244
Abstract of thesis entitled Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and smooth muscle cells attenuates oxidative stress-induced injury Submitted by LI, Xiang for the degree of Doctor of Philosophy jointly at the University of Hong Kong and Imperial College London in June, 2016 Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease characterized by persistent airflow limitation that is not fully reversible and is usually caused by cigarette smoke (CS). The disease is predicted to be the fourth leading cause of death by 2030, but none of the currently available treatments can alleviate the progressive decline in lung function. Mesenchymal stem cells (MSCs) are fibroblast-like multipotent stem cells that can be isolated from various tissues such as bone marrow (BM-MSCs). Despite numerous reports of their efficacy in COPD-related pre-clinical models, BM-MSCs have not demonstrated efficacy in a clinical trial of COPD, highlighting the need for

Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

Abstract of thesis entitled

Mitochondrial transfer from induced pluripotent stem cell-derived

mesenchymal stem cells to airway epithelial and smooth muscle cells

attenuates oxidative stress-induced injury

Submitted by

LI, Xiang

for the degree of Doctor of Philosophy

jointly at the University of Hong Kong and Imperial College London

in June, 2016

Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory

disease characterized by persistent airflow limitation that is not fully reversible and is

usually caused by cigarette smoke (CS). The disease is predicted to be the fourth

leading cause of death by 2030, but none of the currently available treatments can

alleviate the progressive decline in lung function.

Mesenchymal stem cells (MSCs) are fibroblast-like multipotent stem cells that

can be isolated from various tissues such as bone marrow (BM-MSCs). Despite

numerous reports of their efficacy in COPD-related pre-clinical models, BM-MSCs

have not demonstrated efficacy in a clinical trial of COPD, highlighting the need for

Page 2: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

improved MSC-based therapy. The in vitro derivation of MSCs from induced

pluripotent stem cells (iPSCs) has provided a new source of MSCs. Compared to

BM-MSCs, iPSC-derived MSCs (iPSC-MSCs) are a more abundant source, have a

higher expanding capacity and are possibly not subject to the ageing-associated

dysfunction seen in BM-MSCs.

In this study I determined the effects of human iPSC-MSCs in a rat COPD model

using BM-MSCs as comparison. Rats were exposed to CS for 1 hr/day for 56 days.

iPSC-MSCs or BM-MSCs were administrated at days 29 and 43. iPSC-MSCs

demonstrated superior effects over BM-MSCs in attenuating CS-induced lung

airspace enlargement, fibrosis, inflammation and apoptosis. In a mouse model of

ozone-induced lung damage, intravenous administration of iPSC-MSCs 24 hours

before ozone exposure for 3 hours alleviated airway hyper-responsiveness,

inflammation and apoptosis in the lung.

There is increasing evidence demonstrating that mitochondrial dysfunction may

play an important role in COPD pathogenesis, indicating mitochondria as a potential

therapeutic target. Meanwhile, mitochondrial transfer from MSCs to injured airway

cells has been reported as a novel mechanism of action for MSCs.

In this study mitochondrial transfer from iPSC-MSCs to the airway epithelium of

CS-exposed rats was detected. iPSC-MSCs also transferred mitochondria to bronchial

epithelial BEAS-2B cells and primary airway smooth muscle cell (ASMCs) in vitro in

a direct co-culture system, an effect that was enhanced by CS medium (CSM). Direct

Page 3: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

co-culture with iPSC-MSCs alleviated CSM-induced ATP deprivation in BEAS-2B

cells, as well as CSM-induced mitochondrial reactive oxygen species (ROS),

apoptosis and reduction of mitochondrial membrane potential (ΔΨm) in ASMCs.

Administration of iPSC-MSCs also prevented ozone-induced mitochondrial ROS and

ΔΨm reduction in mouse lungs.

The paracrine effects of iPSC-MSCs were also investigated. iPSC-MSC-derived

conditioned medium (iPSC-MSCs-CdM) protected BEAS2-B cells from

CSM-induced apoptosis. The effect was reduced by depleting stem cell factor (SCF)

from iPSC-MSCs-CdM. However, both iPSC-MSCs-CdM and trans-well inserts

containing iPSC-MSCs were only able to alleviate CSM-induced mitochondrial ROS,

but not ΔΨm reduction and apoptosis, in ASMCs.

I demonstrated the capacity of iPSC-MSCs to alleviate oxidative stress-induced

COPD phenotype in vivo. Mitochondrial transfer from iPSC-MSCs was able to

alleviate oxidative stress-induced mitochondrial dysfunction and apoptosis in target

cells. The full capacity of iPSC-MSCs to achieve these effects may rely on a

combination of cell-cell contact and release of paracrine factors. These findings define

iPSC-MSCs as a promising candidate for the development of MSCs-based therapy of

COPD.

Word count: 496

Page 4: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

Mitochondrial transfer from induced pluripotent

stem cell-derived mesenchymal stem cells to airway

epithelial and smooth muscle cells attenuates

oxidative stress-induced injury

By

LI, Xiang

M.Phil. H.K., B.Sc. H.K.

A thesis submitted in fulfilment of the requirements for

the Degree of Doctor of Philosophy

jointly at The University of Hong Kong and Imperial College London

June, 2016

Page 5: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

The copyright of this thesis rests with the author and is made available

under a Creative Commons Attribution Non-Commercial No Derivatives

licence. Researchers are free to copy, distribute or transmit the thesis on

the condition that they attribute it, that they do not use it for commercial

purposes and that they do not alter, transform or build upon it. For any

reuse or redistribution, researchers must make clear to others the licence

terms of this work.

Page 6: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

i

Declaration

I declare that this thesis represents my own work, except where due

acknowledgement is made, and that it has not been previously included in a thesis,

dissertation or report submitted to The University of Hong Kong or Imperial College

London, or to any other institution for a degree, diploma or other qualifications.

LI, Xiang

Page 7: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

ii

Acknowledgements

I would like to express my greatest gratitude to my primary supervisors Dr. Judith

C.W. Mak, The University of Hong Kong (HKU) and Professor Kian Fan Chung,

Imperial College London (ICL) for giving me the opportunity to do this joint Ph.D.

between the two universities, and for their guidance and support during the period of

study. I would also like to thank my co-supervisor Dr. Pankaj K Bhavsar (ICL) for his

precious supports and help especially on the progress management of the projects.

Special thanks are given to my assistant supervisor Dr. Charalambos Michaeloudes

(ICL) for his valuable suggestions on the projects and help on laboratory skills. I

would also like to thank Dr. Coen H Wiegman (ICL) for his generous help on the

animal study.

Sincere thanks are given to Professor Mary S.M. Ip from the Division of

Respiratory Medicine, Dr. Qizhou Lian and Professor Hung-fat Tse from the Division

of Cardiology, HKU for the initiation of the collaborative work on mesenchymal stem

cells and lung diseases in HKU. I would like to thank Dr. Yuelin Zhang from the

Division of Cardiology for his intense collaboration and for providing the

mesenchymal stem cells used in this study.

Grateful thanks are given to my dear labmates in HKU, Mr Dave Yeung, Dr.

Yingmin Liang, Ms Wang Yan, Ms Yuting Cui, Dr. Chunyan Zheng, Dr. Yuanyuan Li

and Dr. Sze-Kwan Lam for their friendship and support.

Finally, I would like to give my deepest thanks to my family. I thank my parents

Page 8: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

iii

for their love and encouragement and my wife Mengke for her trust, patience and love

across the long distance. I would not have achieved this Ph.D without her support.

Page 9: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

iv

Part of the results in this thesis has been published in the following

publications:

Journal article

1. Li X, Zhang Y, Yeung SC, Liang Y, Liang X, Ding Y, Ip MS, Tse HF, Mak JC,

Lian Q. (2014) Mitochondrial transfer of induced pluripotent stem cell-derived

mesenchymal stem cells to airway epithelial cells attenuates cigarette

smoke-induced damage. Am J Respir Cell Mol Biol. 51(3):455-65

Abstracts

1. Li X, Michaeloudes C, Zhang Y, Lian Q, Mak JCW, Bhavsar PK, Chung KF.

(2015) Oxidative Stress-Induced Mitochondria Alteration in Human Airway

Smooth Muscle Cells and Mesenchymal Stem Cells. Am J Respir Crit Care Med

191;2015:A5544 (Abstract)

2. Li X, Zhang Y, Yeung SC, Liang Y, Liang X, Ding Y, Ip MS, Tse HF, Mak JC,

Lian Q. (2014) Mitochondrial transfer of induced pluripotent stem cell-derived

mesenchymal stem cells to airway epithelial cells attenuates cigarette

smoke-induced damage. Am J Respir Crit Care Med 189;2014:A5295 (Abstract)

3. Li X, Zhang Y, Yeung SC, Lian Q, Tse HF, Ip MSM, Mak JCW. (2013) Effects of

induced pluripotent stem cell- and bone marrow-derived mesenchymal stem cells

on cigarette smoke-induced lung damage in rats. Am J Respir Crit Care Med

187;2013:A4850 (Abstract)

Page 10: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

v

Table of Contents

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

1.1. Introduction to Chronic Obstructive Pulmonary Disease (COPD) .................................. 2

1.1.1. Definition and diagnosis of COPD .................................................................................. 2

1.1.2. Epidemiology of COPD .................................................................................................. 5

1.1.3. Risk factors of COPD ...................................................................................................... 7

1.1.4. Pathogenesis of COPD .................................................................................................... 9

1.1.4.1. Oxidative stress ..................................................................................................... 11

1.1.4.2. Chronic airway inflammation ............................................................................... 13

1.1.4.3. Protease and anti-protease imbalance ................................................................... 15

1.1.4.4. Apoptosis .............................................................................................................. 16

1.1.4.5. Ageing ................................................................................................................... 19

1.1.4.6. Bronchial epithelial cells and airway smooth muscle cells in COPD ................... 20

1.1.4.7. Mitochondria and COPD ...................................................................................... 22

1.1.5. Current therapies ........................................................................................................... 27

1.2. Introduction to Stem Cells .................................................................................................. 29

1.2.1. Definition and classification .......................................................................................... 29

1.2.2. Endogenous stem cells in lung ...................................................................................... 30

1.2.3. Mesenchymal stem cells (MSCs) .................................................................................. 32

1.2.4. Induced-pluripotent stem cells (iPSCs) ......................................................................... 33

1.2.5. iPSC-derived MSCs ...................................................................................................... 34

1.3. MSCs-based Therapy .......................................................................................................... 37

1.3.1. Efficacy of MSCs in animal models .............................................................................. 37

1.3.2. Clinical trials ................................................................................................................. 44

1.3.3. Mechanisms ................................................................................................................... 46

1.3.3.1. Engraftment and regeneration ............................................................................... 46

1.3.3.2. Immunoregulation................................................................................................. 47

1.3.3.3. Mitochondrial transfer .......................................................................................... 48

1.4. Aims of Study ....................................................................................................................... 52

Chapter 2 Material and Methods ............................................................................. 54

2.1 Materials ............................................................................................................................... 55

2.1.1 Reagents and chemicals ................................................................................................. 55

2.1.2 Experimental animals ..................................................................................................... 55

2.1.3 Human iPSC-MSCs ........................................................................................................ 56

2.1.4 Human bone marrow – derived MSCs (BM-MSCs) ...................................................... 57

2.1.5 BEAS-2B cells ............................................................................................................... 57

2.1.6 Primary human airway smooth muscle cells (ASMCs).................................................. 58

2.2 Methods ................................................................................................................................. 59

2.2.1 MSCs treatment to cigarette smoke-exposed rats .......................................................... 59

2.2.2 Histology and morphometric analysis of rat lungs ......................................................... 61

2.2.3 iPSC-MSCs treatment to ozone-exposed mice ............................................................... 62

2.2.4 Measurement of airways hyper-responsiveness (AHR) of mice .................................... 65

2.2.5 Measurement of cells and cytokines in BAL of mice ..................................................... 65

Page 11: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

vi

2.2.6 Isolation of intact mitochondria from mouse lungs ........................................................ 66

2.2.7 Generation of cigarette smoke medium (CSM) .............................................................. 67

2.2.8 Co-culture of BEAS-2B cells with MSCs. ..................................................................... 67

2.2.9 Determination of ATP content in BEAS-2B cells following co-culture ......................... 68

2.2.10 BM-MSC and iPSC-MSC conditioned medium treatment of CSM-treated BEAS-2B

cells ......................................................................................................................................... 69

2.2.11 Immunohistochemistry ................................................................................................. 70

2.2.12 Terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL)

assay ........................................................................................................................................ 71

2.2.13 Total protein extraction from cell culture ..................................................................... 72

2.2.14 Western blot .................................................................................................................. 73

2.2.15 Enzyme-linked immunosorbent assay (ELISA) ........................................................... 73

2.2.16 Treatment of CSM or H2O2 to ASMCs or iPSC-MSCs ................................................ 74

2.2.17 Cell viability assay ....................................................................................................... 75

2.2.18 Measurement of mitochondrial ROS ............................................................................ 75

2.2.19 Mitochondrial membrane potential .............................................................................. 76

2.2.20 Annexin V staining ....................................................................................................... 76

2.2.21 Direct co-culture of ASMCs and iPSC-MSCs .............................................................. 77

2.2.22 Detection of mitochondrial transfer in co-culture of ASMCs and iPSC-MSCs ........... 78

2.2.23 iPSC-MSCs-conditioned medium treatment to ASMCs .............................................. 80

2.2.24 Transwell co-culture between ASMCs and iPSC-MSCs .............................................. 80

2.2.25 Statistical analysis ........................................................................................................ 80

Chapter 3. iPSC-MSCs Attenuate CS-induced Damage in Airway Epithelial Cells

through Mitochondrial Transfer and Paracrine Effect .......................................... 82

3.1. Introduction ......................................................................................................................... 83

3.2. Effects of iPSC-MSCs on CS-induced lung damage in rats. ............................................ 85

3.2.1. iPSC-MSCs attenuated CS-induced airspace enlargement, fibrosis and inflammation in

rat lung .................................................................................................................................... 85

3.2.2. iPSC-MSCs attenuated CS-induced apoptosis/proliferation imbalance in rat lung

epithelium. .............................................................................................................................. 90

3.2.3. Mitochondrial transfer from MSCs to adjacent lung cells ............................................ 91

3.3. Mitochondrial Transfer from MSCs to BEAS-2B cells in vitro ....................................... 94

3.3.1. Mitochondrial transfer from MSCs to BEAS-2B cells .................................................. 94

3.3.2. CSM promoted mitochondrial transfer from MSCs to BEAS-2B cells ........................ 96

3.3.3. Co-culture with iPSC-MSCs elevated ATP levels in BEAS-2B cells ........................... 96

3.4. iPSC-MSCs Attenuated CSM-induced Apoptosis and Proliferation Imbalance in

BEAS-2B Cells by Paracrine Effect .......................................................................................... 98

3.4.1. iPSC-MSCs-CdM attenuates CSM-induced apoptosis in BEAS-2B cells .................... 98

3.4.2. Effects of iPSC-MSCs-CdM on apoptosis and proliferation of BEAS-2B cells were

SCF-dependent ...................................................................................................................... 101

3.5. Discussion ........................................................................................................................... 105

Chapter 4. iPSC-MSCs attenuated CSM-induced Mitochondrial Dysfunction in

Human Airway Smooth Muscle Cells .................................................................... 115

4.1. Introduction ....................................................................................................................... 116

Page 12: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

vii

4.2. Oxidative Stress-induced Mitochondrial Dysfunction in ASMCs and iPSC-MSCs .... 119

4.2.1. Oxidative stress reduced the viability of ASMCs ........................................................ 119

4.2.2. Oxidative stress induced mitochondrial ROS in ASMCs ............................................ 121

4.2.3. Oxidative stress induced reduction of ΔΨm in ASMCs .............................................. 123

4.2.4. Oxidative stress reduced cell viability of iPSC-MSCs ................................................ 125

4.2.5. Oxidative stress induced mitochondrial ROS in iPSC-MSCs ..................................... 127

4.2.6. Oxidative stress induced reduction of ΔΨm in iPSC-MSCs ....................................... 129

4.3. Direct Co-culture with iPSC-MSCs Attenuated CSM-induced Mitochondrial

Dysfunction and Apoptosis in ASMCs .................................................................................... 131

4.3.1. iPSC-MSCs prevented and reversed CSM-induced mitochondrial ROS in ASMCs .. 131

4.3.2. iPSC-MSCs prevented but did not reverse CSM-induced reduction in ΔΨm in ASMCs

.............................................................................................................................................. 134

4.3.3. iPSC-MSCs prevented CSM-induced apoptosis of ASMCs ....................................... 136

4.4. Paracrine Effects are only Partly Involved in the Protective Effects of iPSC-MSCs on

CSM-induced Mitochondrial Dysfunction and Apoptosis in ASMCs .................................. 138

4.4.1. Effects of conditioned medium from iPSC-MSCs on CSM-induced mitochondrial

dysfunction in ASMCs .......................................................................................................... 138

4.4.2. Effect of iPSC-MSCs-CdM on CSM-induce apoptosis of ASMCs ............................. 139

4.4.3. Effects of trans-well co-culture with iPSC-MSCs on CSM-induced mitochondrial

alterations in ASMCs ............................................................................................................ 142

4.4.4. Effect of trans-well co-culture with iPSC-MSCs on CSM-induced apoptosis in ASMCs

.............................................................................................................................................. 144

4.5. Mitochondrial Transfer from iPSC-MSCs to ASMCs .................................................... 145

4.5.1. Mitochondrial transfer from iPSC-MSCs to ASMCs .................................................. 145

4.5.2. CSM increased mitochondrial transfer from iPSC-MSCs to ASMCs ......................... 145

4.6. Discussion ........................................................................................................................... 149

Chapter 5. Effects of iPSC-MSCs on Ozone-induced Airway

Hyper-responsiveness, Lung Inflammation, Apoptosis and Mitochondrial

Dysfunction in Mice ................................................................................................. 157

5.1. Introduction ....................................................................................................................... 158

5.2. Results ................................................................................................................................ 159

5.2.1. iPSC-MSCs prevented ozone-induced AHR ............................................................... 159

5.2.2. Effect of iPSC-MSCs on ozone-induced infiltration of inflammatory cells in the lung

.............................................................................................................................................. 161

5.2.3. Effect of iPSC-MSCs on cytokine contents of BALF ................................................. 164

5.2.4. Effects of iPSC-MSCs on apoptosis and proliferation in ozone-exposed lung tissue . 166

5.2.5. Effect of iPSC-MSCs on ozone-induced cellular ROS and mitochondrial dysfunction in

mouse lungs .......................................................................................................................... 169

5.3. Discussion ........................................................................................................................... 172

Chapter 6. General Discussion ................................................................................ 177

6.1. General Discussion ............................................................................................................ 178

6.2. Conclusion .......................................................................................................................... 190

6.3. Future Studies .................................................................................................................... 192

References ................................................................................................................. 194

Page 13: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

viii

List of Figures

Figure 1.1 Cause of airflow limitation in airways of patients with COPD.. ................. 3

Figure 1.2 Measurement of airflow limitation by spirometry. ...................................... 3

Figure 1.3 Prevalence of COPD (GOLD stage 2 or higher) in 2007 ............................ 7

Figure 1.4 Interplay of multiple events leading to emphysema .................................. 10

Figure 1.5 Generation of mitochondrial ROS from electron transport chain. ............ 27

Figure 1.6 Change of potency: from somatic cells to iPSC-MSCs ............................. 37

Figure 1.7 Proposed mechanism of mitochondrial transfer. ....................................... 51

Figure 2.1 Schematic diagram of BM-/iPSC-MSCs treatments in a CS-exposed rat

model ..................................................................................................................... 60

Figure 2.2 The set-up of the ventilated smoking chamber .......................................... 61

Figure 2.3 The set-up of the ventilated chamber for ozone exposure ......................... 63

Figure 2.4 Schematic diagram of iPSC-MSCs treatments in an ozone-exposed mouse

model ..................................................................................................................... 64

Figure 2.5 Protocols of direct co-culture of iPSC-MSCs and ASMCs ....................... 79

Figure 3.1 Human iPSC-MSCs and BM-MSCs alleviated CS-induced airspace

enlargement in rats ................................................................................................ 87

Figure 3.2 BM-MSCs and iPSC-MSCs attenuated CS-induced lung fibrosis in rats . 88

Figure 3.3 BM-MSCs and iPSC-MSCs ameliorated CS-induced inflammation in rat

lungs ...................................................................................................................... 89

Figure 3.4 BM-MSCs and iPSC-MSCs attenuated CS-induced increase in apoptosis

Page 14: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

ix

and reduction of proliferation in rat lung epithelium ............................................ 92

Figure 3.5 Mitochondria transfer from MSCs to lung epithelium in rats ................... 93

Figure 3.6 Mitochondrial transfer from BM-MSCs and iPSC-MSCs to BEAS-2B

cells in co-culture .................................................................................................. 95

Figure 3.7 CSM promoted mitochondrial transfer ...................................................... 97

Figure 3.8 iPSC-MSCs restored intracellular ATP levels of BEAS-2B cells ............. 98

Figure 3.9 CdM from BM-MSCs and iPSC-MSCs ameliorated CSM-induced

apoptosis/proliferation imbalance in BEAS-2B cells. ........................................ 100

Figure 3.10 Secretion of SCF by BM-MSCs and iPSC-MSCs and expression of C-kit

by BEAS-2B cells ............................................................................................... 103

Figure 3.11 The anti-apoptotic and pro-proliferation activity of iPSC-MSCs-CdM was

partly through SCF .............................................................................................. 104

Figure 4.1 Effects of H2O2 and CSM on the viability of ASMCs ............................. 120

Figure 4.2 H2O2 and CSM induced mitochondrial ROS in ASMCs ......................... 122

Figure 4.3 H2O2 and CSM reduced ΔΨm in ASMCs ................................................ 124

Figure 4.4 Effects of H2O2 and CSM on viability of iPSC-MSCs. ........................... 126

Figure 4.5 Effects of H2O2 and CSM on mitochondrial ROS of iPSC-MSCs .......... 128

Figure 4.6 Effects of H2O2 and CSM on ΔΨm of iPSC-MSCs ................................. 130

Figure 4.7 Effects of direct co-culture with iPSC-MSCs on mitochondrial ROS in

ASMCs ................................................................................................................ 133

Figure 4.8 Effects of direct co-culture with iPSC-MSCs on ΔΨm in ASMCs.......... 135

Figure 4.9 Effects of preventive co-culture with iPSC-MSCs on apoptosis in ASMCs

Page 15: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

x

treated with CSM ................................................................................................ 137

Figure 4.10 Effects of iPSC-MSCs-CdM on mitochondrial ROS and ΔΨm in ASMCs

............................................................................................................................. 140

Figure 4.11 Effects of iPSC-MSCs-CdM on CSM-induced apoptosis in ASMCs ... 141

Figure 4.12 Effects of trans-well co-culture with iPSC-MSCs on mitochondrial ROS

and ΔΨm in ASMCs ............................................................................................ 143

Figure 4.13 Effects of trans-well co-culture with iPSC-MSCs on apoptosis in ASMCs

............................................................................................................................. 144

Figure 4.14 Mitochondrial transfer from iPSC-MSCs to ASMCs ............................ 147

Figure 4.15 CSM enhanced mitochondrial transfer from iPSC-MSCs to ASMCs ... 148

Figure 5.1 iPSC-MSCs prevented ozone-induced AHR ........................................... 160

Figure 5.2 Effects of iPSC-MSCs on cell counting of BAL from ozone-exposed mice

............................................................................................................................. 163

Figure 5.3 Effects of iPSC-MSCs on cytokine levels in BALF after ozone exposure

............................................................................................................................. 165

Figure 5.4 Effects of iPSC-MSCs on ozone-induced apoptosis in mouse lung ........ 167

Figure 5.5 Effects of ozone and iPSC-MSCs on cellular proliferation in lung ......... 168

Figure 5.6 Effects of iPSC-MSCs on ozone-induced ROS and mitochondrial

dysfunction in lung ............................................................................................. 171

Figure 6.1 Summary of the effects of iPSC-MSCs on oxidative stress induced lung

cell damage ......................................................................................................... 191

Page 16: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

xi

List of abbreviations

AEC1s type 1 alveolar epithelial cells

AEC2s type 2 alveolar epithelial cells

AHR airway hyper-responsiveness

ASM airway smooth muscle

ASMC airway smooth muscle cell

ATP adenosine triphosphate

BAL bronchoalveolar lavage

BALF bronchoalveolar lavage fluid

bFGF basic fibroblast growth factor

BOLD Burden of Obstructive Lung Disease

BM bone marrow

BM-MSC bone marrow-derived mesenchymal stem cell

BSA bovine serum albumin

cAMP cyclic adenosine monophosphate

CAT COPD assessment test

CC10 clara cell secretory protein 10

CCL chemokine ligand

CdM conditioned medium

COPD chronic obstructive pulmonary disease

CRP C-reactive protein

CS cigarette smoke

CSM cigarette smoke medium

DAB 3,3'-diaminobenzidine

DAMP danger-associated molecular pattern

DAPI 4,6-diamidino-2-phenylindole

DMEM Dulbecco‟s modified Eagle medium

EGF epidermal growth factor

Page 17: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

xii

ELISA enzyme-linked immunosorbent assay

ESC embryonic stem cell

FACS fluorescence-activated cell sorting

FEV₁ forced expiratory volume in 1 s

FISH fluorescent in situ hybridization

FVC forced vital capacity

GBD Global Burden of Disease

GM-CSF granulocyte macrophage-colony stimulating factor

GOLD global initiative for chronic obstructive lung disease

GPx glutathione peroxidase

GRO growth-related oncogene

GSH glutathione

GSSG oxidized glutathione

GVHD graft-versus-host disease

HBSS Hank‟s Balanced Salt Solution

HNA human nuclear antigen

HRP horseradish peroxidase

HSC haematopoietic stem cell

IFN interferon

IL interleukin

IP-10 IFN-γ-inducible protein 10

iPSC induced-pluripotent stem cell

iPSC-MSC induced-pluripotent stem cell-derived mesenchymal stem cell

iPSC-MSCs-CdM iPSC-MSCs-conditioned medium

JC-1 5,5‟,6,6‟-tetrachloro-1,1‟,3,3‟-tetraethylbenzimidazolylcarbocyanineiodide

KC keratinocyte-derived chemokine

KIF5 kinesin motor protein-5

LABA long-acting β2-agonist

Lm mean linear intercept

Page 18: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

xiii

LPS lipopolysaccharide

MCP monocyte chemotactic protein

MDA malondialdehyde

MEF mouse embryonic fibroblast

MHC major histocompatibility complex

MIP macrophage inflammatory protein

Miro mitochondrial Rho GTPase

MMP matrix metalloproteinases

mMRC Modified British Medical Research Council

MPO myeloperoxidase

MSC mesenchymal stem cell

MTT methylthiazolyldiphenyl-tetrazoliumbromide

PGE2 prostaglandin E2

PLATINO Latin American Project for Investigation of Obstructive Lung Disease

PRR pattern-recognition receptor

PT permeability transition

PVDF polyvinylidene fluoride

RL pulmonary resistance

ROS reactive oxygen species

SA sham air

SABA short-acting β2 agonist

SCF stem cell factor

SCID severe combined immunodeficient

SDS sodium dodecyl sulfate

SEM standard error of mean

SFTPC surfactant protein C

SLPI secretory leukoprotease inhibitor

SOD superoxide dismutase

TGF transforming growth factor

Page 19: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

xiv

TIMP tissue inhibitors of MMP

TL telomere length

TMB tetramethylbenzidine

TNF tumor necrosis factor

TNT tunnelling nanotube

TSG-6 tumor necrosis factor-stimulated gene-6

TUNEL terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling

VEGF vascular endothelial growth factor

ΔΨm mitochondrial membrane potential

Page 20: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

1

Chapter 1. Introduction

Page 21: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

2

1.1. Introduction to Chronic Obstructive Pulmonary Disease

(COPD)

1.1.1. Definition and diagnosis of COPD

The Global Initiative for Chronic Obstructive Lung Disease (GOLD) defines

chronic obstructive pulmonary disease (COPD) as „a common preventable and

treatable disease‟ which is „characterized by persistent airflow limitation that is

usually progressive and associated with an enhanced chronic inflammatory response

in the airways and the lung to noxious particles or gases‟ (263). The chronic

inflammation drives small airways disorder named obstructive bronchiolitis leading to

structural alterations and narrowing of small airways, and emphysema characterized

by destruction of parenchyma which causes separation of alveolar from the small

airways and loss of lung elastic recoil (263). Obstructive bronchiolitis and

emphysema together causes the persistent airway flow limitation in patients with

COPD (Figure 1.1), but the contribution from each of them varies within individual

patients (263).

COPD is diagnosed by post-bronchodilator spirometry to measure persistent

airway limitation. Spirometry measures volume of air that a patient can exhale at a

maximal force (as long and as quickly as possible) after a maximal deep inspiration

(208). The total volume of air exhalation with greatest effort of the patient is defined

as forced vital capacity (FVC) (208). The total volume of air exhalation with greatest

effort of the patient in the first second is defined as forced expiratory volume in 1 s

(FEV₁) (208). Persistent airway limitation is confirmed when the ratio of FEV₁ to

Page 22: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

3

FVC is less than 0.7 (263) (Figure 1.2).

Figure 1.2 Measurement of airflow limitation by spirometry. Patients are asked to

exhale as long and as quickly as possible. Exhalation volume from (A) normal lungs

or (B) obstructive lungs is plotted against time. FVC: forced vital capacity; FEV1:

forced expiratory volume in the first second; Airway obstruction is defined if

FEV1/FVC is less than 0.7. The figure is reproduced based on plots by Ranu H et al,

Ulster Med J (208).

Figure 1. 1 Cause of airflow limitation in airways of patients with

COPD. Compared to (A) healthy lungs, lungs with (B) COPD demonstrate

parenchymal destruction leading to emphysema, mucosal and peribronchial

inflammation and fibrosis leading to obliterative bronchiolitis, and mucus

hypersecretion leading to luminal obstruction. As a result, patients with

COPD develop irreversible airflow limitation. Adapted from Barnes PJ, N

Engl J Med (22).

Page 23: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

4

Further assessment of COPD includes determination of levels of symptoms,

severity of airflow limitation, exacerbation risk and comorbidities. The major

symptoms of the disease include chronic and progressive dyspnea, cough and sputum

production. The levels of symptoms can be assessed by questionnaires such as

Modified British Medical Research Council (mMRC) questionnaire on breathlessness

and COPD Assessment Test (CAT). The severity of airflow limitations is graded into

four different stages by GOLD based on the post-bronchodilator FEV₁ (Table 1.1).

The spirometric criteria for Mild, Moderate, Severe and Very Severe severities are

80%, 50% and 30% of predicted FEV₁. Exacerbations refer to short periods (at least

48 hours) of symptom worsening such as increased cough and dyspnea which is

beyond normal variations (65, 263). Exacerbations lead to reduced quality of life,

accelerated disease progression and increased risk of death (65, 71, 236).

Exacerbations usually call for changes in medication by which their severity can be

defined, including change of inhaled treatment (mild exacerbation), medical

intervention such as antibiotic and oral steroids (moderate exacerbation) and

hospitalization (severe exacerbation). A combined assessment of COPD which

integrates levels of symptoms, severity of airway limitation and exacerbation history

is suggested by GOLD. The assessment groups patients with COPD into four

sub-groups demonstrating different burden of symptom and risk of exacerbation

(Group A, low risk and less symptoms; Group B, low risk and more symptoms; Group

C, high risk and less symptoms; Group D, high risk and more symptoms). The burden

of symptoms is defined high or low by mMRC or CAT grade. The risk of

Page 24: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

5

exacerbation is defined high if the severity of airway limitation is GOLD 3-4 or there

are two or more exacerbations in the preceding year. In addition to the combined

assessment approach, assessment of comorbidities is also important. COPD is highly

associated with comorbidities including ischaemic heart disease, diabetes, skeletal

muscle wasting, cachexia, osteoporosis, depression and lung cancer (65, 263). The

comorbidities can increase the risk of admission to hospital and death (234).

Table 1.1 GOLD Severity of airflow limitation in COPD (FEV₁/FVC<0.70)

GOLD stage Severity Post-bronchodilator FEV₁

Stage 1 Mild FEV₁≥ 80% of predicted

Stage 2 Moderate 50% ≤FEV₁< 80% of predicted

Stage 3 Severe 30% ≤FEV₁< 50% of predicted

Stage 4 Very severe FEV₁< 30% of predicted

1.1.2. Epidemiology of COPD

The prevalence of COPD varies among different reports, maybe due to different

assessing criteria and methods of survey and analysis (96). A recent meta-analysis

report estimated that the worldwide COPD cases in people aged 30 or more had

increased from 227.3 million in 1990 to 384.0 million in 2010, while the prevalence

was 10.7% in 1990 and 11.7% in 2010 (3). The actual prevalence may be higher than

the data from epidemiological studies as COPD is considered to be under-diagnosed

in general (259). It is estimated that 60-85% patients with COPD may be undiagnosed,

Page 25: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

6

especially for the mild and moderate severities (GOLD stage 1 or 2) (114).

COPD prevalence varies among different countries and different groups of people

(e.g. gender and smoking history) (Figure 1.3). For example, the estimation of male

population with COPD of GOLD 2 severity or above was estimated as 22.2% in Cape

Town, South Africa, in comparison to 8.5% in Reykjavik, Iceland (168). Based on the

data from two large multinational studies, the Burden of Obstructive Lung Disease

(BOLD) study and the Latin American Project for Investigation of Obstructive Lung

Disease (PLATINO) study, the prevalence of COPD in different countries ranges from

3% to 23% (39, 168). The prevalence of COPD has been stable in some developed

countries, while in the United States, it is the only common cause of death that has

increased over the last 40 year (23). In developing countries such as some African

countries, the prevalence increased more dramatically (65). Variations among nations

were also observed in mortality studies. For example, the death rate of patients with

COPD was estimated to be 130.5/100,000 in China in comparison to 4.4/100,000 in

Japan, although the prevalence is similar between the two countries (168). COPD was

ranked as the 6th leading cause of death worldwide in 1990, and was predicted to be

the 3rd leading cause by 2020. A more recent study estimated COPD as 4th leading

cause of death in 2030 (172). Based on the data of 2010 Global Burden of Disease

(GBD) study, COPD led to 2.9 million deaths, accounting for about 5% of total deaths

globally (161, 181).

The economic burden associated with COPD is huge. For example, in 2003 the

United States spent US$ 32.1 billion in total on COPD, US$ 18.0 billion of which on

Page 26: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

7

direct healthcare costs and the rest on indirect morbidity and mortality costs (168). In

2010 the number increased to US$ 49.9 billion in total and US$ 29.5 billion on direct

healthcare costs (49). In United Kingdom, in 2007 the healthcare cost of COPD is

three fold higher than asthma and the consultation rate of COPD is two to four-fold

higher than ischemic heart disease (23).

Figure 1. 3 Prevalence of COPD (GOLD stage 2 or higher) in 2007. Combined

results from BOLD study and PLATINO study are demonstrated in the figure. The

data were collected from a city or region of the country thus do not represent national

population. Adapted from Mannino DM et al, the Lancet (169).

1.1.3. Risk factors of COPD

COPD results from the interaction between gene and environmental stimuli (65).

The most common genetic risk for COPD is deficiency of α1 antitrypsin, a circulating

inhibitor of serine proteases. Mutation of genes encoding matrix metalloproteinase-12

(MMP-12) (113) and transforming growth factor β1 (47) have also been reported in

association with COPD.

Page 27: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

8

Cigarette smoking is the most common worldwide risk factor for COPD.

