCRP, the pro-inflammatory milieu has been well

documented to be elevated in obese individuals with and without type diabetes; examples of

these inflammatory cytokines include IL-1β, IL-6, IL-15, MCP-1, IP-10, TNFα [11-13, 19,

403, 431]. Previous work have documented significant improvements in low-grade

inflammation following long term exercise training (6 to 12 months), demonstrating the

therapeutic effect of exercise by reduced pro-inflammatory cytokines including hsCRP, IL-1β,

IL-6, IL-18, MCP-1, TNFα and increased anti-inflammatory cytokines including IL-10 and

adiponectin [29, 44]. Despite these findings, previous research have also found no significant

improvements in IL-6, IL-15, MCP-1, hsCRP and TNFα following 3 to 6 months of exercise

training [205, 432]. However, when exercise was accompanied with weight loss and dietary

modification, improvements were seen in pro-inflammatory markers compared to exercise

alone, suggesting a reduction in fat mass may be more important than exercise alone [205].

Following the first training session, acute exercise resulted in increased IP-10. Although acute

exercise did not affect any other inflammatory cytokines measured prior to and immediately

after the first training session, pro- (IL-1β, IP-10, TNFα) and anti-inflammatory (IL-10)

cytokines were increased following the 12th session. This was interesting to note as apart from

IP-10, these markers were only affected following accumulation of the previous 12 exercise

sessions.

Previous work has found that acute exercise leads to an inflammatory response releasing both

pro- and anti-inflammatory cytokines such as IL-6, IL-1β, TNFα and IL-10 [433]. These

cytokines are released as part of an acute phase response as a result of the exercise-induced

suppression of the immune function increasing susceptibility to infection [434, 435]. During

strenuous exercise, cytokines such as IL-6, IL-10, TNFα and IL-1β can be dramatically

increased by up to ~100 fold, 27-fold, ~3-fold, ~2-fold, respectively [436]. The increase in

these inflammatory cytokines is known to be affected following acute exercise which can be

mediated by exercise intensity and duration [47, 278]. In the present study, when blood was

collected post-exercise and cytokines were measured following LPS stimulation and 24 h

incubation, we found that IL-6, IL-10, TNFα and IL-1β increased significantly following

exercise in the 12th exercise session but not the 1st exercise session. The increase seen here

Page 108: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

108

may have been a result of the accumulated training sessions leading to greater activation of

the immune system and subsequently inducing greater functional production of these

cytokines when stimulated with LPS. It is not surprising that IL-6 was increased which may

be explained by the accumulated training period of 9 hours. Increased production of IL-6

following exercise has previously been suggested to be released from the muscle following

contraction [437]. Additionally, the production of IL-6 may also act as an anti-inflammatory

cytokine, stimulating the production of IL-10 [291], as well as inhibiting the production of

TNFα [437]. The production of IL-10 is responsible for its suppressive ability in down-

regulating pro-inflammatory cytokines [438]. The significance of IL-10 as an anti-

inflammatory marker has previously been demonstrated such that IL-10 treatment prevented

lipid-induced insulin resistance and protects skeletal muscle from obesity-associated

macrophage infiltration and the detrimental effect cytokines have on insulin signaling and

glucose metabolism [259]. Additionally, IL-10 also contributes to the expansion of Tregs

which in turn amplifies the production of IL-10 to maintain immune homeostasis [292, 436].

Further production of Tregs has also been demonstrated to be induced by IL-10 under the

presence of TGF-β [267]. Although an increase of IL-10 was seen post-exercise during the

12th session, no such changes were evident in measurement of TGF-β. Previous work has found

that acute moderate-intensity acute exercise resulted in increased TGF-β concentration, but

this was only evident 24h following exercise rather than immediately after exercise [439]. The

lack of change in Tregs was consistent with no alterations found in CD3+CD4+ and

CD3+CD8+ Tcells. The relationship between IL-10 and the therapeutic potential of Tregs has

been shown to be associated with improved insulin sensitivity [258, 440]. Although a lack of

change was seen within circulating Tregs in the present study, previous studies have

demonstrated increased number of Tregs following acute exercise [309, 441] and training

period [310, 311]. Consistent with a previous training study [311], we did not find any changes

with TGF-β. However, in contrast with these findings, others have seen an improvement in

TGF-β 24 h following exercise [439] which has an important role in inducing Tregs and its

suppressive capacity [442]. It must also be stressed that only a limited number of exercise

training related studies have previously been completed in humans [296, 310, 311, 443] and

mice [26, 444]. To the author’s knowledge, this is the first study of intensified exercise training

(12 consecutive days) in a type 2 diabetic population that has attempted to investigate the

relationship of exercise training, Treg production, cytokine response and glucose tolerance.

Little is known with pro-inflammatory marker IP-10 and its relationship with T2DM following

exercise. Elevated IP-10 has previously been demonstrated in obese individuals with [445,

446] and without [447] T2DM compared to healthy individuals; and was positively correlated

with risk markers of T2DM including BMI, fasting glucose fasting insulin, HbA1c and

HOMA-IR, hsCRP and TNFα [448]. Immediately after the 12th exercise session, the

Page 109: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

109

concentration of IP-10 increased by over 50% which suggests a greater pro-inflammatory

environment. Although limited, our finding of increased IP-10 post-exercise is consistent with

others [449] who found elevated IP-10 in post exercise in mice. Pro-inflammatory marker,

RANTES (Regulated upon Activation Normal T cells Expressed and Secreted) has previously

been found to positively correlate with IP-10 [447]. Furthermore, 3 months of exercise training

was found to effectively reduce circulating levels of RANTES in obese humans, but the

changes in IP-10 post-training could not be confirmed as the data was not available [447].

Recent work has also found that treadmill exercise reduced IP-10 in mice, which resulted in

enhanced angiogenesis [450].

Although a number of positive outcomes were found in the present study demonstrating

improved glycaemic control, aerobic capacity, anthropometry and inflammatory status,

several limitations exists which could have further improved the understanding of the changes

seen or lack thereof. An increased number of training sessions over a longer training period

may result in greater improvements in glycaemic control as demonstrated by improved HbA1c

following 2 months of training [451, 452] and improved glucose tolerance following 6 weeks

of training [453]. Consequently, a longer training period of 12 weeks may lead to greater

changes in Treg populations as previously documented [310, 311]. Additionally, the age and

range and menopause may affect the inflammatory results as menopause is associated with

increased concentration of CRP and TNFα [454]. A small sample size in this study may have

obscured some significant outcomes. However, it is important to note that the calculated effect

sizes were fairly small and may demonstrate the importance of adding some dietary

modification into the intervention (beyond asking participants to maintain consistency).

Finally, a control group would have strengthened the study.

In conclusion, the present study demonstrated that 12 days of treadmill exercise training in

individuals with T2DM resulted in some improvements in glycaemic measures and reduced

global inflammation. Future research should consider the inclusion of a larger sample size with

the addition of suitable control group, appropriate dietary control, as well as a longer training

period to determine if a greater training effect is evident. Additionally, extending the training

period together with pre-diabetic and non-diabetic populations should be considered in the

future.

Page 110: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

110

Chapter 7 Thesis summary

The consequence of prolonged sedentary behaviour together with obesity leads to a state of

chronic low grade inflammation which is associated with the development of T2DM [455].

Obesity is associated with an increased pro-inflammatory environment as a result of the

infiltration of classically activated macrophages into the adipose tissue depots. These

inflammatory responses are known to moderate the diurnal glycaemic excursions and expedite

the development of T2DM, as well as increasing the risk of cardiovascular disease and all-

cause mortality [57, 58, 61]. Exercise in contrast, is known to improve the glycaemic responses

by increasing insulin sensitivity, and has been shown to affect the immune response both

acutely and following exercise training. Hence, a better understanding regarding the role of

exercise in managing glycaemic control and reducing chronic inflammation through exercise

is important. The overarching aim of this dissertation was two-fold: i) To assess the role of

exercise intensity on glycaemic control and the inflammatory response in individuals who

were overweight/obese; ii) To determine the impact of 12 consecutive days of exercise on

glycaemic control the inflammatory response both acutely following exercise and in response

to the short-term training period.

Chapter Four investigated the role of exercise intensity on glucose tolerance and inflammatory

markers involved in the IL-6 family. Comparison of exercise intensity was conducted using

continuous moderate-intensity (CME; 65% VO2peak) exercise and high-intensity intermittent

exercise (alternating bouts of high- and low-intensity exercise (HIIE); 100% and 50% VO2peak,

respectively). During each trial, glucose tolerance was assessed on three successive mornings

at the same time of day whilst the inflammatory markers were measured before and

immediately post-, 1 h post- and 25 h post-exercise (completed on second day). Our findings

suggested that HIIE did not result in any additional benefit on glucose tolerance when

compared to CME. This provided a unique contribution to the literature as previous work

comparing between HIIE and CME have been limited by not matching for exercise duration

and work output in overweight/obese males. With this in mind, appropriate interpretation of

inflammatory data can be made comparing between the two exercise interventions. When the

data were pooled, acute exercise (both interventions combined) was effective in reducing

glucose AUC, although to a lesser extent than expected. However, this beneficial effect had

disappeared 25 h post-exercise highlighting the importance of regular physical activity on

managing glycaemic control. The release of muscle-derived IL-6 following acute exercise can

promote glucose uptake [277, 295], with greater IL-6 concentration observed following HIIE

than CME [364]. Although we did not find differences between interventions, IL-6 was

Page 111: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

111

elevated immediately and 1 h post-exercise concurrent with a decrease in soluble receptors

sgp130 and sIL-6R. Thus, the increase in IL-6/IL-6R ratio is consistent with ‘classical

signalling’ which is postulated to contribute to improved glucose tolerance [369].

Adding to the findings in Chapter Four, Chapter Five assessed the response of acute exercise

(CME vs HIIE) in T2DM related markers; matrix metalloproteinase (MMP)-2 and MMP-3, as

well as OPN and adiponectin. These markers were assessed pre-exercise and immediately

post-, 1 h post- and 25 h post-exercise. Glycaemic control under free-living conditions was

also assessed for 72 h via the use of a continuous glucose monitoring system. The use of this

system provided a unique insight into glucose concentrations between CME and HIIE

measured in 5-minute intervals in overweight/obese males. Despite previous work

demonstrating improvements in glucose control throughout the day following acute exercise

[334] in T2DM, neither acute HIIE or CME was sufficient in improving glycaemic control

measured throughout the oral glucose tolerance tests and glucose profile across the day.

Measurement of the inflammatory markers revealed that MMP-2 and adiponectin were not

affected with acute exercise. However, MMP-3 and OPN were acutely increased immediately

post exercise which is consistent with a protective effect of T2DM, although this did not persist

and returned to baseline levels the next day. Although these findings only apply to overweight

individuals, further work is required to determine the response of these markers across

different levels of impaired glucose tolerance, including individuals with T2DM.

Based on the findings from Chapters Four and Five in overweight/obese individuals at risk of

T2DM, Chapter Six focused on the clinical impact exercise training may have on the

combination of glycaemic control and immune-related changes. Specifically, individuals

previously diagnosed with T2DM completed 12 consecutive days of treadmill exercise within

a two-week period. Inflammatory cytokine (unstimulated and LPS-stimulated cytokine)

responses to the first and last (12th) training session were assessed, in addition to measurement

of regulatory T cells via flow cytometry. Markers of glycaemia as well as global inflammation

marker, CRP were also assessed. Although glucose and insulin area under the curve did not

improve post training, fructosamine, an alternative marker to HbA1c demonstrated a

significant reduction highlighting improved glycaemic control following a short-term training

period. Furthermore, a significant reduction in CRP was found following the training period.

This is an important finding as CRP is a risk marker associated with the development of T2DM

[11, 12] in addition to cardiovascular disease [221]. An increase in IP-10, TNFα, IL-1β and

IL-10 were also observed following the last training session. These changes suggest an

accumulative training effect leading to a greater inflammatory response although we expected

a reduction in the pro-inflammatory cytokines and an increase in anti-inflammatory markers

following training. The release of IL-10 in tandem with the pro-inflammatory cytokines is

Page 112: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

112

necessary in order to maintain immune homeostasis which is known for its suppressive ability

in dampening the pro-inflammatory response. Despite the cytokine response, exercise training

did not induce any regulatory T cell responses in contrast to previous research which have

reported increased number of Tregs acutely post-exercise [309] as well as following a training

period [310, 311]. An increase in Tregs as reported in previous research would offer a sensible

explanation to the increase in IL-10 post exercise [296]. Indeed, previous long term (≥ 12

weeks) training studies have reported improved glycaemic control (HbA1c) and reduced

inflammation [29, 235, 284, 456, 457]. The findings in this chapter support a beneficial role

of exercise training in reducing inflammation and improving glycaemic control which are

detrimental to T2DM.

In summary, the results from this thesis indicated that an acute bout of HIIE in

overweight/obese males does not confer additional benefits to CME on glycaemic control.

Although an improvement in glucose AUC was evident immediately following exercise when

data were pooled, the clinical significance of the improvement was relatively small and did

not persist 25 h after exercise. This was also reflected in the lack of improvement in daily

glycaemic control measured by continuous glucose monitoring system. Together with

improved glucose AUC, we found a positive association between glucose AUC and IL-6 post

exercise, accompanied by an increase in IL-6/IL-6R ratio. However, this effect was transient

and returned to baseline 25 h post exercise. This trend was also consistent in the additional

inflammatory markers related to T2DM, MMP-3 and OPN where its protective effect was

acute and did not persist the following day. With this in mind, we investigated the role of short-

term training in previously diagnosed type 2 diabetics to further determine the acute

inflammatory response as well as the impact of exercise training. A significant increase in pro-

and anti-inflammatory markers were evident following the last training session. Although the

measurement of these cytokines was limited to only pre- and immediately post-exercise, it is

plausible that the significant increase in IL-10 may have been crucial in dampening the pro-

inflammatory response. Evidence has shown that Tregs are essential in the production of IL-

10. Unfortunately, the increase in IL-10 was not concurrent with an increase in Tregs which

may have explained for the increased IL-10 concentration. Although interpretation of the acute

inflammatory response is limited, exercise training was indeed effective in reducing CRP, a

global marker of inflammation which was also accompanied with a significant improvement

in fructosamine. Interpretation of these findings were further limited by a restricted selection

of inflammatory markers, where a composite score of a large number of related markers may

be more informative. Furthermore, the inflammatory markers measured in this thesis were

mainly of systemic origin rather than its tissue-specific source. Although systemic markers

may present a ‘snapshot’ of the downstream inflammatory response, cytokine assessment from

Page 113: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

113

adipose tissue and/or skeletal muscle may provide improved interpretation on the markers of

interest to further understand the inflammatory response and its relationship with glycaemic

control.

7.1 Future research directions

The mixed findings in glycaemic control in Chapter Four and Chapter Five pose further

questions on the role acute exercise has on blood glucose management. These chapters

provided clarity such that high-intensity intermittent exercise did not yield greater

improvements in glycaemic control following acute exercise when compared to moderate-

intensity exercise. To the author’s knowledge, these chapters were the first to compare

between two exercise intensities that were matched for exercise duration and workload.

However, a control group was not included which would have further strengthened the studies

in comparing glucose tolerance acutely, as well as the glycaemic changes over the 72 h period.

Whilst insulin and c-peptide were measured, limitations to understanding the complete picture

of glucose metabolism was apparent as we did not measure a number of other substrates and

hormones including free fatty acids, glucagon and epinephrine. Measurement of these analytes

may provide greater clarity on the metabolic and hormonal balance where glucose tolerance

was improved immediately post-exercise, but not 25 h following exercise. Additionally,

frequent sampling of blood markers over a longer period of time (up to 4 hours) post exercise

and additional methods such as isotope-labelled tracer technique may allow improved

interpretation on the acute effects of exercise on glucose disposal.

Chapter Six investigated the effect of 12 days of consecutive training on glycaemic control

and the inflammatory response following acute exercise and following the training period.

Chronic inflammation and type 2 diabetes is an area of research that requires a greater

understanding of how exercise may be beneficial in both reducing the level of inflammation

together with the aim of improving glycaemic control. Chapter Six combined the measures of

regulatory T cells, together with the inflammatory cytokine milieu and metabolic markers pre-

and post-exercise to assess if either acute exercise and/or exercise training modulated these

variables. Despite the measurement of these variables which attempt to clarify the relationship

of exercise, inflammation and glycaemic control, the lack of a control group or another

comparison group further limits the strength of the interpretation of the changes or lack of

changes observed. Although the reduction in CRP and fructosamine were significant, there

were no improvements in glucose tolerance post training which may have been the

consequence of a small sample size. This should be a significant consideration for future

research together with the addition of a control group to strengthen the study. Additionally,

distinguishing differences of exercise intensity within such strict training period is required to

Page 114: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

114

better understand the role of exercise intensity on glycaemic control and adaptations to the

immune system. Although this thesis had primarily measured immune markers from blood,

the assessment of the inflammatory response from key sources such as adipose tissue and

skeletal muscle may lead to a better understanding on the regulation of the inflammatory

response, both locally in the tissue and following a period of exercise. These differences may

further disentangle the anti-inflammatory effect of exercise on adipose-tissue derived

inflammation.

Bibliography

1. Tanamas, S.K. and I. Baker, AusDiab 2012: The Australian Diabetes, Obesity and Lifestyle Study. 2013: Baker IDI Heart and Diabetes Institute.

2. Venables, M.C. and A.E. Jeukendrup, Physical inactivity and obesity: links with insulin resistance and type 2 diabetes mellitus. Diabetes/metabolism research and reviews, 2009. 25(S1): p. S18-S23.

3. Boden, G., et al., Mechanisms of fatty acid-induced inhibition of glucose uptake. The Journal of clinical investigation, 1994. 93(6): p. 2438-2446.

4. Saltiel, A.R. and C.R. Kahn, Insulin signalling and the regulation of glucose and lipid metabolism. Nature, 2001. 414(6865): p. 799.

5. Reaven, G.M., Role of insulin resistance in human disease. Diabetes, 1988. 37(12): p. 1595-1607.

6. Feuerer, M., et al., Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nature medicine, 2009. 15(8): p. 930-939.

7. Weisberg, S.P., et al., Obesity is associated with macrophage accumulation in adipose tissue. Journal of clinical investigation, 2003. 112(12): p. 1796-1808.

8. Shu, C.J., C. Benoist, and D. Mathis. The immune system's involvement in obesity-driven type 2 diabetes. in Seminars in immunology. 2012. Elsevier.

9. Lazar, M.A., How obesity causes diabetes: Not a tall tale. Science, 2005. 307(5708): p. 373-375.

10. Bassuk, S.S., N. Rifai, and P.M. Ridker, High-sensitivity C-reactive protein: clinical importance. Current problems in cardiology, 2004. 29(8): p. 439-493.

11. Pickup, J. and M. Crook, Is type II diabetes mellitus a disease of the innate immune system? Diabetologia, 1998. 41(10): p. 1241-1248.

12. Pradhan, A.D., et al., C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA: the journal of the American Medical Association, 2001. 286(3): p. 327-334.

Page 115: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

115

13. Spranger, J., et al., Inflammatory cytokines and the risk to develop type 2 diabetes results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes, 2003. 52(3): p. 812-817.

14. Hotamisligil, G.S., N.S. Shargill, and B.M. Spiegelman, Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science (New York, NY), 1993. 259(5091): p. 87.

15. Hotamisligil, G., et al., IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha-and obesity-induced insulin resistance. Science (New York, NY), 1996. 271(5249): p. 665-668.

16. Olson, A.L. and J.E. Pessin, Structure, function, and regulation of the mammalian facilitative glucose transporter gene family. Annual review of nutrition, 1996. 16(1): p. 235-256.

17. Blüher, M., et al., Association of interleukin-6, C-reactive protein, interleukin-10 and adiponectin plasma concentrations with measures of obesity, insulin sensitivity and glucose metabolism. Exp Clin Endocrinol Diabetes, 2005. 113(09): p. 534-537.

18. Fernandez-Real, J.-M., et al., Circulating interleukin 6 levels, blood pressure, and insulin sensitivity in apparently healthy men and women. The Journal of Clinical Endocrinology & Metabolism, 2001. 86(3): p. 1154-1159.

19. Yuan, G., et al., Serum CRP levels are equally elevated in newly diagnosed type 2 diabetes and impaired glucose tolerance and related to adiponectin levels and insulin sensitivity. Diabetes research and clinical practice, 2006. 72(3): p. 244-250.

20. Daniele, G., et al., The inflammatory status score including IL-6, TNF-α, osteopontin, fractalkine, MCP-1 and adiponectin underlies whole-body insulin resistance and hyperglycemia in type 2 diabetes mellitus. Acta diabetologica, 2014. 51(1): p. 123-131.

21. Derosa, G., et al., Evaluation of metalloproteinase 2 and 9 levels and their inhibitors in diabetic and healthy subjects. Diabetes Metab, 2007. 33(2): p. 129-134.

22. Derosa, G., et al., Matrix metalloproteinase-2 and-9 levels in obese patients. Endothelium, 2008. 15(4): p. 219-224.

23. Goncalves, I., et al., Elevated plasma levels of MMP-12 are associated with atherosclerotic burden and symptomatic cardiovascular disease in subjects with type 2 diabetes. Arterioscler Thromb Vasc Biol, 2015: p. 1723-1731.

24. Kahles, F., H.M. Findeisen, and D. Bruemmer, Osteopontin: A novel regulator at the cross roads of inflammation, obesity and diabetes. Molecular metabolism, 2014. 3(4): p. 384-393.

25. Pirola, L. and J.C. Ferraz, Role of pro- and anti-inflammatory phenomena in the physiopathology of type 2 diabetes and obesity. World J Biol Chem, 2017. 8(2): p. 120-128.

26. Wang, J., et al., Effect of exercise training intensity on murine T‐regulatory cells and vaccination response. Scandinavian journal of medicine & science in sports, 2012. 22(5): p. 643-652.

27. Boulé, N.G., et al., Effects of exercise on glycemic control and body mass in type 2 diabetes mellitus: a meta-analysis of controlled clinical trials. Jama, 2001. 286(10): p. 1218-1227.

28. Umpierre, D., et al., Volume of supervised exercise training impacts glycaemic control in patients with type 2 diabetes: a systematic review with meta-regression analysis. Diabetologia, 2013. 56(2): p. 242-251.

29. Kadoglou, N.P., et al., The anti-inflammatory effects of exercise training in patients with type 2 diabetes mellitus. European Journal of Cardiovascular Prevention & Rehabilitation, 2007. 14(6): p. 837-843.

Page 116: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

116

30. Bartlett, J.D., et al., High-intensity interval running is perceived to be more enjoyable than moderate-intensity continuous exercise: implications for exercise adherence. Journal of sports sciences, 2011. 29(6): p. 547-553.

31. Tjønna, A.E., et al., Aerobic interval training versus continuous moderate exercise as a treatment for the metabolic syndrome. Circulation, 2008. 118(4): p. 346-354.

32. Gillen, J.B., et al., Acute high‐intensity interval exercise reduces the postprandial glucose response and prevalence of hyperglycaemia in patients with type 2 diabetes. Diabetes, Obesity and Metabolism, 2012.

33. Little, J.P., et al., Low-volume high-intensity interval training reduces hyperglycemia and increases muscle mitochondrial capacity in patients with type 2 diabetes. Journal of Applied Physiology, 2011. 111(6): p. 1554-1560.

34. Babraj, J., et al., Extremely short duration high intensity interval training substantially improves insulin action in young healthy males. BMC Endocrine Disorders, 2009. 9(1): p. 3.

35. Brestoff, J.R.B.J., et al., An acute bout of endurance exercise but not sprint interval exercise enhances insulin sensitivity. Applied Physiology, Nutrition, and Metabolism, 2009. 34(1): p. 25-32.

36. Adams, O.P., The impact of brief high-intensity exercise on blood glucose levels. Diabetes, metabolic syndrome and obesity: targets and therapy, 2013. 6: p. 113.

37. Whyte, L.J., et al., Effects of single bout of very high-intensity exercise on metabolic health biomarkers in overweight/obese sedentary men. Metabolism, 2013. 62(2): p. 212-219.

38. Whyte, L.J., J.M. Gill, and A.J. Cathcart, Effect of 2 weeks of sprint interval training on health-related outcomes in sedentary overweight/obese men. Metabolism, 2010. 59(10): p. 1421-1428.

39. Arciero, P.J., et al., Comparison of short-term diet and exercise on insulin action in individuals with abnormal glucose tolerance. Journal of Applied Physiology, 1999. 86(6): p. 1930-1935.

40. Mikus, C., et al., Glycaemic control is improved by 7 days of aerobic exercise training in patients with type 2 diabetes. Diabetologia, 2012. 55(5): p. 1417-1423.

41. O’gorman, D., et al., Exercise training increases insulin-stimulated glucose disposal and GLUT4 (SLC2A4) protein content in patients with type 2 diabetes. Diabetologia, 2006. 49(12): p. 2983-2992.

42. Beavers, K.M., T.E. Brinkley, and B.J. Nicklas, Effect of exercise training on chronic inflammation. Clinica chimica acta, 2010. 411(11-12): p. 785-793.

43. Donges, C.E., R. Duffield, and E.J. Drinkwater, Effects of resistance or aerobic exercise training on interleukin-6, C-reactive protein, and body composition. Medicine and science in sports and exercise, 2010. 42(2): p. 304-313.

44. Balducci, S., et al., Anti-inflammatory effect of exercise training in subjects with type 2 diabetes and the metabolic syndrome is dependent on exercise modalities and independent of weight loss. Nutrition, Metabolism and Cardiovascular Diseases, 2010. 20(8): p. 608-617.

45. Cipolletta, D., et al., PPAR-[ggr] is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature, 2012. 486(7404): p. 549-553.

46. Pradhan, A.D., et al., C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA, , 2001. 286(3): p. 327-334.

47. Ostrowski, K., P. Schjerling, and B.K. Pedersen, Physical activity and plasma interleukin-6 in humans–effect of intensity of exercise. Eur. J. Appl. Physiol., 2000. 83(6): p. 512-515.

48. Visse, R. and H. Nagase, Matrix metalloproteinases and tissue inhibitors of metalloproteinases. Circulation research, 2003. 92(8): p. 827-839.

Page 117: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

117

49. Bouloumie, A., et al., Adipocyte produces matrix metalloproteinases 2 and 9 - Involvement in adipose differentiation. Diabetes, 2001. 50(9): p. 2080-2086.

50. Wu, Y.Y., et al., High-fat diet-induced obesity regulates MMP3 to modulate depot- and sex-dependent adipose expansion in C57BL/6J mice. Am J Physiol Endocrinol Metab, 2017. 312(1): p. E58-E71.