Cigarette smoking is associated with an increase in prevalence of respiratory

symptoms, lung function abnormalities, annual rate of FEV1 decline, and COPD

mortality rate (136). The association between development of COPD and cigarette

smoking is dose-dependent (292), and smoking cessation reduces rates of morbidity

and mortality in comparison to continued smoking (209). Besides normal cigarette

smoking, passive exposure to cigarette smoke in the environment is also a risk factor

for COPD (73). For example, in China where 30% of the world‟s cigarettes are

consumed (277), 12.1% of male and 51.3% of female nonsmokers are exposed to

environmental smoke at home, while 26.7% of male and 26.2% of female

non-smokers are exposed to environmental smoke in the work place in 2001 (92). In

never-smokers with COPD, the passive exposure to cigarette smoke contributed to 1.9

million additional deaths in China (280). Maternal smoking during pregnancy may

also increase the risk of COPD to the fetus (245). Although cigarette smoking is

recognized as the major risk factor for COPD, COPD does not develop in some

people with the same smoking history (263). In addition, COPD may also develop in

some nonsmokers (48, 141), indicating the importance of other risk factors.

In addition to environmental smoke, other particles and gases in environment can

also contribute to development of COPD, such as occupational exposure to

hazardous dusts and fume (17) and indoor pollutant from biomass burning for family

cooking and heating (36, 75, 191, 207).

Age is commonly considered as a risk factor for COPD as the prevalence,

Page 28: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

9

morbidity and mortality increases along with increased age (38, 126, 137). Gender

was regarded as one risk factor in the past with men presenting higher prevalence and

mortality of COPD than women (233). However, the prevalence of COPD in men and

women from high-income countries has become more similar now (39, 233).

Development of COPD can be also associated with many other risk factors

including impaired lung development, asthma, bronchial hyper-responsiveness,

asthma, infections and social status (168).

1.1.4. Pathogenesis of COPD

The most significant pathophysiological feature of COPD is airflow limitation

resulting from narrowing of the small airway compartment and destruction of

parenchyma, both leading to gas trapping and subsequently hyperinflation (186).

Other pathophysiological changes that may present in patients with COPD include gas

exchange abnormalities, mucus hypersecretion, pulmonary hypertension and other

comorbidities (263). The pathological changes in patients with COPD mainly include

infiltration of various inflammatory cells, structural changes of small airways such as

thickening of walls and mucus overproduction, and destruction of parenchyma (60,

263). The pathogenesis of COPD, including the mechanisms driving the chronic

inflammation, remodeling of small airways and parenchymal destruction is still not

fully understood (65). Current evidence indicates a complex interplay between

oxidative stress, inflammatory responses, protease and anti-protease imbalance,

ageing, and changes in apoptosis, proliferation and tissue repair process (57). A

Page 29: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

10

simple illustration of the factors that leads to emphysema is shown in figure 1.4. The

details are described in following sections.

Figure 1. 4 Interplay of multiple events leading to emphysema. Cigarette smoke

exposure activates airway epithelial cells, which release early inflammatory cytokines

that recruit macrophages and neutrophils. They can also activate dendritic cells and

the subsequent formation of CD8+ or CD4+ T cells. Along the chronic inflammation,

various cell types are activated by inflammatory mediators and in response their

secretory function may also become activated. Their anti-oxidant system may become

defective, while cells such as neutrophils will release ROS, leading to

oxidant/anti-oxidant imbalance. The resulting oxidative stress can induce senescence

and mitochondrial dysfunction. Mitochondrial dysfunction and accelerated ageing can

further amplify the oxidative stress. Apoptosis can be induced by external cytokines

such as perforin and granzymes, as well as by cytochrome c release due to

mitochondrial damage. Both accelerated ageing and apoptosis may lead to airspace

enlargement. The immune cells and structural cells, especially neutrophils and

macrophages can release protease. When the anti-protease of the cells is not enough to

counterbalance such effect, excess protease activity can lead to destruction of alveolar

structures.

Page 30: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

11

1.1.4.1. Oxidative stress

COPD is associated with persistent oxidative stress. Markers of oxidative stress,

such as lipid peroxidation byproducts 4-hydroxy-2-nonenal, 8-isoprostane and

malondialdehyde (MDA), are found increased in lungs of patients with COPD (135,

206). The exhaled breath or breath condensates collected from patients with COPD

also contains increased levels of various markers of oxidative stress including

hydrogen peroxide, carbon monoxide, myeloperoxidase (MPO), 8-isoprostane and

MDA (27, 176, 194, 195). Exacerbations are associated with further enhanced

oxidative stress (263).

Reactive oxygen species (ROS) are free oxygen radicals with unpaired elections

that can oxidize various molecules such as proteins, DNA and lipids (133). Typical

forms of ROS include superoxide radical (O2-), hydroxyl radical (OH

-) and hydrogen

peroxide (H2O2). Under normal conditions, the major sources of ROS are respiration

of mitochondria, xanthine oxidase activity and NADP oxidase activity (167). During

cigarette smoking, the structural and immune cells in the lung are exposed to high

levels of exogenous oxidants from inhaled cigarette smoke (CS). One puff of CS

contains about 1015

oxidants in the gas phase and 1018

oxidants in the tar phase, as

well as 3000 ppm of nitric oxide (NO) (57, 58). Exposure to CS leads to release of

inflammatory mediators and ROS from epithelial cells and immune cells, leading to

release of various inflammatory mediators and ROS (164). Although CS is an obvious

source of oxidants and indeed a major risk factor for COPD, oxidative stress is

persistent after smoking cessation in patients with COPD (160), indicating that the

Page 31: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

12

cellular accumulation and release of ROS is an important source of oxidative stress

during the progression of COPD. Increased release of ROS was reported in alveolar

macrophages and peripheral neutrophils from patients with COPD (115, 220). The

increased production of ROS may be due to the mitochondrial ROS overproduction

and altered xanthine oxidase activity. Increased mitochondrial ROS was reported in

airway smooth muscle cells from patients with COPD (273), and products of xanthine

oxidase activity were detected in bronchoalveolar fluid (BALF) from patients with

COPD (202). Although the increased ROS may be mainly in the form of less potent

radicals such as superoxide anion and hydrogen peroxide, they can be converted to

more potent and destructive ROS such as peroxynitrite (ONOO-) by rapid reaction

between superoxide radical and NO (261), and hypohalous acids (HOCl and HOBr)

by MPO-catalyzed reactions between hydrogen peroxide and Cl- or Br

- (224). MPO

levels in neutrophils are reported to be elevated in patients with COPD (1).

ROS generation is balanced by the activity of anti-oxidant defense in the normal

body. The first anti-oxidant barrier against the noxious particles or gases is the

epithelial lining fluid containing anti-oxidants such as vitamin C, vitamin E and uric

acid (134). Reduced levels of vitamin C and vitamin E in the lung were associated

with declining lung function in patient with COPD (134). Regarding cellular

anti-oxidant activity, one of the most important reactions is the synthesis of

glutathione (GSH), an anti-oxidant that depletes oxidants via conversion to oxidized

glutathione (GSSG). Under acute oxidative stress, epithelial cells from healthy

subjects presented elevated level of glutamate-cysteine ligase which is an enzyme

Page 32: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

13

involved in GSH synthesis (228). Such anti-oxidant defense was impaired in

bronchial epithelial cells and alveolar macrophages from patients with COPD, as

indicated by lower expression of glutamate-cysteine ligase (228). Another

anti-oxidant defense system is mediated through superoxide dismutase (SOD) which

converts superoxide radicals into hydrogen peroxide (167). Hydrogen peroxide can be

further converted to oxygen and water directly by catalase (53) or by reaction with

GSH mediated by glutathione peroxidase (GPx) (232). As mentioned earlier, under

excess ROS production, superoxide anions can react rapidly with NO to form

peroxynitrite. Peroxynitrite can inhibit SOD, thus reducing the anti-oxidant defensive

activity (261). The persistent high ROS burden and defective anti-oxidant defense

lead to oxidative stress of the cells.

1.1.4.2. Chronic airway inflammation

Chronic and persistent airway inflammation is observed in patients with COPD,

and higher grades of COPD severity is associated with higher severities of

inflammation (108). Infiltrated inflammatory cells include neutrophils, macrophages,

B-cells, lymphoid aggregates and T cells (107). In addition to the increased numbers

in the lung, the function of inflammatory cells may also change in line with the

pathogenesis of COPD (109).

The number of macrophages increases in bronchial submucosa along with the

severity of airflow limitation in patients with COPD (108). Macrophages can

contribute to oxidative stress, destruction of structural cells and further progression of

Page 33: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

14

inflammatory responses by releasing ROS, extracellular matrix proteins, leukotrienes,

multiple cytokines, chemokines and matrix metalloproteinases (MMPs) (57). One

primary function of macrophages in normal lungs is bacterial clearance through

phagocytosis, which may be defective in patients with COPD (72). Ex vivo studies

have demonstrated a defective phagocytositic ability against bacteria commonly

detected in airways in both alveolar macrophages (29) and monocyte-derived

macrophages (250) from patients with COPD. Moreover, the secretion of

inflammatory mediators is elevated in alveolar macrophages from smokers and

ex-smokers (101).

Increased number of neutrophils in sputum has been reported to associate with

higher severity of airflow obstruction and faster decline of lung function in patients

with severe COPD (187). Neutrophils are distributed in various sites particularly in

bronchial epithelium, bronchial glands (217) and in the airway smooth muscle (20).

When recruited to the airways, neutrophils become activated and release ROS,

inflammatory cytokines, neutrophil elastase, MMPs and MPO (240). In vitro data

suggest that the phagocytic ability of neutrophils can be impaired by cigarette smoke

extract (242).

In general, infiltrating neutrophils and macrophages are considered to be the

major players in the innate immune response in COPD. However, increased numbers

of eosinophils have also been reported in sputum, BALF and the airway wall (140,

142, 156). Levels of pro-eosinophil cytokines IL-4 and IL-5 are also reported to be

increased in plasma cells of patients with COPD (298). The exact function of

Page 34: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

15

recruited eosinophils and its contribution to the development of COPD remains

unclear (57).

There is also an adaptive immune response to the inflammatory responses in

COPD. The number of dendritic cells has been reported to increase in epithelium and

adventitia of small airways of patients with COPD compared with never-smokers and

healthy smokers and correlated with the severity of airflow limitation (66). Dendritic

cells can present both self-antigens from tissue destruction and foreign antigens to

naive T cells, and numbers of both CD4+ and CD8+ T cells have been reported to

increase in lungs of patients with COPD (175, 213, 216). CD8+ T cells from lungs of

patients with COPD have been shown to release higher levels of inflammatory

mediators including tumor necrosis factor (TNF)-α, interferon-γ and granulocyte

macrophage-colony stimulating factor (GM-CSF).

1.1.4.3. Protease and anti-protease imbalance

Parenchymal destruction is one of the most significant pathological changes in

the lungs of patients with COPD. The resulting loss of elastic recoil leads to the

airflow limitation. Destruction of connective tissue, particularly lung elastin, by

excess protease activity is believed to play an important role in this process (25).

Active neutrophils can release serine proteases including neutrophil elastase,

cathepsin G and proteinase 3 (25). Various cysteine protease cathepsin can also be

released by different cells, including cathepsin S from airway smooth muscle,

cathepsin B, K and L from alveolar macrophages and cathepsin W from CD8+ T cells

Page 35: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

16

(25). Alveolar macrophages can provide another important protease family, MMPs,

particularly MMP-9. The alveolar macrophages from healthy smokers express more

MMP-9 than normal subject, while alveolar macrophages from patients of COPD

express an even higher level (154, 214).

The protease activities are balanced by anti-proteases in normal lungs. Serine

protease activity can be counteracted by two major serine protease inhibitors in the

lung, α1 antitrypsin and secretory leukoprotease inhibitor (SLPI). Genetic variability

of α1 antitrypsin that leads to a deficiency in circulating levels is a risk factor for

COPD. Cigarette smoking may lead to oxidized and impaired α1 antitrypsin activity,

resulting in lack of counterbalance against the activity of neutrophil elastase (46).

SLPI activity may also be reduced by oxidative stress (25). As for the other protease

family MMPs, their activity can be counteracted by tissue inhibitors of MMPs (TIMP).

Alveolar macrophages from normal subjects release increased amounts of TIMPs

after pro-inflammatory stimulation, indicating a counterbalance to the release of

MMPs. The increased release of TIMPs is reduced in the alveolar macrophages from

COPD patients, which may lead to excess activity of MMPs (214).

1.1.4.4. Apoptosis

In addition to the excess protease activity, the parenchymal destruction may also

be contributed to by increased cellular apoptosis in lung (67). Apoptosis and

proliferation are well equilibrated in healthy tissue. Increased apoptosis in endothelial

cells, alveolar epithelial cells, interstitial cells, neutrophils and lymphocytes has been

Page 36: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

17

reported in tissue sections of the lung from patients with COPD compared to healthy

subjects (223). Another study demonstrated that lung tissue from patients with COPD

exhibited increased apoptosis of alveolar epithelial cells, endothelial cells and

mesenchymal cells in association with an increased amount of the active subunit of

caspase-3, a central mediator of apoptosis (116). Moreover, persistent apoptosis of

alveolar epithelial cells and T-lymphocytes in bronchial bushings and bronchoalveolar

lavage has been observed after smoking cessation, as indicated by comparison

between ex-smokers with COPD, current smokers with COPD and healthy

never-smokers (103). Increased apoptosis in alveolar cells in the absence of adequate

compensation by proliferation leads to the gradual destruction of alveolar walls in the

lung (67). Apoptosis of alveolar wall and endothelial cells are reported to induce

emphysema even without infiltration of inflammatory cells in an animal model (67).

The increased apoptosis in the lung may be driven by multiple mechanisms which

are currently not well understood. Oxidative stress-induced attenuation of vascular

endothelial growth factor (VEGF) activity may be a mechanism of the elevated

apoptosis in COPD. It has been reported that increased apoptosis in epithelial and

endothelial alveolar septal cells in patients with emphysema is associated with

reduced expression of VEGF and VEGF receptor 2, which may lead to the endothelial

septal death (127). The level of VEGF in induced sputum from patients with COPD

has also been reported to decrease in severe COPD, in association with increased

levels of oxidative stress (125). In animal models, apoptosis induced by inhibition in

VEGF receptor (128, 199, 255) or VEGF (249) leads to emphysema even without

Page 37: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

18

obvious inflammation. The relationship between oxidative stress and apoptosis may

also be through mitochondrial dysfunction. Mitochondria are the major source of

cellular ROS production of cells under oxidative stress (134). Airway smooth muscle

cells (ASMCs) from patients with COPD have demonstrated increased mitochondrial

ROS compared to healthy subjects (273). In general, mitochondrial dysfunction can

be a potent driving force in inducing apoptosis through the mitochondrial pathways

(90), but more evidence is required to reveal the exact relationship between

mitochondrial damage and apoptosis in COPD. The inflammatory characteristic of

COPD may also contribute to apoptosis. The phagocytic ability of alveolar

macrophages against apoptotic airway epithelial cells is impaired in patients with

COPD (90), leading to insufficient clearance of apoptotic airway epithelial cells. The

recruitment of CD8+ cells into the lung may also induce an increase in apoptosis of

structural cells, by secretion of cytotoxic mediators such as perforins, granzymes and

TNF-α (26, 158). Indeed, increased infiltration of CD8+ T cell to alveolar walls is

associated with increased apoptosis in the lung (166).

Cell death, including apoptosis, is constantly counterbalanced by proliferation

under normal physiological conditions (67). However, whether the proliferative

activity is able to counterbalance increased apoptosis remains unclear (67). Increased

proliferative activity has been reported in the alveolar wall of patients with

emphysema (116, 287), which may indicate a compensatory mechanism to the

increased apoptosis. However, another report demonstrated similar proliferation of the

alveolar septal cells between patients with emphysema and healthy subjects with

Page 38: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

19

increased apoptosis of alveolar epithelial cells in patients with end-stage emphysema

(44).

1.1.4.5. Ageing

Age is a risk factor for COPD. On the one hand, age is associated with longer

exposure history to noxious gas or particles, on the other hand, the process of ageing

may also contribute to the pathogenesis of COPD (119). Lung function starts to

decline approximately after the age of 30 as a result of normal ageing (198), but in

patients with COPD the decline is accelerated (79, 163). The decline in lung function

of ageing lungs may be explained by progressive distal airspace enlargement, which

leads to loss of lung elastic recoil (120). Although the airspace enlargement is not as

destructive as is the case in lungs of patients with COPD (262), the similarity of the

functional and structural changes between ageing lungs and lungs of patients with

COPD suggests a possible relationship between them. COPD is characterized by

persistent oxidative stress, while oxidative stress-induced DNA damage is believed to

play an important role in cellular senescence according to the free radical theory (99).

Senescence of cells is associated with shortening of telomere length (TL) (218).

Indeed, it has been reported that TL of circulating lymphocytes is shortened in

smokers in a dose-dependent manner, but the effect was not amplified in smokers with

COPD (178). Another study demonstrated TL shortening in circulating leucocytes

from patients with COPD compared to healthy subjects of any age range (110, 219).

TL of alveolar epithelial cells, endothelial cells (254) and fibroblasts (180) are also

shortened in patients with emphysema compared with non-emphysematous subjects.

Page 39: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

20

Whereas oxidative stress may drive an accelerated ageing process, the ageing

process itself may also contribute to oxidative stress and inflammation, which is

evident from multiple animal studies. Acute CS exposure of mice has been reported to

induce a five-fold increase of GSH levels in BALF in two-month old mice, indicating

an anti-oxidant response. In mice of increasing age, these effects gradually diminished,

in association with increased inflammatory maker TNF-α and oxidative stress marker

8-hydroxy-2-deoxyuanosine in BALF (88). Another study suggested that older mice

had more neutrophils, inflammatory mediator keratinocyte-derived chemokine (KC, a

mouse homolog of IL-8) and MIP-2 in lungs after 9 days of cigarette smoke (177).

1.1.4.6. Bronchial epithelial cells and airway smooth muscle cells in COPD

Bronchial epithelial cells are the first cellular defensive barrier against noxious

gases and particles in lung. The epithelial injury caused by CS leads to the onset of

oxidative stress and inflammation in the lung. The direct exposure to CS induces the

intracellular release of endogenous molecules named danger-associated molecular

patterns (DAMP) which can be identified by pattern-recognition receptors (PRR) e.g.

Toll-like receptor 2 and 4 (190). This process activates epithelial cells to synthesize

and secrete early inflammatory mediators such as TNF-α, interleukin (IL)-1β and IL-8

(65). The release of these chemokine leads to recruitment of inflammatory cells such

as macrophages and neutrophils. Bronchial epithelial cells can also facilitate the onset

of the adaptive immune response in COPD. Dendritic cell migration, maturation and

activation are reported to be regulated by bronchial epithelial cells by releasing

Page 40: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

21

various chemokines such as macrophage inflammatory protein (MIP)-3α and

chemokine ligand 20 (CCL20) (66, 201). In addition to the initiation of the

inflammatory responses, bronchial epithelial cells also contribute to the persistent

inflammation in COPD by releasing multiple cytokines and chemokines including

IL-1β, TNF-α, IL-6, IL-8 and GM-CSF. For example, epithelial cells from smokers

with COPD express higher levels of mRNA and protein of monocyte chemotactic

protein 1 (MCP-1) and IL-8 compared to the cells from smokers without COPD (64).

Bronchial epithelial cells also play an important role in the anti-oxidant system in the

lung. As mentioned in previous sections, epithelial cells from patients with COPD

demonstrated reduced ability to elevate GSH production in response to oxidative

stress (228). In addition, bronchial epithelial cells also serve as a source of the

anti-protease SLPI (25).

Patients with COPD present with an increased mass of airway smooth muscle

(ASM) in small airways (37, 59), correlating with declining lung function (57). The

thickened airway smooth muscle layer contributes to a thicker airway wall, which

plays an important role in the airflow obstruction of COPD (56). Due to its contractile

properties, ASM is the major modulator of bronchomotor tone in the airway.

Exaggerated constriction of the airways is a feature of patients with COPD, and

relaxation of contracted ASM by bronchodilators is a major therapeutic intervention

in the treatment of COPD (19). The role of ASM in the pathogenesis of COPD is not

limited to its contractile properties. Similar to epithelial cells, ASM cells (ASMCs)

also release inflammatory cytokines and chemokines including IL-6, IL-8, MCP-1,

Page 41: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

22

MCP-2, MCP-3, growth-related oncogene (GRO)-α, IFN-γ-inducible protein (IP)-10

and GM-CSF (56). In addition, increased levels of transforming growth factor-β

(TGF-β) in patients with COPD can lead to reduced expression of catalase and

manganese-SOD in ASMCs, resulting in reduced anti-oxidant activity (174).

Unlike bronchial epithelial cells, ASMCs are usually not directly exposed to

noxious particles or gases, although this may happen when the epithelial barrier is

severely damaged by CS (102, 226). Therefore, the release of inflammatory mediators

from ASMCs is most likely not the result of a direct response to CS, but rather

induced by the effect of cytokines released by neighboring cells, e.g. epithelial cells.

As mentioned earlier, CS causes direct injury to bronchial epithelial cells leading to

release of inflammatory mediators. Amongst them, IL-1β and TNF-α can induce the

release of IL-8 from ASMCs (121), and IFN-γ and TNF-α can induce the release of

IP-10 from ASMCs (98). Moreover, bronchial epithelial cells may also contribute to

the increased TGF-β levels in COPD. An in vitro study demonstrated that epithelial

cells from smokers and patients with COPD secreted higher levels of TGF-β than the

cells from normal subjects (248). As mentioned earlier, high levels of TGF-β can

reduce the anti-oxidant activity in ASMCs. Therefore the secretory activities of

cytokines and a defective anti-oxidant system in ASMCs may partly result from an

interaction with injured epithelial cells.

1.1.4.7. Mitochondria and COPD

Mitochondria are double-membrane organelles with a unique structure and

Page 42: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

23

important function. They are composed of an outer membrane, inner membrane

inter-membrane space and matrix (11). The inner membrane is highly folded into

cristae, and is impermeable to ions. Respiratory chain complex proteins reside on the

inner membrane where they mediate electron transport chain and the subsequent

accumulation of protons in the inter-membrane space (170). The proton gradient

between the two sides of the inner membrane leads to the flux of proton back to

matrix through adenosine triphosphate (ATP) synthase (Complex V) and drives the

synthesis of ATP (170). The electrochemical gradient across the inner membrane is

indicated by the mitochondrial membrane potential (ΔΨm) (170). The electron

leakage from oxidative phosphorylation is believed to be a major source of ROS

production in cells. It has been estimated that 0.2-2.0% of oxygen consumption by

mitochondria results in formation of superoxide radicals (149, 165). Leakage of

electrons from the electron transport chain takes place at Complex I towards the

matrix and at Complex III towards both sides of the inner membrane (97, 149, 165).

Leaked electron leads to partial reduction of oxygen. The resulting superoxide

radicals can act as precursors for many other more active ROS, or they can be

converted to hydrogen peroxide by SOD1 (superoxide dismutase 1) in the matrix or

SOD2 (superoxide dismutase 2) in the inter-membrane space (97, 149, 165).

Superoxide is believed to be short-lived while hydrogen peroxide is more stable and

permeable to other part of mitochondria and cells (149, 170). The level of hydrogen

peroxide has been estimated to be 100-fold higher than the level of superoxide

radicals in mitochondria (43). In general, mitochondria are not rich in catalase, and

Page 43: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

24

the reduction of hydrogen peroxide relies on the reaction with GSH catalyzed by

glutathione peroxidase (149) (Figure 1.5). As the major source of ROS, mitochondria

are also a target of ROS-mediated damage. Excess mitochondrial ROS may induce

damage and mutations of mitochondrial DNA (170, 281). ROS may also induce

carbonylation and dysfunction of various mitochondrial proteins, including the

complex proteins leading to defective mitochondrial respiration, and the

mitochondrial carrier proteins regulating molecular transport across the membrane,

and the protein assembly of permeability transition (PT) pore which may trigger

signals of apoptosis (170).

ΔΨm indicates the electrochemical gradient across the inner membrane, which

results from the activity of the election transport chain (170). As ROS is a byproduct

of the electron transport chain, under the normal conditions where there is an

ROS/anti-oxidant equilibrium, ΔΨm and ROS should be positively correlated. While

this is supported by several studies (149, 165, 188), under conditions of cellular

damage ΔΨm reduction is associated with increased ROS (9, 273). Reduction of ΔΨm

may be a result of impaired proton transfer due to an interrupted electron transport

chain, or be caused by opening of PT pores resulting in increased permeability.

Mitochondria are highly dynamic organelles under constant cycles of fission and

fusion (183). Fission refers to the splitting of mitochondria leading to short and less

branched morphology while fusion is the merging of mitochondria leading to an

elongated and a more branched network (149). These morphological dynamics are

very important for normal function of the mitochondrial network in a cell.

Page 44: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

25

Interruption of either the fusion or fission process causes dysfunction of mitochondria

(183). This might be because the exchange of mitochondrial DNA in matrix during

the fission and fusion can alleviate the effect of mutation (183).

In addition to their role in energy supply and redox regulation in the cells,

mitochondria play a central role in the regulation of apoptosis, senescence and

autophagy (5, 11, 149, 170). Excess ROS may induce mitochondrial PT pores into a

high-conductance state, leading to rising permeability for small molecules (83). This

event is reflected by loss of ΔΨm. As the inner membrane is highly folded, the fluid

influx lead to swelling of the inner membrane, resulting in further opening of

mitochondrial PT pores and sometimes rupture of outer membrane (123). During this

process cytochrome c is released from mitochondria, which triggers activation of

caspase-9 and leads to apoptosis (90). In addition to the effects on PT pores, oxidative

stress can also lead to peroxidation of cardiolipin, a lipid that immobilizes cytochrome

c in the inner membrane, which further promotes the release of cytochrome c from

mitochondria (124, 193).

There is an increasing focus on the involvement of mitochondria in the

pathogenesis of COPD (11). Most direct evidence has come from primary cells from

the patients with COPD. It has been demonstrated that primary bronchial epithelial

cells from ex-smokers with severe COPD exhibited swelling and fragmented

mitochondrial morphology which is similar to that seen in CS medium (CSM)-treated

immortalized bronchial epithelial cells (BEAS2-B) (105). Another study on ASMCs

has been reported by Wiegman and colleagues with a more comprehensive evaluation

Page 45: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

26

of mitochondrial function (273). In this study ASMCs from patients with COPD

demonstrated elevated mitochondrial ROS levels, reduced ΔΨm, reduced amount of

Complex I, III and V compared to healthy controls. Moreover, mitochondrial

respiration was impaired in ASMCs with COPD as indicated by reduced basal and

maximum respiration levels, and respiratory reserve capacity. Healthy smokers also

demonstrated mitochondrial dysfunction compared to healthy controls, but the effects

are less pronounced compared to patients with COPD. Several other studies also

suggested mitochondria of respiratory striated muscle and skeletal muscles are

damaged in patients with COPD (5).

Mitochondrial damage is also observed in other COPD-related animal models or

CSM-treated in vitro models. It has been reported that CS exposure can impair energy

metabolism in mouse lung by switching glucose metabolism to the pentose phosphate

pathway and reducing the substrate supply to mitochondria (4). In a chronic

ozone-induced mouse model of COPD, elevation of mitochondrial ROS and reduction

of ΔΨm is observed, which can be alleviated by the mitochondria-targeted

anti-oxidant MitoQ (273). The alleviation of ozone-induced mitochondrial ROS leads

to attenuation of ozone-induced airway hyper-responsiveness (AHR) and

inflammation, indicating a potential strategy to target mitochondrial ROS for the

treatment of COPD. Another study has demonstrated that wood smoke exposure to

guinea pigs can induce impaired oxygen consumption activity and complex protein

enzyme activity (89). CSM was also reported to alter the fission-fusion activity of

mitochondria in vitro in both airway epithelial cells (105) and ASMCs (10), leading to

Page 46: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

27

mitochondrial fragmentation and increased mitochondrial ROS.

Figure 1. 5 Generation of mitochondrial ROS from electron transport chain.

Leakage of electron from electron transport chain leads to generation of superoxide

radicals, which can be converted to hydrogen peroxide by SOD. I-V: complex

proteins; Q: coenzyme Q; Cyto c: cytochrome c; SOD: Superoxide dismutase, GPx:

Glutathione peroxidase. The figure is plotted based on Li X et al, J Hematol Oncol

(149).

1.1.5. Current therapies

For patients with COPD who are current smokers, smoking cessation is the most

effective approach that can influence the natural history of COPD. Smoking cessation

can be facilitated by nicotine replacement therapies such as nicotine gum, inhaler,

nasal spray, transdermal patch, sublingual tablet and lozenge, as well as supportive

pharmacotherapy including varenicline, bupropion and nortriptyline (263).

Pharmacologic therapies for COPD include bronchodilators, corticosteroid and

Page 47: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

28

phosphodiesterase-4 inhibitors (263). The choice of each medications or the

combination of them is patient specific. Bronchodilators refer to the medications that

induce the dilation of bronchi and bronchioles leading to increase in FEV1 (263).

There are three classes of bronchodilators, β2-adrenorecepter agonist, anticholinergics

and methylxanthine (82). The β2-adrenoreceptor agonists are further classified as

short-acting β2 agonist (SABA) and long-acting β2-agonist (LABA), both working

through induction of cellular cyclic adenosine monophosphate (cAMP).

Bronchodilators are usually administrated through inhalation, and they are the central

medication to manage the symptoms of COPD. Combination of different classes of

bronchodilators may improve efficacy and reduce the side effects compared to a

single class. Corticosteroid treatment is mainly adopted for the severe conditions. Oral

corticosteroids aim to suppress the chronic inflammation, but they are generally

associated with high risks of adverse effects (263). Inhaled corticosteroids in

combination with bronchodilator may improve efficacy compared to single treatment,

but the long-term safety and adverse effects of inhaled corticosteroids is largely

unknown (263). Phosphodiesterase-4 inhibitors are a relatively new medication for

COPD. They are able to reduce inflammation by inhibiting cAMP breakdown, and

should be used with long-acting bronchodilators (263).

COPD is a complex, chronic disease resulting from long term interaction between

genes and environmental stimuli (65). The pathogenesis of the disease is far from

understood and the development of effective pharmacologic therapy has not been easy,

especially for the control of chronic inflammation which is corticosteroid-resistant in

Page 48: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

29

COPD (24). To date, the pharmacologic therapy is able to reduce COPD symptoms,

exacerbations, and improve health status and exercise tolerance (263). However, none

of current treatments is able to alleviate the progressive decline of lung function in

COPD (263).

1.2. Introduction to Stem Cells

1.2.1. Definition and classification

Stem cells are cells that are able to self-renew and differentiate into other cell

types (251). In contrast, somatic cells are usually able to self-renew but cannot

differentiate, while progenitor cells are able to differentiate into somatic cells, but

with poor ability of long-term self-renewal (251). Stem cells and progenitor cells act

as sources of new somatic cells as a normal turnover or as a replenishment to

increased somatic cell death after injury (251).

Stem cells can be classified as totipotent, pluripotent and multipotent according to

their potency that is their capacity to differentiate (2, 251). Totipotent stem cells have

the highest differentiation potential with capacity to develop into new individuals

from a single cell. Although not able to form new individuals, pluripotent stem cells

are capable of differentiating into cell types of all three germ layers (ectoderm,

mesoderm and endoderm), which basically means they can generate almost any cell

types except for extra-embryonic tissues such as trophectoderm (2, 251). Multipotent

stem cells can form limited cell types, usually of one single lineage (2, 251). In

general, the potency of stem cells gradually decreases along the development process

Page 49: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

30

(2, 251). Totipotent stem cells only exist as zygotes and cells from very early embryos,

while stem cells derived from blastocyst named embryonic stem cells (ESCs)

represent the major form of pluripotent stem cells (2, 251). Stem cells in the adult

body, or adult stem cells, are multipotent stem cells (2, 251). They are usually

distributed in specific microenvironments called niches of the parent tissues and

generate somatic cells of the same origin (2, 251). The progenitor cells have similar

differentiation features as the adult stem cells. They also reside at niches of tissues

and differentiate into specific cell types (85).

Utilising stem cells therapeutically is not a recent idea. Haematopoietic stem cells

(HSC) have been used in bone marrow transplantation for treatment of leukaemia for

more than 40 years (41). Other cells that have been tested in clinical trials include

mesenchymal stem cells (MSC) and muscle satellite stem cells (251, 269). When it

comes to stem cell-based therapy, the stem cells can be alternatively classified as

endogenous stem cells which are the adult stem cells in their niches in the body, and

exogenous stem cells which are the isolated and expanded adult stem cells ex vivo

(251).

1.2.2. Endogenous stem cells in lung

The delineation of endogenous stem cells in individual organs is not an easy task.

The most convincing approach is to examine the capacity of the cells to reconstitute

the entire organ. For example, HSCs can rebuild the entire haematopoietic system

after transplantation into irradiated animals (251). However the approach requires

Page 50: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

31

destruction of organs which is rather difficult in most cases. An alternative approach

is to genetically trace the lineage of cells in the organs, which can provide evidence of

the differentiation of specific cell types into other cells. Intestinal stem cells (260) and

skin stem cells (34) were identified through this approach.

The lung is a complex organ with a very low turnover rate during normal

homeostasis compared with intestine or skin tissues (106). The regenerative capacity

of human lung is mostly demonstrated upon injury, such as pneumonia and severe

respiratory infections (28, 42, 131, 138). Whether there is a defective regenerative

function of endogenous lung stem cells in airway diseases is still unclear (268). In fact,

the identity and roles of stem cells and progenitor cells in the lung remains unresolved

due to the lack of markers for each cell type and lineage tracing approaches (268).

Various putative airway and alveolar epithelial stem and progenitor cells are identified

by mounting data from animal and human studies, including airway submucosal

glands, keratin-14 expressing tracheal basal cells, Clara cells in association with

neuroendocrine bodies or bronchoalveolar duct junction, and type 2 alveolar epithelial

cells (AEC2s) (85). However, there is also controversial data from other studies and

there is not yet uniform agreement on the identity of lung stem cells (268).

As COPD is associated with progressive destruction of the alveolar structure, the

identification of stem cells in alveoli, and further investigation of their role in COPD,

may provide valuable information on the disease. Alveoli are mainly composed of

type 1 alveolar epithelial cells (AEC1s) and AEC2s. AEC2s which express high levels

of surfactant protein C (SFTPC) can function as local stem cells (21). Lineage tracing

Page 51: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

32

in mice are demonstrated that AEC2s can differentiate into AEC1s during homeostasis,

and bleomycin-induced injury promoted such activity (21). The self-renewal ability of

SFTPC+ AEC2s was also observed for at least one year. Moreover, when co-cultured

with a mesenchymal population, the SFTPC+ AEC2s can differentiate into alveolar

structures consisting both AEC2s and cells with AEC1 markers (21). In combination

with the self-renewal property in vitro, these findings demonstrated the capacity of

AEC2s for self-renewal and differentiation into other cells both in vivo and in vitro.

1.2.3. Mesenchymal stem cells (MSCs)

Mesenchymal stem cells (MSCs) are multipotent stem cells from stromal origin

(269). Originally described as a fibroblastic-like population of bone marrow (BM)

cells, MSCs now refer to similar cells found in a variety of tissues, including BM,

adipose tissue, cord blood and placenta (269). MSCs can differentiate into

chondrocyte, adipocyte, osteoblast or other lineages such as epithelial and endothelial

cells (269). The general criteria to define MSCs include the cell marker profile,

self-renewal property and the capacity of differentiating into osteoblasts, adipocytes

and chondrocytes (251). Although MSCs isolated from different tissue share common

features regarding these parameters, different gene expression, lineage tendencies and

other properties have been demonstrated amongst them (15, 132). Moreover,

individual MSC types may still present a heterogeneous group of cells of different

subtypes (200). The functional and therapeutic differences between MSCs from

different sources have been increasingly investigated (269).