51. Lund, S.A., et al., Osteopontin mediates macrophage chemotaxis via alpha4 and alpha9 integrins and survival via the alpha4 integrin. J Cell Biochem, 2013. 114(5): p. 1194-202.

52. Philip, S., A. Bulbule, and G.C. Kundu, Osteopontin stimulates tumor growth and activation of promatrix metalloproteinase-2 through nuclear factor-κB-mediated induction of membrane type 1 matrix metalloproteinase in murine melanoma cells. Journal of Biological Chemistry, 2001. 276(48): p. 44926-44935.

53. Baker, J.R., et al., Clinical usefulness of estimation of serum fructosamine concentration as a screening test for diabetes mellitus. Br Med J (Clin Res Ed), 1983. 287(6396): p. 863-867.

54. Sakaguchi, S., et al., Regulatory T cells and immune tolerance. Cell, 2008. 133(5): p. 775-787.

55. Eller, K., et al., Potential role of regulatory T cells in reversing obesity-linked insulin resistance and diabetic nephropathy. Diabetes, 2011. 60(11): p. 2954-2962.

56. Ilan, Y., et al., Induction of regulatory T cells decreases adipose inflammation and alleviates insulin resistance in ob/ob mice. Proceedings of the National Academy of Sciences, 2010. 107(21): p. 9765-9770.

57. Bassuk, S.S. and J.E. Manson, Epidemiological evidence for the role of physical activity in reducing risk of type 2 diabetes and cardiovascular disease. Journal of applied physiology, 2005. 99(3): p. 1193-1204.

58. Gayda, M., et al., Long-term cardiac rehabilitation and exercise training programs improve metabolic parameters in metabolic syndrome patients with and without coronary heart disease. Nutrition, Metabolism and Cardiovascular Diseases, 2008. 18(2): p. 142-151.

59. Ismail, N., et al., Renal disease and hypertension in non–insulin-dependent diabetes mellitus. Kidney international, 1999. 55(1): p. 1-28.

60. Reiber, G.E., E.J. Boyko, and D.G. Smith, Lower extremity foot ulcers and amputations in diabetes. Diabetes in America, 1995. 2: p. 409-27.

61. Laukkanen, J.A., et al., Impaired fasting plasma glucose and type 2 diabetes are related to the risk of out-of-hospital sudden cardiac death and all-cause mortality. Diabetes care, 2013. 36(5): p. 1166-1171.

62. Organization, W.H. 10 facts about diabetes. 2014 November 2014 [cited Accessed 10th May 2015; Available from: http://www.who.int/features/factfiles/diabetes/en/.

63. Tsukui, S., et al., Moderate-intensity regular exercise decreases serum tumor necrosis factor-alpha and HbA1c levels in healthy women. International journal of obesity and related metabolic disorders: journal of the International Association for the Study of Obesity, 2000. 24(9): p. 1207-1211.

64. Thomas, D., E.J. Elliott, and G.A. Naughton, Exercise for type 2 diabetes mellitus. Cochrane Database Syst Rev, 2006. 3(3).

65. Umpierre, D., et al., Physical Activity Advice Only or Structured Exercise Training and Association With HbA(1c) Levels in Type 2 Diabetes A Systematic Review and Meta-analysis. Jama-Journal of the American Medical Association, 2011. 305(17): p. 1790-1799.

66. Metcalfe, R.S., et al., Towards the minimal amount of exercise for improving metabolic health: beneficial effects of reduced-exertion high-intensity interval training. European journal of applied physiology, 2012. 112(7): p. 2767-2775.

Page 118: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

118

67. Richards, J.C., et al., Short‐term sprint interval training increases insulin sensitivity in healthy adults but does not affect the thermogenic response to β‐adrenergic stimulation. The Journal of physiology, 2010. 588(15): p. 2961-2972.

68. Trapp, E., et al., The effects of high-intensity intermittent exercise training on fat loss and fasting insulin levels of young women. International journal of obesity, 2008. 32(4): p. 684-691.

69. MacLeod, S., et al., Exercise lowers postprandial glucose but not fasting glucose in type 2 diabetes: a meta‐analysis of studies using continuous glucose monitoring. Diabetes/metabolism research and reviews, 2013. 29(8): p. 593-603.

70. Douen, A., et al., Exercise induces recruitment of the" insulin-responsive glucose transporter". Evidence for distinct intracellular insulin-and exercise-recruitable transporter pools in skeletal muscle. Journal of Biological Chemistry, 1990. 265(23): p. 13427-13430.

71. Holloszy, J. and P. Hansen, Regulation of glucose transport into skeletal muscle, in Reviews of Physiology Biochemistry and Pharmacology, Volume 128. 1996, Springer. p. 99-193.

72. Garvey, W., et al., Pretranslational suppression of a glucose transporter protein causes insulin resistance in adipocytes from patients with non-insulin-dependent diabetes mellitus and obesity. Journal of Clinical Investigation, 1991. 87(3): p. 1072.

73. Hussey, S., et al., Exercise training increases adipose tissue GLUT4 expression in patients with type 2 diabetes. Diabetes, Obesity and Metabolism, 2011. 13(10): p. 959-962.

74. Coderre, L., et al., Identification and characterization of an exercise-sensitive pool of glucose transporters in skeletal muscle. Journal of Biological Chemistry, 1995. 270(46): p. 27584-27588.

75. Wardzala, L.J. and B. Jeanrenaud, Potential mechanism of insulin action on glucose transport in the isolated rat diaphragm. Apparent translocation of intracellular transport units to the plasma membrane. J Biol Chem, 1981. 256(14): p. 7090-3.

76. Brozinick, J., et al., The effects of muscle contraction and insulin on glucose-transporter translocation in rat skeletal muscle. Biochem. J, 1994. 297: p. 539-545.

77. Sternlicht, E., R.J. Barnard, and G.K. Grimditch, Exercise and insulin stimulate skeletal muscle glucose transport through different mechanisms. Am J Physiol, 1989. 256(2 Pt 1): p. E227-30.

78. Goodyear, L.J., et al., Contractile activity increases plasma membrane glucose transporters in absence of insulin. Am J Physiol, 1990. 258(4 Pt 1): p. E667-72.

79. Hirshman, M.F., et al., Identification of an intracellular pool of glucose transporters from basal and insulin-stimulated rat skeletal muscle. J Biol Chem, 1990. 265(2): p. 987-91.

80. Goodyear, L.J., et al., Skeletal muscle plasma membrane glucose transport and glucose transporters after exercise. J Appl Physiol (1985), 1990. 68(1): p. 193-8.

81. Govers, R., A.C. Coster, and D.E. James, Insulin increases cell surface GLUT4 levels by dose dependently discharging GLUT4 into a cell surface recycling pathway. Mol Cell Biol, 2004. 24(14): p. 6456-66.

82. Lund, S., et al., Contraction stimulates translocation of glucose transporter GLUT4 in skeletal muscle through a mechanism distinct from that of insulin. Proceedings of the National Academy of Sciences, 1995. 92(13): p. 5817-5821.

83. Henriksen, E.J., et al., Glucose Transporter Protein-Content and Glucose-Transport Capacity in Rat Skeletal-Muscles. American Journal of Physiology, 1990. 259(4): p. E593-E598.

84. Ren, J.-M., et al., Exercise induces rapid increases in GLUT4 expression, glucose transport capacity, and insulin-stimulated glycogen storage in muscle. Journal of Biological Chemistry, 1994. 269(20): p. 14396-14401.

Page 119: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

119

85. Cartee, G.D., et al., Stimulation of Glucose-Transport in Skeletal-Muscle by Hypoxia. Journal of Applied Physiology, 1991. 70(4): p. 1593-1600.

86. Constable, S.H., et al., Muscle glucose transport: interactions of in vitro contractions, insulin, and exercise. J Appl Physiol, 1988. 64(6): p. 2329-32.

87. Ahlborg, G., et al., Substrate turnover during prolonged exercise in man: splanchnic and leg metabolism of glucose, free fatty acids, and amino acids. Journal of Clinical Investigation, 1974. 53(4): p. 1080.

88. Nesher, R., I.E. Karl, and D.M. Kipnis, Dissociation of effects of insulin and contraction on glucose transport in rat epitrochlearis muscle. Am J Physiol, 1985. 249(3 Pt 1): p. C226-C232.

89. Gao, J., E. Gulve, and J. Holloszy, Contraction-induced increase in muscle insulin sensitivity: requirement for a serum factor. American Journal of Physiology-Endocrinology And Metabolism, 1994. 266(2): p. E186-E192.

90. Gao, J., et al., Additive effect of contractions and insulin on GLUT-4 translocation into the sarcolemma. Journal of Applied Physiology, 1994. 77(4): p. 1597-1601.

91. Garetto, L.P., et al., Enhanced muscle glucose metabolism after exercise in the rat: the two phases. American Journal of Physiology-Endocrinology And Metabolism, 1984. 246(6): p. E471-E475.

92. Hansen, P.A., et al., Increased GLUT-4 translocation mediates enhanced insulin sensitivity of muscle glucose transport after exercise. Journal of Applied Physiology, 1998. 85(4): p. 1218-1222.

93. Richter, E.A., et al., Contraction-associated translocation of protein kinase C in rat skeletal muscle. FEBS letters, 1987. 217(2): p. 232-236.

94. Young, D.A., et al., Reversal of the exercise-induced increase in muscle permeability to glucose. Am J Physiol, 1987. 253(4 Pt 1): p. E331-E335.

95. Holloszy, J.O. and H. Narahara, Enhanced Permeability to Sugar Associated with Muscle Contraction Studies of the role of Ca++. The Journal of general physiology, 1967. 50(3): p. 551-562.

96. Kjaer, M., et al., Glucoregulation and hormonal responses to maximal exercise in non-insulin-dependent diabetes. Journal of Applied Physiology, 1990. 68(5): p. 2067-2074.

97. Holloszy, J.O. and H. Narahara, Studies of tissue permeability X. Changes in permeability to 3-methylglucose associated with contraction of isolated frog muscle. Journal of Biological Chemistry, 1965. 240(9): p. 3493-3500.

98. Wallberghenriksson, H., et al., Glucose-Transport into Rat Skeletal-Muscle - Interaction between Exercise and Insulin. Journal of Applied Physiology, 1988. 65(2): p. 909-913.

99. Richter, E.A., et al., Muscle glucose metabolism following exercise in the rat: increased sensitivity to insulin. Journal of Clinical Investigation, 1982. 69(4): p. 785.

100. Holloszy, J.O., Exercise-induced increase in muscle insulin sensitivity. Journal of Applied Physiology, 2005. 99(1): p. 338-343.

101. Cartee, G.D., et al., Prolonged increase in insulin-stimulated glucose transport in muscle after exercise. Am J Physiol Endocrinol Metab, 1989. 256: p. E494-E499.

102. Gulve, E.A., et al., Reversal of enhanced muscle glucose transport after exercise: roles of insulin and glucose. American Journal of Physiology-Endocrinology And Metabolism, 1990. 259(5): p. E685-E691.

103. Young, J.C., et al., Carbohydrate feeding speeds reversal of enhanced glucose uptake in muscle after exercise. Am J Physiol, 1983. 245(5 Pt 1): p. R684-8.

104. Cartee, G.D. and J.O. Holloszy, Exercise Increases Susceptibility of Muscle Glucose-Transport to Activation by Various Stimuli. American Journal of Physiology, 1990. 258(2): p. E390-E393.

105. Zinker, B.A., et al., Regulation of glucose uptake and metabolism by working muscle: an in vivo analysis. Diabetes, 1993. 42(7): p. 956-965.

Page 120: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

120

106. Zinker, B.A., et al., Role of glucose and insulin loads to the exercising limb in increasing glucose uptake and metabolism. Journal of Applied Physiology, 1993. 74: p. 2915-2915.

107. Roussel, R., et al., 13C/31P NMR studies of glucose transport in human skeletal muscle. Proc Natl Acad Sci U S A, 1998. 95(3): p. 1313-8.

108. Wahren, J., et al., Glucose metabolism during leg exercise in man. Journal of Clinical Investigation, 1971. 50(12): p. 2715.

109. Bertoldo, A., et al., Quantitative assessment of glucose transport in human skeletal muscle: dynamic positron emission tomography imaging of [O-methyl-11C]3-O-methyl-D-glucose. J Clin Endocrinol Metab, 2005. 90(3): p. 1752-9.

110. Roden, M. and G.I. Shulman, Applications of NMR spectroscopy to study muscle glycogen metabolism in man. Annu Rev Med, 1999. 50: p. 277-90.

111. Katz, A., et al., Leg glucose uptake during maximal dynamic exercise in humans. American Journal of Physiology-Endocrinology And Metabolism, 1986. 251(1): p. E65-E70.

112. Wahren, J., P. Felig, and L. Hagenfeldt, Physical exercise and fuel homeostasis in diabetes mellitus. Diabetologia, 1978. 14(4): p. 213-222.

113. Ploug, T., H. Galbo, and E.A. Richter, Increased muscle glucose uptake during contractions: no need for insulin. Am J Physiol, 1984. 247(6 Pt 1): p. E726-E731.

114. Heinonen, I., et al., Increasing exercise intensity reduces heterogeneity of glucose uptake in human skeletal muscles. PLoS One, 2012. 7(12): p. e52191.

115. Kang, J., et al., Effect of exercise intensity on glucose and insulin metabolism in obese individuals and obese NIDDM patients. Diabetes care, 1996. 19(4): p. 341-349.

116. Ferrannini, E., et al., Effect of fatty acids on glucose production and utilization in man. Journal of Clinical Investigation, 1983. 72(5): p. 1737.

117. Marliss, E.B., et al., Glucoregulatory and hormonal responses to repeated bouts of intense exercise in normal male subjects. Journal of Applied Physiology, 1991. 71(3): p. 924-933.

118. Marliss, E.B. and M. Vranic, Intense exercise has unique effects on both insulin release and its roles in glucoregulation implications for diabetes. Diabetes, 2002. 51(suppl 1): p. S271-S283.

119. Sigal, R.J., et al., Glucoregulation during and after intense exercise: effects of α-adrenergic blockade. Metabolism, 2000. 49(3): p. 386-394.

120. Mikines, K.J., et al., Effect of physical exercise on sensitivity and responsiveness to insulin in humans. American Journal of Physiology-Endocrinology And Metabolism, 1988. 254(3): p. E248-E259.

121. Baynard, T., et al., Effect of a single vs multiple bouts of exercise on glucose control in women with type 2 diabetes. Metabolism, 2005. 54(8): p. 989-994.

122. Larsen, J., et al., The effect of moderate exercise on postprandial glucose homeostasis in NIDDM patients. Diabetologia, 1997. 40(4): p. 447-453.

123. Roberts, S., et al., Glycemic response to carbohydrate and the effects of exercise and protein. Nutrition, 2013. 29(6): p. 881-885.

124. Venables, M.C., et al., Effect of acute exercise on glucose tolerance following post-exercise feeding. European journal of applied physiology, 2007. 100(6): p. 711-717.

125. Araujo-Vilar, D., et al., Influence of moderate physical exercise on insulin-mediated and non—insulin-mediated glucose uptake in healthy subjects. Metabolism, 1997. 46(2): p. 203-209.

126. Bonen, A., M. Ball-Burnett, and C. Russel, Glucose tolerance is improved after low-and high-intensity exercise in middle-age men and women. Canadian journal of applied physiology, 1998. 23(6): p. 583-593.

127. Brun, J., et al., Influence of short-term submaximal exercise on parameters of glucose assimilation analyzed with the minimal model. Metabolism, 1995. 44(7): p. 833-840.

Page 121: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

121

128. Oberlin, D.J., et al., One bout of exercise alters free-living postprandial glycemia in type 2 diabetes. Medicine and science in sports and exercise, 2014. 46(2): p. 232.

129. Rynders, C.A., et al., Effects of exercise intensity on postprandial improvement in glucose disposal and insulin sensitivity in prediabetic adults. J Clin Endocrinol Metab, 2014. 99(1): p. 220-8.

130. King, D.S., et al., Time course for exercise-induced alterations in insulin action and glucose tolerance in middle-aged people. Journal of Applied Physiology, 1995. 78(1): p. 17-22.

131. O’Connor, A.M., et al., The gastroenteroinsular response to glucose ingestion during postexercise recovery. American Journal of Physiology-Endocrinology and Metabolism, 2006. 290(6): p. E1155-E1161.

132. Rose, A.J., et al., Effect of prior exercise on glucose metabolism in trained men. American Journal of Physiology-Endocrinology and Metabolism, 2001. 281(4): p. E766-E771.

133. Knudsen, S.H., et al., The immediate effects of a single bout of aerobic exercise on oral glucose tolerance across the glucose tolerance continuum. Physiological reports, 2014. 2(8): p. e12114.

134. Sylow, L., et al., Exercise-stimulated glucose uptake - regulation and implications for glycaemic control. Nat Rev Endocrinol, 2017. 13(3): p. 133-148.

135. Minuk, H.L., et al., Glucoregulatory and metabolic response to exercise in obese noninsulin-dependent diabetes. Am J Physiol, 1981. 240(5): p. E458-E464.

136. Cartee, G.D., Mechanisms for greater insulin-stimulated glucose uptake in normal and insulin-resistant skeletal muscle after acute exercise. American Journal of Physiology-Endocrinology and Metabolism, 2015. 309(12): p. E949-E959.

137. Trefts, E., A.S. Williams, and D.H. Wasserman, Exercise and the Regulation of Hepatic Metabolism. Prog Mol Biol Transl Sci, 2015. 135: p. 203-25.

138. Wasserman, D.H., Four grams of glucose. Am J Physiol Endocrinol Metab, 2009. 296(1): p. E11-21.

139. Kindermann, W., et al., Catecholamines, growth hormone, cortisol, insulin, and sex hormones in anaerobic and aerobic exercise. European journal of applied physiology and occupational physiology, 1982. 49(3): p. 389-399.

140. Eigler, N., L. Sacca, and R.S. Sherwin, Synergistic interactions of physiologic increments of glucagon, epinephrine, and cortisol in the dog: a model for stress-induced hyperglycemia. J Clin Invest, 1979. 63(1): p. 114-23.

141. Kjaer, M., Hepatic glucose production during exercise. Adv Exp Med Biol, 1998. 441: p. 117-27.

142. Kjaer, M., et al., Regulation of hepatic glucose production during exercise in humans: role of sympathoadrenergic activity. Am J Physiol, 1993. 265(2 Pt 1): p. E275-83.

143. Hirsch, I., et al., Insulin and glucagon in prevention of hypoglycemia during exercise in humans. American Journal of Physiology-Endocrinology And Metabolism, 1991. 260(5): p. E695-E704.

144. Wasserman, D.H., et al., Glucagon is a primary controller of hepatic glycogenolysis and gluconeogenesis during muscular work. Am J Physiol, 1989. 257(1 Pt 1): p. E108-17.

145. Wasserman, D.H., et al., Hepatic nerves are not essential to the increase in hepatic glucose production during muscular work. Am J Physiol, 1990. 259(2 Pt 1): p. E195-203.

146. Coker, R.H., et al., Hepatic alpha- and beta-adrenergic receptors are not essential for the increase in R(a) during exercise in diabetes. Am J Physiol Endocrinol Metab, 2000. 278(3): p. E444-51.

147. Moates, J.M., et al., Metabolic role of the exercise-induced increment in epinephrine in the dog. Am J Physiol, 1988. 255(4 Pt 1): p. E428-36.

Page 122: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

122

148. Sherwin, R.S., et al., Epinephrine and the regulation of glucose metabolism: effect of diabetes and hormonal interactions. Metabolism, 1980. 29(11 Suppl 1): p. 1146-54.

149. Han, X.-X. and A. Bonen, Epinephrine translocates GLUT-4 but inhibits insulin-stimulated glucose transport in rat muscle. American Journal of Physiology-Endocrinology And Metabolism, 1998. 274(4): p. E700-E707.

150. Williams, R.S., M.G. Caron, and K. Daniel, Skeletal muscle beta-adrenergic receptors: variations due to fiber type and training. American Journal of Physiology-Endocrinology And Metabolism, 1984. 246(2): p. E160-E167.

151. Hunt, D.G. and J.L. Ivy, Epinephrine inhibits insulin-stimulated muscle glucose transport. Journal of Applied Physiology, 2002. 93(5): p. 1638-1643.

152. Lager, I., et al., Studies on the insulin-antagonistic effect of catecholamines in normal man. Evidence for the importance of beta 2-receptors. Diabetologia, 1986. 29(7): p. 409-16.

153. Hartley, L.H., et al., Multiple hormonal responses to graded exercise in relation to physical training. Journal of Applied Physiology, 1972. 33(5): p. 602-606.

154. Rizza, R.A., L.J. Mandarino, and J.E. Gerich, Effects of growth hormone on insulin action in man: mechanisms of insulin resistance, impaired suppression of glucose production, and impaired stimulation of glucose utilization. Diabetes, 1982. 31(8): p. 663-669.

155. Schwarz, J.-M., et al., Effects of recombinant human growth hormone on hepatic lipid and carbohydrate metabolism in HIV-infected patients with fat accumulation. The Journal of Clinical Endocrinology & Metabolism, 2002. 87(2): p. 942-945.

156. Svensson, J., et al., Effects of seven years of GH-replacement therapy on insulin sensitivity in GH-deficient adults. The Journal of Clinical Endocrinology & Metabolism, 2002. 87(5): p. 2121-2127.

157. Kilgour, E., S. Baldwin, and D. Flint, Divergent regulation of rat adipocyte GLUT1 and GLUT4 glucose transporters by GH. Journal of endocrinology, 1995. 145(1): p. 27-33.

158. Marcus, C., et al., Effects of growth hormone on lipolysis in humans. Acta Paediatr Suppl, 1994. 406: p. 54-8; discussion 59.

159. Pontiroli, A.E., et al., Plasma free fatty acids and serum insulin in subjects feeding at 12-hour intervals; effects of methionyl growth hormone and of acipimox, an inhibitor of lipolysis. J Endocrinol Invest, 1992. 15(2): p. 85-91.

160. Sakharova, A.A., et al., Role of growth hormone in regulating lipolysis, proteolysis, and hepatic glucose production during fasting. J Clin Endocrinol Metab, 2008. 93(7): p. 2755-9.

161. Stefan, N., et al., Effect of the pattern of elevated free fatty acids on insulin sensitivity and insulin secretion in healthy humans. Horm Metab Res, 2001. 33(7): p. 432-8.

162. Segerlantz, M., et al., Inhibition of lipolysis during acute GH exposure increases insulin sensitivity in previously untreated GH-deficient adults. Eur J Endocrinol, 2003. 149(6): p. 511-9.

163. Randle, P., E. Newsholme, and P. Garland, Regulation of glucose uptake by muscle. 8. Effects of fatty acids, ketone bodies and pyruvate, and of alloxan-diabetes and starvation, on the uptake and metabolic fate of glucose in rat heart and diaphragm muscles. Biochemical Journal, 1964. 93(3): p. 652.

164. Garland, P., E. Newsholme, and P. Randle, Regulation of glucose uptake by muscle. 9. Effects of fatty acids and ketone bodies, and of alloxan-diabetes and starvation, on pyruvate metabolism and on lactate/pyruvate and l-glycerol 3-phosphate/dihydroxyacetone phosphate concentration ratios in rat heart and rat diaphragm muscles. Biochemical Journal, 1964. 93(3): p. 665.

165. Garland, P. and P. Randle, Regulation of glucose uptake by muscle. 10. Effects of alloxan-diabetes, starvation, hypophysectomy and adrenalectomy, and of fatty acids, ketone bodies and pyruvate, on the glycerol output and concentrations of free fatty

Page 123: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

123

acids, long-chain fatty acyl-coenzyme A, glycerol phosphate and citrate-cycle intermediates in rat-heart and diaphragm muscles. Biochemical Journal, 1964. 93(3): p. 678.

166. Rennie, M.J., W.W. Winder, and J. Holloszy, A sparing effect of increased plasma fatty acids on muscle and liver glycogen content in the exercising rat. Biochem. J, 1976. 156: p. 647-655.

167. Costill, D., et al., Effects of elevated plasma FFA and insulin on muscle glycogen usage during exercise. J Appl Physiol, 1977. 43(4): p. 695-699.

168. Kjaer, M., et al., Increased epinephrine response and inaccurate glucoregulation in exercising athletes. Journal of Applied Physiology, 1986. 61(5): p. 1693-1700.

169. Kotchen, T.A., et al., Renin, norepinephrine, and epinephrine responses to graded exercise. Journal of Applied Physiology, 1971. 31(2): p. 178-184.

170. Tidgren, B., et al., Renal neurohormonal and vascular responses to dynamic exercise in humans. Journal of Applied Physiology, 1991. 70(5): p. 2279-2286.

171. Goodwin, M.L., Blood glucose regulation during prolonged, submaximal, continuous exercise: a guide for clinicians. J Diabetes Sci Technol, 2010. 4(3): p. 694-705.

172. Coggan, A.R., Plasma glucose metabolism during exercise in humans. Sports Med, 1991. 11(2): p. 102-24.

173. Hargreaves, M., Carbohydrates and exercise. J Sports Sci, 1991. 9 Spec No: p. 17-28. 174. Hearris, M.A., et al., Regulation of Muscle Glycogen Metabolism during Exercise:

Implications for Endurance Performance and Training Adaptations. Nutrients, 2018. 10(3).

175. Jeukendrup, A.E., Nutrition for endurance sports: marathon, triathlon, and road cycling. J Sports Sci, 2011. 29 Suppl 1: p. S91-9.

176. Sahlin, K., Metabolic factors in fatigue. Sports Med, 1992. 13(2): p. 99-107. 177. Kraniou, G.N., D. Cameron-Smith, and M. Hargreaves, Acute exercise and GLUT4

expression in human skeletal muscle: influence of exercise intensity. Journal of applied physiology, 2006. 101(3): p. 934-937.

178. Houmard, J.A., et al., Effect of the volume and intensity of exercise training on insulin sensitivity. Journal of Applied Physiology, 2004. 96(1): p. 101-106.

179. Boulé, N., et al., Meta-analysis of the effect of structured exercise training on cardiorespiratory fitness in Type 2 diabetes mellitus. Diabetologia, 2003. 46(8): p. 1071-1081.

180. Little, J.P., et al., An acute bout of high-intensity interval training increases the nuclear abundance of PGC-1α and activates mitochondrial biogenesis in human skeletal muscle. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 2011. 300(6): p. R1303-R1310.

181. Burgomaster, K.A., et al., Divergent response of metabolite transport proteins in human skeletal muscle after sprint interval training and detraining. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 2007. 292(5): p. R1970-R1976.