Page 52: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

33

Cells with features of MSCs have been isolated from adult mouse lungs, human

nasal mucosa, and lungs of human neonates and lung transplant recipients (269). The

functions of these cells in the lung remain unclear. They have been reported to play a

potential role in fibrotic diseases and in the bronchiolitis obliterans following lung

transplantation (14, 61, 210, 264).

MSCs express very low levels of major histocompatibility complex (MHC) class

I and class II molecules, which makes them highly immunologically-tolerant (80, 132,

144, 269). Exogenous administration of allogeneic MSCs usually lead to low levels of

immune rejections and does not require immunosuppression (251). MSCs are

commonly used to develop exogenous stem cell-based therapy for lung diseases.

There are numerous studies on this topic, which will be introduced in the later

sections.

1.2.4. Induced-pluripotent stem cells (iPSCs)

As described earlier, in nature, cells demonstrate a unidirectional reduction in

differentiation potency with development. Therefore, the access to pluripotent stem

cells had relied on the isolation of ESCs until the discovery of induced-pluripotent

stem cells (iPSCs) (247). iPSCs are pluripotent stem cells derived from somatic cells

through transcriptional factor-induced reprogramming in vitro (e.g. through induction

by Oct4, Sox2, Nanog and Lin28). Deriving iPSCs from somatic cells was first

achieved by Takahashi and Yamanaka using mouse fibroblasts in 2006 (247). Human

iPSCs were later generated also from fibroblasts in 2007 (246, 289). In theory, this

Page 53: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

34

robust approach can generate iPSCs from any somatic cell type.

Compared to the isolation of adult stem cells from the relevant tissue, the

reprogramming of somatic cells is less restricted to the type of somatic tissue.

Moreover, the high proliferation capacity of iPSCs reduces the amount of somatic

tissue required. Therefore, iPSC provides an abundant supply of stem cells from each

particular individual. However, the therapeutic application of iPSCs is limited by

pluripotency. They are so potent that direct administration of iPSCs may lead to

formation of teratomas (122).

Although iPSCs may not be a very good candidate as a direct therapy for airway

disease, they can serve as a great source to generate other cell types that may be

effective in airway diseases. Indeed, the derivation of lung progenitor cells or

epithelial cells from iPSCs has been reported by several studies (91, 111, 179, 278,

279). AEC2 is the most common and prominent cell type derived from iPSCs, while

AEC1 can also be generated. The iPSC-derived epithelial cells can be used in ex vivo

tissue engineering of lung, providing cellular source to re-populate decellularized

whole lung scaffolds (86, 159). However, information of their effects as direct source

of cell-based therapy for airway disease is still lacking.

1.2.5. iPSC-derived MSCs

Given the numerous studies on the therapeutic potential of MSCs in airway

disease models, deriving MSCs from iPSCs provides a promising strategy. This task

was achieved by Lian and colleagues in 2010 (153) (Figure 1.6). The derivation was

Page 54: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

35

through incubation of iPSCs in pro-MSC induction medium (detailed methodology

can be found in Chapter 2). MSC-like cells were identified as CD24- and CD105+

cells. The resulting cell culture exhibited fibroblast-like morphology which is typical

of MSCs. To acquire homogenous populations, single-cell derived colonies were

generated from these cells. In addition to the fibroblast-like morphology, the derived

cells demonstrated other features of MSC. Firstly, they presented an MSC-like surface

marker profile (CD44+, CD49a+ CD49e+, CD73+, CD105+, CD166+, CD34−,

CD45−, and CD133−). Secondly, the differentiation capacity was demonstrated by

efficient adipogenesis, osteogenesis and chondrogenesis. Lastly, they were able to

self-renew without loss of differentiation potential. In fact, they were far more

expandable than BM-MSCs, with an ability to keep their differentiation capacity up to

passage 40. As they fulfil all criteria of MSCs, these cells were named iPSC-derived

MSCs (iPSC-MSCs). Compared to iPSCs, iPSC-MSCs demonstrated reduced

expression of pluripotency-associated genes such as Oct4, Nanog and Sox2. When

subcutaneously injected into combined immunodeficient (SCID) mice, iPSC induced

teratoma while no teratoma was induced by iPSC-MSCs for up to four months.

The in vitro generation of iPSC-MSCs from iPSCs brings potential opportunities

to overcome several limitations of the adult MSCs as seen in BM-MSCs. The amount

of BM-MSCs available from BM is limited which makes ex vivo expansion important

(269). However, BM-MSCs become senescent after 20 doublings during ex vivo

expansion with loss of differentiation potential (69). Their genetically stability in vitro

also draws concerns as they may develop abnormal karyotypes during expansion

Page 55: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

36

(148). The limited in vitro expansion capacity restricted the resource for cell therapy.

In addition, one of the future directions of MSCs-based therapy is to genetically

manipulate the cells. As MSCs can be recruited to injured tissue, by this approach

they can express and release desired molecules at the site of the injured tissue (63,

122). Such genetic manipulation also calls for high stability and expanding capacity

of the MSCs. In contrast to BM-MSCs, iPSC-MSCs demonstrated normal karyotypes

during expansion (153). They can be expanded for 120 doublings with stable surface

marker profile for MSCs (153). Another limitation of BM-MSCs is that when isolated

from aged subject or patients with ageing-related disorders, they may present reduced

survival and differentiation ability (87, 100, 267, 294). In theory, as the safety of

allogeneic MSCs has been well demonstrated, this issue can be avoided by selection

of the donor. Nevertheless, availability of personalized MSCs is appealing for future

development of MSCs-based therapy. iPSC-MSCs may hold hope as a way of

overcoming the ageing-associated impairment of BM-MSCs. When generating

iPSC-MSCs, somatic cells were firstly reprogrammed to an embryonic stem cell-like

state, which may help to avoid the problem of senescence. According to the same

study by Lian and Zhang et al., the telomerase activity of iPSC-MSCs was 10 times

higher compared to BM-MSCs (153).

As a relatively new type of MSCs, beneficial effects of iPSC-MSCs have already

been demonstrated in rodent models of limb ischemia (153), allergic airway

inflammation (244) and cardiomyopathy (293).

Page 56: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

37

1.3. MSCs-based Therapy

1.3.1. Efficacy of MSCs in animal models

MSCs are the most studied candidates for the development of cell-based therapy

against lung diseases. Efficacy of MSCs was demonstrated in a broad spectrum of

animal models including acute lung injury and bacterial lung infection, COPD,

asthma, bronchiolitis obliterans, bronchopulmoary dysplasia, fibrosing pulmonary

injury, pulmonary hypertension, pulmonary ischemia re-perfusion injury, obstructive

sleep apnea, radiation-induced lung injury, sepsis and burns (269). The studies of

MSCs in COPD-related models are listed in Table 1.2. In general, these models can

Figure 1. 6 Change of potency: from somatic cells to iPSC-MSCs. Inducing

iPSCs from somatic cells provides great resource of stem cells. However, iPSCs are

too potent for some applications, and differentiation of them into multipotent stem

cells can generate cell types that may be more useful as treatment of diseases.

Page 57: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

38

exhibit histological changes (primarily airspace enlargement) in the lung mimicking

the lungs of patients with COPD, which are alleviated by MSCs from various sources

including BM, adipose tissue, lung and amniotic fluid. The major approaches for

inducing emphysema in animals are by administration of elastase and by chronic

exposure to CS. Elastase can cause excess protease activity rapidly leading to direct

destruction of alveolar structures, while CS exposure can induce inflammation very

quickly but with the airspace enlargement developing much later (usually months

after exposure). However, as cigarette smoking is the major risk factor of COPD, the

CS-exposed model may be more relevant to the pathogenesis of COPD. CS-exposed

animals have been demonstrated to develop inflammation, oxidative stress and excess

protease activity both in lung and systemically (51, 52). The capacity of alleviating

CS-induced inflammation is demonstrated in MSCs from BM (94, 112), amniotic

fluid (151) and adipose tissue (222). Moreover, the MSCs from BM and adipose

tissue also inhibited CS-induced apoptosis in the lung.

Page 58: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

39

Table 1.2 Studies of MSCs treatment in COPD-related animal models.

Studies Experimental Model

Route, and Timing of Treatment Source of MSCs Outcome Compared to Injury Effects

Potential

Mechanisms

Shigemura

2006(230)

Rat: elastase.

Direct topical application of ASC-

seeded polyglycolic acid at lung

volume reduction surgery.

Rat adipose

Plastic adherent • Improved histological repair HGF secretion

Shigemura

2006 (229)

Rat: elastase.

i.v. ASCs 1 week after elastase.

Rat adipose

Plastic adherent

• Decreased apoptosis, improved

histological repair

• Improved gas exchange and exercise

tolerance

HGF secretion

Yuhgetsu

2006 (290)

Rabbit: elastase.

i.t. MSCs 24 hours after elastase.

Rat BM - mononuclear

cells from Ficoll gradient of

total BM

• Improved histology and lung function

• Decreased BALF inflammation

• Decreased tissue MMP2 and MMP9

expression

None specified

(soluble mediators)

Zhen

2008 (295)

Rat: papain.

i.v. MSCs after papain (timing not

specified).

Rat BM

Plasticadherentmononucelar

cells from Percoll gradient

• Decreased histological injury

• Decreased alveolar cell apoptosis None specified

Page 59: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

40

Zhen

2010 (296)

Rat: papain.

i.v. MSCs 2 hours after papain

Rat BM

Plastic adherent

mononucelar cells from

Percoll gradient

• Decreased histological injury

• Partial restoration of VEGF

expression in lung homogenates

TNF-α–stimulated

VEGF secretion by

MSCs

Katsha

2011 (130)

Mouse: i.t. elastase.

i.t. MSCs 14 day after elastase.

Mouse BM

Plastic Adherent

• Decreased histological injury

• Decreased IL-1β

• Transient increase in lung EGF, HGF,

and SLPI

None clarified

(soluble mediators)

Huh

2011 (112)

Rat: CS exposure for 6 months.

Retrobulbar MSCs at the beginning

of smoke cessation.

Rat BM

• Decreased histological injury

• Increased numbers of small vessels

and reduced RVSP

• Increased proliferation of AEC2 cells

and endothelial cells

• Decreased apoptosis

Soluble mediators

Akt signaling

suspected

Page 60: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

41

Schweitzer

2011 (222)

Mouse: CS exposure for 4 months.

i.v. ASCs every other week for 3rd

and 4th months of CS exposure.

Human or mouse adipose

plastic adherent

• Decreased histological injury and

apoptosis (caspase activation)

• Decreased BALF inflammation

• Decreased CS-induced MAPK signal

transduction

• Decreased weight loss

• Decreased BM suppression

None specified

Immunodeficient NOD-SCID

mouse: VEGFR blockage induced

emphysema.

Human ASCs 3 days after VEGFR

blockade

Human adipose plastic

adherent

• Human ASCs decreased histological

injury and caspase 3 activation

following VEGFR blockade

• Emboli if used > 5 × 105 MSCs or if >

passage 3

None specified

Hoffman

2011 (104)

Mouse: i.t. elastase

i.v. MSCs 6-7 weeks after elastase

Mouse BM

Lung-MSCs from primary

explants cultures

• Higher retention of lung MSCs

• Decreased histological injury None specified

Page 61: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

42

Ingenito

2012 (117)

Sheep: i.t. elastase (5

doses/20weeks)

Endoscopically transplanted in

biological delivering scaffold

Sheep lung

• Increased lung tissue mass and

perfusion

• Increased histological cellularity, cell

retention and extracellular matrix

• Improved lung mechanics

Paracrine effects.

Increased epithelial

proliferation in in

vitro co-culture.

Guan

2013 (94)

Rat: CS exposure for 11 weeks,

sacrificed after 4 weeks cessation.

i.t. MSCs on the first day of week

7

Rat BM

• Decreased histological injury

• Increased lung function

• Decreased lung apoptosis

• Decreased TNF-α, IL-1β, MCP-1,

IL-6, MMP9 and MMP12 in lung

• Increased VEGF, VEGF receptor-2

and TGFβ-1 in lung

Non specified

Song

2014 (237)

Rats: CS exposure for 7 weeks,

sacrificed after 4 weeks cessation.

i.t. MSCs at week 8

Rat BM

• Increased histological injury

• Increased lung function

• Decreased inflammatory cells in BAL

• Decreased levels of IL-1β, IL-6,

TNF-α, MDA and increased TGFβ-1 in

lung

Non specified

Tibboel

2014 (252)

Mouse: i.t. elastase

i.t. or i.v. MSCs before or after

elastase

Mouse BM

• Increased lung function only by i.v.

injection prior elastase

• No significant histological difference

Non specified

Page 62: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

43

Li X

2014 (150)

Rat: CS exposure for 56 days

i.v. MSCs at day 29 and 43 Human iPSC-MSCs • Decreased histological injury

Mitochondrial

transfer

Li Y

2014 (151)

Rat: CS exposure for 12 weeks

plus i.t. LPS at week 4 and 8.

i.t. MSCs after CS exposureperiod

Rat amniotic fluid

• Decreased histological injury

• Increased surfactant protein A and C

• Decreased AEC2 apoptosis

Differentiation into

AEC2-like cells

Anti-apoptotic

mechanism

unknown

Gu

2015 (93)

Rat: CS exposure for 12 weeks

sacrificed after 4 weeks cessation.

i.t. MSCs started at week 7,

twice/week for 5 weeks

Rat BM

• Decreased histological injury

• Decreased inflammation

• Decreased PGE-2 and IL-6 and

increased IL-10 in BALF and serum

• Decreased COX-2 and PGE2 in lung

Inhibition of

COX-2/PGE2 in

alveolar

macrophages by p38

MAPK pathways

Abbreviations: AEC2: Type 2 Alveolar epithelial cell; ASC: adipose stem cell; BALF: bronchoalveolar lavage fluid; BM: bone marrow;

COX-2: cyclooxygenase-2; CS: cigarette smoke; EGF: epidermal growth factor; HGF: hepatocyte growth factor; IL: interleukin; iPSC-MCS:

induced pluripotent stem cell-derived mesenchymal stem cells; i.t.: intratracheal; i.v.: intravenous; MAPK: mitogen-activated protein kinase;

MCP-1: monocyte chemoattractant protein-1; MDA: malondialdehyde; MMP: matrix metalloproteinases; PGE2: prostaglandin E2; RVSP:

right ventricle systolic pressure; SLPI: secretory leukoprotease inhibitor; TGF-β: transforming growth factor-β; TNF-α: tumor necrosis factor;

VEGF(R): vascular endothelial growth factor (receptor).

The list before (including) 2012 is adapted from two reviews by Weiss et al (269, 270); studies after 2012 are described from corresponding

references.

Page 63: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

44

1.3.2. Clinical trials

The safety and efficacy of MSCs have been tested in acute myocardial infarction

and several inflammatory and autoimmune diseases such as graft-versus-host disease

(GVHD), autoimmune diabetes and Crohn‟s disease (269). The first licensed

MSCs-based therapy has been approved in Canada, as a medication for GVHD (8,

269). The intensive clinical study in these fields showed an important fact that MSCs

may be well tolerated even by patients with very severe illness (8, 269).

The mounting evidence supporting the efficacy of MSC in COPD-related animal

models led to the initiation of the first multicenter, double-blind, placebo-controlled

phase II clinical trial of a systemic-administrated BM-MSC preparation

(PROCHYMAL, Osiris Therapeutic Inc, Columbia, MD, USA) for moderate or

severe COPD (271). 62 patients participated in the clinical trial, during which they

received 4 monthly infusions of BM-MSCs (100x106

cells/infusion) or vehicle

controls and were monitored in a subsequent 2-year period. End points included

comprehensive safety evaluation, pulmonary function testing (PFT), and

quality-of-life indicators including questionnaires, 6-minute walk test, and

assessments of systemic inflammation. The trial proved the safety of BM-MSCs in

patients with moderate or severe COPD. No pulmonary emboli formation or other

infusional toxicity was observed for every single infusion among all the 4 infusions of

all patients. No mortality or serious adverse effects were observed in the two-year

follow-up. However, the administration of BM-MSCs did not improve the lung

function of the patients compared to the placebo, as well as the quality of life

Page 64: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

45

indicators such as 6-minute-walk evaluation and quality of life questionnaire. The

only effect observed was on the systemic C-reactive protein (CRP) levels, which may

indicate a modulation of systemic inflammation. Among the patients with high

systemic CRP levels (above 4 mg/L) at the start of the trial, infusion of BM-MSCs

significantly reduced the systemic CRP levels within the first month and the trend

persisted for 2 years although the statistical significance was lost over time.

A multicentre, open-label, dose-escalation, phase I clinical trial on treatment with

BM-MSCs in moderate to severe acute respiratory distress syndrome (ARDS) in USA

has been reported recently (276). Nine patients were included, and three dosages were

tested (106, 5x10

6, 10x10

6 cells/kg predicted body weight). The findings demonstrated

safety of the single infusion into patients with ARDS, with no adverse effects or

mortality attributable to the infusion. The trial has entered a phase II stage for

examination of efficacy. Another placebo-controlled study demonstrated safety of

adipose-derived MSCs infused in patients with ARDS (297). However, no significant

efficacy was observed in this study.

To date two non-randomized phase I clinical trials in patients with idiopathic

pulmonary fibrosis have been performed. In the first placenta-derived MSCs were

administrated intravenously (50) and in the second endobronchial infused adipose

derived stromal cells-stromal vascular fraction were used (256). Both these phase I

trials demonstrated good safety profile of MSCs in patients with idiopathic pulmonary

fibrosis.

Although the efficacy of MSCs in lung diseases has not been demonstrated in any

Page 65: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

46

studies to date, the above studies proved strong evidence of the safety of MSCs

infusions. There are many variables regarding the potential functions of MSCs,

including dosage, preparation, and tissue source, and more investigation is required to

define its clinical role. The lack of efficacy in the clinical trial in contrast with the

numerous reports on the efficacy in animal models also calls for better pre-clinical

models and more information about the mechanisms of action of MSCs.

1.3.3. Mechanisms

1.3.3.1. Engraftment and regeneration

While intratracheal administration definitely leads to the delivery of MSCs to the

lung, systemic-administrated MSCs have also been reported to localize in the lung

vascular bed initially, and the number of localized cells can be enhanced by lung

injury (269). The retention ability of systemic-administrated MSCs in lung varies in

different studies. The effects of the MSC properties such as size, expression of

specific integrins and proteoglycan patterns on the retention ability remain unclear.

MSCs from different sources present different retention capacities. For example,

MSCs from umbilical cord blood have been reported to be more easily cleared by the

lung compared to BM-MSCs (185). In CS-exposed models, adipose stem cells have

been reported to retain in lung tissue after 7 days or 21 days (222), while another

study only detected 2-13 male BM cells per entire lung from 1 day to 1 month by

fluorescent in situ hybridization (FISH) (112). Higher retention rate of iPSC-MSCs in

the lung compared to BM-MSCs has also been reported 14 days after administration

Page 66: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

47

(150).

The capacity of BM-MSCs and adipose-MSCs to differentiate into cells

expressing markers of alveolar or epithelial cells has been demonstrated in vitro (162,

171, 243, 282), which represents an early motivation for research on MSCs-based

therapy: to let the MSCs differentiate into progenitor or somatic cell types, replenish

the damaged cells and thus regenerate the injured tissue. Indeed, early reports

provided evidence of structural-engrafted HSCs, MSCs or endothelial progenitor cells

differentiating into airway and alveolar cells or pulmonary vascular or interstitial cells

(129, 270). However, later research suggested some major technical limitations of

these early studies and current understanding regarding the issue is that the

differentiation of MSCs into structural cells in the lung is rare and with unlikely

physiological significance (270).

Although the retention of MSCs does not lead to differentiation, it may induce

other activities. The retention is possible to modulate paracrine function of MSCs,

such as secretion of the anti-inflammatory cytokine tumor necrosis factor-stimulated

gene-6 (TSG-6) (145). The retention may also facilitate the direct contact between

MSCs and local cell types, which may induce the onset of other mechanism such as

mitochondrial transfer.

1.3.3.2. Immunoregulation

Immunoregulation is a very significant function of MSCs. As mentioned above,

treatment of the autoimmune disease GVHD using BM-MSCs has already been

Page 67: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

48

approved for clinical use.

The role of endogenous MSCs in immunoregulation is still largely unknown.

Endogenous MSCs are found to reside perivascularly, and they may represent a

portion of the pericytes (182). At this site they can interact with HSC and may affect

immune responses. The immunoregulating capacity of exogenous MSCs has been

widely reported in vitro. They have been demonstrated to inhibit various functions of

immune cell such as dendritic cell differentiation, B cell proliferation, cytolytic

potential of natural killer cell, and proliferation and activation of T cell (33, 235, 269,

270). MSCs also facilitate the development of the M2 phenotype of macrophages,

which is believed to be anti-inflammatory (81, 266, 286). In addition, MSCs are able

to reduce the release of inflammatory mediators from these immune cells such as

IFN-γ and promote secretion of anti-inflammatory cytokines such as IL-10. The full

mechanism of the immunomodulation is unclear but the paracrine release of

immunomodulating agents including of TGF-β1, HGF, prostaglandin E2 (PGE2), IL-10

and nitric oxide from MSCs may play an important role in the process (2, 235, 270).

1.3.3.3. Mitochondrial transfer

Mitochondrial transfer was first observed in vitro in 2006 by Spees and

colleagues (238). In their study they damaged mitochondrial DNA in a lung epithelial

cell-line A549 by pre-treatment with ethidium bromide, leading to impaired aerobic

respiration and proliferation. The mutated A549 cells only survive in a permissive

medium containing uridine and pyruvate used to supplement anaerobic glycolysis.

Page 68: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

49

However, after co-culture with BM-MSCs, these cells re-acquired the capacity to

proliferate in normal medium, indicating restoration of aerobic respiration.

Mitochondrial function was recovered as indicated by ATP levels, and mitochondrial

transfer was identified from BM-MSCs to mutated A549 cells both by DNA

measurement and microscopy. Following this study, mitochondrial transfer from

BM-MSCs to other cell types including endothelial cells, cardiomyocytes and

osteosarcoma cells in vitro has also been reported (55, 192, 204).

In 2012, Islam and colleagues reported the first in vivo study demonstrating the

occurrence and functional significance of mitochondrial transfer from exogenous

BM-MSCs to host airway epithelial cells (118). In the study acute lung injury was

induced by intratracheal infusion of lipopolysaccharide (LPS), followed by

intratracheal infusion of BM-MSCs. They found BM-MSCs attached to the alveolar

epithelia at connexin 43-containing gap junctions. Mitochondrial transfer was

observed within 24 hours from BM-MSCs to alveolar epithelial cells, which is

mediated by connexin 43-dependent generation of nanotubes and microvesicles. The

mitochondrial transfer elevated alveolar ATP concentrations. The therapeutic efficacy

of BM-MSCs on the acute lung injury, including reduction of alveolar leukocytosis

and protein leak, elevation of surfactant secretion and reduction of mortality, were

eliminated in BM-MSCs with mutated connexin 43 or dysfunctional mitochondria.

Current understanding on the mechanisms of mitochondrial transfer is still very

limited. Tunnelling nanotubes (TNTs) are identified as routes through which

mitochondria move intracellularly in most in vitro studies. TNTs were first discovered

Page 69: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

50

in vitro by Rustom and colleagues in 2004 as thin structures connecting single cells

over long distance. TNTs are characterized with continuity of membrane connection

between the two cells as well as continued cytoskeleton in the tubes (13). F-actin is

found in most TNTs spanning along the entire length, and inhibition of F-actin

polymerization lead to blockage of TNT formation (40). Machinery of intracellular

movements of mitochondria in neuronal cells has been studied for many years. In

neuronal cells, mitochondrial Rho GTPase 1 (Miro-1, also named Rhot-1), a

calcium-sensitive adaptor protein, can bind mitochondria to kinesin motor protein-5

(KIF5) with the aid of accessory proteins Milton/TRAK (155). Kinesin can then carry

the mitochondria to move along microtubes (155). It has been proposed that this

mechanism may also mediate the intercellular movement of mitochondria along the

TNTs (Figure 1.7). A recent study demonstrated that over-expressing of Miro-1 in

BM-MSCs can promote mitochondrial transfer to epithelial cells and enhance efficacy

in vitro, while Miro-1 knock-down leads to lack of efficacy (6). Over-expression of

Miro-1 also enhanced the efficacy to attenuate rotenone-induced lung injury and

allergic airway inflammation in vivo. This finding provided a possible approach to

selectively block mitochondrial transfer, which may be very helpful for future

research about the role of mitochondrial transfer in MSCs action. Nevertheless, the

mechanisms regulating TNT formation or the Miro-1-involved movement of

mitochondria remain unclear. Further understanding on the topic may help address the

important feature of mitochondria transfer such as the direction selectivity and

enhanced activity upon injury.

Page 70: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

51

Figure 1. 7 Proposed mechanism of mitochondrial transfer. BM-MSCs transfer

mitochondria to damaged epithelial cells through tunneling nanotubes (TNTs). Miro-1

loads mitochondria on KIF5 with aid of accessory protein Milton/Trak. KIF-5 moves

along microtubes in the TNTs and thus transfers mitochondria between cells. The

image showing the enlarged region is adapted from Sheng ZH et al, Nat Rev Neurosci

(227).

Page 71: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

52

1.4. Aims of Study

The overall hypothesis for this thesis is that iPSC-MSCs are able to alleviate

oxidative stress (CS or ozone)-induced lung damage (histological changes,

hyper-responsiveness, inflammation, apoptosis and mitochondrial dysfunction) in

vivo as well as mitochondrial dysfunction and apoptosis in bronchial epithelial cells

and ASMCs in vitro, through multiple mechanisms including paracrine effect and

mitochondrial transfer.

Based on previous sections, the background of the study can be summarized as

follows:

1. COPD is a chronic and complex disease. It is a rising cause of death associated

with huge social and economic burden. There are no current medications that can

slow the progression of the disease. Cigarette smoking is the major cause of

COPD. Age is also an important risk factor.

2. Mitochondrial dysfunction may play an important role in the pathogenesis of

COPD. Targeting mitochondria may be a novel strategy to develop new treatment

for the disease.

3. Efficacy of MSCs isolated from BM, adipose tissue and cord blood on

CS-induced emphysema models has been demonstrated. BM-MSCs have been

proven to be safe in clinical trials but without efficacy in COPD patients.

4. iPSC-MSCs have several advantages over BM-MSCs including more abundant

resource, high capacity of expanding without loss of differentiation potency and

Page 72: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

53

potential to overcome ageing-associated impairment.

5. The mechanisms of action of MSCs are not fully understood. Paracrine effects and

mitochondrial transfer represents two possible mechanisms.

Based on the above knowledge, my Ph.D study focuses on the effects of

iPSC-MSCs on airway cells in a COPD or oxidative stress-related context. The aims

of the study include:

1. To compare effects of iPSC-MSCs and BM-MSCs on CS-induced damage in

the rat lung in vivo and in human bronchial epithelial cells in vitro with an

emphasis on apoptosis. Both mitochondrial transfer and paracrine effects

were examined.

2. To study the capacity of iPSC-MSCs to alleviate mitochondrial dysfunction in

CS-treated human ASMCs in vitro and to examine the relevant contribution of

mitochondrial transfer and paracrine effects to the efficacy.

3. To investigate the capacity of iPSC-MSCs to alleviate oxidative

stress-induced AHR, lung inflammation, mitochondrial dysfunction and

apoptosis in an acute ozone-exposed mouse model.

These studies should provide evidence to demonstrate the potential of

iPSC-MSCs as a candidate for cell-based therapy for COPD.

Page 73: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

54

Chapter 2 Material and Methods

Page 74: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

55

2.1 Materials

2.1.1 Reagents and chemicals

Chemicals, reagents and recombinant proteins were supplied by Thermo Fisher

Scientific (Carlsbad, CA, USA), Sigma-Aldrich (St. Louis, MO, USA), Amresco

(Solon, OH, USA), Bio-Rad Laboratories (Richmond, CA, USA), Cayman Chemical

(Ann Arbor, MI, USA), Life Technologies (Eugene, OR, USA) and Roche (Mannheim,

Germany). Cell culture reagents were purchased from Invitrogen (Thermo Fisher

Scientific) and Sigma-Aldrich. Cigarettes including Camel (11 mg TAR and 0.8 mg

nicotine, R.J. Reynolds, Winston-Salem, NC, USA) or Marlboro Red (10 mg TAR

and 0.8 mg nicotine, Philip Morris International, New York City, NY, United States)

were obtained from commercial retailers.

2.1.2 Experimental animals

For the in vivo study performed in Hong Kong, the procedures of the project and

the personnel involved were under licenses from Department of Health, Hong Kong

SAR and approved by the Committee on the Use of Live Animals in Teaching and

Research (CULATR) of the University of Hong Kong. Male Sprague-Dawley (SD)

rats (weighted 170-200 g) were purchased from and kept in the Laboratory Animal

Unit, the University of Hong Kong.

For the in vivo study performed at Imperial College London, the procedures of

the project and the personnel involved were under a Project License and Personal

Licenses from the British Home Office, United Kingdom, under the Animals

Page 75: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

56

(Scientific Procedures) Act 1986. Male C57BL/6 mice (6 weeks old) were purchased

from Harlan UK Ltd (Wyton, UK) and kept in Central Biomedical Services, Imperial

College London.

The living environment of all animals was monitored and controlled with respect

to temperature, humidity, and day/night cycle. The animals‟ general conditions were

monitored daily.

2.1.3 Human iPSC-MSCs

The iPSC-MSCs were kindly provided by Dr. Lian Q and Professor Tse HF from

the Cardiology Division, Department of Medicine, LKS Faculty of Medicine, HKU.

The protocol to generate iPSC-MSCs includes two steps: inducing iPSCs from

somatic cells, and generating iPSC-MSCs from the iPSCs. Firstly, iMR90 fibroblast

cells (CCL-186, American Type Culture Collection, Manassas, VA, USA) were

transduced with human OCT4, SOX2, NANOG and LIN28 genes through lentiviral

vectors (Plasmid 16577-80, Addgene, Cambridge, MA, USA) overnight. The

transduction gene mixture was then replaced with human embryonic stem cell culture

medium and cells transferred to inactivated mouse embryonic fibroblast (MEF) feeder.

20 days later, cell colonies with human embryonic stem cell morphology were

identified as iPSC-MSCs and propagated. The resulting iPSC lines were differentiated

into iPSC-MSCs as previously described (153). iPSCs were harvested and placed on a

gelatinized 10-cm dish containing knockout Dulbecco‟s modified Eagle medium

(DMEM) (Invitrogen) supplemented with 10% serum replacement medium

Page 76: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

57

(Invitrogen), 10 ng/mL basic fibroblast growth factor (bFGF) (Invitrogen), 10 ng/mL

platelet-derived growth factor AB (Peprotech, Rocky Hill, NH, USA), and 10 ng/mL

epidermal growth factor (EGF) (Peprotech) to induce differentiation. Cells were left

to differentiate for 7 days after which they were harvested and incubated with

CD24-phycoerythrin and CD105-FITC (BD PharMingen, San Diego, CA, USA). The

CD24-CD105

+ population were separated by fluorescence-activated cell sorting

(FACS). The CD24-CD105

+ cells were diluted and seeded in 96-well plates to select

wells containing a single cell. Upon reaching 70% confluence, the single cell-derived

clones were serially sub-cultured to confluence in a 175-cm2 tissue culture flask. They

were then trypinised, aliquoted and frozen as stocks in liquid nitrogen for future

experiments.

2.1.4 Human bone marrow – derived MSCs (BM-MSCs)

Human BM-MSCs were obtained from a commercial supplier (Cat. No. PT-2501,

Cambrex Bioscience, Rockland, ME, USA). Both iPSC-MSCs and BM-MSCs were

cultured in DMEM (Invitrogen) supplemented with 10% fetal bovine serum

(Invitrogen), 5 ng/mL bFGF (R&D Systems, Minneapolis, MN, USA), 10 ng/mL EGF

(R&D Systems), 55 μM 2-mercaptoethanol (Invitrogen), 20 units/ml penicillin and 20

μg/ml streptomycin.

2.1.5 BEAS-2B cells

The BEAS-2B cells were virus-transformed immortalized human bronchial

Page 77: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

58

epithelial cells from autopsy of non-cancerous individuals. The BEAS-2B cells in this

study were from a commercial supplier (American Type Culture Collection, Rockville,

MD, USA). They were cultured in keratinocyte serum free media K-SFM (Invitrogen)

supplemented with 5 ng/ml EGF and 50 mg/ml bovine pituitary extract. Passages

42-50 were used for experiments.

2.1.6 Primary human airway smooth muscle cells (ASMCs)

ASMCs were isolated from biopsies of bronchi or tracheas of healthy transplant

donor lungs as previously described (174). Briefly, biopsies were cut into small pieces

(<1 mm2) and placed in DMEM supplemented with 10% FBS, 4 mM L-glutamine, 20

units/ml penicillin, 20 μg/ml streptomycin and 2.5 μg/ml amphotericin B. ASMCs

were confirmed by the characteristic “hill and valley” morphology under light

microscopy. Cells were sub-cultured to reach confluence in 175-cm2 tissue culture

flasks and then frozen as stock. Passages 3 to 7 were used for experiments. When

serum-free medium was required in the experiments, it was prepared as phenol-free

DMEM supplemented with 1 mM sodium pyruvate, 4 mM L-glutamine, 1:100

non-essential amino acids, 0.1% BSA, 20 units/ml penicillin, 20 μg/ml streptomycin

and 2.5 μg/ml amphotericin B.

All of the above cell types were maintained in a humidified incubator of 5% CO2

and 37C and fed with medium every 2-3 days.

Page 78: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

59

2.2 Methods

2.2.1 MSCs treatment to cigarette smoke-exposed rats

Male Sprague-Dawley rats were exposed to 4% (v/v) CS for one hour daily for 56

days (Figure 2.1) using a same protocol as described in a previous study (52). Briefly,

a maximum of 8 rats were placed into a ventilated 20 liter-chamber. 800 ml of CS was

injected into the chamber through a syringe to raise the initial CS concentration to

4%.The filters of the cigarettes were removed beforehand. The chamber was then

ventilated with a steady inflow of 4% CS at 1 L/min – a gas mixture of 100% CS (40

ml/min) and normal air (960 ml/min) from two separate peristaltic pumps (Masterflex,

Cole-Parmer Instruments Co, Niles, IL, USA) (Figure 2.2). The control group was

exposed to sham air (SA) using the same protocol. The exposure was performed in a

chemical fume hood at the same time of each day. At day 29 and day 43, CS-exposed

rats were intravenously injected with plain PBS, 3x106 human BM-MSCs or 3x10

6

human iPSC-MSCs into tail vein (Figure 2.1). The SA group was injected with PBS.

Daily check of general conditions showed no obvious adverse reactions towards the

injections.

Rats were sacrificed 24 hours after the last CS or air exposure by intraperitoneal

injection of over-dosed pentobarbitone (100 mg/kg body weight). The larger lobe of

left lungs was firstly inflated with 1 ml formalin and then fixed, dehydrated and

paraffin embedded. The paraffin block was cut into 5 μm sections. The rest of the lung

tissue was frozen and stored at -80℃.

Page 79: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

60

Figure 2. 1 Schematic diagram of BM-/iPSC-MSCs treatments in a cigarette

smoke (CS)-exposed rat model. Male Sprague-Dawley rats were exposed to 4% CS

for 1 hour daily for 56 days. At Days 29 and 43, 3x106

human iPSC-MSCs (n = 8) or

BM-MSCs (n = 8) were injected intravenously through tail veins. A sham air (SA)

group was included in which the rats were exposed to fresh air. At Day 57, the rats

were sacrificed, and the larger lobe of the left lungs were fixed and sectioned for

histological studies.

Air Exposure (1 hour/day, 56 days)

PBS

4% CS Exposure (1 hour/day, 56 days)

4% CS Exposure (1 hour/day, 56 days)

4% CS Exposure (1 hour/day, 56 days)

PBS

Sacrifice

PBS

PBS

BM-MSCs

Day 29 Day 43 Day 57

BM-MSCs

iPSC-MSCs

iPSC-MSCs

Sacrifice

Sacrifice

Sacrifice

SA

CS

iPSC-MSCs

BM-MSCs

Male SD rats

(170-200 g)

Page 80: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

61

Figure 2. 2 The set-up of the ventilated smoking chamber. In the right chamber, the

fresh air and CS were mixed in ratio of 96:4 to generate constant flow of 4% CS at

1000 ml/min. In the left chamber, only fresh air was pumped. The whole set-up was

placed in a chemical fume hood.

2.2.2 Histology and morphometric analysis of rat lungs

The paraffin sections were always deparaffinised and dehydrated before staining.

The slides were incubated at 40℃ in an oven for one hour to melt the paraffin,

followed by incubation twice in xylene for 15 minutes. A serial rehydration was then

applied by incubation in 100%, 95%, 75% ethanol and double distilled water (ddH2O)

successively, each for 10 minutes, twice.

Mean linear intercept (Lm) was measured to evaluate airspace enlargement based

on a modified method (62). Briefly, the paraffin sections from the largest lobes of rat

Page 81: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

62

lungs were rehydrated and stained with hematoxylin and eosin. The slides were then

mounted in a xylene-based mounting medium, observed using a brightfield

microscope at 10x magnification and analysed using AxioVision (Zeiss, Germany). A

100 μm-length interval was inserted in the field of a randomly selected region of the

lung sections. The interval passed through several alveoli, resulting in multiple

alveolar intercepts. Lm was calculated as 100 μm divided by the number of alveolar

intercepts. 10 fields per section, 8 rats per group were analysed.

Masson's trichrome staining was performed to measure the pulmonary fibrosis

according to the standard protocol. The pulmonary fibrosis was assessed by

semi-quantitative Ashcroft score, which gave each field a score ranging from 0

(normal lung), 1 (minimal fibrous thickening of alveolar or bronchiolar walls), 3

(moderate thickening of walls without obvious damage to lung architecture), 5

(increased fibrosis with definite damage to lung structure and formation of fibrous

bands or small fibrous masses), 7 (severe distortion of structure and large fibrous

areas) to 8 (total fibrous obliteration of the field) (12). Scores from 5 fields per section,

total 6 rats, were assessed under 20x magnification.

2.2.3 iPSC-MSCs treatment to ozone-exposed mice

Male C57BL/6 mice were put into a chamber ventilated with gas mixture of

normal air and ozone produced by an ozone generator (model 500 Ozoniser, Sander,

Wuppertal, Germany). The concentration of ozone in the chamber was monitored by

an ozone detector and maintained at 3 ppm by tuning the ozone generator (Figure 2.3).

Page 82: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

63

Control groups were exposed to normal air. The exposure lasted for 3 hours.

iPSC-MSCs or PBS were administrated to mice by intravenous injection through

tail vein. The administration was performed either 24 hours prior or 6 hours after the

exposure (Figure 2.4). There were 5 groups of mice in total: Air, Air with iPSC-MSCs

administrated 24 hours prior (-24 hr), Ozone, Ozone with iPSC-MSCs administrated

24 hours prior-exposure (-24 hr) and Ozone with iPSC-MSCs administrated 6 hour

post-exposure (+6 hr). The Mice from all groups were anaesthetised for AHR

measurements 21 hours after the exposure. They were sacrificed immediately after the

AHR measurements by intraperitoneal injection of over-dosed pentobarbitone (100

mg/kg body weight). Bronchoalveolar lavage (BAL) was collected before the

collection of the lungs. The right lobes were frozen for mitochondria extraction while

left lobes were inflated with and fixed in paraformaldehyde. The larger lobe of the

fixed lungs were later embedded into paraffin blocks and sectioned into 5

μm-sections.

Figure 2. 3 The set-up of the ventilated chamber for ozone exposure. Ozone

generated from an ozone generator and normal air from an air pump was ventilated

into the chamber. The ozone concentration was constantly monitored by an ozone

detector and maintained at 3 ppm by tuning the ozone generator.

Page 83: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

64

Figure 2. 4 Schematic diagram of iPSC-MSCs treatments in an ozone-exposed

mouse model. Male C57BL/6 mice were exposed to ozone (3 ppm) for 3 hours.

Control groups were exposed to fresh air. 106

iPSC-MSCs were intravenously injected

into the tail vein either 24 hours prior to the ozone exposure, or 6 hours after the

exposure. Airway hyper-responsiveness (AHR) was measured 21 hours after the

exposure. The mice were then sacrificed and BAL and lung tissues collected.

Page 84: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

65

2.2.4 Measurement of airways hyper-responsiveness (AHR) of mice

The AHR was measured as previously described (272). Before the measurement

of AHR, mice were anaesthetised by intraperitoneal injection of

ketamine/xylazine/saline mixture. The trachea was then opened and inserted with a

catheter through which the mouse was ventilated (MiniVent type 845, Hugo Sach

Electronic, Germany) at 250 breaths/minute and a tidal volume of 250 μl. The animal

was continuously monitored in a whole body plethysmograph with a

pneumotachograph connected to a transducer (EMMS, Hants, UK). Another

transducer was connected to an esophageal catheter to evaluate the intrapleural

pressure. The transpulmonary pressure was assessed based on the pressure

measurements from the two transducers. Increasing concentrations (4-256 mg/ml) of

acetylcholine in PBS were then administered into airway via a nebulizer (Aeroneb

Lab Micropump Nebulizer, EMMS) and the pulmonary resistance (RL) was recorded

for 3 minute periods during each dose. Baseline RL was defined as the RL with

nebulised PBS. A concentration-response curve was plotted for each animal and the

concentration of acetylcholine that induced 100% elevation of RL from baseline was

derived (PC100). The value of -log PC100 was taken as a measurement of AHR.

2.2.5 Measurement of cells and cytokines in BAL of mice

Bronchoalveolar lavage (BAL) was collected by rinsing the lungs three times

with 0.8 ml cold PBS through an endotracheal tube. After centrifugation at 3000 rpm,

4°C for 5 min, the supernatant was collected as BAL fluid (BALF) while the cell

Page 85: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

66

pellets were resuspended in 200 μl PBS and counted on a haemocytometer for total

cell number. BAL cell samples were centrifuged again onto glass slides by a cytospin

device at 30g for 6 minutes (Shandon Cytospin 4; Thermo Electron Corporation,

Waltham, MA, US). The resulting cell slides were air-dried overnight and stained

using Diff-Quick kit (Reagena, Toivala, Finland) according to manufacturer‟s

instructions. Differential count of white blood cells (neutrophil, macrophage,

lymphocyte and eosinophil) was performed under an optical microscope (Olympus

BH2, Olympus).

The cytokine levels in BALF of mice were determined using a MAGPIX

Luminex multiplexing analyser (Merck Millipore, Darmstadt, Germany), which

simultaneously measured levels of multiple analytes, including MCP-1, Eotaxin, IL-6,

IL-5 and macrophage inflammatory proteins (MIP)-1α.

2.2.6 Isolation of intact mitochondria from mouse lungs

Intact mitochondria were isolated from mouse lungs using a Mitochondria

Isolation Kit for Tissue (Thermo Fisher Scientific) and Dounce tissue grinder set

(Sigma) following the manufacturer‟s instructions. In brief, approximately 20 mg of

lung tissue was washed with PBS supplemented with EDTA-free protease inhibitors

(Roche). The tissue was homogenized in corresponding reagent with 5 strokes of

Dounce A followed by 20 strokes of Dounce B. The homogenates were centrifuged

firstly at 700xg for 10 minutes at 4°C, to sediment large organelle fractions containing

nucleus, plasma membrane, endoplasmic reticulum, Golgi and so on. The

Page 86: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

67

supernatants were centrifuged again at 12,000xg for 5 minutes. The supernatants and

pellets were collected as cytosolic fractions and mitochondrial fractions, respectively.

2.2.7 Generation of cigarette smoke medium (CSM)

CSM was prepared as previously described (143). Briefly, cigarette smoke from

two filter-removed cigarettes was bubbled into 20 ml of serum free medium. The

medium was then filtered through a 0.22 μm filter. The optical density (OD) at

wavelength 320 nm was measured.

For the study with BEAS-2B cells which was performed at Hong Kong university,

the cigarettes smoke was from Camel cigarettes (11 mg TAR and 0.8 mg nicotine),

and pumped into serum free medium manually by syringe. 100% CSM was generated

by adjusting the OD at 320 nm with serum free medium to 3.5.

For the study of ASMCs which was performed at Imperial College London, the

cigarette smoke was from Marlboro Red cigarettes (10 mg TAR and 0.8 mg nicotine)

and pumped into serum free medium through a peristaltic pump, which resulted in a

lower OD read-out than the manual method. Thus the 100% CSM was generated by

adjusting the OD at 320 nm with serum free medium to 1.1. The advantage of

peristaltic pump is a very consistent output for each batch of CSM.

After preparation, the 100% CSM was aliquoted and stored at -80℃.

2.2.8 Co-culture of BEAS-2B cells with MSCs.

BEAS-2B cells were pre-stained with CellTrace Violet (Invitrogen) which

Page 87: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

68

fluorescently labelled the cells for several generations while iPSC-MSCs or

BM-MSCs were pre-stained with MitoTracker Green (Invitrogen), which specifically

stained the mitochondria in the cells. In brief, BEAS-2B cells were harvested, counted,

incubated with 5 µM Celltrace Violet for 20 minutes, while MSCs were harvested,

counted and incubated with 200 nM Mitotracker Green for 30 minutes. They were

centrifuged at 1000 rpm for 5 min and incubated in complete medium for 10 min after

the staining to wash away the remaining dye. After another centrifugation, they were

re-suspended in 1:1 mixed medium and co-cultured at a 1:1 ratio in a 24-well plate.

Approximately 5 hours later, when the cells attached to the plates, 100% CSM was

added into the medium to make the final concentration 2%. After 24 hours, cells were

visualized by motorized inverted microscope (IX81-ZDC2; Olympus, Shinjuku,

Tokyo City, Tokyo, Japan) or analyzed by flow cytometry with a BD FACS Aria I

Cell Sorter (BD Biosciences, Oxford, UK).

2.2.9 Determination of ATP content in BEAS-2B cells following co-culture

BEAS-2B cells in the co-culture were sorted as CellTrace positive cells by a BD

FACS Aria I Cell Sorter. ATP was extracted with boiling distilled water according to a

one-step method previously described (284). The ATP content was measured by a

luciferase-catalysed luminescent assay using an ATP Determination Kit (Invitrogen)

according to the manufacturer‟s instruction.

Page 88: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

69

2.2.10 BM-MSC and iPSC-MSC conditioned medium treatment of CSM-treated

BEAS-2B cells

The conditioned medium (CdM) was prepared as previously described (294).

Briefly, BM-MSCs and iPSC-MSCs were re-fed with DMEM without serum and

supplements. After 24 hr, the medium was collected and concentrated via

centrifugation through ultrafiltration conical tubes (Amicon Ultra-15 with membranes

selective for 5 kDa). The final concentration was adjusted to 20 times that of the

collected CdM.

Alternatively, to investigate the effects of stem cell factor (SCF), SCF was

depleted from iPSC-MSCs-CdM as previously described (139). Briefly, 0.5 μg of

anti-SCF (AF-255-NA; R&D) or normal human IgG control antibody (1-001-A; R&D)

were mixed with protein G-agarose beads in PBS at 4°C for 1 hour with intermittent

shaking. After centrifugation, beads were washed three times and used for

immunedepletion of SCF. iPSC-MSCs-CdM was incubated with protein G-agarose

beads immobilized with anti-SCF antibodies or human IgG control antibody for 1

hour at 4°C. The beads with immune complexes attached were precipitated by

centrifugation. The supernatants collected were either SCF-depleted

iPSC-MSCs-CdM or control iPSC-MSCs-CdM.

The BEAS-2B cells were cultured on top of coverslips in 24-well plates. The

medium was replaced by keratinocyte medium with no supplements 24 hr before the

treatment. They were then treated with 2% (v/v) CSM. BM-MSCs-CdM or

iPSC-MSCs-CdM containing 3 μg of total protein was added at the same time. After

Page 89: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

70

24 hours, the supernatant was removed and the cells fixed for immunohistochemical

tests or terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling

(TUNEL) assay. To investigate role of SCF, the cells were treated with either

SCF-depleted iPSC-MSCs-CdM or recombinant SCF (255-SC, R&D) with 2% CSM.

2.2.11 Immunohistochemistry

To detect expression and localization of various proteins in lung sections and cell

culture, standard immunohistochemistry procedures were applied. Briefly, lung

sections were serially rehydrated and antigen retrieved by boiling for 5 minutes at

120C in Sodium Citrate Buffer (10 mM sodium citrate, 0.05% Tween 20, pH 6.0).

They were then blocked by 5% bovine serum albumin (BSA) for 30 minutes at room

temperature followed by overnight incubation in primary antibodies at 4C. The

primary antibodies included human nuclear antigen (HNA) (MAB1281, Chemicon),

human/rat clara cell secretory protein 10 (CC10) (SC-9772, Santa Cruz, CA, USA),

human/rat/mouse Ki-67 (ab15580, Abcam, Cambridge, UK), human/rat Complex I

(SC-271387, Santa Cruz), human COX4 (SC-133478, Santa Cruz), rat/mouse CD68

(SC-7084, Santa Cruz), mouse/rat/human C-kit (SC-1494, Santa Cruz). After 3 washs

with PBS, the sections were incubated in secondary antibodies including horseradish

peroxidase-conjugated or fluorescent-conjugated anti-mouse IgG, anti-rat IgG and

anti-rabbit IgG for 1 hour at room temperature. For HRP-conjugated secondary

antibodies, the targets were recognized by an intense brown colour after developing

with 3,3'-diaminobenzidine (DAB) substrate solution. For fluorescent-conjugated

Page 90: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

71

secondary antibodies, the sections were mounted with mounting medium (ProLong

Gold antifade reagent with DAPI, Invitrogen) containing

4,6-diamidino-2-phenylindole (DAPI) to visualize nuclei. At least five fields per

section were randomly captured under a conventional fluorescent microscope

(IX81-ZDC2; Olympus) at the desired magnification and analyzed using AxioVision

(Zeiss, Oberkochen, Germany).

For cell culture, the cells were seeded and treated on coverslips in 24-well dishes.

They were fixed with 4% paraformaldehyde in PBS for 15 min, blocked with 5%

BSA for 30 min and permeabilized with 0.1% Triton-100 in PBS for 3 min, all at

room temperature. The cells were incubated in primary antibodies including C-kit

(SC-1494, Santa Cruz) and cytochrome c (SC-13156, Santa Cruz) at room

temperature for 1 hr and then in fluorescent-conjugated secondary antibodies. The

coverslips were taken out from the wells, flipped and put on a drop of mounting

medium containing DAPI on glass slides. Images of 5 fields for each slide were

captured randomly by a motorized inverted microscope (Olympus) and analyzed

using AxioVision (Zeiss). The ratio of positively stained cells in the total cell

population was calculated as the rate of cytochrome c translocation, apoptosis or

proliferation.

2.2.12 Terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling

(TUNEL) assay

TUNEL assay detects cell apoptosis by labelling the terminal end of nucleic acids

Page 91: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

72

which increase in abundance in cells only if the DNA becomes fragmented. Apoptosis

of BEAS-2B cells and mouse lung sections were determined via TUNEL assay using

the In Situ Cell Death Detection Kit, POD (Roche Applied Science, Mannheim,

Germany).

For analysis of the lung sections, they were rehydrated as per the

immunohistochemistry protocol, and heated in citrate buffer in a microwave oven for

5 minutes. They were then washed with PBS and incubated with TUNEL reaction

mixture for 1 hour. Nucleus of apoptotic cells were stained green by the reaction.

For the BEAS-2B cells, they were fixed with 4% paraformaldehyde in PBS,

blocked with 3% H2O2 in methanol, permeabilized with 0.1% Triton-100 in 0.1%

sodium citrate and washed with PBS. TUNEL reaction mixture was then applied for a

one-hour incubation at 37 ℃.

All samples were mounted in mounting medium containing DAPI. Images of 5

fields for each slide were captured randomly by a motorized inverted microscope

(Olympus) and analysed using AxioVision (Zeiss).

2.2.13 Total protein extraction from cell culture

To extract total protein from cultured cells, cells in 6-well plates were first

washed with ice-cold PBS and then frozen at -80℃. The cells were then scraped with

approximately 50 μl/well T-PER Cell Protein Extraction Reagent (Thermo Fisher

Scientific) supplemented with protease/phosphatase inhibitor cocktail (Thermo Fisher

Scientific) and 100 mM PMSF. The cell/reagent mixture were vortexed, put on ice for

Page 92: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

73

30 min and centrifuged at 16000xg at 4℃ for 30 minutes. The supernatant was

collected as cell lysates and stored at -80℃. Protein concentrations were determined

by Bradford protein assay using bovine BSA as standard.

2.2.14 Western blot

Cell lysates containing the comparable amounts of total protein (20-40 μg) were

denatured and reduced by boiling at 95℃ for 5 minutes in Laemmli buffer with

sodium dodecyl sulfate (SDS) and 2-mercaptoethanol. The proteins in the samples

were separated by size by running through an SDS–polyacrylamide gel. They were

transferred onto a polyvinylidene fluoride (PVDF) membrane (Bio-Rad Laboratories)

which was then blocked in 5% skimmed milk in Tris-buffered saline (pH 7.4) with 0.1%

Tween-20. The membranes were incubated with diluted anti-C-kit (1:500) (SC-1494,

Santa Cruz) or anti-β-actin (1:5000) antibodies at 4°C overnight, washed three times,

and then incubated with horseradish peroxide-conjugated anti-goat antibody (1:2000)

or anti-mouse antibody (1:2000) (Dako, Danmark) for 1 h at room temperature. After

three further washes and a one-minute incubation with enhanced chemiluminescence

(ECL), the membranes were exposed to x-ray films (Fujifilm, Tokyo, Japan) on which

the target proteins were developed as dark bands.

2.2.15 Enzyme-linked immunosorbent assay (ELISA)

The concentration of SCF in CdM was measured by Human SCF DuoSet ELISA

Kit (R&D) according to manufacturer‟s instructions. In general, samples and serially

Page 93: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

74

diluted standards were added into 96-well plates coated with capture antibody. After

adequate washing, the captured target proteins were further incubated with detecting

antibody and streptavidin conjugated with horseradish peroxidase (HRP),

consecutively. HRP catalysed a colorimetric reaction of tetramethylbenzidine (TMB)

substrates. The reaction was stopped by adding 2 N H2SO4, and the OD was measured

at wavelength 450/570 nm in a microplate reader (FLUOstar Optima, BMG Labtech).

A standard curve was generated according to OD values of the standards, and SCF

levels of CdM were calculated from the standard curve.

2.2.16 Treatment of CSM or H2O2 to ASMCs or iPSC-MSCs

ASMCs were cultured in 6-well plates or 96-well plates. On reaching 90%

confluence, the medium was replaced with serum-free medium for 24 hours. The cells

were then treated with different concentrations of H2O2 (50, 100 and 200 μM) or CSM

(10, 25, 50 and 75%) for 2, 4 and 24 hours. A cessation group was also included, in

which after 4 hours of treatment H2O2 or CSM was replaced with fresh serum-free

medium for the next 20 hours. The iPSC-MSCs were similarly treated for 4 hours and

24 hours. The cell viability, mitochondria membrane potential and mitochondrial ROS

levels were measured by methylthiazolyldiphenyl-tetrazoliumbromide (MTT) assay,

JC-1 (5,5‟,6,6‟-tetrachloro-1,1‟,3,3‟-tetraethylbenzimidazolylcarbocyanineiodide) and

MitoSOX staining, respectively.

Page 94: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

75

2.2.17 Cell viability assay

MTT assay is a commonly used cell viability assay. In brief, MTT

(Sigma-Aldrich) was dissolved in 37 ℃ serum free medium at 1 mg/ml. ASMCs or

iPSC-MSCs cultured in 96-well plates were incubated with 100 μl MTT solution at

37 ℃ after treatments. After 15 min, the cells were checked every 5 minutes using

microscope to observe formation of purple color within. When the color is visible but

not too intense, the MTT solution was replaced with 50 μl DMSO. After shaking for 5

minutes, the OD at wavelength 550 nm was measured.

2.2.18 Measurement of mitochondrial ROS

Mitochondrial ROS levels were assessed by staining with MitoSOX Red

Mitochondrial Superoxide Indicator (Invitrogen). 50 μg MitoSOX powder was

dissolved in 13 μl DMSO to make a 5 mM stock solution and further diluted to 5 μM

with relevant buffers to make working solutions.

For the in vitro study of ASMCs, cells in 6-well plates were washed with

modified Hank‟s Balanced Salt Solution (HBSS) (containing Ca/Mg) (Sigma-Aldrich),

and incubated with 5 μM MitoSOX diluted in modified HBSS for 30 minutes. The

cells were then washed with HBSS (Sigma-Aldrich) and incubated with 500 µl/well

accutase solution (Sigma-Aldrich) for up to 5 min at 37 °C and 5% CO2. One ml/well

DMEM with 4 mM L-glutamine and 10% FBS was then added to the detached cells.

After centrifugation and resuspension in HBSS, the intensity of red fluorescence was

measured at 510/580 nm by a FACSCanto II flow cytometer (BD Biosciences).

Page 95: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

76

For the in vivo study of ozone-exposed mice, the intact mitochondria were

isolated from lungs, and then incubated with 5 μM MitoSOX in 96-well plates for 30

minutes at 37°C. The intensity of red fluorescence was measured at 510/580 nm using

a fluorescence plate reader.

2.2.19 Mitochondrial membrane potential

Mitochondrial membrane potential (ΔΨm) was determined by staining with JC-1

dye (Invitrogen). The dye exists as monomers with green fluorescence, but when

they enter live mitochondria they form J-aggregates which exhibit red fluorescence.

The accumulation of the dye in mitochondria is potential-dependent, making the ratio

of red to green fluorescence as an indicator of ΔΨm. ASMCs or intact mitochondria

from mouse lungs were incubated with 2 µM of JC-1 for 30 minutes at 37°C. The

fluorescent signal was determined either using a FACSCanto II flow cytometer (BD

Biosciences) for the living cells, or a fluorescence plate reader (Synergy HT Biotek,

Winooski, VT, USA) for the isolated mouse mitochondria. The green fluorescence

from JC-1 monomers was measured at excitation/emission ratios of 485/535 nm and

the red fluorescence from J-aggregates at 560/595 nm.

2.2.20 Annexin V staining

The apoptosis of ASMCs/iPSC-MSCs co-culture was detected by Annexin V

staining through flow cytometry. Annexin V detects apoptosis by binding to the

externalized phosphatidylserine in apoptotic cells. Briefly, cells were harvested and

Page 96: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

77

resuspended at a density of 106 cells/ml in assay buffer. 5 μl of FITC-conjugated

Annexin V (Invitrogen) was added into 100 μl of cell suspension. After 15

minutes-incubation at room temperature, the intensity of Annexin V was measured by

a FACSCanto II flow cytometer (BD Biosciences).

2.2.21 Direct co-culture of ASMCs and iPSC-MSCs

To study the effects of direct interaction between ASMCs and iPSC-MSCs on

mitochondrial ROS, ΔΨm and apoptosis, ASMCs and iPSC-MSCs were co-cultured

in using two different procedures, one as a prophylactic study and the other as

therapeutic (Figure 2.5).

For the prophylactic study, the ASMCs and iPSC-MSCs were cultured in T150

flasks separately before the experiments. The ASMCs were trypsinized, resuspended

and counted. They were washed with HBSS by centrifugation and resuspension,

followed by 20 minutes-incubation with CellTrace Violet (1:1000 in HBSS, 1 ml/106

cells). After centrifugation, they were incubated with complete medium for 10

minutes to wash and neutralize the dye. Meanwhile, the iPSC-MSCs were trypsinized,

resuspended and counted. The two cell types were mixed and seeded in 6-well plates

at a density of 105 ASMCs plus 1.5x10

5 iPSC-MSCs per well. Single cell culture

groups were included as comparison, in which only 105 ASMCs or 1.5x10

5

iPSC-MSCs were seeded into each well. After 20 hours, the medium was replaced

with 10% or 25% CSM for 4 hours. The mitochondrial ROS, ΔΨm and apoptosis

were then assessed by MitoSOX, JC-1 and Annexin V staining through flow

Page 97: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

78

cytometry.

For the therapeutic study, 105 ASMCs were first seeded in each well of 6-well

plates. 20 hours later, they were incubated with CellTrace Violet for 20 minutes

followed by treatment with 10% or 25% CSM for 4 hours. The CSM were then

removed and 1.5x105

iPSC-MSCs were added into each well. After another 24 hours,

the mitochondrial ROS, mitochondrial membrane potential and apoptosis were

measured through flow cytometry.

2.2.22 Detection of mitochondrial transfer in co-culture of ASMCs and

iPSC-MSCs

To detect mitochondrial transfer in the co-culture of ASMCs and iPSC-MSCs,

ASMCs pre-stained with CellTrace Violet and iPSC-MSCs pre-stained with

MitoTracker Red (for fluorescent microscope) or MitoTracker Green (for flow

cytometry) were mixed and seeded into 6-well plates at a density of 105 ASMCs plus

1.5x105

iPSC-MSCs per well. 20 hours later, they were treated with 10% or 25%

CSM and analyzed by flow cytometry. The percentage of ASMCs that were

MitoTracker positive was subsequently calculated. For fluorescent microscopy, the

cells were fixed in 3.7% formaldehyde (Sigma-Aldrich) for 10 minutes, permeabilized

in 0.1% Triton X-100 (Sigma-Aldrich) for 5 minutes and stained by Alexa Fluor 488

phalloidin (Invitrogen) in PBS with 1% BSA. Phalloidin is a probe that labels

filamentous actin selectively. The cells were then visualized by a motorized inverted

fluorescent microscope.

Page 98: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

79

A

B

Figure 2. 5 Protocols of direct co-culture of iPSC-MSCs and ASMCs. (A)

Prophylactic co-culture with iPSC-MSCs. 1.5x 105 ASMCs were pre-stained with

CellTrace Violet and seeded in 6-well plates either with or without 105

iPSC-MSCs. 20 hours later they were treated with CSM for 4 hours. MitoSOX ,

JC-1 and Annexin V staining were applied to measure mitochondrial ROS, ΔΨm

and apoptosis. (B) Therapeutic co-culture with iPSC-MSCs. 1.5x 105 ASMCs were

labeled with CellTrace Violet and treated with CSM for 4 hours. CSM was

removed and either 105 iPSC-MSCs or serum free medium was added. After 20

hours of co-culture, the mitochondrial ROS and membrane potential were

measured.

Page 99: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

80

2.2.23 iPSC-MSCs-conditioned medium treatment to ASMCs

The iPSC-MSCs-CdM was prepared as described above. The ASMCs were

pretreated with 20-fold diluted iPSC-MSCs-CdM for 4 hours. The CSM were then

added on top of the iPSC-MSCs-CdM to make 10% or 25% CSM. Serum free

medium was added to the control group or 10% CSM group to keep the total volume

of each well the same. 4 hours later, the mitochondrial ROS, ΔΨm and apoptosis were

measured.

2.2.24 Transwell co-culture between ASMCs and iPSC-MSCs

To study the effects of paracrine crosstalk between ASMCs and iPSC-MSCs, they

were co-cultured in a transwell co-culture system. In brief, 105 ASMCs were seeded

in 6-well plates while 1.5x105

iPSC-MSCs were seeded in a cell-culture insert with

pores sized 0.4 μm (Falcon, Corning, NY, USA). They were incubated overnight and

then combined. Single-culture groups were included with empty inserts or empty

wells. After 20 hours‟ co-culture, they were treated with 10% or 25% CSM for 4

hours, and assessed for the mitochondrial ROS, ΔΨm and apoptosis.

2.2.25 Statistical analysis

Numerical data are presented as mean±standard error of mean (SEM). The

statistical analysis was performed using software Prism 5.0 (Graphpad, San Diego,

CA, USA). Differences between groups were evaluated by student t-test or one-way

ANOVA followed by Newman-Keuls test when appropriate. Significant difference

was defined as when p-value < 0.05. The number of animals required for the study

Page 100: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

81

was determined by power calculations (78) using data from previous studies

conducted in the department. In particular, for CS-exposed rats, the Lm were

31.28±1.75 μm vs. 19.79±0.81 μm for CS and SA groups (n=9) in a previous report

(52). Based on this change, the maximal number of animals to attain statistical

significance of p<0.01 with a 95% probability equated to 6. Considering possible loss

of animals during experimental period, n=8 was chosen for the rat experiments. For

the ozone-exposed mice, -logPC100 was reported to be 1.059±0.127 vs. 2.088±0.136

for ozone and air groups (n=8) respectively (275). Based on these data, the maximal

number of animals to attain statistical significance of p<0.01 with a 95% probability

equated to 5. n=5-6 was chosen for the mouse study.

Page 101: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

82

Chapter 3. iPSC-MSCs Attenuate CS-induced

Damage in Airway Epithelial Cells through

Mitochondrial Transfer and Paracrine Effect

Page 102: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

83

3.1. Introduction

Cigarette smoke (CS) is the primary cause of chronic obstructive pulmonary

disease (COPD) (23, 39). In the first part of this study a rat model of chronic exposure

to CS was used to study the effects of BM-MSCs and iPSC-MSCs on CS-induced

alterations of lung regarding emphysema, fibrosis, inflammation, apoptosis and

proliferation. Male SD rats were exposed to 4% CS for 1 hour/day for 56 consecutive

days. 3x106 of BM-MSCs or iPSC-MSCs were intravenously administrated at day 29

and 43. A control group exposed to sham air (SA) was included.

Despite numerous reports of MSCs‟ effectiveness in diverse disease models, the

mechanism of MSCs‟ action remains unresolved (270). A major breakthrough was the

report of mitochondrial transfer from BM-MSCs to pulmonary alveoli which

protected mice against acute lung injury (118). In this study I hypothesized that a

similar phenomenon also existed in the CS-exposed model. The results demonstrated

mitochondrial transfer in vivo from BM-MSCs or iPSC-MSCs to the local cells of

CS-exposed lung tissue. The in vivo findings were followed by in vitro studies on

such activity in a co-culture of MSCs and bronchial epithelial cells (BEAS-2B)

treated with CSM. The mitochondrial transfer was determined through fluorescent

microscope and flow cytometry, and the effect on ATP depletion was also examined.

Although mitochondrial transfer may be a potent way to rescue damaged cells,

such action requires direct cell contact and is therefore restricted to small regions

around the MSCs. On the other hand, MSCs have been reported to induce paracrine

effects via release of various immunomodulators, such as TGF-β1, hepatic growth

Page 103: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

84

factor, prostaglandin E2, interleukin (IL)-10 and nitric oxide (2, 235, 270). The

paracrine effect can be effective in a large radius from MSCs as it does not rely on

direct cell-cell contact. Therefore I further investigated the effects of the paracrine

factors released from MSCs in an in vitro model using bronchial epithelial cells. In

particular, the role of stem cell factor (SCF) secreted from MSCs, a growth factor that

can mediate cell survival, migration, and proliferation was examined (146). BEAS-2B

cells were treated with conditioned medium (CdM) from BM-MSCs or iPSC-MSCs to

test a general paracrine effect on apoptosis and proliferation. SCF was then depleted

from CdM for a more specific investigation on the role of SCF in the CdM.

The aims of the study include:

1. To examine the effect of iPSC-MSCs on CS-induced lung damage in a chronic

CS-exposed rat model compared with BM-MSCs.

2. To determine whether there is mitochondrial transfer from iPSC-MSCs to

CS-damaged airway epithelial cells both in vivo and in vitro.

3. To investigate the involvement of paracrine regulation on the protective effects of

iPSC-MSCs on CSM-induced apoptosis in bronchial epithelial cells by using

conditioned medium (CdM), and to specifically determine the role of a potential

functional molecule, namely stem cell factor (SCF).

Page 104: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

85

3.2. Effects of iPSC-MSCs on CS-induced lung damage in

rats.

3.2.1. iPSC-MSCs attenuated CS-induced airspace enlargement, fibrosis and

inflammation in rat lung

Emphysema is one of the major characteristics of COPD. In this study, airspace

enlargement in the CS-exposed rat lung was assessed by histological examination

after haematoxylin and eosin staining. CS exposure caused obvious alveolar

destruction (Figure 3.1A), leading to a much longer mean linear intercept (Lm) (60 ±

4.6 μm) compared to the SA group (31.0±2.3 μm, p<0.001) (Figure 3.1B). The

airspace enlargement was attenuated by the BM-MSCs treatment (Figure 3.1A), with

a significantly reduced Lm compared with the CS group (48.0±4.0 μm, p<0.01)

(Figure 3.1B). Treatment with iPSC-MSCs also attenuated alveolar wall destruction

(Figure 3.1A), leading to significant reduction in Lm compared to the CS group

(40.0±2 μm, p<0.05). Moreover, the treatment with iPSC-MSCs was more effective

than BM-MSCs, as indicated by a shorter Lm value (p < 0.05) (Figure 3.1B).

CS also induced airway fibrosis compared to the SA group, as shown by the

Masson's Trichrome staining (Figure 3.2) (5±0.13% vs. 0.98±0.09%, p < 0.05).

Fibrosis was reduced by both BM-MSCs (3.85±0.17%, p < 0.05) and iPSC-MSCs

treatment (2.37±0.12%, p < 0.05), and iPSC-MSCs demonstrated superior effect

compared to BM-MSCs (p < 0.05) (Figure 3.2).

The inflammation induced by CS in lung tissues was illustrated by

immunohistochemistry staining against CD68, a transmembrane glycoprotein highly

Page 105: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

86

expressed by monocytes and tissue macrophages (26) (Figure 3.3A). CS exposure

significantly elevated the number of CD68-positive cells compared to SA group

(21.3±1.9 vs. 2.0±0.41%; p<0.01) (Figure 3.3B). Both treatments of iPSC-MSCs and

BM-MSCs significantly reduced the number of CD68-positive cells compared to the

CS group (4.3±0.8% for iPSC-MSCs and 12.3±1.3% for BM-MSCs respectively;

p<0.01) with a superior effect seen iPSC-MSCs (p<0.01) (Figure 3.3B).

Page 106: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

87

Figure 3.1 Human iPSC-MSCs and BM-MSCs alleviated CS-induced airspace

enlargement in rats. (A) Histological assessment of lung sections stained with

haematoxylin and eosin at 10x magnification. Scale bar = 100 μm. (B) Morphometric

analysis of mean linear intercept (Lm) (n=8). *p<0.05 compared with SA group;

#p<0.05 compared with CS group;

†p<0.05 compared with BM-MSCs group.

Page 107: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

88

Figure 3.2 BM-MSCs and iPSC-MSCs attenuated CS-induced lung fibrosis in

rats. (A) Masson's trichrome staining of lung sections. Blue staining indicates site of

fibrosis. 40x magnification. Scale bar = 50 μm. (B) Percentage of bronchioles with

fibrosis (n=8). *p<0.05 compared to SA group; #p<0.05 compared to CS group;

†p<0.05 compared to BM-MSCs group.