182. Mikus, C.R., et al., Seven days of aerobic exercise training improves conduit artery blood flow following glucose ingestion in patients with type 2 diabetes. Journal of Applied Physiology, 2011. 111(3): p. 657-664.

183. Mikus, C.R., et al., Lowering physical activity impairs glycemic control in healthy volunteers. Medicine and science in sports and exercise, 2012. 44(2): p. 225.

184. Rogers, M.A., et al., Improvement in glucose tolerance after 1 wk of exercise in patients with mild NIDDM. Diabetes Care, 1988. 11(8): p. 613-618.

185. Skleryk, J., et al., Two weeks of reduced‐volume sprint interval or traditional exercise training does not improve metabolic functioning in sedentary obese men. Diabetes, Obesity and Metabolism, 2013. 15(12): p. 1146-1153.

Page 124: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

124

186. Babraj, J., et al., Extremely short duration high intensity interval training substantially improves insulin action in young healthy males. BMC. Endocr. Disord., 2009. 9(1): p. 3.

187. Whyte, L.J., J.M. Gill, and A.J. Cathcart, Effect of 2 weeks of sprint interval training on health-related outcomes in sedentary overweight/obese men. Metab. Clin. Exp., 2010. 59(10): p. 1421-1428.

188. Kirwan, J.P., et al., Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. American Journal of Physiology-Endocrinology And Metabolism, 2009. 297(1): p. E151-E156.

189. de Heredia, F.P., S. Gómez-Martínez, and A. Marcos, Obesity, inflammation and the immune system. Proceedings of the Nutrition Society, 2012. 71(2): p. 332-338.

190. Donath, M.Y. and S.E. Shoelson, Type 2 diabetes as an inflammatory disease. Nature Reviews Immunology, 2011. 11(2): p. 98.

191. Shoelson, S.E., J. Lee, and A.B. Goldfine, Inflammation and insulin resistance. Journal of clinical investigation, 2006. 116(7): p. 1793-1801.

192. Fox, C.S., et al., Abdominal visceral and subcutaneous adipose tissue compartments: association with metabolic risk factors in the Framingham Heart Study. Circulation, 2007. 116(1): p. 39-48.

193. Bigornia, S., et al., Relation of depot-specific adipose inflammation to insulin resistance in human obesity. Nutrition & diabetes, 2012. 2(3): p. e30.

194. Koster, A., et al., Body fat distribution and inflammation among obese older adults with and without metabolic syndrome. Obesity, 2010. 18(12): p. 2354-2361.

195. Xu, H., et al., Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. Journal of clinical investigation, 2003. 112(12): p. 1821.

196. Hurlimann, J., G. Thorbecke, and G. Hochwald, The liver as the site of C-reactive protein formation. Journal of Experimental Medicine, 1966. 123(2): p. 365-378.

197. Pickup, J.C., et al., Plasma interleukin-6, tumour necrosis factor α and blood cytokine production in type 2 diabetes. Life sciences, 2000. 67(3): p. 291-300.

198. Lumeng, C.N., J.L. Bodzin, and A.R. Saltiel, Obesity induces a phenotypic switch in adipose tissue macrophage polarization. Journal of Clinical Investigation, 2007. 117(1): p. 175.

199. Unoki, H., et al., Macrophages regulate tumor necrosis factor-α expression in adipocytes through the secretion of matrix metalloproteinase-3. International journal of obesity, 2008. 32(6): p. 902-911.

200. Ahmad, R., et al., Interaction of osteopontin with IL-18 in obese individuals: implications for insulin resistance. PloS one, 2013. 8(5): p. e63944.

201. Esposito, K., et al., Meal modulation of circulating interleukin 18 and adiponectin concentrations in healthy subjects and in patients with type 2 diabetes mellitus. The American journal of clinical nutrition, 2003. 78(6): p. 1135-1140.

202. Wagner, N.M., et al., Circulating regulatory T cells are reduced in obesity and may identify subjects at increased metabolic and cardiovascular risk. Obesity, 2013. 21(3): p. 461-468.

203. van Exel, E., et al., Low production capacity of interleukin-10 associates with the metabolic syndrome and type 2 diabetes. Diabetes, 2002. 51(4): p. 1088-1092.

204. Ostrowski, K., et al., Pro-and anti-inflammatory cytokine balance in strenuous exercise in humans. J. Physiol., 1999. 515(1): p. 287-291.

205. Christiansen, T., et al., Exercise training versus diet-induced weight-loss on metabolic risk factors and inflammatory markers in obese subjects: a 12-week randomized intervention study. American Journal of Physiology - Endocrinology And Metabolism, 2010. 298(4): p. E824-E831.

Page 125: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

125

206. Leggate, M., et al., Determination of inflammatory and prominent proteomic changes in plasma and adipose tissue after high-intensity intermittent training in overweight and obese males. Journal of Applied Physiology, 2012. 112(8): p. 1353-1360.

207. Mikus, C.R., et al., Lowering physical activity impairs glycemic control in healthy volunteers. Med Sci Sports Exerc, 2012. 44(2): p. 225.

208. Gomez-Ambrosi, J., et al., Plasma osteopontin levels and expression in adipose tissue are increased in obesity. The Journal of Clinical Endocrinology & Metabolism, 2007. 92(9): p. 3719-3727.

209. Ehses, J., et al., IL-1 antagonism reduces hyperglycemia and tissue inflammation in the type 2 diabetic GK rat. Proceedings of the National Academy of Sciences, 2009. 106(33): p. 13998-14003.

210. Werner, E.D., et al., Insulin resistance due to phosphorylation of insulin receptor substrate-1 at serine 302. Journal of Biological Chemistry, 2004. 279(34): p. 35298-35305.

211. Xi, L., et al., C‐reactive protein impairs hepatic insulin sensitivity and insulin signaling in rats: Role of mitogen‐activated protein kinases. Hepatology, 2011. 53(1): p. 127-135.

212. Eder, K., et al., The major inflammatory mediator interleukin-6 and obesity. Inflammation Research, 2009. 58(11): p. 727.

213. Morohoshi, M., et al., Glucose-dependent interleukin 6 and tumor necrosis factor production by human peripheral blood monocytes in vitro. Diabetes, 1996. 45(7): p. 954-959.

214. Esposito, K., et al., Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans. Circulation, 2002. 106(16): p. 2067-2072.

215. Mohamed-Ali, V., et al., Production of soluble tumor necrosis factor receptors by human subcutaneous adipose tissue in vivo. Am. J. Physiol. Endocrinol. Metab., 1999. 277(6): p. E971-E975.

216. Carey, A. and M. Febbraio, Interleukin-6 and insulin sensitivity: friend or foe? Diabetologia, 2004. 47(7): p. 1135-1142.

217. Bastard, J.-P., et al., Adipose tissue IL-6 content correlates with resistance to insulin activation of glucose uptake both in vivo and in vitro. The Journal of Clinical Endocrinology & Metabolism, 2002. 87(5): p. 2084-2089.

218. Yudkin, J.S., et al., C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arteriosclerosis, thrombosis, and vascular biology, 1999. 19(4): p. 972-978.

219. Pickup, J., et al., NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia, 1997. 40(11): p. 1286-1292.

220. Wellen, K.E. and G.S. Hotamisligil, Obesity-induced inflammatory changes in adipose tissue. J Clin Invest, 2003. 112(12): p. 1785.

221. Ridker, P.M., et al., C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. New England Journal of Medicine, 2000. 342(12): p. 836-843.

222. Hotamisligil, G.S., et al., Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. Journal of clinical investigation, 1995. 95(5): p. 2409.

223. Kern, P.A., et al., Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. American Journal of Physiology-Endocrinology And Metabolism, 2001. 280(5): p. E745-E751.

224. Saghizadeh, M., et al., The expression of TNF alpha by human muscle. Relationship to insulin resistance. Journal of Clinical Investigation, 1996. 97(4): p. 1111.

Page 126: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

126

225. Park, H.S., J.Y. Park, and R. Yu, Relationship of obesity and visceral adiposity with serum concentrations of CRP, TNF-α and IL-6. Diabetes research and clinical practice, 2005. 69(1): p. 29-35.

226. Rosen, O.M., et al., Phosphorylation activates the insulin receptor tyrosine protein kinase. Proceedings of the National Academy of Sciences, 1983. 80(11): p. 3237-3240.

227. Dandona, P., et al., Tumor necrosis factor-α in sera of obese patients: fall with weight loss. The Journal of Clinical Endocrinology & Metabolism, 1998. 83(8): p. 2907-2910.

228. Sims, J.E. and D.E. Smith, The IL-1 family: regulators of immunity. Nature Reviews Immunology, 2010. 10(2): p. 89.

229. Bendtzen, K., et al., Cytotoxicity of human pI 7 interleukin-1 for pancreatic islets of Langerhans. Science, 1986. 232(4757): p. 1545-1547.

230. Maedler, K., et al., Glucose-induced β cell production of IL-1β contributes to glucotoxicity in human pancreatic islets. The Journal of clinical investigation, 2002. 110(6): p. 851-860.

231. Dinarello, C.A., M.Y. Donath, and T. Mandrup-Poulsen, Role of IL-1β in type 2 diabetes. Current Opinion in Endocrinology, Diabetes and Obesity, 2010. 17(4): p. 314-321.

232. Maedler, K., et al., Leptin modulates β cell expression of IL-1 receptor antagonist and release of IL-1β in human islets. Proceedings of the National Academy of Sciences of the United States of America, 2004. 101(21): p. 8138-8143.

233. Cancello, R., et al., Increased infiltration of macrophages in omental adipose tissue is associated with marked hepatic lesions in morbid human obesity. Diabetes, 2006. 55(6): p. 1554-1561.

234. Kolak, M., et al., Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes, 2007. 56(8): p. 1960-1968.

235. Kadoglou, N., et al., Exercise ameliorates serum MMP-9 and TIMP-2 levels in patients with type 2 diabetes. Diabetes & metabolism, 2010. 36(2): p. 144-151.

236. van der Leeuw, J., et al., Novel biomarkers to improve the prediction of cardiovascular event risk in type 2 diabetes mellitus. Journal of the American Heart Association, 2016. 5(6): p. e003048.

237. Hansson, G.K., Inflammation, atherosclerosis, and coronary artery disease. New England Journal of Medicine, 2005. 352(16): p. 1685-1695.

238. Death, A.K., et al., High glucose alters matrix metalloproteinase expression in two key vascular cells: potential impact on atherosclerosis in diabetes. Atherosclerosis, 2003. 168(2): p. 263-269.

239. Wang, K.X. and D.T. Denhardt, Osteopontin: role in immune regulation and stress responses. Cytokine & growth factor reviews, 2008. 19(5-6): p. 333-345.

240. Giachelli, C.M., et al., Osteopontin is elevated during neointima formation in rat arteries and is a novel component of human atherosclerotic plaques. Journal of Clinical Investigation, 1993. 92(4): p. 1686.

241. Nomiyama, T., et al., Osteopontin mediates obesity-induced adipose tissue macrophage infiltration and insulin resistance in mice. J Clin Invest, 2007. 117(10): p. 2877.

242. Nakamachi, T., et al., PPARα agonists suppress osteopontin expression in macrophages and decrease plasma levels in patients with type 2 diabetes. Diabetes, 2007. 56(6): p. 1662-1670.

243. Sponder, M., et al., Physical inactivity increases endostatin and osteopontin in patients with coronary artery disease. Heart and vessels, 2016. 31(10): p. 1603-1608.

244. Shoelson, S.E., L. Herrero, and A. Naaz, Obesity, inflammation, and insulin resistance. Gastroenterology, 2007. 132(6): p. 2169-2180.

Page 127: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

127

245. Lindsay, R.S., et al., Adiponectin and development of type 2 diabetes in the Pima Indian population. The Lancet, 2002. 360(9326): p. 57-58.

246. Maeda, N., et al., Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nature medicine, 2002. 8(7): p. 731-737.

247. Stefan, N., et al., Plasma adiponectin concentration is associated with skeletal muscle insulin receptor tyrosine phosphorylation, and low plasma concentration precedes a decrease in whole-body insulin sensitivity in humans. Diabetes, 2002. 51(6): p. 1884-1888.

248. Ouchi, N., et al., Obesity, adiponectin and vascular inflammatory disease. Current opinion in lipidology, 2003. 14(6): p. 561-566.

249. Fernandez-Real, J.M., et al., Novel interactions of adiponectin with the endocrine system and inflammatory parameters. The Journal of Clinical Endocrinology & Metabolism, 2003. 88(6): p. 2714-2718.

250. Ouchi, N., et al., Adipokines in inflammation and metabolic disease. Nature Reviews Immunology, 2011. 11(2): p. 85-97.

251. Combs, T.P., et al., Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. Journal of Clinical Investigation, 2001. 108(12): p. 1875.

252. Yamauchi, T., et al., The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nature medicine, 2001. 7(8): p. 941-946.

253. Kumada, M., et al., Adiponectin specifically increased tissue inhibitor of metalloproteinase-1 through interleukin-10 expression in human macrophages. Circulation, 2004. 109(17): p. 2046-2049.

254. Wolf, A.M., et al., Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res Commun, 2004. 323(2): p. 630-635.

255. Maynard, C.L. and C.T. Weaver, Diversity in the contribution of interleukin‐10 to T‐cell‐mediated immune regulation. Immunological reviews, 2008. 226(1): p. 219-233.