Page 108: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

89

Figure 3.3 BM-MSCs and iPSC-MSCs ameliorated CS-induced inflammation in

rat lungs. (A) Illustration of monocytes/macrophages as CD68 positive cells with

brown staining. (B) Number of CD68 positive cells in lung sections (n=6-8).

**p<0.01 compared to SA group; ##

p<0.01 compared to CS group; ††

p<0.01 compared

to BM-MSCs group.

Page 109: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

90

3.2.2. iPSC-MSCs attenuated CS-induced apoptosis/proliferation imbalance in

rat lung epithelium.

To illustrate apoptosis in the epithelium, lung sections were co-stained with the

TUNEL assay which labels apoptotic cells, and Clara cell 10-kDa protein (CC-10)

which is a marker of epithelium (Figure 3.4A). CS exposure induced increased

apoptosis in lung epithelium (5.2±0.7% vs. 0.4±0.2%; p<0.01), which was attenuated

by both BM-MSCs (3.4±0.5%; p<0.01 in comparison to CS group) and iPSC-MSCs

(2.0±0.6%; p<0.01 in comparison to CS group) (Figure 3.4B). The treatment of

iPSC-MSCs was more effective than BM-MSCs group (p<0.01) (Figure 3.4B).

CS exposure also led to reduction of proliferation in the epithelium, which was

shown by co-staining against CC-10 and Ki-67, a marker of cell proliferation (221)

(Figure 3.4A). CS exposure significantly reduced the number of proliferating cells in

the epithelium (5.5±0.5% vs. 25.8±2.8%; p<0.01), which was improved by the

treatment with either BM-MSCs (11.2±0.8%; p<0.01 in comparison to CS group) or

iPSC-MSCs (15.2±2.3%; p<0.01 in comparison to CS group) (Figure 3.4B). The

iPSC-MSCs group demonstrated higher proliferation activity than the BM-MSCs

group (p<0.01) (Figure 3.4B).

Page 110: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

91

3.2.3. Mitochondrial transfer from MSCs to adjacent lung cells

The epithelium in the rat lung was stained with CC-10, and mitochondria in both

MSCs and host cells were labeled with an antibody against human/rat Complex I.

Meanwhile, human mitochondria were detected by staining using a human specific

Cox-4 antibody (Figure 3.5A&B). For both BM-MSCs and iPSC-MSCs treatments,

human mitochondria were detected inside some of the CC-10-expressing cells

(Figure 3.5A&B), indicating events of mitochondrial adoption by the bronchial

epithelium from the foreign human MSCs. With the same number of cells injected,

rats treated with iPSC-MSCs demonstrated a higher number of cells receiving

MSC-derived mitochondria than those treated with BM-MSCs (number/40x field:

8.4 ± 1.6 vs. 4.6 ± 0.9, p< 0.05; Figure 3.5C).

Page 111: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

92

Figure 3.4 BM-MSCs and iPSC-MSCs attenuated CS-induced increase in

apoptosis and reduction of proliferation in rat lung epithelium. (A) Apoptotic

cells in airway epithelium. Red (CC-10): airway epithelium; green (TUNEL):

apoptotic cells; blue (DAPI): nuclei. (B) Proliferative cells in airway epithelium.

Green (CC-10): airway epithelium, Red (Ki-67): proliferative cells; Blue (DAPI):

nuclei. (C) Number of apoptotic cells in airway epithelium (n=6-8). (D) Number of

proliferating cells in airway epithelium. **p<0.01 compared to SA; ##

p<0.01

compared to CS; ††

p<0.01 compared to BM-MSCs.

Page 112: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

93

Figure 3.5 Mitochondria transfer from MSCs to lung epithelium in rats. Lung

sections treated with (A) BM-MSCs and (B) iPSC-MSCs were co-stained against

CC-10 (red) marking lung epithelium, human/rat Complex I (green) labeling

human/rat mitochondria and human Cox-4 (violet) detecting human mitochondria

specifically. Co-localization of three signals indicates receiving of human

mitochondria by lung epithelium of rats. Scale bar = 50 μm. (C) Number of cells

receiving foreign mitochondria in 40x fields. *p<0.05 compared between iPSC-MSCs

and BM-MSCs groups.

Page 113: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

94

3.3. Mitochondrial Transfer from MSCs to BEAS-2B cells in

vitro

3.3.1. Mitochondrial transfer from MSCs to BEAS-2B cells

Mitochondrial transfer was further investigated in an in vitro co-culture model in

the context of CSM treatment. According to a previous report, treatment of 2% CSM

for 24 hours induces a significant inflammatory response and oxidative stress without

loss of viability in human bronchial epithelial BEAS-2B cells (143). This condition

was adopted in the co-culture study. BEAS-2B cells were pre-labeled with CellTrace

Violet while mitochondria of BM-MSCs or iPSC-MSCs were pre-labeled with

MitoTracker Green. BEAS-2B cells were then co-cultured with either BM-MSCs or

iPSC-MSCs in a 1:1 ratio and treated with 2% CSM for 24 hours. The

MitoTracker-labeled mitochondria which originated from the MSCs were observed in

neighboring BEAS-2B cells, indicating that mitochondria transferred from MSCs to

BEAS-2B cells within 24 hours (Figure 3.6A&B). Moreover, formation of tunneling

nanotube (TNT)-like structures between the MSCs and BEAS-2B cells was identified

after 24 hours (Figure 3.6A&B). The formation of TNTs was reported to be blocked

by Cytochalasin B at nanomolar concentrations without affecting endocytosis and

phagocytosis (40). In this study, when 300 nM of cytochalasin B was added to the

co-culture together with the CSM, the formation of TNTs was highly inhibited and

few MSCs-derived mitochondria were observed in BEAS-2B cells (Figure 3.6 C&D),

indicating that that mitochondrial transfer may be mediated through TNTs.

Page 114: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

95

Figure 3.6 Mitochondrial transfer from BM-MSCs and iPSC-MSCs to BEAS-2B

cells in co-culture. (A) BM-MSCs or (B) iPSC-MSCs pre-stained with MitoTracker

Green and BEAS-2B cells pre-labeled by CellTrace Violet were co-cultured in a 1:1

ratio and treated with 2% CSM for 24 hours. The observation of MitoTracker Green

in the BEAS-2B cells (Blue) indicated the adoption of MSCs-derived mitochondria by

BEAS-2B cells. Connection between the cells through TNT-like structures was

observed. Treatment of Cytochalasin B suppressed the formation of TNT-like

structures and mitochondrial transfer in both (C) BM-MSCs and (D) iPSC-MSCs

group. Scale bar = 20 μm. B2B: BEAS-2B cells; DIC: differential interference

contrast. A representative image from three independent experiments is shown.

Page 115: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

96

3.3.2. CSM promoted mitochondrial transfer from MSCs to BEAS-2B cells

In addition to fluorescent microscopy, mitochondrial transfer in the co-culture

was further evidenced by flow cytometry at different time-points. The percentage of

CellTrace/MitoTracker double positive cells in the whole CellTrace positive

population was defined as the transfer rate (Figure 3.7A). There were three findings

from this experiment. First, the transfer rate gradually increased along the co-culture

period in both BM-MSCs and iPSC-MSCs groups. Second, after 24 hours, the

iPSC-MSCs group demonstrated a higher transfer rate compared with the BM-MSCs

group (38.8±2% vs. 27.8±1.6%, p<0.05), suggesting a higher potential for

mitochondrial transfer between iPSC-MSCs and BEAS-2B cells. Lastly, at the 24

hour time-point, CSM treatment significantly increased the transfer rate compared to

the untreated group, both for the BM-MSCs co-culture (27.8±1.6% vs. 14.4±1%, p<

0.05) and iPSC-MSCs co-culture (38.8±2% vs. 18±1.9%, p<0.05), suggesting that

mitochondrial transfer is promoted in the presence of CSM.

3.3.3. Co-culture with iPSC-MSCs elevated ATP levels in BEAS-2B cells

Given the observation of mitochondrial transfer in the co-culture, the effect of

co-culture with BM-MSCs and iPSC-MSCs on intracellular ATP levels in BEAS-2B

cells was investigated. BEAS-2B cells were identified from the co-culture followed

by determination of intracellular ATP levels. The 2% CSM treatment for 24 hours

significantly reduced the intracellular ATP level (percentage of control: 76.5 ±3.5% vs.

100 ±10.1%, p<0.05) (Figure 3.8). The drop of ATP level was restored by co-culture

Page 116: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

97

with iPSC-MSCs (percentage of control: 97.8 ±9.1% vs. 76.5 ±3.5% for iPSC-MSCs

and CSM group respectively, p<0.05), but not the BM-MSCs (percentage of control:

80.7 ± 7.1%) (Figure 3.8).

Figure 3.7 CSM promoted mitochondrial transfer. (A) Representative flow

cytometry outcome of single culture of MSCs (left), BEAS-2B cells (middle) and

co-culture (right). BM-MSCs or iPSC-MSCs pre-labeled with MitoTracker Green and

BEAS-2B cells pre-labeled with CellTrace Violet were co-cultured at a 1:1 ratio in

the presence of 2% CSM. (B) Effects of time, CSM and cell type on mitochondrial

transfer rate (n=3).Transfer rate was defined as the percentage of BEAS-2B cells

containing MSCs-derived mitochondria in the total BEAS-2B population. W/O stands

for CSM untreated group while other groups were treated with 2% CSM for 24 hours.

*p<0.05 represents statistical difference as indicated.

Page 117: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

98

Figure 3.8 iPSC-MSCs restored intracellular ATP levels of BEAS-2B cells.

BEAS-2B cells were co-cultured with MSCs with 2% CSM for 24 hours before sorted

out through flow cytometry and determination of intracellular ATP levels (n=3).

*p<0.05 compared to control group; #p<0.05 compared to CSM group;

†p<0.05

compared to BM-MSCs group.

3.4. iPSC-MSCs Attenuated CSM-induced Apoptosis and

Proliferation Imbalance in BEAS-2B Cells by Paracrine

Effect

3.4.1. iPSC-MSCs-CdM attenuates CSM-induced apoptosis in BEAS-2B cells

Conditioned medium (CdM) from BM-MSCs and iPSC-MSCs contains paracrine

factors released from the MSCs accumulated over 24 hours. Thus the effects of

MSCs-CdM on BEAS-2B cells were investigated in order to study the paracrine effect.

BEAS-2B cells were treated with 2% CSM and CdM for 24 hr. The release of

cytochrome c from mitochondria and its translocation to nucleus (Figure 3.9A) is a

major event in triggering the onset of apoptosis (184, 211). CSM elevated the

percentage of cells with cytochrome c translocation compared to the untreated group

Page 118: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

99

(73.4±6.8%vs. 1.88±1%; p<0.01), which was attenuated by both BM-MSCs-CdM

(41.2±6.3%; p<0.01 in comparison to CSM group) and iPSC-MSCs-CdM (16.6±5%;

p<0.01 in comparison to CSM group) (Figure 3.9D). iPSC-MSCs-CdM exhibited a

superior effect compared with to BM-MSCs-CdM (p<0.01) (Fig. 3.9D). In addition,

cellular apoptosis was identified by TUNEL staining (Figure 3.9B). CSM elevated the

percentage of apoptotic cells (16.2±2% vs. 1.4±0.4%; p<0.01), which was attenuated

by both BM-MSCs-CdM (9.3±0.7%; p<0.01 in comparison to CSM group) and

iPSC-MSCs-CdM (5.5±0.5%; p<0.01 in comparison to CSM group) (Figure 3.9E).

There were fewer apoptotic cells in the iPSC-MSCs-CdM group compared to

BM-MSCs-CdM group (p<0.01), suggesting a better anti-apoptotic activity of

iPSC-MSCs-CdM.

While apoptosis was enhanced, the percentage of proliferating cells was reduced

by the 2% CSM (3.4±0.5% vs. 19.2±0.8%; p<0.01) as determined by staining against

Ki-67 (Figure 3.9C&F). This reduction was improved by both BM-MSCs-CdM

(8.2±0.6%; p<0.01 in comparison to CSM group) and iPSC-MSCs-CdM (13.4±0.5%;

p<0.01 in comparison to CSM group) (Figure 3.9F), in which iPSC-MSCs-CdM

demonstrated a more potent effect (p<0.1) (Figure 3.9F), indicating a higher ability to

promote cell proliferation.

Page 119: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

100

Figure 3.9 CdM from BM-MSCs and iPSC-MSCs ameliorated CSM-induced

apoptosis/proliferation imbalance in BEAS-2B cells. BEAS-2B cells were treated

with 2% CSM and BM-MSCs-CdM or iPSC-MSCs-CdM for 24hr. (A) Cytochrome c

translocation in BEAS-2B cells. Green: cytochrome c; blue: DAPI. Cells with

cytochrome c translocation to nuclei are marked by red arrows. (B) Apoptotic

BEAS-2B cells as stained by TUNEL. Green: TUNEL; blue: DAPI. (C) Proliferative

BEAS-2B cells as marked by Ki-67. Red: Ki-67; blue: DAPI. (D) Percentage of

BEAS-2B cells with cytochrome c translocation (n=3). (E) Percentage of apoptotic

BEAS-2B cells (n=3). (F) Percentage of proliferative BEAS-2B cells. **p<0.01

compared to control group; ##

p<0.01 compared to CSM group; ††

p<0.01 compared to

BM-MSCs-CdM group.

Page 120: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

101

3.4.2. Effects of iPSC-MSCs-CdM on apoptosis and proliferation of BEAS-2B

cells were SCF-dependent

Given the superior effects of iPSC-MSCs-CdM over BM-MSCs-CdM regarding

apoptosis and proliferation, the active components responsible for such effects were

further investigated. In particular, the role of SCF was examined in my model.

First of all, the levels of SCF in CdM were measured, showing that

iPSC-MSCs-CdM contains a much higher level of SCF than BM-MSCs-CdM

(518±57 vs. 113±13 pg/mg protein; p<0.01) (Figure 3.10A). The expression of SCF

receptor, C-kit, by BEAS-2B cells was confirmed by both immunohistochemistry

staining and Western blotting (Figure 3.10B) which showed that the level of C-kit was

not changed by CSM treatment (Figure 3.10B).

To investigate the role of SCF, SCF-deprived iPSC-MSCs-CdM was generated by

incubation with immobilized anti-SCF antibody. Effects of iPSC-MSCs-CdM,

SCF-deprived iPSC-MSCs-CdM and recombinant SCF were evaluated on BEAS-2B

cells treated with 2% CSM for 24 hours. All three treatments attenuated CSM-induced

morphological change (Figure 3.11A), cytochrome c translocation (22.0±2.1%,

42.4±2.5% and 41.8±1.9% respectively, p<0.01 in comparison to CSM group,

74.8±2.9%) (Figure 3.11BE), apoptosis (5.6±0.5%, 10.4±0.5% and 10.2±1.0%

respectively, p<0.01 in comparison to CSM group, 20±1.7%) (Figure 3.11CF) and

reduction of proliferation (15.4±0.5%, 11.0±0.5% and 11.0±0.9% respectively, p<0.01

in comparison to the CSM group, 5.8±0.4%) (Figure 3.11DG). Compared to the

normal iPSC-MSCs-CdM treatment, depletion of SCF in the CdM resulted in

Page 121: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

102

significantly weakened activity, as shown by more cytochrome c translocation

(p<0.01), more apoptosis (p<0.01) and less proliferation (p<0.01). This finding, in

combination with the result that recombinant SCF was able to induce considerable

effects by itself, indicated that SCF contributed to the activity of iPSC-MSCs-CdM.

Similarly, the recombinant SCF were less effective than iPSC-MSCs-CdM regarding

cytochrome c translocation (p<0.01), apoptosis (p<0.01) and proliferation (p<0.01).

Given that the deprived iPSC-MSCs-CdM still retained some capacity to restore all

three parameters, this finding indicated that in addition to SCF, there were other

components promoting similar effects. In summary, the data suggested that the action

of iPSC-MSCs-CdM to alleviate CSM-induced apoptosis/ proliferation imbalance was

partly through SCF.

Page 122: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

103

Figure 3.10 Secretion of SCF by BM-MSCs and iPSC-MSCs and expression of

C-kit by BEAS-2B cells. (A) SCF levels in CdM from BM-MSCs or iPSC-MSCs as

measured by ELISA (n=3). (B) Expression of C-kit in BEAS-2B cells. C-kit

expression was identified by immunohistostaining (i) with or (ii) without 2% CSM

treatment for 24 hours, and by (iii) Western blotting. Red: C-kit; blue: DAPI.

**p<0.01 compared to BM-MSCs-CdM.

A

Page 123: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

104

Figure 3.11 The anti-apoptotic and pro-proliferation activity of

iPSC-MSCs-CdM was partly through SCF. Immobilized anti-SCF antibody was

used to scavenge SCF from iPSC-MSCs-CdM. BEAS-2B cells were treated with 2%

CSM and iPSC-MSCs-CdM for 24 hours before fixation and TUNEL staining or

immunostaining against cytochrome c and Ki-67 (n=3). (A) The morphological

change of the BEAS-2B cells. DIC: differential interference contrast. (B) Cytochrome

c translocation in BEAS-2B cells. Green: cytochrome c; blue: DAPI. Cells with

cytochrome c translocation to nuclei are marked by red arrows. (C) Apoptotic

BEAS-2B cells as stained by TUNEL. Green: TUNEL; blue: DAPI. (D) Proliferative

BEAS-2B cells as marked by Ki-67. Red: Ki-67; blue: DAPI. (E) The percentage of

BEAS-2B cells with cytochrome c translocation to nuclei (n=3). (F) The apoptotic

rate of BEAS-2B cells (n=3). (G) Percentage of proliferative BEAS-2B cells (n=3).

**p<0.01 compared to control group; ##

p<0.01 compared to CSM group; ††

p<0.01

compared to normal iPSC-MSCs-CdM group.

Page 124: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

105

3.5. Discussion

This study elucidated that iPSC-MSCs are more effective than BM-MSCs in

attenuating CS-induced alterations in rat lungs. The higher efficacy may be explained

by a higher activity of mitochondrial transfer, which was evidenced in vivo and

further confirmed by in vitro co-culture study. Meanwhile, the higher efficacy may

also be attributed to more potent paracrine effects, partly through SCF.

MSCs isolated from BM (94, 112), amniotic fluid (151) and adipose tissue (222)

have been reported to attenuate CS-induced emphysema in murine models. These

MSCs are isolated directly from adult tissue, hence relevant patient tissues such as

bone marrow and adipose tissue need to be obtained to collect them. The technique to

differentiate iPSCs into MSCs provides a different source of MSCs, which has several

advantages over classic MSCs. Firstly, large quantities of iPSC-MSCs can be

generated from a small number of somatic cells which can be of a number cell types

such as fibroblasts. In other words, the supply of iPSC-MSCs is unlimited and

personalised. Secondly, iPSC-MSCs have a strong capacity of proliferation and

differentiation, with differentiation potential maintained for over 120 population

doublings (152), while normal MSCs may reach senescence after 20 doublings (69).

Thirdly, impaired survival and differentiation ability may be associated with MSCs

isolated from aged subject or patients with ageing-related disorders (87, 100, 267,

294). However, iPSC-MSCs may hold hope to overcome these issues because when

deriving iPSC-MSCs, somatic cells are firstly reprogrammed to an embryonic stem

cell-like state, which may provide the cells with a rejuvenated phenotype. Therefore

Page 125: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

106

iPSC-MSCs are valuable candidates for the development of cell therapy against

COPD whose prevalence, morbidity, and mortality are all highly associated with age

(38, 126, 137). Regarding airway disease models, iPSC-MSCs have been reported to

attenuate allergic airway inflammation in mice (244).

The first part of this study evaluated a general effect of iPSC-MSCs on

CS-induced lung alterations in rats. The parameters included emphysema, fibrosis,

inflammation, apoptosis and proliferation. My results show that, iPSC-MSCs

demonstrated a better effect regarding each of the parameters in comparison to those

elicited in response to treatment with BM-MSCs. In fact, as far as iPSC-MSCs can

produce comparable effects to BM-MSCs, due to iPSC-MSCs‟ advantages regarding

the resource and ageing issue, they would at the outset be a better candidate than the

BM-MSCs. The superior efficacy over BM-MSCs further defined iPSC-MSCs as a

promising candidate. Moreover, for the BM-MSCs, a placebo-controlled, randomized

clinical trial in COPD subjects has already been reported (271). The trial observed no

toxicity caused by the treatment. However, there were also no effects regarding both

pulmonary function and indicators of quality-of-life. It did reveal some potential

anti-inflammatory activity though, as the treatment decreased C-reactive protein (CRP)

levels at an early stage. There were some limitations of the study including the

determination of doses, but it still pointed out to the possibility that BM-MSCs might

not work in the clinical setting, despite the clear efficacy demonstrated in animal

models. From our study, iPSC-MSCs are much more effective than BM-MSCs in the

rat model. Therefore the possible failure of BM-MSCs in the clinical context will not

Page 126: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

107

necessarily indicate a failure of iPSC-MSCs. Instead, it will call for improved MSCs

therapies, which makes study of iPSC-MSCs even more worthwhile and important.

However, on the other hand, the clinical study also revealed the difference between

animal models and diseased subjects, thus the effects of iPSC-MSCs in the clinical

context are not predictable.

Despite numerous reports of MSCs‟ effectiveness in diverse disease models, the

mechanism of MSCs‟ action remains unresolved (270). A major breakthrough is the

report of that in vivo mitochondrial transfer from BM-MSCs into pulmonary alveoli

protect the lung against lipopolysaccharide-induced acute lung injury (118),

illustrating a new mechanism of MSCs‟ action. However, mitochondrial transfer in the

context of CS-induced lung damage was unknown. This study demonstrated that

BM-MSCs and iPSC-MSCs could effectively transfer mitochondria to CS-damaged

lung epithelial cells, similar to sending in a “Trojan horse”. This is one of the very

early demonstrations of such activity following the first report in the acute lung injury

model mentioned above. Although the signaling that drives the MSCs to transfer

mitochondria for rescue of lung epithelial cell was unknown, in vitro formation of

TNTs was observed between MSCs and bronchial epithelial cells. The interruption of

TNT formation by cytochalasin B almost completely blocked mitochondrial transfer,

suggesting that mitochondria might be transferred via TNTs in vitro. Such

TNT-mediated mitochondrial transfer between MSCs and damaged lung epithelial

cells might be an important mechanism through which the exhausted epithelial

cellular bioenergetics can be restored and damaged lung epithelium be rejuvenated.

Page 127: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

108

Bronchial epithelial cells were chosen for the in vitro study because they act as

the first barrier against CS exposure. The inflammation and oxidative stress in COPD

was initiated by the bronchial epithelial cells‟ response to CS-exposure such as

expression of cell injury–associated endogenous molecules and secretion of

inflammatory mediators including TNF-α and IL-8 (190). Mitochondria are mostly

known as the powerhouse of the cell. In patients with COPD, decreased energy

metabolism is associated with declining lung function (285). Given the observation of

mitochondrial transfer from MSCs to bronchial epithelial cells, the effect of MSCs on

the cellular bioenergetics was studied by measuring the ATP level. The CSM

treatment reduced ATP levels of bronchial epithelial cells, which was alleviated by

co-culture with iPSC-MSCs, indicating improvement in bioenergetics. Such activity

was coupled with CSM-induced enhancement of mitochondrial transfer from

iPSC-MSCs to bronchial epithelial cells, which may be the source of the restored

bioenergetics. Moreover, iPSC-MSCs have demonstrated a more active role in

mitochondrial transfer in both in vivo and in vitro experiments, which may contribute

to the higher efficacy of iPSC-MSCs in attenuating CS-induced lung alterations.

The limitation of mitochondrial transfer is its working radius. Relying on direct

contact between iPSC-MSCs and pulmonary cells, the primary effects of

mitochondrial transfer might be only effective within a limited distance from the

iPSC-MSCs. Although the retention of the iPSC-MSCs were identified even two

weeks after injection based on our previous study (150), it is unlikely that all the cells

in the lungs would have the opportunity to directly adopt mitochondria from them.

Page 128: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

109

The full activity of the MSCs may be better explained with a complementary

mechanism that overcomes the restriction imposed by the distance between cells,

namely, the paracrine effect. The paracrine effect of MSCs has been documented in

various studies, with different active factors identified, such as vascular endothelial

growth factor, TGF-β1, hepatic growth factor, bFGFr and platelet derived growth

factor (2, 235, 270). In this study the potential of paracrine effects was demonstrated

by treating bronchial epithelial cells with CdM in vitro. The treatment of CSM led to

increased apoptosis and reduced proliferation in bronchial epithelial cells in vitro,

resulting in an imbalance between apoptosis and proliferation. CdM restored the

imbalance by reducing the apoptotic rate and promoting proliferation. The

iPSC-MSCs-CdM was superior than BM-MSC-CdM regarding all the observed

effects. From the in vivo data discussed earlier, iPSC-MSCs have shown superior

effects than BM-MSCs in attenuating apoptosis and promoting proliferation in rat

lung epithelium. The in vivo and in vitro findings together suggested a better

anti-apoptotic and pro-proliferation capacity of iPSC-MSCs through paracrine action.

There are two reasons for studying the effects on apoptosis. Firstly, most studies

on paracrine effects have focused on the immunoregulatory role (e.g. suppression of

inflammation) of MSCs, which is therefore well demonstrated by previous reports (2,

235, 270). In contrast, the modulation of apoptosis by the CdM is less well

investigated. Secondly, there is mounting data suggesting that the gradual alveolar

destruction in COPD pathogenesis may be partly driven by imbalance between

apoptosis and proliferation (67), although classically it is explained by the imbalance

Page 129: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

110

of protease and anti-protease activity (68, 76, 77, 189). Apoptosis and proliferation are

well equilibrated in healthy tissue. The induction of apoptosis by CS has been

reported in various cell types of lung, such as endothelial cells, alveolar epithelial

cells, fibroblasts and immune cells (16, 103, 116, 127, 166, 223). Excess apoptosis in

alveolar cells without compensation from proliferation led to the gradual destruction

of alveolar walls in lung (67). Apoptosis of alveolar wall and endothelial cells was

reported to induce emphysema even without infiltration of inflammatory cells in an

animal model (67). Therefore the anti-apoptotic and pro-proliferation potential might

identify a key capacity of iPSC-MSCs as a promising candidate for cell therapy

development against COPD.

Given the effects of CdM on apoptosis and inflammation, the active component

of CdM was the next research question. SCF, a dimeric protein, triggers

homodimerization and autophosphorylation of its receptor c-Kit upon binding, which

further activates diverse pathways involving cell survival, migration, and proliferation

(146). In addition to its ability to induce proliferation, SCF/c-Kit may also affect

apoptosis through down-stream activation of the serine/threonine kinase Akt (35). Akt

subsequently phosphorylates and inhibits pro-apoptotic protein Bad which regulates

the cytochrome c release from the mitochondria through Bcl-2/Bax signalling (35,

265, 283). Despite of a lack of understanding regarding the mechanism, the SCF/c-Kit

signalling has been proved essential to maintain normal alveolar structure in mouse

lung. Deficiency in SCF/c-Kit signalling was reported to cause spontaneous alveolar

destruction, increased ex vivo lung compliance and enlarged residual volume in mice

Page 130: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

111

(157). Given its important role in proliferation/apoptosis regulation and maintaining

alveolar structure, this study targeted SCF as a potential active component in CdM.

SCF alone was able to attenuate CSM-induced cytochrome c translocation, apoptosis

and reduction in proliferation, indicating its anti-apoptotic and pro-proliferative

activity. Meanwhile, SCF depletion led to a weakened ability of iPSC-MSCs-CdM to

attenuate CSM-induced cytochrome c translocation, apoptosis and reduction in

proliferation, indicating that SCF played an important role in the action of

iPSC-MSCs-CdM. However, the data also showed that SCF alone was less effective

than iPSC-MSCs-CdM, and that SCF depletion only weakened but did not eliminate

the efficacy of iPSC-MSCs-CdM, suggesting that the action of iPSC-MSC-CdM was

only partly dependent on SCF. Moreover, iPSC-MSCs secreted much higher levels of

SCF than BM-MSCs, which may explain its superior anti-apoptotic and

pro-proliferation effects through paracrine action.

From the above discussion, this study has demonstrated two possible mechanisms

through which the iPSC-MSCs protect lung epithelial cells against CS-induced

damage. However, the relative importance of each mechanism was not evaluated in

the study. As mitochondrial transfer was limited by distance, the total therapeutic

effects of the treatment strongly rely on the number of MSCs retained in the lung

tissue. However, the ability of the MSCs to be retained in the lung varies between

different reports. From our previous study in the CS-exposed model, clusters of MSCs

were identified even two weeks after injection. Similarly, adipose stem cells have

been reported to be retained in lung tissue in CS-exposed model after 7 days or 21

Page 131: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

112

days (222). However, in another study using BM cells in CS-exposed female rats,

fluorescent in situ hybridization (FISH) only found 2-13 male BM cells per entire

lung from 1 day to 1 month (112). The factors that determine this retention ability

remain unknown, and such factors would also be key to give mitochondrial transfer

higher importance in therapeutic action of MSCs. The paracrine effect, on the other

hand, may be more effective when the retained MSCs are less abundant, as the effects

might be initiated by low level secretion of relevant paracrine factors but amplified by

signalling pathways in the responding cells. The MSCs in the circulation might also

possess systemic effects through release of molecules into the circulation. However,

regarding acute responses, the direct delivery of mitochondria seemed to be a more

straightforward and immediate rescue, which may be why the mitochondrial transfer

has been identified as such an important mechanism in the context of acute lung

injury models where bioenergetics is rapidly impaired.

There are nonetheless several limitations of this study, and more future research

questions can be raised from current findings.

First of all, for the general advantages discussed earlier, although the iPSC-MSCs

may be able to overcome the age-related impairment of normal MSCs, its phenotype

when derived from patients with genetic mutations/inherited diseases remains unclear.

About 1-3% of COPD patients are associated with a genetic alteration which leads to

deficiency of serine protease α-1 antitrypsin (241). There are also other genes

suspected to be associated, such as TGF-β1 (47).

The mitochondrial transfer was proposed as an important mechanism, but the

Page 132: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

113

TNT may also facilitate the transfer of other organelles, proteins and compounds. For

example, whether the ATP was directly transferred from MSCs to bronchial epithelial

cells was not determined. In addition, other mechanisms for the transfer of material

between cells may also play important roles, such as gap junction and microvesicles.

Lastly, despite the identification of mitochondrial transfer via TNTs in vitro, the route

of mitochondrial transfer in vivo was not demonstrated and requires further

investigation.

Regarding the paracrine effects, the CdM was collected from normal culture of

BM-MSCs or iPSC-MSCs without any interactions with BEAS-2B cells. However, it

is possible that CSM can induce BEAS-2B cells to release cytokines, which may act

on MSCs and modulate secretory functions of MSCs. A trans-well co-culture with

both cells in the same system may overcome this limitation in vitro.

SCF has been identified to contribute to the anti-apoptosis and pro-proliferation

action of iPSC-MSCs-CdM, but the depletion of SCF did not eliminate the effects.

Therefore more active molecules remain to be determined.

In summary, this study demonstrated that iPSC-MSCs attenuate CS-induced lung

damage and observed evidence of mitochondrial transfer from iPSC-MSCs to lung

epithelial cells. The higher activity to transfer mitochondria to CS-damaged bronchial

epithelial cells in vitro and in vivo, in line with the higher anti-apoptosis and

pro-proliferation capacity by paracrine effect partly due to more abundant SCF

secretion, together explained the superior effects of iPSC-MSCs over BM-MSCs. This

Page 133: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

114

study defines iPSC-MSCs as a promising candidate for the development of cellular

therapy against COPD.

Page 134: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

115

Chapter 4. iPSC-MSCs attenuated

CSM-induced Mitochondrial Dysfunction in

Human Airway Smooth Muscle Cells

Page 135: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

116

4.1. Introduction

There is an increasing interest in the role of mitochondria in the pathogenesis of

airway diseases. Mitochondria, most well-known as the powerhouse of the cells, also

play crucial roles in other cellular functions in addition to bioenergetic regulation,

such as calcium metabolism, reactive oxygen species (ROS) modulation and onset of

apoptosis (45, 90, 95, 212). As the site of oxidative phosphorylation, mitochondria are

an important intracellular source of ROS. Defective oxidative phosphorylation may

lead to overwhelming generation of ROS in mitochondria. Increased external and

internal ROS production, without sufficient protection from anti-oxidants, leads to

cellular oxidative stress, which is a key player in the pathogenesis of COPD (57). The

effectiveness of MSCs has been demonstrated in multiple airway disease models

including emphysema and allergic airway inflammation, but the underlining

mechanisms remain unresolved (270). Given that mitochondria are a potential

therapeutic target for airway diseases, investigating the effects of MSCs on oxidative

stress-induced mitochondrial damage in airway cells may be of value. In this study I

hypothesized that interaction with iPSC-MSCs can attenuate cigarette smoke medium

(CSM)-induced mitochondrial damage in ASMCs.

Recently, mitochondrial dysfunction has been reported in airway smooth muscle

cells (ASMCs) isolated from patients with COPD (273), indicating that mitochondria

may be a promising therapeutic target for COPD. Airway smooth muscle plays an

important role in airway remodelling and inflammation (56). In addition to their

contractile properties, ASMCs are also involved in airway inflammation by releasing

Page 136: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

117

pro-inflammatory cytokines (10). The first part of this chapter aims to evaluate

changes in mitochondrial function in normal ASMCs in response to oxidative stress.

Cigarette smoke (CS), the primary cause of COPD, is a significant source of oxidants.

Cigarette smoke extract was reported to alter the morphology of mitochondria in vitro

in both airway epithelial cells (105) and ASMCs (10), leading to mitochondrial

fragmentation. Therefore CSM was used in this study to induce mitochondrial damage.

In addition, H2O2 was used as a direct source of oxidants. Mitochondrial ROS levels

and ΔΨm were examined to evaluate changes in mitochondrial function.

Mitochondria play a key role in the regulation of cell fate. The depolarization of

mitochondria is regarded as an early event triggering mitochondrial apoptosis (90).

Therefore, in the second part of this chapter I investigated the effects of co-culture

with iPSC-MSCs on CSM-induced mitochondrial dysfunction and apoptosis in

ASMCs.

Paracrine regulation by MSCs is a widely reported mechanism through which

MSCs can suppress inflammation (2, 235, 270) or attenuate apoptosis, as suggested in

the previous chapter. The third part of this chapter aims to determine whether the

effects of iPSC-MSCs on CSM-induced mitochondrial alterations were through

secretion of paracrine factors. To investigate this, the effects of

iPSC-MSCs-conditioned medium (iPSC-MSCs-CdM) on CSM-induced

mitochondrial alterations were firstly examined. The contents of the

iPSC-MSCs-CdM represented the paracrine factors secreted by iPSC-MSCs under

normal conditions. However, in a real interaction, ASMCs may affect the iPSC-MSCs

Page 137: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

118

and modulate the latter‟s synthetic and secretory functions. CSM may also affect the

paracrine effects of iPSC-MSCs as well as the interaction between iPSC-MSCs and

ASMCs. Therefore a study using a trans-well co-culture system was carried out in

which the two cell types are separated by a membrane that is permeable to the

paracrine factors but prevents direct cell-cell contact.

Another recently reported mechanism of the action of MSCs is through

mitochondrial transfer from MSCs to recipient cells, which was firstly described in

vivo in an acute lung injury rodent model (118). In the previous chapter, mitochondrial

transfer from iPSC-MSCs to airway epithelial cells was observed in CS-exposed rat

lungs and CSM-treated bronchial epithelial cells. However, the effect of

mitochondrial transfer on markers of mitochondrial damage was not studied. In the

final part of this chapter, I studied mitochondrial transfer between ASMCs and

iPSC-MSCs using fluorescent microscopy and flow cytometry.

The aims of this chapter were:

1. To evaluate oxidative stress-induced mitochondrial dysfunction in human primary

ASMCs

2. To examine the capacity of iPSC-MSCs to reduce CSM-induced mitochondrial

dysfunction and apoptosis in ASMCs

3. To evaluate the relative contributions of paracrine effects and cell-cell contact in

the protective effects of iPSC-MSC‟s on mitochondrial dysfunction and apoptosis

in ASMCs

4. To investigate mitochondrial transfer from iPSC-MSCs to ASMCs

Page 138: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

119

4.2. Oxidative Stress-induced Mitochondrial Dysfunction in

ASMCs and iPSC-MSCs

4.2.1. Oxidative stress reduced the viability of ASMCs

Normal ASMCs isolated from bronchi or tracheas of healthy transplant donor

lungs were treated with H2O2 (50, 100 and 200 μM) or CSM (10, 25, 50 and 75%) for

2, 4 and 24 hours. In the 4/20 hour group cells were stimulated for 4 hours and then

incubated with serum-free medium for 20 hours. Cell viability was measured by MTT

assay. H2O2 led to a concentration-dependent reduction in viability at each time point

(Maximum reduction at 200 μM, fold-change vs. controls: 2 hours, 0.70±0.05,

p<0.001; 4 hours, 0.62 ±0.02, p<0.001; 24 hours, 0.62 ±0.06, p<0.01; 4/24 hours, 0.59

±0.06, p<0.001) (Figure 4.1A). A similar effect was observed at all time points.

CSM also caused loss of viability in a concentration-dependent manner at 2 hours,

4 hours and 24 hours (Maximum effects at 75% CSM, fold-change vs. controls: 2

hours, 0.88±0.04, p<0.01; 4 hours, 0.81±0.07, p<0.01; 24 hours, 0.57±0.13, p<0.05)

(Figure 4.1B). However, no significant reduction in viability was observed in the 4/20

hours group (fold to control, 75% CSM: 0.93±0.15). Moreover, the 75% CSM

treatment led to a significantly lower viability at 24 hours compared with the viability

at 2 hours (p<0.05) or 4 hours (p<0.05), indicating that the effect of 75% CSM was

time-dependent.

Page 139: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

120

MTT assay is based on NAD(P)H-dependent reductive reactions catalyzed by

oxidoreductases in cells (30-32). Mitochondria are one of the major carriers of such

enzymes, thus the reduction of MTT readout may be a reflection of mitochondrial

dysfunction.

2hr 4hr 24hr 4/20hr0.0

0.5

1.0

1.5

*

*** ***

* *

*****

Control

50 M

100 M

200 M

[H2O2]

Cell Viability (n=5)

Cel

l via

bili

ty

(Fo

ld c

han

ge

to c

on

tro

l)

A

2hr 4hr 24hr 4/20hr0.0

0.5

1.0

1.5

** ***

*

Control

10%

25%

50%

75%

CSM%

#

#

#

Cell Viability (n=4)

Cel

l via

bili

ty

(Fo

ld c

han

ge

to c

on

tro

l)

B

Figure 4. 1 Effects of H2O2 and CSM on the viability of ASMCs. Viability of

ASMC was measured by MTT assay after treatment with (A) H2O2 (50, 100 and

200 μM) or (B) CSM (10, 25, 50 and 75%) for 2 hours, 4 hours or 24 hours. In the

4/20 hr group ASMCs were stimulated for 4 hours and then incubated with

serum-free medium for 20 hours. *p<0.05, **p<0.01 and ***p<0.001 compare

corresponding groups with relevant control groups; #p<0.05 compares groups as

indicated.

Page 140: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

121

4.2.2. Oxidative stress induced mitochondrial ROS in ASMCs

Mitochondrial ROS levels were measured by staining with MitoSOX, a dye that

targets the mitochondria of live cells and emits red fluorescence when oxidized by

superoxide. H2O2 treatment increased the mitochondrial ROS levels in a

concentration-dependent manner at 2 hours (Maximum induction at 200 μM, fold to

control, 1.61±0.16, p<0.05 vs. control) and 4 hours (Maximum induction at 200 μM,

fold to control, 1.75±0.15, p<0.01vs. control). After 24 hours only 200 μM H2O2

increased mitochondrial ROS levels (fold to controls: 24 hours, 1.6±0.25, p<0.05 vs.

control) (Figure 4.2A). Treatment for 24 hours did not show a more potent induction

of mitochondrial ROS than the 2 hour or 4 hour treatments. The induction of

mitochondrial ROS persisted up to 20 hours after the removal of H2O2 (Maximum

induction at 200 μM, fold to control, 1.56±0.22, p<0.01 vs. control).

Mitochondrial ROS was highly induced by CSM treatment in a

concentration-dependent manner (Max induction at 75% CSM, fold to controls: 2

hours, 8.54±1.99, p<0.001; 4 hours, 8.70±1.17, p<0.001; 24 hours, 11.98±2.85,

p<0.001; p-value compared to control of each time point) (Figure 4.2B). The increase

in mitochondrial ROS persisted up to 20 hour after the removal of CSM (Max

induction at 75% CSM, 8.74±2.87, p<0.001).

Page 141: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

122

A

B

2hr 4hr 24hr 4/20hr0.0

0.5

1.0

1.5

2.0

2.5

** *** **

[H2O2]

Ctrl

50M

100M

200M

#

Mito-ROS (n=5)

Mito-R

OS

(fold

chan

ge

to c

ontr

ol)

2hr 4hr 24hr 4/20hr0

5

10

15

20

*

*** ***

***

***

** ***

*

*

Control

10%

25%

50%

75%

CSM%

Mito-ROS (n=5)

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

Figure 4. 2 H2O2 and CSM induced mitochondrial ROS in ASMCs.

Mitochondrial ROS in ASMCS was measured by staining with MitoSOX after

treatment with (A) H2O2 (50, 100 and 200 μM) or (B) CSM (10, 25, 50 and 75%)

for 2, 4 or 24 hours. In the 4/20 hr group ASMCs were stimulated for 4 hours and

then incubated with serum-free medium for 20 hours. *p<0.05, **p<0.01 and

***p<0.001 compare corresponding groups with relevant control groups;

#p<0.05 compares groups as indicated.

Page 142: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

123

4.2.3. Oxidative stress induced reduction of ΔΨm in ASMCs

Maintenance of the mitochondrial membrane potential (ΔΨm) is very important

for the normal function of mitochondria. Mitochondrial depolarization can lead to an

interruption in the action of the respiratory chain and triggers apoptosis (90). In this

study ΔΨm was measured by staining with JC-1. A decreased ratio of red/green

fluorescence indicates loss of ΔΨm.

The ΔΨm was reduced by H2O2 treatment in a concentration-dependent manner at

each time point (Maximum effects at 200 μM, fold to controls: 2 hours, 0.88±0.02,

p<0.05; 4 hours, 0.89±0.01, p<0.05; 24 hours, 0.70±0.10, p<0.01; p-value compared

to control of each time point) (Figure 4.3A). The reduction of ΔΨm was observed

even 20 hours after the removal of H2O2 (Maximum effects at 200 μM, fold to

controls: 0.64±0.09, p<0.01).

There was a trend showing a decrease in ΔΨm after 2 hours treatment with CSM,

which did not reach statistical significance. Significant reduction in ΔΨm was

observed in a concentration-dependent manner at the 4 hour and 24 hour time points

(Maximum effects at 75% CSM, fold to controls: 4 hours, 0.62±0.04, p<0.001; 24

hours, 0.39±0.07, p<0.001) (Figure 4.3B). Moreover, 75% CSM treatment led to

significantly lower ΔΨm at 24 hours compared to the 2 hours (p<0.001) and 4 hours

(p<0.001) time points. The reduction of ΔΨm was still evident even 20 hours after the

removal of CSM (Maximum effects at 75% CSM, fold to controls: 0.67±0.07,

p<0.01)

Page 143: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

124

2hr 4hr 24hr 4hr/20hr0.0

0.5

1.0

1.5

********

***

**

Control

10%

25%

50%

75%

CSM%

## #

#########

Mitochondria membrane potential (n=5)

m

(Fo

ld c

ha

ng

e t

o c

on

tro

l)

2hr 4hr 24hr 4hr/20hr0.0

0.5

1.0

1.5

* ****

**

Ctrl

50 M

100 M

200 M

[H2O2]

Mitochondria membrane potential (n=6)

m

(Fo

ld c

han

ge t

o c

on

tro

l)

B

A

Figure 4. 3 H2O2 and CSM reduced ΔΨm in ASMCs. ΔΨm in ASMCS was

measured by staining with JC-1 after treatment with (A) H2O2 (50, 100 and 200

μM) or (B) CSM (10, 25, 50 and 75%) for 2, 4 or 24 hours. In the 4/20 hr group

ASMCs were stimulated for 4 hours and then incubated with serum-free medium

for 20 hours. *p<0.05, **p<0.01 and ***p<0.001 compare corresponding groups

with relevant control groups; #p<0.05, ##p<0.01, ###p<0.001 compare groups as

indicated.

Page 144: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

125

4.2.4. Oxidative stress reduced cell viability of iPSC-MSCs

To examine the effects of oxidative stress on iPSC-MSCs, iPSC-MSCs were

treated with H2O2 (50, 100 and 200 μM) or CSM (10, 25, 50 and 75%) for 4 and 24

hours. The effects were compared with those observed in ASMCs.

H2O2 reduced the viability of iPSC-MSCs in a concentration-dependent manner

after 4 and 24 hours (Max effect at 200 μM, fold to controls: 4 hours, 0.43 ±0.14,

p<0.05; 24 hours, 0.44 ±0.05, p<0.001; p-value compared to control of each time

point), in line with the effects seen in ASMCs (Figure 4.4A).

CSM also reduced the viability of iPSC-MSCs in a concentration-dependent

manner after 4 and 24 hours. At 4 hours, significant reduction was observed with 50%

CSM (fold to controls: 0.60 ±0.12, p<0.05 vs. control) and 75% CSM (fold to controls:

0.49 ±0.09, p<0.01 vs. controls; Figure 4.4B). At 24 hours, significant reduction was

observed with 25% CSM (fold to controls: 0.76 ±0.09, p<0.05 vs. control), 50% CSM

(0.18 ±0.02, p<0.001 vs. control) and 75% CSM (0.16±0.02, p<0.001 vs. control).

The viability of iPSC-MSCs was significantly lower than ASMCs (p<0.05 at each

treatment and time point), indicating that iPSC-MSCs are more sensitive to high

concentrations of CSM than ASMCs with respect to their viability.

Page 145: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

126

4hr H2O2 24hr H2O2

4hr CSM 24hr CSM

Figure 4. 4 Effects of H2O2 and CSM on viability of iPSC-MSCs. Changes in the

viability of iPSC-MSCs were measured by MTT assay after treatment with (A) H2O2

or (B) CSM for 4 hours and 24 hours. Changes in the viability of ASMCs were shown

for comparison. *p<0.05, **p<0.01 and ***p<0.001 compare corresponding groups

with relevant control groups; #p<0.05 compares groups as indicated.

0.0

0.5

1.0

1.5

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=6) iPSC-MSC (n=3)

*

****

Cell v

iab

ilit

y

(Fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=6) iPSC-MSC (n=3)

*****

Cell v

iab

ilit

y

(Fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

CSM%

*

******

*

# ##

Ctrl 10% 25% 50% 75%

ASMC (n=4) iPSC-MSC (n=3)

Cell v

iab

ilit

y

(Fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

CSM%

***

* **

# #

Ctrl 10% 25% 50% 75%

ASMC (n=4) iPSC-MSC (n=3)

Cell v

iab

ilit

y

(Fo

ld c

han

ge t

o c

on

tro

l)

A

B

Page 146: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

127

4.2.5. Oxidative stress induced mitochondrial ROS in iPSC-MSCs

H2O2 increased the mitochondrial ROS levels in iPSC-MSCs in a

concentration-dependent manner at both 4 and 24 hours (Maximum effect at 200 μM,

fold to controls: 4 hours, 4.12±1.73, p<0.05; 24 hours, 4.46±0.81, p<0.05; p-value

compared to control of each time point) (Figure 4.5A). The increase in mitochondrial

ROS levels induced by CSM (200 μM) after 24 hours was greater in iPSC-MSCs

compared to ASMCs (p<0.01.)

CSM also induced mitochondrial ROS in iPSC-MSCs, in a

concentration-dependent manner after 4 and 24 hours (Maximum effect at 75% CSM,

fold to controls: 4 hours, 6.36±1.94, p<0.05; 24 hours, 10.33±3.29, p<0.001; p-value

compared to control of each time point) (Figure 4.5B). No significant difference was

observed between the response of iPSC-MSCs and ASMCs.

Page 147: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

128

Figure 4. 5 Effects of H2O2 and CSM on mitochondrial ROS of iPSC-MSCs.

Mitochondrial ROS levels of iPSC-MSCs were measured by MitoSOX staining after

treatment with (A) H2O2 or (B) CSM for 4 and 24 hours. Changes in the mitochondrial

ROS levels of ASMCs were shown for comparison. *p<0.05, **p<0.01 and

***p<0.001 compare corresponding groups with relevant control groups; ##p<0.01

compares groups as indicated.

0

2

4

6

8

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=5) iPSC-MSC (n=4)

** **

*

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

0

2

4

6

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=5) iPSC-MSC (n=4)

*

*

##

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

0

5

10

15

ASMC (n=5) iPSC-MSC (n=4)

CSM%

*

*

***

***

*

Ctrl 10% 25% 50% 75%

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

0

5

10

15

20

ASMC (n=5) iPSC-MSC (n=4)

CSM%

****

*

Ctrl 10% 25% 50% 75%

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

A

B

4hr H2O2 24hr H2O2

4hr CSM 24hr CSM

Page 148: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

129

4.2.6. Oxidative stress induced reduction of ΔΨm in iPSC-MSCs

H2O2 reduced the ΔΨm in iPSC-MSCs in a concentration-dependent manner at

both 4 hours and 24 hours (Maximum effect at 200 μM, fold to controls: 4 hours,

0.80±0.09, p<0.05; 24 hours, 0.41±0.09, p<0.001; p-value compared to control of

each time point) (Figure 4.6A). The reduction in ΔΨm by H2O2 (200 μM) was

stronger in iPSC-MSCs compared to ASMCs but it did not reach statistical

significance.

CSM also reduced the ΔΨm in iPSC-MSCs in a concentration-dependent manner

at both 4 and 24 hours (Maximum effect at 75% CSM, fold to controls: 4 hours,

0.23±0.06, p<0.001; 24 hours, 0.21±0.01, p<0.001; p-value compared to control of

each time point) (Figure 4.6B). The reduction in ΔΨm was significantly stronger in

iPSC-MSCs compared with ASMCs after treatment with 50% and 75% CSM for 4

hours or with any concentration of CSM for 24 hours.

Page 149: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

130

Figure 4. 6 Effects of H2O2 and CSM on ΔΨm of iPSC-MSCs. The ΔΨm of

iPSC-MSCs was measured by JC-1 assay after treatment with H2O2 (A) or CSM (B)

for 4 hours and 24 hours. Changes in the ΔΨm of ASMCs were shown for

comparison. *p<0.05, **p<0.01 and ***p<0.001 compare corresponding groups

with relevant control groups; #p<0.05, ##p<0.01 compare groups as indicated.

A 4hr H2O2 24hr H2O2

0.0

0.5

1.0

1.5

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=6) iPSC-MSC (n=5)

*

***

**

m

(Fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

ASMC (n=5) iPSC-MSC (n=5)

CSM%

*******

**

******

# ##

Ctrl 10% 25% 50% 75%

m

(Fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

ASMC (n=5) iPSC-MSC (n=5)

CSM%

***

*****

***

***

***

# ## # #

Ctrl 10% 25% 50% 75%

m

(Fo

ld c

han

ge t

o c

on

tro

l)4hr CSM 24hr CSM

B

0.0

0.5

1.0

1.5

[H2O2]Ctrl 50 M 100 M 200 M

ASMC (n=6) iPSC-MSC (n=5)

** *

m

(Fo

ld c

han

ge

to

co

ntr

ol)

Page 150: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

131

4.3. Direct Co-culture with iPSC-MSCs Attenuated

CSM-induced Mitochondrial Dysfunction and Apoptosis in

ASMCs

In order to determine whether iPSC-MSCs are capable of preventing

CSM-induced mitochondrial dysfunction in ASMCs, in a prophylactic protocol

iPSC-MSCs were directly co-cultured with CellTrace-labeled ASMCs for 20 hours

and then treated with CSM (10 and 25%) for 4 hours. As a control, single cultures of

ASMCs were stained with Cell Trace, incubated for 20 hrs and then treated with CSM

for 4 hours (Figure 2.5 A). In a therapeutic protocol, CellTrace-labeled ASMCs were

treated with CSM (10 and 25%) for 4 hours before iPSC-MSCs were added to the

culture and incubated for a further 20 hours. As a control, single cultures of ASMCs

were stained with Cell Trace, treated with CSM for 4 hours and then incubated in the

absence of stimulation for 20 hrs (Figure 2.5B).

4.3.1. iPSC-MSCs prevented and reversed CSM-induced mitochondrial ROS in

ASMCs

The two cell types in the co-cultures were distinguished by the CellTrace staining

using flow cytometry (Figure 4.7A). The mitochondrial ROS levels were measured in

the gated ASMC (CellTrace-positive) population and compared with the ASMCs in

the single-culture (Figure 4.7B). 10% CSM significantly increased the levels of

mitochondrial ROS in ASMCs in the single culture (fold to control: 3.26±0.33,

p<0.01 in comparison to control), which was prevented by co-culture with

Page 151: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

132

iPSC-MSCs (fold to control: 2.12±0.17, p<0.05 in comparison to single-culture). 25%

CSM further increased the level of mitochondrial ROS in single-culture compared to

10% CSM (fold to control: 6.71±0.75, p<0.001), which was also reduced by

iPSC-MSCs (fold to control: 4.01±0.44, p<0.05 in comparison to single-culture).

However, a concentration-dependent increase of mitochondrial ROS levels of ASMCs

in the co-culture was still observed, indicating the effects of CSM were only

alleviated but not eliminated.

In addition, a therapeutic protocol was also performed, in which the CSM was

removed from the ASMCs and replaced by either serum free medium or iPSC-MSCs

(Figure 4.7C). Under these conditions only 25% CSM led to a significant increase in

mitochondrial ROS (fold to control: 3.42±0.46, p<0.01 in comparison to control). A

small but significant reduction in mitochondrial ROS was observed in the co-culture

group compared to the single culture group treated with 25% CSM (fold to control:

2.70±0.48, p<0.05), indicating therapeutic recovery from CSM-induced mitochondrial

ROS by iPSC-MSCs. Again the concentration-dependent elevation of mitochondrial

ROS was still evident among the co-culture groups, indicating the effects of CSM

were not eliminated.

Page 152: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

133

0

2

4

6

8

*

*

Control 10% CSM 25% CSM

*****

***

****

***

ASMC only

ASMC+iPSC-MSC

MitoROS of ASMCs (n=7)

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

B

A

C

Figure 4. 7 Effects of direct co-culture with iPSC-MSCs on mitochondrial

ROS in ASMCs. (A) Flow cytometry histograms showing CellTrace positive

and negative populations corresponding to ASMCs and iPSC-MSCs,

respectively. (B) Prophylactic effects of iPSC-MSCs on CSM-induced

mitochondrial ROS in ASMCs. iPSC-MSCs were co-cultured with

CellTrace-labeled ASMCs for 20 hours and then treated with CSM (10 and

25%) for 4 hours. Single ASMCs cultures were used as controls. Mitochondrial

ROS levels were determined using MitoSOX staining. (C) Therapeutic effects

of iPSC-MSCs on CSM-induced mitochondrial ROS in ASMCs.

CellTrace-labeled ASMCs were treated with CSM (10 and 25%) for 4 hours.

CSM was then removed and iPSC-MSCs were loaded. Single ASMCs cultures

were used as controls. *p<0.05, **p<0.01, ***p<0.001 compare groups as

indicated.

0

1

2

3

4

Control 10% CSM 25% CSM

**** *

***

ASMC only

ASMC+iPSC-MSC

MitoROS of ASMCs (n=6)

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

Page 153: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

134

4.3.2. iPSC-MSCs prevented but did not reverse CSM-induced reduction in ΔΨm

in ASMCs

In the prophylactic protocol, CSM induced reduction of ΔΨm at both 10% (fold

to control: 0.80±0.04, p<0.01) and 25% concentration (fold to control: 0.52±0.07,

p<0.001). 25% CSM led to lower ΔΨm than 10% CSM (p<0.001). Co-culture with

iPSC-MSCs improved the ΔΨm under both 10% CSM treatment (fold to control:

0.95±0.04, p<0.01) and 25% CSM treatment (fold to control: 0.78±0.10, p=0.059)

(Figure 4.8A). CSM still led to a trend showing a reduction in ΔΨm in ASMCs in the

co-culture, but the difference was not statistically-significant.

In the therapeutic protocol, after recovery for 20 hours, single-cultures of ASMCs

still showed significantly reduced ΔΨm in both 10% CSM-treated group (fold to

control: 0.84±0.07, p<0.01) and 25% CSM-treated group (fold to control: 0.75±0.59,

p<0.001) (Figure 4.8B). There is a trend showing an increase in mitochondrial

potential in the co-culture groups compared to the single culture at both 10% (fold to

control: 0.94±0.04) and 25% CSM treatments (fold to control: 0.87±0.11), which did

not reach statistical significance.

Page 154: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

135

0.0

0.5

1.0

1.5

Control 10% CSM 25% CSM

******** p=0.059

**

ASMC only

ASMC+iPSC-MSC

Mitochondrial Membrane Potential (n=5)

m

(Fo

ld c

han

ge t

o c

on

tro

l)

A

0.0

0.5

1.0

1.5

Control 10% CSM 25% CSM

*

* ASMC only

ASMC+iPSC-MSC

Mitochondrial Membrane Potential (n=3)

m

(Fo

ld c

han

ge t

o c

on

tro

l)

B

Figure 4. 8 Effects of direct co-culture with iPSC-MSCs on ΔΨm in ASMCs.

(A) Prophylactic effects of iPSC-MSCs on CSM-induced reduction in ΔΨm in

ASMCs. iPSC-MSCs were co-cultured with CellTrace-labeled ASMCs for 20

hours and then treated with CSM (10 and 25%) for 4 hours. Single ASMCs

cultures were used as controls. ΔΨm was determined using JC-1 staining. (B)

Therapeutic effects of iPSC-MSCs on CSM-induced reduction in ΔΨm in ASMCs.

CellTrace-labeled ASMCs were treated with CSM (10 and 25%) for 4 hours. CSM

was then removed and iPSC-MSCs were loaded. Single ASMCs cultures were used

as controls. *p<0.05, **p<0.01, ***p<0.001 compares groups as indicated.

Page 155: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

136

4.3.3. iPSC-MSCs prevented CSM-induced apoptosis of ASMCs

Based on the observations of a protective role of iPSC-MSCs against

CSM-induced mitochondrial dysfunction in ASMCs, further investigations were

carried out on whether such mitochondrial improvement led to improved function of

the cells. In particular apoptosis, as a possible outcome of severe mitochondrial

damage, was investigated in the co-culture using the prophylactic protocol (Figure

4.9). Results from Annexin V staining confirmed induction of apoptosis by CSM in

single-cultured ASMCs at 25% CSM (46.6±1.3% vs. 17.4±2.9%, p<0.001) and 50%

CSM (86.8±3.8% vs. 17.4±2.9%, p<0.001). In the absence of stimulation, the

iPSC-MSCs-treated ASMCs demonstrated higher percentage of apoptosis than those

in the single-culture (30.7±1.4% vs. 17.4±2.9%, p<0.05), which indicates that

introducing iPSC-MSCs to the system may induce stress to the ASMCs at baseline.

However, iPSC-MSCs led to a significant reduction in apoptosis induced by 25%

(34.7±1.5% vs. 46.6±1.3%, p<0.05) and 50% CSM (49.5±4.2% vs. 86.8±3.8%,

p<0.001), indicating that interaction with iPSC-MSCs can protect ASMCs from

CSM-induced mitochondrial dysfunction and apoptosis.

Page 156: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

137

Figure 4. 9 Prophylactic effects of iPSC-MSCs on CSM-induced apoptosis

in ASMCs. iPSC-MSCs were co-cultured with CellTrace-labeled ASMCs for

20 hours and then treated with CSM (10 and 25%) for 4 hours. Single ASMCs

cultures were used as controls. Apoptosis was detected by Annexin V staining.

*p<0.05, **p<0.01, ***p<0.001 compare groups as indicated.

0

20

40

60

80

100 ASMC only

ASMC+iPSC-MSC

Control 10%CSM 25%CSM 50%CSM

***

*

******

*****

***

***

*

Apoptotic rate in ASMCs (n=5)

An

nexin

V p

osit

ive c

ells (

%)

Page 157: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

138

4.4. Paracrine Effects are only Partly Involved in the

Protective Effects of iPSC-MSCs on CSM-induced

Mitochondrial Dysfunction and Apoptosis in ASMCs

4.4.1. Effects of conditioned medium from iPSC-MSCs on CSM-induced

mitochondrial dysfunction in ASMCs

In order to examine the role of paracrine effects in the protective effects of

iPSC-MSCs on CSM-induced mitochondrial damage in ASMCs, the effects of

conditioned medium from iPSC-MSCs (iPSC-MSCs-CdM) on CSM-induced

mitochondrial damage in ASMCs were investigated. CdM was a concentrate of all

paracrine factors released over a 24 hour period from confluent cultures of

iPSC-MSCs. The ASMCs were pre-treated with iPSC-MSCs-CdM for 4 hours

followed by CSM treatment for a further 4 hours in the presence of CdM. The

evaluation of mitochondrial ROS revealed very similar results with the direct

co-culture system (Figure 4.10A). CSM treatments increased mitochondrial ROS in a

concentration-dependent manner in both untreated ASMCs (fold to controls: 10%

CSM, 2.96±0.34, p<0.05; 25% CSM, 7.76±0.06, p<0.001; p-value compared to

relevant controls) and ASMCs treated with iPSC-MSCs-CdM (fold to controls: 10%

CSM, 2.56±0.29, p<0.01; 25% CSM, 5.81±0.49, p<0.001; p-value compared to

relevant controls). However, compared to untreated ASMCs, ASMCs treated with

iPSC-MSCs-CdM demonstrated significantly lower levels of mitochondrial ROS in

both 10% CSM group (p<0.05) and 25% CSM group (p<0.05).

The reduction of mitochondrial ROS by iPSC-MSCs-CdM was not associated

Page 158: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

139

with prevention of ΔΨm reduction. CSM treatments reduced ΔΨm in the untreated

ASMCs (fold to controls: 10% CSM, 0.84±0.03, p<0.01; 25% CSM, 0.59±0.05,

p<0.001; p-value compared to relevant controls) as well as in the ASMCs treated with

iPSC-MSCs-CdM (fold to controls: 10% CSM, 0.87±0.02, p<0.05; 25% CSM,

0.59±0.05, p<0.001; p-value compared to relevant controls; Figure 4.10B). The effect

of CSM on ΔΨm was similar in untreated and iPSC-MSCs-CdM-treated ASMCs.

These findings suggest that paracrine factors are only partly involved in the capacity

of iPSC-MSCs to protect ASMCs from CSM-induced mitochondrial damage

4.4.2. Effect of iPSC-MSCs-CdM on CSM-induce apoptosis of ASMCs

The effect of iPSC-MSCs-CdM on CSM-induced ASMC apoptosis was also

determined. CSM increased the percentage of apoptotic ASMCs in a

concentration-dependent manner (Maximum induction by 50% CSM: 64.8±9.2% vs.

12.6±1.4%, p<0.001; p-value compared to control). Within the iPSC-MSCs-CdM

treated groups, a similar induction was observed (Maximum induction by 50% CSM:

67.8±9.0% vs. 13.8±1.3%, p<0.001; p-value compared to control) (Figure 4.11).

Treatment with iPSC-MSCs-CdM did not affect the baseline or CSM-induced

apoptosis.

Page 159: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

140

0

2

4

6

8

10Without CdM

With CdM

******

*****

***

Control 10% CSM 25% CSM

*

*

*

Mitochondrial-ROS (n=6)

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5Without CdM

With CdM

Control 10% CSM 25% CSM

******

****

***

**

Mitochondrial Membrane Potential (n=4)

m

(Fo

ld c

han

ge t

o c

on

tro

l)

B

A

Figure 4. 10 Effects of iPSC-MSCs-CdM on mitochondrial ROS and

ΔΨm in ASMCs. ASMCs were pre-treated with iPSC-MSCs-CdM for 4

hours before stimulating with CSM for 4 hours. (A) Mitochondrial ROS

was determined by MitoSOX staining and (B) ΔΨm by JC-1 staining.

*p<0.05, **p<0.01, ***p<0.001 compare groups as indicated.

Page 160: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

141

0

20

40

60

80

100Without CdM

With CdM

Control 10%CSM 25%CSM 50%CSM

******

*****

**

******

*****

**

Apoptotic rate of ASMCs (n=4)

An

nexin

V p

osit

ive c

ells (

%)

Figure 4. 11 Effects of iPSC-MSCs-CdM on CSM-induced apoptosis in

ASMCs. ASMCs were pre-treated with iPSC-MSCs-CdM for 4 hours before

stimulating with CSM for 4 hours. Apoptosis was determined by Annexin V

staining. **p<0.01, ***p<0.001 compare groups as indicated.

Page 161: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

142

4.4.3. Effects of trans-well co-culture with iPSC-MSCs on CSM-induced

mitochondrial alterations in ASMCs

To better replicate the crosstalk between the ASMCs and iPSC-MSCs through

paracrine effects, trans-well co-culture was performed using cell culture inserts with

0.4 μm pores. ASMCs in 6-well plates were incubated with either blank inserts or

inserts containing iPSC-MSCs for 20 hours before being treated with CSM (10 or

25%) for 4 hours (Figure 4.12A). Mitochondrial ROS levels were increased by 10%

CSM (fold to control: 3.55±0.59, p<0.01) and 25% CSM (fold to control: 8.35±0.83,

p<0.001) in the ASMCs treated with blank inserts. In the trans-well co-culture group,

mitochondrial ROS levels in ASMCs were also elevated by 10% CSM (fold to control:

3.12±0.57, p<0.05) and 25% CSM (fold to control: 7.15±0.84, p<0.001), but the

mitochondrial ROS induced by 25% CSM was significant reduced compared to the

blank insert group (p<0.05) (Figure 4.12B).

Significant reduction of ΔΨm was induced by 25% CSM at both blank insert

group (fold to control: 0.39±0.06, p<0.01) and iPSC-MSCs-treated group (fold to

control: 0.49±0.12, p<0.01). No significant difference was observed between the

iPSC-MSCs treatment and blank insert (Figure 4.12C). These results are in line with

the findings from the iPSC-MSCs-CdM experiment.

Page 162: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

143

A

Figure 4. 12 Effects of co-culture with iPSC-MSCs using the trans-well

system on mitochondrial ROS and ΔΨm in ASMCs. (A) Illustration of the

trans-well setup. Inserts with or without iPSC-MSCs were placed into the wells of

6-well tissue culture plates, containing ASMCs, for 20 hours. The whole system

was treated with CSM for 4 hours. In the ASMCs mitochondrial ROS was

determined by (B) MitoSOX staining and (C) ΔΨm by JC-1 staining. *p<0.05,

**p<0.01, ***p<0.001 compare groups as indicated.

0

2

4

6

8

10

Ctrl 10%CSM 25%CSM

****

*****

*******

Blank Insert

iPSC-MSCs

Mitochondrial-ROS (n=4)

Mit

o-R

OS

(fo

ld c

han

ge t

o c

on

tro

l)

0.0

0.5

1.0

1.5

Ctrl 10%CSM 25%CSM

Blank Insert

iPSC-MSCs

****

Mitochondrial Membrane Potential (n=4)

m

(Fo

ld c

han

ge t

o c

on

tro

l)

C

B

Page 163: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

144

4.4.4. Effect of trans-well co-culture with iPSC-MSCs on CSM-induced apoptosis

in ASMCs

The effect of co-culture using a trans-well system on ASMC apoptosis was also

examined. 25% CSM induced a significant increase in ASMC apoptosis in both the

blank insert group (48.6±10.9% vs. 11.7±2.6%, p<0.05) and the iPSC-MSC-treated

group (54.6±8.3% vs. 16.7±3.3%, p<0.01) (Figure 4.13). No difference was observed

in the iPSC-MSCs-treated group in comparison with the blank group.

0

20

40

60

80

Ctrl 10%CSM 25%CSM

iPSC-MSCs

Blank Insert

***

Apoptotic rate of ASMCs (n=4)

An

nexin

V p

osit

ive c

ells (

%)

Figure 4. 13 Effects of trans-well co-culture with iPSC-MSCs on apoptosis in

ASMCs. Inserts with or without iPSC-MSCs were placed into the wells of 6-well

tissue culture plates, containing ASMCs, for 20 hours. The whole system was treated

with CSM for 4 hours. In the ASMCs apoptosis was determined by Annexin V

staining.*p<0.05, **p<0.01 compares corresponding groups with relevant controls.

Page 164: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

145

4.5. Mitochondrial Transfer from iPSC-MSCs to ASMCs

4.5.1. Mitochondrial transfer from iPSC-MSCs to ASMCs

A possible mechanism underlining the protective effects of iPSC-MSCs against

CSM-induced mitochondrial dysfunction in ASMCs is mitochondrial transfer. To

investigate whether mitochondrial transfer from iPSC-MSCs to ASMCs takes place,

ASMCs were pre-stained with CellTrace Violet while iPSC-MSCs were pre-stained

with MitoTracker Red, a mitochondrial-targeted fluorescent dye. They were

co-cultured for 20 hours followed by 4 hours stimulation with 25% CSM. The cells

were fixed and stained with phalloidin which selectively labels F-actin. Using

fluorescent microscopy, iPSC-MSCs-derived MitoTracker-labeled mitochondria were

observed in CellTrace-labeled ASMCs, indicating transfer of mitochondria from

iPSC-MSCs to ASMCs (Figure 4.14). In addition, tunneling nanotube (TNT)-like

structures, containing iPSC-MSC mitochondria, were observed connecting

iPSC-MSCs and ASMCs. Actin filaments were identified in the TNTs by phalloidin

staining confirming a connection between the cytoskeleton systems of iPSC-MSCs

and ASMCs, which may provide a path through which the mitochondria are

transferred.

4.5.2. CSM increased mitochondrial transfer from iPSC-MSCs to ASMCs

Mitochondrial transfer was quantified by determining the percentage of

MitoTracker-positive cells in the ASMC population using flow cytometry.

Mitochondrial transfer was detected after co-culture for 24 hours without CSM

Page 165: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

146

stimulation or after stimulation with 10% or 25% CSM during the last 4 hours of the

co-culture period. 10% CSM enhanced mitochondrial transfer, as shown by a

significant elevation of the transfer rate (48.9±6.8% vs. 18.8±4.1%, p<0.05). 25%

CSM further increased the transfer rate compared to 10% CSM (75.5±8.5% vs.

48.9±6.8%, p<0.05), suggesting that CSM promoted mitochondrial transfer from

iPSC-MSCs to ASMCs in a concentration-dependent manner (Figure 4.15).