256. Chernoff, A.E., et al., A randomized, controlled trial of IL-10 in humans. Inhibition of inflammatory cytokine production and immune responses. The Journal of Immunology, 1995. 154(10): p. 5492-5499.

257. de Vries, J.E., Immunosuppressive and anti-inflammatory properties of interleukin 10. Annals of medicine, 1995. 27(5): p. 537-541.

258. Cintra, D.E., et al., Interleukin-10 is a protective factor against diet-induced insulin resistance in liver. Journal of hepatology, 2008. 48(4): p. 628-637.

259. Hong, E.-G., et al., Interleukin-10 prevents diet-induced insulin resistance by attenuating macrophage and cytokine response in skeletal muscle. Diabetes, 2009. 58(11): p. 2525-2535.

260. Berg, D.J., et al., Interleukin-10 is a central regulator of the response to LPS in murine models of endotoxic shock and the Shwartzman reaction but not endotoxin tolerance. Journal of Clinical Investigation, 1995. 96(5): p. 2339.

261. Mallat, Z., et al., Protective role of interleukin-10 in atherosclerosis. Circulation research, 1999. 85(8): p. e17-e24.

262. Vieira, P.L., et al., IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+ CD25+ regulatory T cells. The Journal of Immunology, 2004. 172(10): p. 5986-5993.

263. Jagannathan-Bogdan, M., et al., Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes. The Journal of Immunology, 2011. 186(2): p. 1162-1172.

264. Winer, S., et al., Normalization of obesity-associated insulin resistance through immunotherapy. Nature medicine, 2009. 15(8): p. 921-929.

Page 128: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

128

265. Zeng, C., et al., The imbalance of Th17/Th1/Tregs in patients with type 2 diabetes: relationship with metabolic factors and complications. Journal of molecular medicine, 2012. 90(2): p. 175-186.

266. Han, J.M., et al., Insulin inhibits IL-10–mediated regulatory T cell function: Implications for obesity. The Journal of Immunology, 2014. 192(2): p. 623-629.

267. Hsu, P., et al., IL-10 potentiates differentiation of human induced regulatory T cells via STAT3 and Foxo1. The Journal of Immunology, 2015. 195(8): p. 3665-3674.

268. Knowler, W., E. Barrett-Connor, and S. Fowler, E.; Hamman R, F.; Lachin, JM; Walker, EA; Diabetes Prevention Program Research Group. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med, 2002. 346: p. 393-403.

269. Manson, J.E., et al., Walking compared with vigorous exercise for the prevention of cardiovascular events in women. New England Journal of Medicine, 2002. 347(10): p. 716-725.

270. Blair, S.N., Y. Cheng, and J.S. Holder, Is physical activity or physical fitness more important in defining health benefits? Medicine & Science in Sports & Exercise, 2001. 33(6): p. S379-S399.

271. King, P.A., et al., Glucose transport in skeletal muscle membrane vesicles from control and exercised rats. American Journal of Physiology-Cell Physiology, 1989. 257(6): p. C1128-C1134.

272. Ivy, J. and J. Holloszy, Persistent increase in glucose uptake by rat skeletal muscle following exercise. American Journal of Physiology-Cell Physiology, 1981. 241(5): p. C200-C203.

273. Richter, E.A., et al., Enhanced muscle glucose metabolism after exercise: modulation by local factors. American Journal of Physiology-Endocrinology And Metabolism, 1984. 246(6): p. E476-E482.

274. Group, U.P.D.S., Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). The Lancet, 1998. 352(9131): p. 854-865.

275. Liubaoerjijin, Y., et al., Effect of aerobic exercise intensity on glycemic control in type 2 diabetes: a meta-analysis of head-to-head randomized trials. Acta diabetologica, 2016. 53(5): p. 769-781.

276. Ostrowski, K., et al., A trauma‐like elevation of plasma cytokines in humans in response to treadmill running. J. Physiol., 1998. 513(3): p. 889-894.

277. Helge, J.W., et al., The effect of graded exercise on IL‐6 release and glucose uptake in human skeletal muscle. J. Physiol., 2003. 546(1): p. 299-305.

278. Nieman, D.C., et al., Variance in the acute inflammatory response to prolonged cycling is linked to exercise intensity. Journal of Interferon & Cytokine Research, 2012. 32(1): p. 12-17.

279. Scott, J.P.R., et al., Effect of exercise intensity on the cytokine response to an acute bout of running. Medicine & Science in Sports & Exercise, 2011. 43(12): p. 2297-2306.

280. Cullen, T., et al., Interleukin-6 and associated cytokine responses to an acute bout of high intensity interval exercise: the effect of exercise intensity and volume. Applied Physiology, Nutrition, and Metabolism, 2016.

281. Nieman, D.C., et al., Immune and inflammation responses to a 3-day period of intensified running versus cycling. Brain, behavior, and immunity, 2014. 39: p. 180-185.

282. Bruunsgaard, H., et al., Exercise‐induced increase in serum interleukin‐6 in humans is related to muscle damage. The Journal of physiology, 1997. 499(3): p. 833-841.

283. Peake, J.M., et al., Plasma cytokine changes in relation to exercise intensity and muscle damage. European journal of applied physiology, 2005. 95(5-6): p. 514-521.

Page 129: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

129

284. Kadoglou, N.P., et al., Exercise training ameliorates the effects of rosiglitazone on traditional and novel cardiovascular risk factors in patients with type 2 diabetes mellitus. Metabolism-Clinical and Experimental, 2010. 59(4): p. 599-607.

285. Kasapis, C. and P.D. Thompson, The effects of physical activity on serum C-reactive protein and inflammatory markers: a systematic review. Journal of the American College of Cardiology, 2005. 45(10): p. 1563-1569.

286. Stensvold, D., S.A. Slørdahl, and U. Wisløff, Effect of Exercise Training on Inflammation Status Among People with Metabolic Syndrome. Metabolic Syndrome and Related Disorders, 2012. 10(4): p. 267-272.

287. Dekker, M.J., et al., An exercise intervention without weight loss decreases circulating interleukin-6 in lean and obese men with and without type 2 diabetes mellitus. Metabolism, 2007. 56(3): p. 332-8.

288. Adamopoulos, S., et al., Physical training reduces peripheral markers of inflammation in patients with chronic heart failure. European heart journal, 2001. 22(9): p. 791-797.

289. Goldhammer, E., et al., Exercise training modulates cytokines activity in coronary heart disease patients. International journal of cardiology, 2005. 100(1): p. 93-99.

290. Mattusch, F., et al., Reduction of the plasma concentration of C-reactive protein following nine months of endurance training. International journal of sports medicine, 2000. 21(01): p. 21-24.

291. Steensberg, A., et al., IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. Am. J. Physiol. Endocrinol. Metab., 2003. 285(2): p. E433-E437.

292. Steensberg, A., et al., Acute interleukin‐6 administration does not impair muscle glucose uptake or whole‐body glucose disposal in healthy humans. The Journal of physiology, 2003. 548(2): p. 631-638.

293. Tilg, H., et al., Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood, 1994. 83(1): p. 113-118.

294. Petersen, E., et al., Acute IL-6 treatment increases fatty acid turnover in elderly humans in vivo and in tissue culture in vitro. American Journal of Physiology-Endocrinology and Metabolism, 2005. 288(1): p. E155-E162.

295. Carey, A.L., et al., Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes, 2006. 55(10): p. 2688-2697.

296. Handzlik, M.K., et al., The influence of exercise training status on antigen-stimulated IL-10 production in whole blood culture and numbers of circulating regulatory T cells. European journal of applied physiology, 2013. 113(7): p. 1839-1848.

297. Pedersen, B.K. and C.P. Fischer, Beneficial health effects of exercise–the role of IL-6 as a myokine. Trends in pharmacological sciences, 2007. 28(4): p. 152-156.

298. Jamurtas, A.Z., et al., The effects of acute exercise on serum adiponectin and resistin levels and their relation to insulin sensitivity in overweight males. Eur J Appl Physiol, 2006. 97(1): p. 122-126.

299. Kelly, K.R., et al., A 7-d exercise program increases high-molecular weight adiponectin in obese adults. Medicine and science in sports and exercise, 2012. 44(1): p. 69-74.

300. Kriketos, A.D., et al., Exercise increases adiponectin levels and insulin sensitivity in humans. Diabetes Care, 2004. 27(2): p. 629-630.

301. Racil, G., et al., Effects of high vs. moderate exercise intensity during interval training on lipids and adiponectin levels in obese young females. European journal of applied physiology, 2013. 113(10): p. 2531-2540.

302. Esposito, K., et al., Association of low interleukin-10 levels with the metabolic syndrome in obese women. The Journal of Clinical Endocrinology & Metabolism, 2003. 88(3): p. 1055-1058.

Page 130: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

130

303. Kadoglou, N., et al., The anti-inflammatory effects of exercise training promote atherosclerotic plaque stabilization in apolipoprotein E knockout mice with diabetic atherosclerosis. Eur J Histochem, 2013. 57(1).

304. You, J.S., et al., Serum osteopontin concentration is decreased by exercise-induced fat loss but is not correlated with body fat percentage in obese humans. Molecular medicine reports, 2013. 8(2): p. 579-584.

305. Duggan, C., et al., Effect of a 12-month Exercise Intervention on Serum Biomarkers of Angiogenesis in Postmenopausal Women: a randomized controlled trial. Cancer Epidemiology and Prevention Biomarkers, 2014: p. cebp. 1155.2013.

306. Lee, D.H., et al., Effects of a 12-week home-based exercise program on the level of physical activity, insulin, and cytokines in colorectal cancer survivors: a pilot study. Supportive Care in Cancer, 2013. 21(9): p. 2537-2545.

307. Venojärvi, M., et al., 12 Weeks’ aerobic and resistance training without dietary intervention did not influence oxidative stress but aerobic training decreased atherogenic index in middle-aged men with impaired glucose regulation. Food and chemical toxicology, 2013. 61: p. 127-135.

308. Bertola, A., et al., Elevated expression of osteopontin may be related to adipose tissue macrophage accumulation and liver steatosis in morbid obesity. Diabetes, 2009. 58(1): p. 125-133.

309. Wilson, L.D., et al., Circulating T-regulatory cells, exercise and the elite adolescent swimmer. Pediatric exercise science, 2009. 21(3): p. 305-317.

310. Yeh, S., et al., Regular tai chi chuan exercise enhances functional mobility and CD4CD25 regulatory T cells. British journal of sports medicine, 2006. 40(3): p. 239-243.

311. Yeh, S.-H., et al., Regular Tai Chi Chuan exercise improves T cell helper function of patients with type 2 diabetes mellitus with an increase in T-bet transcription factor and IL-12 production. British journal of sports medicine, 2009. 43(11): p. 845-850.

312. Wenning, P., et al., Endurance exercise alters cellular immune status and resistin concentrations in men suffering from non-insulin-dependent type 2 diabetes. Experimental and Clinical Endocrinology & Diabetes, 2013. 121(08): p. 475-482.

313. Moldoveanu, A.I., R.J. Shephard, and P.N. Shek, Exercise elevates plasma levels but not gene expression of IL-1β, IL-6, and TNF-α in blood mononuclear cells. Journal of Applied Physiology, 2000. 89(4): p. 1499-1504.

314. Oberbach, A., et al., Effect of a 4 week physical training program on plasma concentrations of inflammatory markers in patients with abnormal glucose tolerance. Eur J Endocrinol, 2006. 154(4): p. 577-85.

315. Omori, K., et al., High glucose enhances interleukin-6-induced vascular endothelial growth factor 165 expression via activation of gp130-mediated p44/42 MAPK-CCAAT/enhancer binding protein signaling in gingival fibroblasts. Journal of Biological Chemistry, 2004. 279(8): p. 6643-6649.

316. Vozarova, B., et al., Circulating interleukin‐6 in relation to adiposity, insulin action, and insulin secretion. Obesity, 2001. 9(7): p. 414-417.

317. Lee, S.W., et al., Alterations in peripheral blood levels of TIMP-1, MMP-2, and MMP-9 in patients with type-2 diabetes. Diabetes research and clinical practice, 2005. 69(2): p. 175-179.

318. Rullman, E., et al., Endurance exercise activates matrix metalloproteinases in human skeletal muscle. Journal of applied physiology, 2009. 106(3): p. 804-812.

319. Žakovičova, E., et al., Circulating Serum Matrix Metalloproteinase-3 and Metalloproteinase-9 Are Not Associated with Echocardiographic Parameters of Diastolic Function in Asymptomatic Type 2 Diabetic Patients. Journal of International Medical Research, 2010. 38(6): p. 2093-2099.

Page 131: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

131

320. Straczkowski, M., et al., Changes in tumor necrosis factor-alpha system and insulin sensitivity during an exercise training program in obese women with normal and impaired glucose tolerance. European Journal of Endocrinology, 2001. 145(3): p. 273-280.

321. Kim, H.-J., et al., Differential effects of interleukin-6 and-10 on skeletal muscle and liver insulin action in vivo. Diabetes, 2004. 53(4): p. 1060-1067.

322. Dorneles, G.P., et al., High intensity interval exercise decreases IL-8 and enhances the immunomodulatory cytokine interleukin-10 in lean and overweight–obese individuals. Cytokine, 2016. 77: p. 1-9.

323. Hayashi, Y., et al., A single bout of exercise at higher intensity enhances glucose effectiveness in sedentary men. J. Clin. Endocrinol. Metab., 2005. 90(7): p. 4035-4040.