Page 166: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

147

Figure 4. 14 Mitochondrial transfer from iPSC-MSCs to ASMCs. ASMCs were

pre-stained with CellTrace and iPSC-MSCs with MitoTracker. Cells were co-cultured

for 20 hours and then treated with 25% CSM for 4 hours. Phalloidin staining was

applied after fixation. The presence of MitoTracker-labeled mitochondria was

observed in CellTrace-labeled cells, indicating transfer of iPSC-MSC-derived

mitochondria to ASMCs. Tunneling nanotube–like structures between iPSC-MSCs

and ASMCs can be observed. Scale bar=25 μm.

Page 167: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

148

Ctrl 10% CSM 25% CSM0

20

40

60

80

100 ****

*

Percentage of ASMCs that recievedMSCs-derived mitochondria (n=3)

Mit

oT

racker+

AS

MC

s (

%)

Figure 4. 15 Effect of CSM on the rate of mitochondrial transfer from

iPSC-MSCs to ASMCs. ASMCs were pre-stained with CellTrace and

iPSC-MSCs with MitoTracker. Cells were co-cultured for 20 hours and then

treated with 25% CSM for 4 hours. (A) Mitochondrial transfer from

iPSC-MSCs (MitoTracker-positive) to ASMCs (CellTrace-positive) was

determined by flow cytometry. The ASMC population is indicated by the gate

in the plots of co-culture. (B) The rate of mitochondrial transfer was evaluated

by the percentage of MitoTracker-positive ASMCs in total ASMC population.

*p<0.05, ***p<0.001 compare groups as indicated.

A

B

Page 168: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

149

4.6. Discussion

This study demonstrated increased mitochondrial ROS and reduced ΔΨm in

healthy ASMCs, in response to H2O2 or CSM. Direct contact with iPSC-MSCs

protected the ASMCs from CSM-induced mitochondrial ROS and reduction of ΔΨm,

which may explain the attenuation of CSM-induced apoptosis. When the interaction

between the two cell types was restricted to paracrine effects, only CSM-induced

mitochondrial ROS but not CSM-induced ΔΨm loss and apoptosis in ASMCs were

ameliorated, indicating that the paracrine effects can only partly explain the protective

effects of iPSC-MSCs. Under direct interaction, mitochondria were transferred from

iPSC-MSCs to ASMCs, which was enhanced by CSM treatment. Formation of TNTs

between iPSC-MSCs and ASMCs was observed in the co-culture. The prophylactic

effects of iPSC-MSCs on CSM-induced mitochondrial damage in ASMCs were lost

when the co-culture was carried out using a trans-well system. As the trans-well

inserts would block nanotube formation, whilst allowing only paracrine

communication, this suggests an important role of mitochondrial transfer in the

protective effects of iPSC-MSCs. These findings highlight that the full protective

capacity of iPSC-MSCs may rely on direct contact with target cells through which

mitochondrial transfer can take place. This study suggested that iPSC-MSCs may be

promising candidates for cell-based treatments targeting mitochondrial damage in

patients with COPD.

Targeting mitochondrial damage provides a new promising strategy for

developing treatments for COPD. Mitochondrial dysfunction was recently

Page 169: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

150

demonstrated in ASMCs from patients with COPD, as characterized by reduced ATP

levels, mitochondrial complex protein expression and ΔΨm, as well as increased

mitochondrial ROS (273). The study also demonstrated mitochondrial damage, as

reflected by the same parameters, in an ozone-induced mouse model of airway

inflammation and hyper-responsiveness. Mitochondrial damage as well as airway

inflammation and hyper-responsiveness were attenuated by the mitochondrial-

targeted antioxidant MitoQ (273). In an earlier report, swelling and fragmented

mitochondria with depleted cristae were observed in airway epithelial cells from

patients with stage 4 COPD (105). In the present study, oxidative stress-induced

mitochondrial damage was identified by increased mitochondrial ROS and loss of

ΔΨm in ASMCs, which provided a model in which to study the effects of

iPSC-MSCs.

CS, the major cause of COPD, contains numerous oxidants. One puff of cigarette

smoke contains approximately 1015

oxidants in the gas phase and 1018

oxidants in the

tar phase, together with 3000 ppm of nitrite oxide (57, 58). Acute exposure to CS is

reported to impair energy metabolism and mitochondrial protein expression in mouse

lungs, leading to switch of glucose metabolism to pentose phosphate pathway and

reduction of substrate supply to mitochondrial respiration (4). Cigarette smoke

medium was reported to alter the morphology of mitochondria in vitro, in both airway

epithelial cells (105) and ASMCs (10), inducing mitochondrial fragmentation in both

cell types and mitochondrial ROS elevation in ASMCs. However, the effect on ΔΨm

was not examined in these studies. In the present study CSM was demonstrated to

Page 170: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

151

induce mitochondrial ROS, ΔΨm loss and apoptosis in normal ASMCs from healthy

subjects. Although inhalation of CS primarily affects the epithelium, CS-derived

chemicals could also affect airway smooth muscle directly via the circulation (205) or

indirectly through inducing the production of epithelial cell-derived inflammatory

mediators such as IL-1β, IFN-γ and TNF-α which affect ASMC function (70, 231).

Moreover, chronic exposure to CS can lead to a defective epithelial barrier function

(102, 226), which may result in a direct exposure of CS to ASMCs. Hence the in vitro

model of CSM-induced ASMC dysfunction is physiologically relevant. On the other

hand, it is to my interest to establish a model of oxidative stress-induced

mitochondrial damage, through which mitochondrial-targeted therapeutic approaches

can be tested. The concentrations of CSM chosen for the co-culture experiments were

based on the findings of concentration response experiments performed on single

cultures of ASMCs, as it would be impossible to determine physiologically-relevant

concentrations of cigarette smoke constituents. CSM-induced apoptosis was observed

in association with mitochondrial dysfunction. Induction of apoptosis by CS has also

been demonstrated in other cell types in the lung including endothelial cells, alveolar

epithelial cells, fibroblasts and immune cells (16, 103, 116, 127, 166, 223). The

anti-apoptotic role of iPSC-MSCs demonstrated in the ASMC model may also be

important in other cell types in the lung.

MSCs have been reported to attenuate lung emphysema and allergic airway

inflammation in murine models (94, 150, 151, 244). Although MSCs are considered

as putative cell-based therapies for COPD, their potential mechanisms of their action

Page 171: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

152

remain unclear. It is possible that their effects are at least partly mediated by

improving mitochondrial function in COPD lungs. Indeed, MSCs have been shown to

rescue defective mitochondria and impaired bioenergetics of mitochondria in an

animal model of lipopolysaccharide-induced acute lung injury (118), as well as in an

in vitro model of mitochondrial DNA mutation (238). In my study, the effects of

iPSC-MSCs on CSM-induced mitochondrial damage in ASMCs were examined.

Using a prophylactic protocol, I have shown that interaction with iPSC-MSCs

prevents CSM-induced mitochondrial ROS and reduction in ΔΨm in ASMCs.

CSM-induced apoptosis in ASMCs was also attenuated by iPSC-MSCs, which may

be a result of improved mitochondrial function. Using the therapeutic protocol, I

observed significant attenuation of CSM-induced mitochondrial ROS but not reversal

of CSM-induced ΔΨm reduction. The therapeutic effects of iPSC-MSCs appear to be

less pronounced, possibly due to the partial recovery of mitochondrial function in

absence of CSM stimulation, or the progression of mitochondrial damage to an

irreversible stage. Therefore the prophylactic protocol was chosen for later

experiments. These findings support the hypothesis that iPSC-MSCs can attenuate

CSM-induced mitochondrial damage, suggesting their potential capacity to repair

mitochondrial dysfunction in patients with COPD.

MSCs have been reported to induce paracrine effects by releasing

immunoregulatory cytokines such as VEGF, TGF-β1, HGF, bFGF and platelet derived

growth factor (2, 235, 270). In the previous chapter iPSC-MSCs-CdM was found to

attenuate CSM-induced apoptosis in bronchial epithelial cells in vitro. However, in

Page 172: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

153

the present study, iPSC-MSCs-CdM was only able to ameliorate CSM-induced

mitochondrial ROS in ASMCs without any effectiveness on ΔΨm or apoptosis. As

iPSC-MSCs were not in close proximity to ASMCs or exposed to CSM treatment

before the collection of CdM, the lack of effect may indicate that the protective effects

of iPSC-MSCs on ASMCs may be induced by the CSM or mediators produced by the

ASMCs themselves. However, in a trans-well co-culture system where there is

paracrine crosstalk between the two cell types, just as in the direct co-culture system,

the iPSC-MSCs prevented CSM-induced mitochondrial ROS production but not the

reduction of ΔΨm or apoptosis in ASMCs.

Therefore, the protective role of iPSC-MSCs against CSM-induced mitochondrial

depolarization and apoptosis in ASMCs seems to be dependent on the direct contact

between the two cell types, which may also involve mitochondrial transfer.

Mitochondrial transfer was firstly identified in co-cultures of BM-MSCs and lung

epithelial cells with defective mitochondria (238). Since then, mitochondrial transfer

from BM-MSCs to epithelial cells, endothelial cells, cardiomyocytes and

osteosarcoma cells has been demonstrated (55, 192, 204, 238). Mitochondrial transfer

from MSCs to epithelial cells was also observed in vivo in rodent models of acute

lung injury (118) and CS-induced emphysema (150). In this study mitochondrial

transfer from iPSC-MSCs to ASMCs was demonstrated. Moreover, CSM significantly

enhanced the transfer in a concentration-dependent manner. A previous study reported

that CS exposure can lead to a reduction in the supply of substrates to the electron

transport chain, leading to increased mitochondrial workload to maintain normal

Page 173: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

154

energy supply (4). The transfer of healthy mitochondria may alleviate such burden

and thus relieve the mitochondrial stress as indicated by the reversal of mitochondrial

depolarization. In this study the formation of TNTs connecting the cytoskeleton

systems of the two cells were evident by staining the actin filaments. TNTs are highly

sensitive nanotubular structures between cells which can facilitate the selective

transfer of membrane vesicles and organelles (215). TNTs have been reported to

mediate mitochondrial transfer from BM-MSCs to epithelial cells, from endothelial

cells to cancer cells and from vascular smooth muscle cells to MSCs (196, 238, 258).

ASMCs are also reported to form TNTs with CD4+ T cells in vitro, leading to

enhanced T cell survival (7). The formation of TNTs between iPSC-MSCs and

ASMCs may provide a path through which mitochondria can be transferred, but the

mechanisms underlining the regulation of TNT formation by the two cell types or by

CSM remain unknown. The mechanism by which cells regulate mitochondrial

transfer through the TNTs is also unclear. A recent study demonstrated that

mitochondria are carried by a mitochondrial Rho-GTPase, Miro1, which can move

along the actin filament in the TNTs between two cell types (6). Possible mechanisms

of regulation of Miro1 by CS should be further investigated in the future.

Despite the evidence indicating iPSC-MSC-mediated protection of ASMCs from

CSM-induced mitochondrial damage, some of my findings need to be considered

carefully. Firstly, although MSCs have been reported to rescue injured cells in various

models of inflammation and oxidative stress, their own response to oxidative stress is

often neglected. In this study I have demonstrated that iPSC-MSCs are not more

Page 174: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

155

resistant to oxidative stress than ASMCs. Therefore caution is required in the

development of MSC-based therapies as MSCs may also show mitochondrial damage

under oxidative stress. Secondly, in the absence of CSM treatment, the co-culture with

iPSC-MSCs increased the apoptosis of ASMCs suggesting that iPSC-MSCs may

induce stress in the target cells at baseline. Such data imply that the safety of

iPSC-MSCs should be carefully examined if they are to be used therapeutically in

man.

There are several limitations to this study and therefore further work is required

to better understand the protective effects of iPSC-MSCs and the mechanisms

mediating these effects. Firstly, the ASMCs used in my study were isolated from

healthy subjects. A better understanding of the therapeutic potential of iPSC-MSCs in

COPD would result from a study of ASMCs from COPD patients, in which

mitochondria are known to be already impaired (273). Secondly, although the

paracrine effects were not shown to mediate the full protective effect of iPSC-MSCs,

protection from CSM-induced mitochondrial ROS was still evident. The molecules

mediating these effects require further investigation. Thirdly, the formation of TNTs

may also facilitate the exchange of other cellular content, which may also protect the

ASMCs from mitochondrial damage. Lastly, the mechanism of CSM‟s effect on

promotion of mitochondrial transfer need to be further addressed.

In summary, this study demonstrated that the capacity of iPSC-MSCs to attenuate

CSM-induced mitochondrial damage in ASMCs relies highly on direct cell-cell

contact. Mitochondrial transfer through the TNTs may play an important role in this

Page 175: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

156

process. I elucidated the potential of iPSC-MSCs to target mitochondrial dysfunction

in patients with COPD, thus further highlighting iPSC-MSCs as a promising

candidate for development of cell-based therapies in COPD.

Page 176: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

157

Chapter 5. Effects of iPSC-MSCs on

Ozone-induced Airway

Hyper-responsiveness, Lung Inflammation,

Apoptosis and Mitochondrial Dysfunction

in Mice

Page 177: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

158

5.1. Introduction

Ozone (O3) is a potent oxidizing pollutant. High concentration of ozone in the

environment have been reported to be associated with worsening of symptoms in

patients with COPD and asthma (18, 74, 84, 173, 197, 239, 257). In mouse models,

exposure to ozone is reported to induce airway oxidative stress, airway

hyper-responsiveness (AHR) and inflammation (54, 253, 272, 274, 275). Repeated

exposure to ozone can destroy alveolar structures in mice, finally leading to

emphysema (203, 253).

Beneficial effects of iPSC-MSCs have been reported in rodent models of allergic

airway inflammation and CS-exposed emphysema (150, 244). However, their effects

on AHR have not been investigated yet. In this study, a previously reported mouse

model of acute ozone-exposure was used (274, 275) in order to examine the effects of

iPSC-MSCs on oxidative stress-induced AHR and airway inflammation.

Although in the last chapter the capacity of iPSC-MSCs to prevent oxidative

stress-induced mitochondrial dysfunction in ASMCs was demonstrated in vitro, the

capacity of iPSC-MSCs to modulate oxidative stress-induced mitochondrial

dysfunction in the lung in vivo remains unknown. Mitochondrial dysfunction in lung

tissue has been documented in the ozone-exposed mouse model (273). Therefore the

other aim of the study was to examine the effects of iPSC-MSCs on oxidative

stress-induced mitochondrial dysfunction in the ozone-exposed mouse model. The

hypothesis of the study is that intravenous administration of iPSC-MSCs can

prophylactically prevent and/or therapeutically reverse ozone-induced AHR, airway

Page 178: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

159

inflammation, apoptosis and mitochondrial dysfunction in the lung.

The aims of this chapter were:

1. To evaluate the effects of iPSC-MSCs on AHR and lung inflammation in an acute

ozone-exposed mouse model.

2. To examine effects of iPSC-MSCs on mitochondrial dysfunction and apoptosis in

the lung, in an acute ozone-exposed mouse model.

5.2. Results

5.2.1. iPSC-MSCs prevented ozone-induced AHR

Male C57BL/6 mice were exposed to normal air or ozone (3 ppm) for 3 hours.

1x106 iPSC-MSCs or PBS were administrated to mice through intravenous injection

either 24 hours prior or 6 hours after the exposure. There were 5 groups of mice in

total: the Air/saline (n=5), the Air with iPSC-MSCs administrated 24 hours

prior-exposure (Air/-24hr) (n=5), the Ozone/saline (n=6), the Ozone with iPSC-MSCs

administrated 24 hours prior-exposure (Ozone/-24hr) (n=6) and the Ozone with

iPSC-MSCs administrated 6 hour post-exposure (Ozone/+6hr) (n=6).

Pulmonary resistance (RL) to increasing concentration of acetylcholine was

measured 24 hours after exposure (Figure 5.1A). The injection of iPSC-MSCs 24

hours prior to air exposure did not show significant effect on AHR compared to the

Air/saline group, as indicated by -log PC100 (2.01±0.06 vs. 2.15±0.06, p>0.05) Figure

5.1B). Ozone/saline group demonstrated significantly increased AHR compared with

Air/saline group, as indicated by significantly reduced -log PC100 (1.68±0.06 vs.

Page 179: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

160

2.15±0.06, p<0.01). The treatment of iPSC-MSCs 24 hours prior-exposure

significantly reduced ozone-induced AHR as compared with Ozone/saline group (-log

PC100: 1.99±0.08 vs.1.68±0.06, p<0.05). The Ozone/+6hr group, on the other hand,

did not exhibit a difference in AHR compared to the Ozone group (-log PC100:

1.75±0.07 vs.1.68±0.06, p>0.05). The data suggest that intravenously injected

iPSC-MSCs can prevent rather than reverse ozone-induced AHR in mice.

0.0 0.5 1.0 1.5 2.0 2.50

50

100

150

200

air -24hr

ozone

ozone -24hr

air

ozone +6hr

Log[ACh] (mg/ml)

Ch

an

ge in

RL(%

)

A

air air ozone ozone ozone0

1

2

3

salinesaline MSCs

-24hrMSCs

+6hr

MSCs

-24hr

**

B

-lo

gP

C1

00

Figure 5. 1 Effect of iPSC-MSCs on ozone-induced AHR. Mice were exposed to

ozone (3 ppm) or air for 3 hours. iPSC-MSCs were intravenously injected 24 hrs

before or 6 hours post exposure. (A) RL in response to increasing concentration of

acetylcholine. (B) -log PC100 derived from plot. **p<0.01 compared to air/saline; †p<0.05 compared to ozone/saline.

Page 180: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

161

5.2.2. Effect of iPSC-MSCs on ozone-induced infiltration of inflammatory cells in

the lung

The total cell number in bronchoalveolar lavage (BAL) was determined as

indication of inflammatory cell infiltration in the lung (Figure 5.2A). The

Ozone/saline group demonstrated significantly higher total cell numbers in the BAL

compared to the Air/saline group (51.5±6.7x105 vs. 13.8±4.8x10

5, p<0.01). The

Ozone/-24hr group also demonstrated higher total cell numbers in the BAL compared

to Air/-24hr group (31.9±4.3x105 vs. 12.4±2.1x10

5, p<0.01). However, the total cell

numbers in Ozone/-24hr group was significantly lower compared to Ozone/saline

group (p<0.05). The Ozone/+6hr group did not show any significant difference in

total cell numbers compared to the Ozone/Saline group.

Diff-Quik staining was performed on the BAL cells after cytospin. The numbers

of neutrophils, macrophages, eosinophils and lymphocytes were determined. In the

saline groups, ozone exposure significantly increased the neutrophil number in the

BAL compared to air exposure (20.6±3.5x105 vs. 0.44±0.20x10

5, p<0.05). Treatment

with iPSC-MSCs 24 hours before the ozone exposure significantly reduced the

neutrophil number compared to the ozone/saline group (7.6±1.3x105 vs. 20.6±3.5x10

5,

p<0.01), but the level was still higher than the air/-24hr group (7.6±1.3x105 vs.

0.26±0.07x105, p<0.05). The Ozone/+6hr group did not show any significant

difference compared to Ozone/Saline group. Macrophage number was also elevated in

Ozone/saline group compared to Air/saline (21.6±1.7x105 vs. 1.58±0.67x10

5, p<0.01).

There was a trend of decrease in Ozone/-24hr compared to Ozone/saline but not

Page 181: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

162

reaching significance (14.6±2.8x105 vs. 21.6±1.7x10

5). The Ozone/+6hr group did not

show trend of difference compared to Ozone/Saline group (21.2±3.1x105 vs.

21.6±1.7x105). Similarly, eosinophil number in the BAL was increased in

Ozone/saline group compared with Air/saline (2.58±0.63x105 vs. 0.16±0.09x10

5,

p<0.01). There was a trend of decreased eosinophil number in Ozone/-24hr group

compared with Ozone/saline though not reaching significance (0.98±0.27x105 vs.

2.58±0.63x105). Similar trend existed in Ozone/+6hr group as compared with

Ozone/Saline group, also not reaching significance (1.31±0.52x105 vs.

2.58±0.63x105). As for number of lymphocytes, the induction by ozone is significant

(5.76±1.21x105 vs. 1.06±0.41x10

5, p<0.05). There were trends of decrease in both

Ozone/-24hr (4.44±0.86x105) and Ozone/+6hr groups (4.13±1.21x10

5), neither of

which reached significance.

The results suggested that iPSC-MSCs were able to prevent but not reverse

ozone-induced recruitment of inflammatory cells into mouse lung.

Page 182: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

163

air air ozone ozone ozone0

20

40

60

80

**##

**

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

To

tal C

ells

(cells

x 1

05)

air air ozone ozone ozone0

10

20

30

40

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

*

#

**

††

Neu

tro

ph

ils (

cells x

10

5)

air air ozone ozone ozone0

10

20

30

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

**##

**

Macro

ph

ag

es (

cells x

10

5)

air air ozone ozone ozone0

2

4

6

8

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

*##

Lym

ph

ocyte

s (

cells x

10

5)

air air ozone ozone ozone0

1

2

3

4

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

*

#

Eo

sin

op

hils

(cells

x 1

05)

A

B

Figure 5. 2 Effects of iPSC-MSCs on total and differential cell counts in BAL

from ozone-exposed mice. BAL was collected 24 hours after ozone exposure. (A)

The number of total cells was determined. (B) Differential counts of cells in BAL.

BAL cells were subjected to a cytospin protocol and stained by Diff-Quik.

Neutrophils, macrophages, eosinophils and lymphocytes were identified, based on

their color and morphology, and counted. *p<0.05, **p<0.01 compared to air/saline;

#p<0.05, ##p<0.01 compared to air/-24hr, †p<0.05,

††p<0.01 compared to

ozone/saline.

Page 183: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

164

5.2.3. Effect of iPSC-MSCs on cytokine contents of BALF

The BAL fluid (BALF) was collected as the non-cellular compartment of the

BAL. Levels of the inflammatory mediators MCP-1, eotaxin, IL-6, IL-5 and MIP-1α

in BALF were measured by MAGPIX Luminex multiplexing analyzer. Compared

with the Air/saline group, ozone exposure significantly elevated the levels of MCP-1

(181.5±13.3 pg/ml vs. 151.9±1.6 pg/ml, p<0.05), eotaxin (48.6±2.2 pg/ml vs.

30.2±2.7 pg/ml, p<0.01), IL-6 (76.1±9.8 pg/ml vs. 37.4±2.7 pg/ml, p<0.01), IL-5

(11.9±2.6 pg/ml vs. 1.9±0.07 pg/ml, p<0.05) and MIP-1α (9.2±1.6 pg/ml vs. 2.2±0.1

pg/ml, p<0.05). Prophylactic administration of iPSC-MSCs significantly reduced IL-6

(45.1±3.2 pg/ml, p<0.05) and IL-5 (4.66±0.60 pg/ml, p<0.01) levels compared with

the Ozone/saline group. Trends of reduction in eotaxin (40.8±2.3 pg/ml, p=0.053) and

MIP-1α (5.4±0.8 pg/ml, p=0.055) in the BALF were also demonstrated in the

Ozone/-24hr group compared with Ozone/saline group. Therapeutic treatment with

iPSC-MSCs reversed the ozone-induced increase in eotaxin (36.8±2.5 pg/ml, p<0.01),

IL-6 (47.8±3.6 pg/ml, p<0.05), IL-5 (4.6±0.9 pg/ml, p<0.05) and MIP-1α (4.8±0.6

pg/ml, p<0.05) in the BALF. Both prophylactic and therapeutic treatments induced a

trend of reduction in MCP-1 levels which did not reach statistical significance

(Ozone/-24hr, 160.0±1.0 pg/ml; Ozone/+6hr, 157.9±2.8 pg/ml).

The results indicated that iPSC-MCSs prevented and reversed ozone-induced

elevation in inflammatory mediator levels in BALF, including eotaxin, IL-6, IL-5 and

MIP-1α.

Page 184: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

165

air air ozone ozone ozone0

50

100

150

200

250

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

*M

CP

-1 (

pg

/ml)

air air ozone ozone ozone0

20

40

60

80

100

†**

p=0.053

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

Eo

taxin

(p

g/m

l)

air air ozone ozone ozone0

50

100

150

200

**†

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

IL-6

(p

g/m

l)

air air ozone ozone ozone0

10

20

30

*

##

*†

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

IL-5

(p

g/m

l)

air air ozone ozone ozone0

10

20

30

40

*# †

*

p=0.055

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

MIP

-1

(p

g/m

l)

Figure 5. 3 Effects of iPSC-MSCs on inflammatory mediator levels in BALF after

ozone exposure. Levels of MCP-1, eotaxin, IL-6, IL-5 and MIP-1α in BALF were

measured using a MAGPIX Luminex multiplexing analyser. *p<0.05, **p<0.01

compares to air/saline; #p<0.05, ##p<0.01 compared to air/-24hr, †p<0.05,

††p<0.01

compares to ozone/saline.

Page 185: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

166

5.2.4. Effects of iPSC-MSCs on apoptosis and proliferation in ozone-exposed

lung tissue

Apoptosis in lung sections was detected using TUNEL staining (Figure 5.4A).

Apoptotic cells were labeled green and the nuclei were labeled blue. The Ozone/saline

group demonstrated significantly increased number of apoptotic cells in lung sections

in comparison with the air/saline group (number/20x field: 21.7±2.9 vs. 3.7±0.4,

p<0.05) (Figure 5.4B). Both Ozone/-24hr and Ozone/+6hr groups showed

significantly reduced number of apoptotic cells compared with Ozone/saline group

(number/20x field: 9.0±1.2, p<0.01 and 9.3±1.3, p<0.01 respectively) (Figure 5.4B).

Proliferative cells in lung sections were identified by immunofluorescent staining

against Ki-67, a marker of proliferation (Figure 5.5A). Ozone exposure significantly

reduced the number of proliferating cells in lung sections (number/20x field: 54.6±5.5

vs. 77.2±5.9, p<0.05) (Figure 5.5B). The Ozone/-24hr group did not show a

significant difference in proliferating cell number compared to Air/-24hr group

(number/20x field: 66.4±7.9, vs.71.5±6.4), indicating that negative effect of ozone on

proliferation might be prevented by treatment with iPSC-MSCs (Figure 5.4B).

However, the Ozone/-24hr group only showed a trend of increased proliferating cell

number compared to Ozone/saline group which did not reach statistical significance.

The Ozone/+6hr group also did not show a significant difference in the number of

proliferating cells compared with the Ozone/saline group.

Page 186: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

167

air air ozone ozone ozone0

10

20

30

*

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

† †† †

Ap

op

toti

c C

ells

(n

um

ber/

20x f

ield

)

Figure 5. 4 Effects of iPSC-MSCs on ozone-induced apoptosis in mouse lungs. (A)

Apoptotic cells were identified by TUNEL staining (green) indicated by the arrows.

The nuclei were visualized by DAPI staining (blue). (B) Number of apoptotic cells in

lung sections. Apoptotic cells were counted in five randomly selected 20x fields for

each mouse using a fluorescent microscope. *p<0.05 compared to air/saline; ††

p<0.01

compares to ozone/saline.

A

B

Page 187: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

168

air air ozone ozone ozone0

20

40

60

80

100

salinesaline SC

-24hr

SC

+6hr

*

SC

-24hr

Pro

life

rati

ve C

ells

(n

um

ber/

20x f

ield

)

Figure 5. 5 Effects of ozone and iPSC-MSCs on cellular proliferation in mouse

lungs. (A) Detection of proliferating cells by Ki-67 staining (red). The nuclei were

visualized by DAPI staining (blue). (B) Number of proliferative cells in lung sections.

Proliferative cells were counted in five randomly selected 20x fields for each mouse

using a fluorescent microscope. *p<0.05 compared to air/saline.

A

B

Page 188: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

169

5.2.5. Effect of iPSC-MSCs on ozone-induced cellular ROS and mitochondrial

dysfunction in mouse lungs

Intact mitochondria and cytoplasmic fractions were extracted from lung tissue.

The ROS levels in the cytoplasm fractions were measured by DCF assay (Figure

5.6A). Ozone exposure significantly elevated the cytoplasmic ROS levels (RFU/mg

protein: 3131±269.8 vs. 2061±123.9, p<0.01). When treated with iPSC-MSCs 24

hours prior to ozone exposure, the cytoplasmic ROS levels were still significantly

higher compared with the Air/-24hr group (RFU/mg protein: 2787±171.9 vs.

2030±178.9, p<0.05). The Ozone/+6hr group also showed significantly higher level of

cytoplasmic ROS compared with Air/saline group (RFU/mg protein: 2909±194.4 vs.

2061±123.9, p<0.05). No significant reduction was observed in both Ozone/-24hr and

Ozone/+6 groups in comparison with the Ozone/saline group.

The mitochondrial ROS levels in the intact mitochondria were measured by

MitoSOX staining (Figure 5.6B). Mitochondrial ROS level was significantly elevated

in the Ozone/saline group compared with the Air/Saline group (RFU/mg protein:

166.4±9.5 vs. 119.9±8.8, p<0.05). Both Ozone/-24hr and Ozone/+6hr groups

demonstrated significantly reduced mitochondrial ROS levels compared to

Ozone/saline group (RFU/mg protein: 122.8±10.0, p<0.05 and 62.9±10.3, p<0.01

respectively). In addition, the mitochondrial ROS level in Ozone/+6hr group is

significantly lower than the Ozone/-24hr group (p<0.01).

Mitochondrial membrane potential (ΔΨm) of the intact mitochondria was

accessed by JC-1 staining (Figure 5.6C). Exposure to ozone without treatment of

Page 189: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

170

iPSC-MSCs reduced the ΔΨm in comparison to the Air/saline group (fold to

Air/saline: 0.692±.0.057 vs. 1.00±0.041, p<0.01). The treatment of iPSC-MSCs 24

hours prior to the ozone-exposure significantly improved the ΔΨm compared with the

Ozone/saline group (fold to Air/saline: 1.004±.0.076 vs. 0.692±.0.057, p<0.01).

However, the Ozone/+6hr group didn‟t show significant difference as compared with

Ozone/saline group, and the ΔΨm was still significantly lower than the Air/saline

group (fold to Air/saline: 0.778±.0.060 vs. 1.00±0.041, p<0.01).

Page 190: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

171

air air ozone ozone ozone0

1000

2000

3000

4000

5000

**# *

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

Cyto

pla

sm

ic

RO

S

(RF

U/m

g p

rote

in)

air air ozone ozone ozone0

100

200

300

*

**

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

††

§§

Mit

och

on

dri

al R

OS

(RF

U/m

g p

rote

in)

air air ozone ozone ozone0.0

0.5

1.0

1.5

** *

salinesaline MSCs

-24hr

MSCs

+6hr

MSCs

-24hr

††

m (

Fo

ld t

o A

ir/s

ali

ne)

A

B

C

Figure 5. 6 Effects of iPSC-MSCs on ozone-induced ROS and mitochondrial

dysfunction in mouse lungs. Intact mitochondria and cytoplasmic fractions were

isolated from mouse lungs. (A) Cytoplasmic ROS was measured by DCF staining. (B)

Mitochondrial ROS was measured in intact mitochondria by MitoSOX staining. (C)

ΔΨm was determined in intact mitochondria by JC-1 staining. *p<0.05, **p<0.01

compared to air/saline; #p<0.05 compared to air/-24hr, †p<0.05,

††p<0.01 compared to

ozone/saline; §§

p<0.01 compared to ozone/-24hr.

Page 191: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

172

5.3. Discussion

This study demonstrated that intravenously administrated iPSC-MSCs were able

to prevent ozone-induced AHR, airway inflammation, apoptosis, mitochondrial ROS

and reduction in ΔΨm. The therapeutic administration of iPSC-MSCs failed to reverse

ozone-induced AHR, airway inflammation and reduction in ΔΨm, but the

ozone-induced apoptosis and mitochondrial ROS were reversed. Although the data

also showed lack of effects on ozone-induced cytoplasmic ROS and reduction in

proliferation, the findings suggested a protective role of iPSC-MSCs on

ozone-induced lung damage.

Ozone is a potent oxidant. High concentration of environmental ozone was

reported to be associated with worsening of symptoms in patients with COPD and

asthma (18, 74, 84, 173, 197, 239, 257). In mouse models, exposure to ozone is

reported to induce airway oxidative stress, AHR and inflammation (54, 253, 272, 274,

275). Oxidative stress-associated airway inflammation is believed to be a key

component of the pathogenesis of COPD (65). Indeed, repeated exposure to ozone can

lead to airway inflammation accompanied by destruction of alveolar structure, leading

to an emphysema-like phenotype (203, 253). In the present study mice were exposed

to ozone (3 ppm) for a single period of 3 hours. The recruitment of inflammatory cells

to the lungs was robust after 24 hours, as indicated by increased cell counts in the

BAL. Increased levels of inflammatory mediators including MCP-1, eotaxin, IL-6,

IL-5 and MIP-1α were also observed in the BALF. These findings confirmed the

induction of inflammation by ozone in the model. The prophylactic injection of

Page 192: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

173

iPSC-MSCs prevented ozone-induced inflammation, as indicated by the reduction of

both total cell counts in the BAL and levels of inflammatory mediators including

eotaxin, IL-6, IL-5 and MIP-1α, in the BALF. On the other hand, administration of

iPSC-MSCs after ozone exposure did not reverse the recruitment of inflammatory

cells. However, it did reverse the induction of several inflammatory mediators in the

lungs, including eotaxin, IL-6, IL-5 and MIP-1α, which suggested that iPSC-MSCs

still had an anti-inflammatory effect. Failure to reverse inflammatory cell infiltration 6

hours after exposure may be due to a stronger inflammatory response or due to the

shorter length of treatment time in the therapeutic protocol (18 hours) compared to the

prophylactic protocol (48 hours). These findings are in agreement with my results

showing lack of therapeutic effects of iPSC-MSCs on AHR. Although, the

anti-inflammatory role of iPSC-MSCs has been demonstrated by previous studies in

allergic (244) and CS-induced airway inflammation models (as shown in previous

chapters), the ability of iPSC-MSCs to ameliorate AHR has never been reported. AHR

is a major clinical feature of asthma, however it is also observed in some patients with

COPD (291). A previous study in an acute ozone-exposed mouse model demonstrated

that ozone-induced AHR results from the activation of p38 mitogen-activated protein

kinase (147). The exact mechanism of the action of iPSC-MSCs in this model requires

further investigation.

Excessive protease activity associated with oxidative stress and chronic

inflammation is believed to lead to alveolar destruction in patients with COPD (68, 76,

77, 189). However, accumulating evidence suggest that the imbalance between

Page 193: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

174

apoptosis and proliferation may also contribute to the gradual alveolar destruction

(67). In this study increased lung apoptosis and reduced proliferation was observed

after ozone exposure, which is consistent with the imbalance between apoptosis and

proliferation in the chronic CS-exposed model described in Chapter 3. Treatment with

iPSC-MSCs, either before or after ozone exposure, reduced ozone-induced apoptosis,

but the ozone-induced reduction in proliferation was not alleviated. These findings

indicate a possibly higher anti-apoptotic ability of iPSC-MSCs than pro-proliferative

ability in a context of oxidative stress.

Mitochondrial dysfunction has been characterized in the ozone-exposed mouse

model in a previous report (273). Ozone exposure can lead to reduced ATP levels,

mitochondrial complex proteins expression and ΔΨm as well as increased

mitochondrial ROS. The mitochondria-targeted antioxidant, MitoQ, was able to

attenuate mitochondrial dysfunction in the model, as well as the ozone-induced AHR

and inflammation, indicating the AHR and inflammation may be partly driven by

mitochondrial dysfunction (273). Moreover, mitochondrial dysfunction can trigger the

cellular apoptosis (90). Therefore, the effects of iPSC-MSCs on AHR, inflammation

and apoptosis may all relate to its effects on ozone-induced mitochondrial dysfunction

in lung. Indeed, the prophylactic administration of iPSC-MSCs reduced

ozone-induced mitochondrial ROS and reduction in ΔΨm, in line with the effects on

ozone-induced inflammation, AHR and apoptosis. The administration after ozone

exposure only reversed the ozone-induced mitochondrial-ROS level, but not the

reduction of ΔΨm. The findings are in agreement with the results of our co-culture

Page 194: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

175

experiments described in the Chapter 4. There I show that iPSC-MSCs can prevent

and reverse CSM-induced mitochondrial ROS, whereas they can prevent but not

reverse CSM-induced ΔΨm loss.