324. Katz, A., et al., Leg glucose uptake during maximal dynamic exercise in humans. Am. J. Physiol. Endocrinol. Metab., 1986. 251(1): p. E65-E70.

325. Wahren, J., et al., Glucose metabolism during leg exercise in man. J. Clin. Invest., 1971. 50(12): p. 2715.

326. Wahren, J., P. Felig, and L. Hagenfeldt, Physical exercise and fuel homeostasis in diabetes mellitus. Diabetologia,, 1978. 14(4): p. 213-222.

327. Ahlborg, G., et al., Substrate turnover during prolonged exercise in man: splanchnic and leg metabolism of glucose, free fatty acids, and amino acids. J. Clin. Invest., 1974. 53(4): p. 1080-1090.

328. Sylow, L., et al., Exercise-stimulated glucose uptake - regulation and implications for glycaemic control. Nat. Rev. Endocrinol., 2017. 13(3): p. 133-148.

329. Holloszy, J.O., Exercise-induced increase in muscle insulin sensitivity. J Appl Physiol (1985), 2005. 99(1): p. 338-43.

330. Baynard, T., et al., Effect of a single vs multiple bouts of exercise on glucose control in women with type 2 diabetes. Metab. Clin. Exp., 2005. 54(8): p. 989-994.

331. Larsen, J., et al., The effect of moderate exercise on postprandial glucose homeostasis in NIDDM patients. Diabetologia,, 1997. 40(4): p. 447-453.

332. Roberts, S., et al., Glycemic response to carbohydrate and the effects of exercise and protein. Nutrition,, 2013. 29(6): p. 881-885.

333. Venables, M.C., et al., Effect of acute exercise on glucose tolerance following post-exercise feeding. Eur. J. Appl. Physiol., 2007. 100(6): p. 711-717.

334. Oberlin, D.J., et al., One bout of exercise alters free-living postprandial glycemia in type 2 diabetes. Med. Sci. Sports, Exerc., 2014. 46: p. 232-238.

335. Mikines, K.J., et al., Effect of physical exercise on sensitivity and responsiveness to insulin in humans. Am. J. Physiol. Endocrinol. Metab., 1988. 254(3): p. E248-E259.

336. Brun, J., et al., Influence of short-term submaximal exercise on parameters of glucose assimilation analyzed with the minimal model. Metab. Clin. Exp., 1995. 44(7): p. 833-840.

337. Bonen, A., M. Ball-Burnett, and C. Russel, Glucose tolerance is improved after low-and high-intensity exercise in middle-age men and women. Can. J. Appl. Physiol., 1998. 23(6): p. 583-593.

338. Araujo-Vilar, D., et al., Influence of moderate physical exercise on insulin-mediated and non—insulin-mediated glucose uptake in healthy subjects. Metabolism,, 1997. 46(2): p. 203-209.

339. Rynders, C.A., et al., Effects of exercise intensity on postprandial improvement in glucose disposal and insulin sensitivity in prediabetic adults. J. Clin. Endocrinol. Metab., 2013. 99(1): p. 220-228.

340. King, D.S., et al., Time course for exercise-induced alterations in insulin action and glucose tolerance in middle-aged people. J. Appl. Physiol., 1995. 78(1): p. 17-22.

Page 132: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

132

341. O’Connor, A.M., et al., The gastroenteroinsular response to glucose ingestion during postexercise recovery. Am. J. Physiol. Endocrinol. Metab., 2006. 290(6): p. E1155-E1161.

342. Rose, A.J., et al., Effect of prior exercise on glucose metabolism in trained men. Am. J. Physiol. Endocrinol. Metab., 2001. 281(4): p. E766-E771.

343. Knudsen, S.H., et al., The immediate effects of a single bout of aerobic exercise on oral glucose tolerance across the glucose tolerance continuum. Physiol. Rep., 2014. 2(8): p. e12114.

344. Mendham, A.E., et al., Effects of mode and intensity on the acute exercise-induced IL-6 and CRP responses in a sedentary, overweight population. Eur J Appl Physiol, 2011. 111(6): p. 1035-45.

345. Pedersen, B.K., IL-6 signalling in exercise and disease. Biochem. Soc. Trans., 2007. 35(Pt 5): p. 1295-7.

346. Harder-Lauridsen, N.M., et al., Effect of IL-6 on the insulin sensitivity in patients with type 2 diabetes. Am. J. Physiol. Endocrinol. Metab., 2014. 306(7): p. E769-78.

347. Wang, X., et al., Inflammatory markers and risk of type 2 diabetes. Diabetes Care,, 2013. 36(1): p. 166-175.

348. Kado, S., T. Nagase, and N. Nagata, Circulating levels of interleukin-6, its soluble receptor and interleukin-6/interleukin-6 receptor complexes in patients with type 2 diabetes mellitus. Acta. Diabetol., 1999. 36(1-2): p. 67-72.

349. Hunter, C.A. and S.A. Jones, IL-6 as a keystone cytokine in health and disease. Nat Immunol, 2015. 16(5): p. 448-57.

350. Kraakman, M.J., et al., Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance. Cell Metab, 2015. 21(3): p. 403-416.

351. Gillen, J.B., et al., Acute high‐intensity interval exercise reduces the postprandial glucose response and prevalence of hyperglycaemia in patients with type 2 diabetes. Diabetes. Obes. Metab., 2012.

352. Little, J.P., et al., An acute bout of high-intensity interval training increases the nuclear abundance of PGC-1α and activates mitochondrial biogenesis in human skeletal muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol., 2011. 300(6): p. R1303-R1310.

353. Brestoff, J.R.B.J., et al., An acute bout of endurance exercise but not sprint interval exercise enhances insulin sensitivity. Appl. Physiol. Nutr. Metab., 2009. 34(1): p. 25-32.

354. Borg, G., Perceived exertion as an indicator of somatic stress. Scand. J. Rehabil. Med., 1970. 2: p. 92-98.

355. Stork, A.D., et al., Comparison of the accuracy of the HemoCue glucose analyzer with the Yellow Springs Instrument glucose oxidase analyzer, particularly in hypoglycemia. Eur. J. Endocrinol., 2005. 153(2): p. 275-281.

356. Sim, A., et al., High-intensity intermittent exercise attenuates ad-libitum energy intake. Int. J. Obes., 2014. 38(3): p. 417-422.

357. Kanaley, J.A., et al., Alteration of postprandial glucose and insulin concentrations with meal frequency and composition. Br. J. Nutr., 2014. 112(9): p. 1484-93.

358. Cohen, J., Statistical power analysis. Current directions in psychological science, 1992. 1(3): p. 98-101.

359. Garetto, L.P., et al., Enhanced muscle glucose metabolism after exercise in the rat: the two phases. Am. J. Physiol. Endocrinol. Metab., 1984. 246(6): p. E471-E475.

360. Yale, J.-F., L.A. Leiter, and E.B. Marliss, Metabolic Responses to Intense Exercise in Lean and Obese Subjects. J. Clin. Endocrinol. Metab. , 1989. 68(2): p. 438-445.

Page 133: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

133

361. Marliss, E.B. and M. Vranic, Intense exercise has unique effects on both insulin release and its roles in glucoregulation implications for diabetes. Diabetes,, 2002. 51(suppl 1): p. S271-S283.

362. Marliss, E.B., et al., Glucoregulatory and hormonal responses to repeated bouts of intense exercise in normal male subjects. J. Appl. Physiol., 1991. 71(3): p. 924-933.

363. Ellingsgaard, H., et al., Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat. Med., 2011. 17(11): p. 1481-1489.

364. Leggate, M., et al., The response of interleukin-6 and soluble interleukin-6 receptor isoforms following intermittent high intensity and continuous moderate intensity cycling. Cell Stress Chaperones,, 2010. 15(6): p. 827-833.

365. Steensberg, A., et al., Production of interleukin‐6 in contracting human skeletal muscles can account for the exercise‐induced increase in plasma interleukin‐6. j. Physiol., 2000. 529(1): p. 237-242.

366. Ulven, S.M., et al., An acute bout of exercise modulate the inflammatory response in peripheral blood mononuclear cells in healthy young men. Arch. Physiol. Biochem., 2015. 121(2): p. 41-49.

367. Zwetsloot, K.A., et al., High-intensity interval training induces a modest systemic inflammatory response in active, young men. Journal of inflammation research, 2014. 7: p. 9.

368. Sigal, R.J., et al., Effects of aerobic training, resistance training, or both on glycemic control in type 2 diabetes: a randomized trial. Ann. Intern. Med., 2007. 147(6): p. 357-369.

369. Pal, M., M.A. Febbraio, and M. Whitham, From cytokine to myokine: the emerging role of interleukin-6 in metabolic regulation. Immunol. Cell. Biol., 2014. 92(4): p. 331-9.

370. Asghar, A. and N. Sheikh, Role of immune cells in obesity induced low grade inflammation and insulin resistance. Cell Immunol, 2017. 315: p. 18-26.

371. Wensveen, F.M., et al., The "Big Bang" in obese fat: Events initiating obesity-induced adipose tissue inflammation. Eur J Immunol, 2015. 45(9): p. 2446-56.

372. Bagi, Z., Z. Broskova, and A. Feher, Obesity and coronary microvascular disease - implications for adipose tissue-mediated remote inflammatory response. Curr Vasc Pharmacol, 2014. 12(3): p. 453-61.

373. Knight, S.F. and J.D. Imig, Obesity, insulin resistance, and renal function. Microcirculation, 2007. 14(4-5): p. 349-62.

374. Van Gaal, L.F., I.L. Mertens, and C.E. De Block, Mechanisms linking obesity with cardiovascular disease. Nature, 2006. 444(7121): p. 875.

375. Fox, C.S., et al., Increasing cardiovascular disease burden due to diabetes mellitus. Circulation, 2007. 115(12): p. 1544-1550.

376. Meshkani, R. and S. Vakili, Tissue resident macrophages: Key players in the pathogenesis of type 2 diabetes and its complications. Clin Chim Acta, 2016. 462: p. 77-89.

377. Wang, T., C. He, and X. Yu, Pro-Inflammatory Cytokines: New Potential Therapeutic Targets for Obesity-Related Bone Disorders. Curr Drug Targets, 2017. 18(14): p. 1664-1675.

378. Tong, K.M., et al., Adiponectin increases MMP‐3 expression in human chondrocytes through adipor1 signaling pathway. J Cell Biochem, 2011. 112(5): p. 1431-1440.

379. Verheggen, R.J., et al., A systematic review and meta-analysis on the effects of exercise training versus hypocaloric diet: distinct effects on body weight and visceral adipose tissue. Obes Rev, 2016. 17(8): p. 664-90.

380. Jaoude, J. and Y. Koh, Matrix metalloproteinases in exercise and obesity. Vasc Health Risk Manag, 2016. 12: p. 287-295.

Page 134: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

134

381. Kiefer, F.W., et al., Neutralization of osteopontin inhibits obesity-induced inflammation and insulin resistance. Diabetes, 2010. 59(4): p. 935-946.

382. Urso, M.L., et al., Effects of exercise training on the matrix metalloprotease response to acute exercise. Eur J Appl Physiol, 2009. 106(5): p. 655-663.

383. Numao, S., et al., Influence of acute aerobic exercise on adiponectin oligomer concentrations in middle-aged abdominally obese men. Metabolism, 2011. 60(2): p. 186-194.

384. Magkos, F., B.S. Mohammed, and B. Mittendorfer, Enhanced insulin sensitivity after acute exercise is not associated with changes in high-molecular weight adiponectin concentration in plasma. Eur J Endocrinol, 2010. 162(1): p. 61-66.

385. Association, A.D., Postprandial blood glucose. Clin Diabetes, 2001. 19(3): p. 127-130. 386. Bonora, E. and M. Muggeo, Postprandial blood glucose as a risk factor for

cardiovascular disease in type II diabetes: the epidemiological evidence. Diabetologia, 2001. 44(12): p. 2107-2114.

387. Manders, R., J. Van Dijk, and L. Van Loon, Low-intensity exercise reduces the prevalence of hyperglycemia in type 2 diabetes. Med Sci Sports Exerc, 2010. 42(2): p. 219-25.

388. Van Dijk, J.-W., et al., Exercise therapy in type 2 diabetes. Diabetes care, 2012. 35(5): p. 948-954.

389. Kovatchev, B., et al., Comparison of the numerical and clinical accuracy of four continuous glucose monitors. Diabetes care, 2008. 31(6): p. 1160-1164.

390. Melki, V., et al., Value and limitations of the Continuous Glucose Monitoring System in the management of type 1 diabetes. Diabetes & metabolism, 2006. 32(2): p. 123-129.

391. Vashist, S.K., Continuous glucose monitoring systems: a review. Diagnostics, 2013. 3(4): p. 385-412.

392. Shu, C.J., C. Benoist, and D. Mathis, The immune system's involvement in obesity-driven type 2 diabetes. Semin Immunol, 2012. 24(6): p. 436-42.

393. Levings, M.K., et al., The role of IL-10 and TGF-β in the differentiation and effector function of T regulatory cells. International archives of allergy and immunology, 2002. 129(4): p. 263-276.

394. Kang, Y.E., et al., The Roles of Adipokines, Proinflammatory Cytokines, and Adipose Tissue Macrophages in Obesity-Associated Insulin Resistance in Modest Obesity and Early Metabolic Dysfunction. PLoS One, 2016. 11(4): p. e0154003.

395. Schmidt, F.M., et al., Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS One, 2015. 10(3): p. e0121971.

396. Connelly, M.A., et al., GlycA, a marker of acute phase glycoproteins, and the risk of incident type 2 diabetes mellitus: PREVEND study. Clinica Chimica Acta, 2016. 452: p. 10-17.

397. Kanda, H., et al., MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. The Journal of clinical investigation, 2006. 116(6): p. 1494-1505.

398. Sartipy, P. and D.J. Loskutoff, Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proceedings of the National Academy of Sciences, 2003. 100(12): p. 7265-7270.

399. Chang, C.-C., et al., Interferon gamma-induced protein 10 is associated with insulin resistance and incident diabetes in patients with nonalcoholic fatty liver disease. Scientific reports, 2015. 5: p. 10096.

400. Kim, C., et al., Circulating levels of MCP-1 and IL-8 are elevated in human obese subjects and associated with obesity-related parameters. International journal of obesity, 2006. 30(9): p. 1347.

Page 135: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

135

401. Quinn, L.S., et al., Interleukin-15 stimulates adiponectin secretion by 3T3-L1 adipocytes: evidence for a skeletal muscle-to-fat signaling pathway. Cell biology international, 2005. 29(6): p. 449-457.

402. Barra, N., et al., Interleukin‐15 treatment improves glucose homeostasis and insulin sensitivity in obese mice. Diabetes, Obesity and Metabolism, 2012. 14(2): p. 190-193.

403. Wang, X., et al., Inflammatory markers and risk of type 2 diabetes. Diabetes care, 2013. 36(1): p. 166-175.

404. American Diabetes, A., 4. Lifestyle Management: Standards of Medical Care in Diabetes-2018. Diabetes Care, 2018. 41(Suppl 1): p. S38-S50.

405. Richter, E.A., et al., Effect of exercise on insulin action in human skeletal muscle. J Appl Physiol, 1989. 66(2): p. 876-885.

406. Oberlin, D.J., et al., One bout of exercise alters free-living postprandial glycemia in type 2 diabetes. Med. Sci. Sports Exerc, 2014. 46: p. 232-238.

407. Praet, S.F., et al., Influence of acute exercise on hyperglycemia in insulin-treated type 2 diabetes. Medicine & science in Sports & Exercise, 2006. 38(12): p. 2037-2044.

408. Snowling, N.J. and W.G. Hopkins, Effects of different modes of exercise training on glucose control and risk factors for complications in type 2 diabetic patients: a meta-analysis. Diabetes care, 2006. 29(11): p. 2518-2527.

409. Thomas, D.E., E.J. Elliott, and G.A. Naughton, Exercise for type 2 diabetes mellitus. Cochrane Database Syst Rev, 2006(3): p. CD002968.

410. Ploeger, H.E., et al., The effects of acute and chronic exercise on inflammatory markers in children and adults with a chronic inflammatory disease: a systematic review. Exerc Immunol Rev, 2009. 15: p. 6-41.

411. Brown, W.M., et al., A Systematic Review of the Acute Effects of Exercise on Immune and Inflammatory Indices in Untrained Adults. Sports Med Open, 2015. 1(1): p. 35.

412. Moura, B.P., et al., Effect of a short-term exercise program on glycemic control measured by fructosamine test in type 2 diabetes patients. Diabetology & metabolic syndrome, 2014. 6(1): p. 16.

413. DeFronzo, R.A. and M. Matsuda, Reduced time points to calculate the composite index. Diabetes Care, 2010. 33(7): p. e93-e93.

414. Abdul-Ghani, M.A., et al., Muscle and liver insulin resistance indexes derived from the oral glucose tolerance test. Diabetes Care, 2007. 30(1): p. 89-94.

415. Cederholm, J. and L. Wibell, Insulin release and peripheral sensitivity at the oral glucose tolerance test. Diabetes Res Clin Pract, 1990. 10(2): p. 167-75.

416. Jansen, K., et al., Polychromatic flow cytometric high-throughput assay to analyze the innate immune response to Toll-like receptor stimulation. Journal of immunological methods, 2008. 336(2): p. 183-192.

417. Croghan, C. and P. Egeghy, Methods of dealing with values below the limit of detection using SAS. Southern SAS User Group, 2003: p. 22-24.

418. Gleeson, M., et al., The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat. Rev. Immunol., 2011. 11(9): p. 607.

419. Kosaryan, M., et al., Credibility of measurement of fructosamine and hemoglobin A1C in estimating blood glucose level of diabetic patients with thalassemia major. Open Journal of Hematology, 2012. 3(1).

420. Baynard, T., et al., Short‐term Training Effects on Diastolic Function in Obese Persons With the Metabolic Syndrome. Obesity, 2008. 16(6): p. 1277-1283.

421. Patarrão, R.S., W.W. Lautt, and M.P. Macedo, Assessment of methods and indexes of insulin sensitivity. Revista Portuguesa de Endocrinologia, Diabetes e Metabolismo, 2014. 9(1): p. 65-73.

Page 136: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

136

422. Kim, K.B., Effect of different training mode on Interleukin-6 (IL-6) and C-reactive protein (CRP) in type 2 diabetes mellitus (T2DM) patients. Journal of exercise nutrition & biochemistry, 2014. 18(4): p. 371.

423. Kohut, M., et al., Aerobic exercise, but not flexibility/resistance exercise, reduces serum IL-18, CRP, and IL-6 independent of β-blockers, BMI, and psychosocial factors in older adults. Brain, behavior, and immunity, 2006. 20(3): p. 201-209.

424. Akinkuolie, A.O., et al., Novel protein glycan side-chain biomarker and risk of incident type 2 diabetes mellitus. Arteriosclerosis, thrombosis, and vascular biology, 2015: p. ATVBAHA. 115.305635.

425. Higai, K., et al., Altered glycosylation of α1-acid glycoprotein in patients with inflammation and diabetes mellitus. Clinica chimica acta, 2003. 329(1-2): p. 117-125.

426. Schmidt, M.I., et al., Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. The Lancet, 1999. 353(9165): p. 1649-1652.

427. Singh-Manoux, A., et al., Association between inflammatory biomarkers and all-cause, cardiovascular and cancer-related mortality. Canadian Medical Association Journal, 2017. 189(10): p. E384-E390.

428. Poortmans, J. and R.W. Jeanloz, Quantitative immunological determination of 12 plasma proteins excreted in human urine collected before and after exercise. The Journal of clinical investigation, 1968. 47(2): p. 386-393.

429. Bartlett, D.B., et al., Association of the Composite Inflammatory Biomarker GlycA, with Exercise-Induced Changes in Body Habitus in Men and Women with Prediabetes. Oxidative medicine and cellular longevity, 2017. 2017.

430. Stewart, L.K., et al., The influence of exercise training on inflammatory cytokines and C-reactive protein. Medicine & Science in Sports & Exercise, 2007. 39(10): p. 1714-1719.

431. Masters, S.L., et al., Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nature immunology, 2010. 11(10): p. 897.

432. Zoppini, G., et al., Effects of moderate-intensity exercise training on plasma biomarkers of inflammation and endothelial dysfunction in older patients with type 2 diabetes. Nutrition, metabolism, and cardiovascular diseases: NMCD, 2006. 16(8): p. 543.

433. Louis, E., et al., Time course of proteolytic, cytokine, and myostatin gene expression after acute exercise in human skeletal muscle. Journal of applied physiology, 2007. 103(5): p. 1744-1751.

434. Nieman, D., et al., The effects of moderate exercise training on natural killer cells and acute upper respiratory tract infections. International journal of sports medicine, 1990. 11(06): p. 467-473.

435. Nieman, D.C., Exercise, infection, and immunity. International journal of sports medicine, 1994. 15(3): p. S131.

436. Ostrowski, K., et al., Pro-and anti-inflammatory cytokine balance in strenuous exercise in humans. The Journal of physiology, 1999. 515(1): p. 287-291.

437. Febbraio, M.A. and B.K. Pedersen, Muscle-derived interleukin-6: mechanisms for activation and possible biological roles. The FASEB Journal, 2002. 16(11): p. 1335-1347.

438. Boonstra, A., et al., Macrophages and myeloid dendritic cells, but not plasmacytoid dendritic cells, produce IL-10 in response to MyD88-and TRIF-dependent TLR signals, and TLR-independent signals. The Journal of Immunology, 2006. 177(11): p. 7551-7558.

439. Rosa, L., et al., Moderate acute exercise (70% VO2 peak) induces TGF‐β, α‐amylase and IgA in saliva during recovery. Oral diseases, 2014. 20(2): p. 186-190.

Page 137: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

137

440. Straczkowski, M., et al., Plasma interleukin-10 concentration is positively related to insulin sensitivity in young healthy individuals. Diabetes care, 2005. 28(8): p. 2036-2037.

441. Weinhold, M., et al., Physical exercise modulates the homeostasis of human regulatory T cells. Journal of Allergy and Clinical Immunology, 2016. 137(5): p. 1607-1610. e8.

442. Zheng, S.G., et al., Natural and induced CD4+ CD25+ cells educate CD4+ CD25− cells to develop suppressive activity: the role of IL-2, TGF-β, and IL-10. The Journal of Immunology, 2004. 172(9): p. 5213-5221.

443. Yeh, S.-H., et al., Tai chi chuan exercise decreases A1C levels along with increase of regulatory T-cells and decrease of cytotoxic T-cell population in type 2 diabetic patients. Diabetes Care, 2007. 30(3): p. 716-718.

444. Lowder, T., et al., Repeated bouts of aerobic exercise enhance regulatory T cell responses in a murine asthma model. Brain, behavior, and immunity, 2010. 24(1): p. 153-159.

445. Kajitani, N., et al., Microinflammation is a common risk factor for progression of nephropathy and atherosclerosis in Japanese patients with type 2 diabetes. Diabetes research and clinical practice, 2010. 88(2): p. 171-176.

446. Sajadi, S., et al., Plasma levels of CXCL1 (GRO-alpha) and CXCL10 (IP-10) are elevated in type 2 diabetic patients: evidence for the involvement of inflammation and angiogenesis/angiostasis in this disease state. Clin Lab, 2013. 59(1-2): p. 133-7.

447. Baturcam, E., et al., Physical exercise reduces the expression of RANTES and its CCR5 receptor in the adipose tissue of obese humans. Mediators of inflammation, 2014. 2014.

448. Chang, C.-J., et al., Evidence in obese children: contribution of hyperlipidemia, obesity-inflammation, and insulin sensitivity. PloS one, 2015. 10(5): p. e0125935.

449. Griffin, T.M., et al., Induction of osteoarthritis and metabolic inflammation by a very high‐fat diet in mice: Effects of short‐term exercise. Arthritis & Rheumatology, 2012. 64(2): p. 443-453.

450. Ishiuchi, Y., et al., Skeletal muscle cell contraction reduces a novel myokine, chemokine (CXC motif) ligand 10 (CXCL10): potential roles in exercise-regulated angiogenesis. Bioscience, biotechnology, and biochemistry, 2017: p. 1-9.

451. Hansen, D., et al., Continuous low-to moderate-intensity exercise training is as effective as moderate-to high-intensity exercise training at lowering blood HbA1c in obese type 2 diabetes patients. Diabetologia, 2009. 52(9): p. 1789-1797.

452. Mourier, A., et al., Mobilization of visceral adipose tissue related to the improvement in insulin sensitivity in response to physical training in NIDDM: effects of branched-chain amino acid supplements. Diabetes care, 1997. 20(3): p. 385-391.

453. Trovati, M., et al., Influence of physical training on blood glucose control, glucose tolerance, insulin secretion, and insulin action in non-insulin-dependent diabetic patients. Diabetes Care, 1984. 7(5): p. 416-420.

454. Sites, C.K., et al., Menopause-related differences in inflammation markers and their relationship to body fat distribution and insulin-stimulated glucose disposal. Fertility and sterility, 2002. 77(1): p. 128-135.

455. Gleeson, M., et al., The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nature Reviews Immunology, 2011. 11(9): p. 607.

456. Jorge, M.L.M.P., et al., The effects of aerobic, resistance, and combined exercise on metabolic control, inflammatory markers, adipocytokines, and muscle insulin signaling in patients with type 2 diabetes mellitus. Metabolism-Clinical and Experimental, 2011. 60(9): p. 1244-1252.

Page 138: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

138

457. Sigal, R.J., et al., Effects of aerobic training, resistance training, or both on glycemic control in type 2 diabetes: a randomized trial. Annals of internal medicine, 2007. 147(6): p. 357-369.

Page 139: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

139

Appendix A Murdoch University Ethics

Approval

Page 140: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

140

A.1 Murdoch University Ethics Approval

(Chapters 4 and 5)

Page 141: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

141

A.2 Murdoch University Ethics Approval

(Chapters 6)

Page 142: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

142

Page 143: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

143

Appendix B Information Letters

Page 144: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

144

B.1 Information Letters (Chapter 4 and 5)

Page 145: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

145

Page 146: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

146

Page 147: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

147

Page 148: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

148

B.2 Information Letters (Chapter 6)

Page 149: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

149

Page 150: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

150

Page 151: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

151

Page 152: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

152

Appendix C Adult Pre-Exercise Screening

Tool

Page 153: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

153

Page 154: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

154

Page 155: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

155

Page 156: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

156

Appendix D 24-h Food Diary

Page 157: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

157

Appendix E Borg Scale

Page 158: The role of exercise in glycaemic control and inflammation in … · 2018. 10. 10. · The role of exercise in glycaemic control and inflammation in overweight/obese individuals with

158

Appendix F CONSORT Diagram