Despite the reduction in ozone-induced mitochondrial ROS by iPSC-MSCs,

ozone-induced cytoplasmic ROS levels were not modulated by iPSC-MSCs. Such

result may indicate a lack of anti-oxidant effects by iPSC-MSCs in the model. Further

investigation on oxidative stress and anti-oxidant mechanisms in the lung should

better address this issue. On the other hand, the data also suggested that mitochondrial

and cytoplasmic ROS are differentially regulated in cells. Although the mechanisms

by which iPSC-MSCs modulate mitochondrial ROS are unclear, the prevention of

ΔΨm loss and the lack of effect on cytoplasmic ROS suggest that this possibly occurs

through mechanisms that specifically target the mitochondria, such as mitochondrial

transfer.

This study has several limitations and further investigations are required. Firstly,

although the acute exposure of ozone can induce AHR and inflammation, a model

involving multiple ozone exposures would be more relevant to COPD (253).

Investigation of effects of iPSC-MSCs on ozone-induced emphysema, AHR,

inflammation and mitochondrial dysfunction in such model would be an important

experiment to perform. Secondly, the mechanisms by which iPSC-MSCs prevent

ozone-induced mitochondrial dysfunction and the associated inflammation, apoptosis

and AHR remain unclear. Lastly, the effects of iPSC-MSCs on the oxidant and

anti-oxidant balance in the lungs can be further investigated.

Page 195: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

176

In summary, iPSC-MSCs prevented ozone-induced mitochondrial dysfunction as

well as lung inflammation and apoptosis, and AHR. In the therapeutic protocol,

iPSC-MSCs showed no effect on AHR and airway inflammation but reversed

apoptosis and mitochondrial dysfunction in response to ozone. I demonstrate for the

first time the capacity of iPSC-MSCs to prevent mitochondrial dysfunction and AHR

in the lungs, highlighting iPSC-MSCs as promising candidates for the development of

cell-based therapies for COPD.

Page 196: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

177

Chapter 6. General Discussion

Page 197: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

178

6.1. General Discussion

In this study, I demonstrated a superior efficacy of iPSC-MSCs to attenuate

chronic CS-exposure-induced lung emphysema, inflammation and apoptosis in rats

compared to BM-MSCs, which was associated with a higher capacity for

mitochondrial transfer from iPSC-MSCs to lung epithelium. In vitro studies also

demonstrated a higher ability of iPSC-MSCs to transfer mitochondria to and attenuate

CSM-induced ATP depletion in CSM-treated bronchial epithelial cells. In addition,

iPSC-MSCs-CdM demonstrated better efficacy in attenuating CSM-induced apoptosis

and reducing proliferation in bronchial epithelial cells compared to BM-MSCs, which

was partly mediated by SCF. Therefore, iPSC-MSCs demonstrated higher capacity of

both mitochondrial transfer and paracrine release of anti-apoptotic cytokines to

bronchial epithelial cells. I then investigated whether such activities were able to

restore mitochondrial health under oxidative stress. This study was carried out in vitro

first in a CSM-treated model of ASMCs. Direct co-culture with iPSC-MSCs protected

ASMCs from CSM-induced reduction in ΔΨm and elevation of mitochondrial ROS

and apoptosis. However, when direct cell-cell contact was prevented, iPSC-MSCs

only reduced CSM-induced mitochondrial ROS but not reduction of ΔΨm and

increase of apoptosis in ASMCs. Mitochondrial transfer from iPSC-MSCs to ASMCs

was observed in direct co-culture which was enhanced by CSM treatment. In line with

the in vitro findings, in a mouse model of acute ozone exposure, intravenous

administration of iPSC-MSCs protected lung tissue from ozone-induced

mitochondrial dysfunction and apoptosis. Ozone-induced AHR and inflammation was

Page 198: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

179

also reduced by iPSC-MSCs. In summary, I demonstrated that mitochondrial transfer

activity from iPSC-MSCs was able to alleviate oxidative-stress-induced ATP

depletion, mitochondrial dysfunction and apoptosis in target cells. The full capacity of

iPSC-MSCs to achieve these effects may rely on combined mechanisms of both direct

cell-cell contact and release of paracrine factors (Figure 6.1). The capacity of

iPSC-MSCs to transfer their mitochondria to and protect airway cells from oxidative

stress-induced damage is superior to that of BM-MSCs.

The study was initially motivated by three observations regarding current status

of respiratory medicine and stem cells. First of all, current medications for COPD can

only reduce symptoms, exacerbations, and improve health status and exercise

tolerance, and no current medication is able to attenuate the progressive decline of

lung function in COPD (263). As a disease predicted to be the 4th leading cause of

death by 2030 (172) associated with enormous economic and social burden, seeking

new treatments for COPD is an urgent and important task in respiratory medicine.

Secondly, there have been numerous reports on the efficacy of MSCs in animal

models of lung disease (269, 270). Despite a lack of knowledge of the underlining

mechanisms, MSCs from BM, adipose or cord blood have been demonstrated to

attenuate emphysema induced by CS, elastase or VEGF deficiency (94, 112, 151, 222,

269, 270). However, in a clinical trial, intravenously infused BM-MSCs failed to

modulate quality of life and lung function in patients with COPD (271). Although

there are multiple variables that may contribute to the lack of efficacy such as dose

and timing, the results still point out the potential gap between the pre-clinical models

Page 199: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

180

and patients, and the possibility that current findings of therapeutic effects of MSCs in

COPD-related models may not be necessarily translated into treatments for patients

with COPD. Therefore, despite current studies already having demonstrated adequate

efficacy of MSCs in pre-clinical models, the search for improved MSC therapies with

more profound effects in pre-clinical models should not stop. After all, the clinical

trial demonstrated extraordinary tolerance and safety of intravenously infused

BM-MSCs in patients with severe COPD, leaving the door open for future trials.

Thirdly, the in vitro derivation of iPSC-MSCs from iPSCs has provided a new

opportunity to develop improved MSC therapy against COPD. Compared to

BM-MSCs, iPSC-MSCs are more readily available in abundance and possess a higher

ability of proliferation without loss of differentiation potential and telomerase activity

(153). They may hold the hope of overcoming ageing-associated dysfunction of

BM-MSCs by the induction of pluripotency during their generation. As age is an

important risk factor for COPD, iPSC-MSCs may be a good candidate for getting one

own source of MSCs. Moreover, the process of inducing somatic cells into iPSCs and

differentiating iPSCs into iPSC-MSCs provided an opportunity to genetically

manipulate the cells at different stages. Using MSCs as a vehicle for gene therapy

may also be a future option to develop improved therapy against COPD (122). In

summary, iPSC-MSCs may represent a novel and improved candidate for cell therapy

of COPD. From the data presented in Chapter 3, my study demonstrated better

efficacy of iPSC-MSCs than BM-MSCs in attenuating CS-induced lung emphysema,

inflammation and apoptosis, possibly via higher activity of mitochondrial transfer and

Page 200: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

181

paracrine secretion of cytokines such as SCF. My study has demonstrated the potential

of iPSC-MSCs as an improved MSC therapy, hence providing a new option that may

lead to a better treatment for COPD.

One striking observation from the study of CS-exposed rats is the mitochondrial

transfer from iPSC-MSCs to epithelial cells, which led to focusing my attention to the

other theme of the thesis, namely the oxidative stress-induced mitochondrial

dysfunction in pulmonary cells. Developing new treatments for COPD relies on the

understanding of the pathogenesis of COPD. One of the recent advances is the

demonstration of mitochondrial dysfunction associated with COPD. Primary

bronchial epithelial cells from ex-smokers with severe COPD have been shown to

exhibit swelling and fragmented mitochondrial morphology which is similar to

CSM-treated BEAS-2B cells (105). Primary ASMCs from patients with COPD were

shown to have elevated mitochondrial ROS levels, and reduced ΔΨm, complex

protein levels and respiratory function (273). Moreover, the mitochondria-targeted

anti-oxidant MitoQ was able to reduce chronic ozone exposure-induced mitochondrial

dysfunction, inflammation and AHR in mice, highlighting mitochondria as a potential

target for COPD treatment (273). Meanwhile, despite a large number of reports on the

efficacy of MSCs in lung disease models, their mechanisms of action remain largely

unknown. One of the latest advances is the in vivo observation of mitochondrial

transfer from BM-MSCs to pulmonary epithelial cells in an acute lung injury model

(118). In other words, mitochondria are a novel target for COPD treatment as well as

a novel player in the action of MSCs. Therefore, the second half of my study aimed to

Page 201: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

182

investigate whether mitochondria for lung cells are damaged by oxidative stress,

whether this damage can be prevented or reversed by iPSC-MSCs, and whether

iPSC-MSCs exert such protective actions through mitochondrial transfer. The results

demonstrated that CSM led to reduced ATP contents in BEAS-2B cells, and increased

mitochondrial ROS, apoptosis and reduced ΔΨm in primary ASMCs in vitro. Acute

ozone exposure also led to increased mitochondrial ROS and reduced ΔΨm in mouse

lungs in vivo. This oxidative stress-induced mitochondrial dysfunction was alleviated

by iPSC-MSCs either through direct co-culture in vitro, or through intravenous

administration in vivo. Mitochondrial transfer was demonstrated from iPSC-MSCs to

airway epithelium in the CS-exposed model in vivo, and from iPSC-MSCs to

BEAS-2B cells and ASMCs in vitro. CSM enhanced mitochondrial transfer from

iPSC-MSCs to both BEAS-2B cells and ASMCs. The enhanced mitochondria transfer

may be driven by the need for repair, and may be responsible for the alleviation of

CSM-induced mitochondrial dysfunction. Therefore this study demonstrated that

oxidative stress can induce mitochondrial dysfunction of pulmonary cells in vitro and

in vivo, and iPSC-MSCs are able to transfer mitochondria to these damaged cells in

response to the oxidative stress-induced damage and alleviate the mitochondrial

dysfunction.

Improved mitochondrial function may bring other potential benefits to the cell.

Mitochondria serve as the powerhouse of the cells. It has been reported that CS

exposure led to altered lung energy metabolism, with a switch of glucose metabolism

toward pentose phosphate pathway and a reduced supply of substrate to mitochondria.

Page 202: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

183

The transfer of mitochondria into the lung cells may help maintain the energy supply

(4). In addition, as described in Chapter 1.1.4.7, mitochondria play an important role

in regulating ROS generation and cell fate. Oxidant/anti-oxidant imbalance plays an

important role in the pathogenesis of COPD, with the process of oxidative

phosphorylation in mitochondria being a major source of intracellular ROS (149, 165).

Severe mitochondrial damage may lead to depolarization of mitochondria and release

of cytochrome c, and to mitochondrial pathways of cell apoptosis (90). Mitochondrial

damage can also mediate autophagy and senescence, processes that could contribute

to the development of COPD (11).

In general, mitochondrial dysfunction can be targeted using three different

strategies: repair via scavenging excess ROS, reprogramming via regulatory pathways,

and replacement by exogenous functional mitochondria (5). Mitochondria-targeted

anti-oxidant such as MitoQ is a direct way to reduce mitochondrial ROS, which may

subsequently improve other mitochondrial functions such as ΔΨm and respiration.

This has been demonstrated in the chronic ozone-exposed mouse model (273).

Nutrient overload and metabolic changes associated with obesity are common causes

of mitochondrial stress (5) and improvement of mitochondrial function by exercise,

diet and possibly pharmacotherapy such as metformin has been reported (5). Clinical

trials on COPD have demonstrated the potential of metformin to reduce dyspnea and

increase exercise capacity, but more comprehensive studies are awaited to confirm its

efficacy (225). Both repair and reprogramming relies on the recovery of the current

defective mitochondria, which may not be easy for cells of severely-damaged

Page 203: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

184

mitochondria. Also, if mitochondrial DNA has been damaged, such strategies may not

work. In such a situation, replacement of damaged mitochondria with fresh

mitochondria may be a better strategy. Mitochondrial replacement can be achieved by

microinjection, incubation with mitocytoplasts or intact purified mitochondria, and

the cell-cell contact-dependent mitochondrial transfer from MSCs (225). The latter

method used in my thesis, namely mitochondrial transfer, is the only way that does

not involve any disturbance of the cells or involve isolating mitochondria and

injecting the cells but involves a natural method of transferring organelles such as

mitochondria between cells. It provides a perfect approach to deliver exogenous

mitochondria into damaged cells. My study confirmed that mitochondrial transfer can

occur in vivo following its first report in 2012 (118). Moreover, as iPSC-MSCs have a

higher expanding ability than BM-MSCs, the approach we used therefore provided a

more abundant source of mitochondrial donor.

Although mitochondrial transfer was demonstrated both in vivo and in vitro in

this study, it should be noted that the recipient cells were human bronchial epithelial

or ASMCs in the in vitro experiments, while in the in vivo study mitochondria were

transferred from human MSCs into rodent lung cells. The functional compatibility

between the human mitochondria and rodent lung cells was not tested in the current

study. Previous reports have demonstrated the transfer of human mitochondria from

human BM-MSCs to rat cardiomyocytes in vitro (204) and to AEC2s in mouse lungs

(118), but the functional compatibility was not discussed. In a further study, despite

reporting efficient mitochondrial fusion between mouse and human mitochondria

Page 204: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

185

within 4 hours (288), the study demonstrated low compatibility between mouse

nuclear and human mitochondrial genomes. Therefore the functional significance of

the mitochondrial transfer observed in vivo in this thesis is largely unknown.

Two animal models have been used in this thesis to study the rescuing effect of

MSCs. The first one is a rat model exposed to CS for 1 hour/day for 56 days, while

the second one is a mouse model with a single ozone exposure for 3 hours. The two

models are related in some aspects but also different. Both CS and ozone are potent

sources of oxidants. Therefore, both models can be referred to as oxidative

stress-induced lung damage, which makes them distinct from some other

COPD-related models, such as elastase-induced emphysema in which oxidative stress

is not the direct driving force. Cigarette smoking is the major risk factor of COPD,

and chronic CS-exposure is a classic way to induce COPD-like phenotype such as

airspace enlargement and lung inflammation. Although ozone is not a major cause of

COPD, elevated ozone levels have been reported to increase hospital admission due to

COPD (173). Exposure to ozone twice per week for 6 weeks can induce COPD-like

phenotype in mice including emphysema, chronic inflammation, cell infiltration and

steroid resistance (253, 272). Moreover, the ozone-exposed mouse model has been

reported to exhibit a gene expression signature similar to that seen in human COPD

(273). The major difference between the two models in the present study is that the

CS-exposed model is a chronic model with multiple exposures, while the

ozone-exposed model is an acute model with a single exposure period. The acute

exposure of ozone cannot induce airspace enlargement, but infiltration of neutrophils

Page 205: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

186

and other inflammatory cells was demonstrated and, as such, could be taken as a

model mimicking an exacerbation of COPD which is a frequent manifestation of

advanced COPD. In general, acute exposure indicates that the animals were

responding to the stimulation based on a normal physiological condition, whereas in

chronic models, along the progression of the damage, the animals respond to the

stimulation based on a damaged, abnormal condition. While the efficacy of

iPSC-MSCs in the chronic CS-exposed model specifically defines iPSC-MSCs as a

promising candidate for MSC therapy of COPD, the efficacy of iPSC-MSCs in the

acute ozone-induced model may provide a more general implication on an acute

oxidative stress insult such as during an exacerbation. Although the acute

ozone-exposed model did not exhibit airspace enlargement, this model has been used

to investigate some parameters that have not been measured in the CS-exposed model,

including AHR, cytosolic ROS, mitochondrial ROS and ΔΨm. Nevertheless, both

models in the study have demonstrated inflammation and apoptosis in the lung. Using

the two different models, I demonstrated the capacity of iPSC-MSCs to alleviate

oxidative stress-induced lung damage in both acute and chronic setups.

The in vitro study involved two different airway cell types. Although both cell

types were treated with CSM, it should be noted that the treatments were optimized

based on different parameters. In the study on bronchial epithelial BEAS-2B cells, the

concentration (2%) and length of treatment (24 hours) was adapted from a previous

study, with an induction of IL-8 release without reduction of viability. As

demonstrated in Chapter 3, such condition was sufficient to induce apoptosis and

Page 206: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

187

reduce ATP content. In the study of primary ASMCs, the concentration of CS (10% or

25%) and length of treatment (4 hours) was determined by the induction of

mitochondrial dysfunction as measured by mitochondrial ROS and ΔΨm. The CSM

stimulation was more acute and strong in the ASMCs study, leading to a more acute

induction of apoptosis. Such difference may explain why the released cytokines from

iPSC-MSCs (iPSC-MSCs-CdM) were able to attenuate CSM-induced apoptosis in

BEAS-2B cells but not in ASMCs in the study. On the other hand, enhanced

mitochondrial transfer has been detected from iPSC-MSCs to both BEAS-2B cells

and ASMCs under the corresponding CSM treatments. Mitochondrial transfer and

paracrine effects may serve as two different mechanisms of iPSC-MSCs‟ action. By

using apoptosis as a functional output, it is clear that the relative contribution of

mitochondrial transfer and paracrine effects varies among different target cell types

and treatments. In the 24 hour-treated BEAS-2B cells, paracrine effects from

iPSC-MSCs were sufficient to reduce apoptosis. By contrast, in the 4 hour-treated

ASMCs, the anti-apoptotic efficacy fully relied on cell-cell contact that facilitated the

mitochondrial transfer which is a more direct mechanism to rescue mitochondrial

dysfunction. Such differences demonstrated the complexity and the dynamics of

mechanisms of action of iPSC-MSCs.

The in vivo and in vitro data from different chapters complement each other very

well in this study. The mitochondrial transfer from iPSC-MSCs to damaged cells in

vivo in the CS-exposed model was also observed in vitro for both BEAS-2B cells and

ASMCs. The anti-apoptotic effect of iPSC-MSCs in both CS- and ozone-exposed

Page 207: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

188

animal models was also demonstrated in both BEAS-2B cells and ASMCs. The

attenuation of mitochondrial dysfunction by iPSC-MSCs was demonstrated in vivo in

ozone-exposed mice as well as in vitro in ASMCs. Moreover, both studies

demonstrated time-dependent effects. In the CSM-treated ASMCs, iPSC-MSCs

prevented but not reversed the CSM-induced mitochondrial dysfunction. Similarly, in

the ozone-exposed mice, iPSC-MSCs prevented but not reversed the ozone-induced

inflammation, AHR and reduction of ΔΨm in the lung. This may be explained by the

fact that ASMCs or mice were treated with iPSC-MSCs for more time in the

preventive protocols, or that the oxidative stress-induced damage has progressed to a

stage that cannot be reversed by iPSC-MSCs. Interestingly, in the ozone-exposed

mice, although the preventive treatment did not attenuate ozone-induced reduction of

ΔΨm, it attenuated ozone-induced mitochondrial ROS in the lung. This is very similar

to the effects of iPSC-MSCs-CdM or iPSC-MSCs in trans-well inserts in vitro, which

also demonstrated reduction of CSM-induced mitochondrial ROS but no change in

ΔΨm in ASMCs. In theory, excess mitochondrial ROS can lead to defective election

transport chain which is responsible for proton transfer across the inner mitochondrial

membrane and defective permeability transition pore which controls the permeability

of inner membrane (170). Both effects lead to ΔΨm reduction. The reduction of

mitochondrial ROS without effects on ΔΨm in the study may be because the reduced

mitochondrial ROS levels were still high enough to induce a reduction in ΔΨm, or

because there are other mechanisms regulating ΔΨm which were less targeted by

iPSC-MSCs, such as cytosolic ROS levels.

Page 208: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

189

The most direct clinical implication of the study is that intravenously-infused

iPSC-MSCs may exert a stronger effect in patients with COPD compared to

BM-MSCs which have not been shown to be effective in the clinical trials. However,

it is of concern that the genetic reprogramming may also change the safety profile of

iPSC-MSCs compared to BM-MSCs. In a more general perspective, iPSC-MSCs may

be effective in other oxidative stress-mediated airway diseases such as asthma. In

addition, the higher mitochondria transfer capacity and abundant resource also

suggests that iPSC-MSCs may serve as a useful mitochondrial donor to target

mitochondrial dysfunction in various diseases.

There are several limitations of the work in my thesis. Firstly, due to time

limitations, not all parameters were investigated in all models. As mitochondrial

dysfunction was not the initial focus of the study, mitochondrial ROS and ΔΨm were

not measured in the CS-exposed rat model and CSM-treated BEAS-2B cells.

Secondly, although inflammation was investigated in the in vivo models, there was no

direct measurement of inflammation in the in vitro models. A common inflammatory

measurement used in cell culture studies is measurement of released inflammatory

mediators. However, as both cell types in the co-culture are from human, the secretory

function of one particular cell type is difficult to ascertain. Thirdly, there are other

potential mechanisms involved in iPSC-MSCs action which may also contribute to

the efficacy observed in the study. For example, formation of TNTs may also facilitate

transfer of other organelles such as lysosomes. iPSC-MSCs may also secrete

protective microvesicles to lung cells. Lastly, mitochondrial ROS and ΔΨm may not

Page 209: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

190

represent all the functions of mitochondria. A measurement of mitochondrial

respiration profile would have been ideal to address the issue.

6.2. Conclusion

I demonstrated the superior effects of iPSC-MSCs in attenuating CS-induced

emphysema, inflammation and apoptosis in rat lung compared to BM-MSCs, which

was attributed to higher mitochondrial transfer and paracrine release of cytokines

including SCF. iPSC-MSCs can also alleviate oxidative stress-induced AHR,

inflammation, mitochondrial dysfunction and apoptosis in mouse lungs.

Mitochondrial transfer from iPSC-MSCs was able to alleviate

oxidative-stress-induced ATP depletion, mitochondrial dysfunction and apoptosis in

target cells. The full capacity of iPSC-MSCs to achieve this efficacy may rely on

combined mechanisms of both direct cell-cell contact and release of paracrine factors.

These findings support iPSC-MSCs as a promising candidate for the development

of MSCs-based therapy against COPD, which may be driven by multiple

mechanisms.

Page 210: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

191

Figure 6. 1 Summary of the effects of iPSC-MSCs on oxidative stress-induced

lung cell damage. The full capacity of iPSC-MSCs to alleviate oxidative

stress-induced mitochondrial dysfunction and apoptosis may rely on combined

mechanisms of both direct cell-cell contact and release of paracrine factors. SCF: stem

cell factor; Mito-ROS: mitochondrial ROS; iPSC-MSCs: induced pluripotent stem

cell-derived mesenchymal stem cells; ASMCs: airway smooth muscle cells; CS:

cigarette smoke; AHR: airway hyper-responsiveness.

Page 211: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

192

6.3. Future Studies

Several future investigations can be extended from this study.

In this study, the effects of iPSC-MSCs on mitochondrial dysfunction in animal

models with COPD-like phenotypes have not been demonstrated. It would be

important to further investigate effects of iPSC-MSCs on mitochondria of the lung in

a chronic CS-/ozone-exposed models focusing particularly on the development of

emphysema.

In the study I used cytochalasin B or trans-well membrane to block the formation

of TNTs between iPSC-MSCs and target cells. However, such methods are not

selective blockers of mitochondrial transfer. A recent study has demonstrated that

mitochondrial transfer is mediated by Miro-1 (6). Knock-down or over-expression of

Miro-1 in BM-MSCs leads to reduced or enhanced mitochondrial transfer activity

respectively. This would provide a more selective approach to block mitochondrial

transfer without interruption of TNT formation. Regarding my study, the following

investigations could be proposed. First, whether Miro-1 is also responsible for the

mitochondrial transfer in iPSC-MSCs can be examined. Determination of efficacy of

iPSC-MSCs and mitochondrial transfer after Miro-1 knock-down and over-expression

would address the question. Second, how does CSM enhance mitochondrial transfer?

One hypothesis is that CSM treatment can directly regulate capacity of mitochondrial

transfer of iPSC-MSCs. This can be investigated by measurement of Miro-1

expression in response to CSM and identification of the possible pathways involved,

such as mitogen-activated protein kinase pathways, using specific inhibitors of these

Page 212: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

193

pathways. On the other hand, the enhanced mitochondrial transfer may be a response

to signals from injured target cells. How the recipient cells regulate the mitochondrial

transfer will also require further investigation.

Another important follow-up study is to determine whether iPSC-MSCs can

improve mitochondrial function of primary ASMCs from patients with COPD.

ASMCs from patients with COPD already demonstrated mitochondrial dysfunction

compared to normal controls. How this would change the mitochondrial transfer

between the ASMCs and iPSC-MSCs should also be investigated.

Page 213: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

194

References

1. Aaron SD, Angel JB, Lunau M, Wright K, Fex C, Le Saux N, and Dales RE.

Granulocyte inflammatory markers and airway infection during acute exacerbation of

chronic obstructive pulmonary disease. Am J Respir Crit Care Med 163: 349-355,

2001.

2. Abreu SC, Antunes MA, Pelosi P, Morales MM, and Rocco PR. Mechanisms

of cellular therapy in respiratory diseases. Intensive Care Med 37: 1421-1431, 2011.

3. Adeloye D, Chua S, Lee C, Basquill C, Papana A, Theodoratou E, Nair H,

Gasevic D, Sridhar D, Campbell H, Chan KY, Sheikh A, et al. Global and regional

estimates of COPD prevalence: Systematic review and meta-analysis. J Glob Health 5:

020415, 2015.

4. Agarwal AR, Zhao L, Sancheti H, Sundar IK, Rahman I, and Cadenas E.

Short-term cigarette smoke exposure induces reversible changes in energy

metabolism and cellular redox status independent of inflammatory responses in mouse

lungs. Am J Physiol Lung Cell Mol Physiol 303: L889-898, 2012.

5. Agrawal A, and Mabalirajan U. Rejuvenating cellular respiration for

optimizing respiratory function: targeting mitochondria. Am J Physiol Lung Cell Mol

Physiol 310: L103-113, 2016.

6. Ahmad T, Mukherjee S, Pattnaik B, Kumar M, Singh S, Kumar M, Rehman

R, Tiwari BK, Jha KA, Barhanpurkar AP, Wani MR, Roy SS, et al. Miro1

regulates intercellular mitochondrial transport & enhances mesenchymal stem cell

rescue efficacy. EMBO J 33: 994-1010, 2014.

7. Al Heialy S, Zeroual M, Farahnak S, McGovern T, Risse PA, Novali M,

Lauzon AM, Roman HN, and Martin JG. Nanotubes connect CD4+ T cells to

airway smooth muscle cells: novel mechanism of T cell survival. J Immunol 194:

5626-5634, 2015.

8. Ankrum JA, Ong JF, and Karp JM. Mesenchymal stem cells: immune evasive,

not immune privileged. Nat Biotechnol 32: 252-260, 2014.

9. Aon MA, Cortassa S, and O'Rourke B. Redox-optimized ROS balance: a

Page 214: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

195

unifying hypothesis. Biochim Biophys Acta 1797: 865-877, 2010.

10. Aravamudan B, Kiel A, Freeman M, Delmotte P, Thompson M, Vassallo R,

Sieck GC, Pabelick CM, and Prakash YS. Cigarette smoke-induced mitochondrial

fragmentation and dysfunction in human airway smooth muscle. Am J Physiol Lung

Cell Mol Physiol 306: L840-854, 2014.

11. Aravamudan B, Thompson MA, Pabelick CM, and Prakash YS.

Mitochondria in lung diseases. Expert Rev Respir Med 7: 631-646, 2013.

12. Ashcroft T, Simpson JM, and Timbrell V. Simple method of estimating

severity of pulmonary fibrosis on a numerical scale. J Clin Pathol 41: 467-470, 1988.

13. Austefjord MW, Gerdes HH, and Wang X. Tunneling nanotubes: Diversity in

morphology and structure. Commun Integr Biol 7: e27934, 2014.

14. Badri L, Murray S, Liu LX, Walker NM, Flint A, Wadhwa A, Chan KM,

Toews GB, Pinsky DJ, Martinez FJ, and Lama VN. Mesenchymal stromal cells in

bronchoalveolar lavage as predictors of bronchiolitis obliterans syndrome. Am J

Respir Crit Care Med 183: 1062-1070, 2011.

15. Baer PC, and Geiger H. Adipose-derived mesenchymal stromal/stem cells:

tissue localization, characterization, and heterogeneity. Stem Cells Int 2012: 812693,

2012.

16. Baglole CJ, Bushinsky SM, Garcia TM, Kode A, Rahman I, Sime PJ, and

Phipps RP. Differential induction of apoptosis by cigarette smoke extract in primary

human lung fibroblast strains: implications for emphysema. Am J Physiol Lung Cell

Mol Physiol 291: L19-29, 2006.

17. Balmes J, Becklake M, Blanc P, Henneberger P, Kreiss K, Mapp C, Milton D,

Schwartz D, Toren K, Viegi G, Environmental, and Occupational Health

Assembly ATS. American Thoracic Society Statement: Occupational contribution to

the burden of airway disease. Am J Respir Crit Care Med 167: 787-797, 2003.

18. Balmes JR, Aris RM, Chen LL, Scannell C, Tager IB, Finkbeiner W,

Christian D, Kelly T, Hearne PQ, Ferrando R, and Welch B. Effects of ozone on

normal and potentially sensitive human subjects. Part I: Airway inflammation and

responsiveness to ozone in normal and asthmatic subjects. Res Rep Health Eff Inst

Page 215: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

196

1-37; discussion 81-99, 1997.

19. Banerjee A, and Panettieri R, Jr. Vitamin D modulates airway smooth muscle

function in COPD. Curr Opin Pharmacol 12: 266-274, 2012.

20. Baraldo S, Turato G, Badin C, Bazzan E, Beghe B, Zuin R, Calabrese F,

Casoni G, Maestrelli P, Papi A, Fabbri LM, and Saetta M. Neutrophilic infiltration

within the airway smooth muscle in patients with COPD. Thorax 59: 308-312, 2004.

21. Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, Stripp BR,

Randell SH, Noble PW, and Hogan BL. Type 2 alveolar cells are stem cells in adult

lung. J Clin Invest 123: 3025-3036, 2013.

22. Barnes PJ. Chronic obstructive pulmonary disease. N Engl J Med 343: 269-280,

2000.

23. Barnes PJ. Chronic Obstructive Pulmonary Disease: A Growing but Neglected

Global Epidemic. PLoS Med 4: e112, 2007.

24. Barnes PJ, and Adcock IM. Glucocorticoid resistance in inflammatory diseases.

Lancet 373: 1905-1917, 2009.

25. Barnes PJ, Shapiro SD, and Pauwels RA. Chronic obstructive pulmonary

disease: molecular and cellular mechanisms. Eur Respir J 22: 672-688, 2003.

26. Barry M, and Bleackley RC. Cytotoxic T lymphocytes: all roads lead to death.

Nat Rev Immunol 2: 401-409, 2002.

27. Bartoli ML, Novelli F, Costa F, Malagrino L, Melosini L, Bacci E, Cianchetti

S, Dente FL, Di Franco A, Vagaggini B, and Paggiaro PL. Malondialdehyde in

exhaled breath condensate as a marker of oxidative stress in different pulmonary

diseases. Mediators Inflamm 2011: 891752, 2011.

28. Beers MF, and Morrisey EE. The three R's of lung health and disease: repair,

remodeling, and regeneration. J Clin Invest 121: 2065-2073, 2011.

29. Berenson CS, Garlipp MA, Grove LJ, Maloney J, and Sethi S. Impaired

phagocytosis of nontypeable Haemophilus influenzae by human alveolar

macrophages in chronic obstructive pulmonary disease. J Infect Dis 194: 1375-1384,

2006.

30. Bernas T, and Dobrucki J. Mitochondrial and nonmitochondrial reduction of

Page 216: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

197

MTT: interaction of MTT with TMRE, JC-1, and NAO mitochondrial fluorescent

probes. Cytometry 47: 236-242, 2002.

31. Berridge MV, Herst PM, and Tan AS. Tetrazolium dyes as tools in cell biology:

new insights into their cellular reduction. Biotechnol Annu Rev 11: 127-152, 2005.

32. Berridge MV, and Tan AS. Characterization of the cellular reduction of

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT): subcellular

localization, substrate dependence, and involvement of mitochondrial electron

transport in MTT reduction. Arch Biochem Biophys 303: 474-482, 1993.

33. Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG, Simmons PJ, and Wang

CY. The meaning, the sense and the significance: translating the science of

mesenchymal stem cells into medicine. Nat Med 19: 35-42, 2013.

34. Blanpain C, and Fuchs E. Epidermal homeostasis: a balancing act of stem cells

in the skin. Nat Rev Mol Cell Biol 10: 207-217, 2009.

35. Blume-Jensen P, Janknecht R, and Hunter T. The kit receptor promotes cell

survival via activation of PI 3-kinase and subsequent Akt-mediated phosphorylation

of Bad on Ser136. Curr Biol 8: 779-782, 1998.

36. Boman C, Forsberg B, and Sandstrom T. Shedding new light on wood smoke:

a risk factor for respiratory health. Eur Respir J 27: 446-447, 2006.

37. Bosken CH, Wiggs BR, Pare PD, and Hogg JC. Small airway dimensions in

smokers with obstruction to airflow. Am Rev Respir Dis 142: 563-570, 1990.

38. Buchman AS, Boyle PA, Wilson RS, Gu L, Bienias JL, and Bennett DA.

Pulmonary function, muscle strength and mortality in old age. Mech Ageing Dev 129:

625-631, 2008.

39. Buist AS, McBurnie MA, Vollmer WM, Gillespie S, Burney P, Mannino DM,

Menezes AM, Sullivan SD, Lee TA, Weiss KB, Jensen RL, Marks GB, et al.

International variation in the prevalence of COPD (the BOLD Study): a

population-based prevalence study. Lancet 370: 741-750, 2007.

40. Bukoreshtliev NV, Wang X, Hodneland E, Gurke S, Barroso JF, and Gerdes

HH. Selective block of tunneling nanotube (TNT) formation inhibits intercellular

organelle transfer between PC12 cells. FEBS Lett 583: 1481-1488, 2009.

Page 217: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

198

41. Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, Wen Y, Rapp JA,

and Kessler J. Clinical applications of blood-derived and marrow-derived stem cells

for nonmalignant diseases. JAMA 299: 925-936, 2008.

42. Butler JP, Loring SH, Patz S, Tsuda A, Yablonskiy DA, and Mentzer SJ.

Evidence for adult lung growth in humans. N Engl J Med 367: 244-247, 2012.

43. Cadenas E, and Davies KJ. Mitochondrial free radical generation, oxidative

stress, and aging. Free Radic Biol Med 29: 222-230, 2000.

44. Calabrese F, Giacometti C, Beghe B, Rea F, Loy M, Zuin R, Marulli G,

Baraldo S, Saetta M, and Valente M. Marked alveolar apoptosis/proliferation

imbalance in end-stage emphysema. Respir Res 6: 14, 2005.

45. Camougrand N, and Rigoulet M. Aging and oxidative stress: studies of some

genes involved both in aging and in response to oxidative stress. Respir Physiol 128:

393-401, 2001.

46. Carp H, and Janoff A. Possible mechanisms of emphysema in smokers. In vitro

suppression of serum elastase-inhibitory capacity by fresh cigarette smoke and its

prevention by antioxidants. Am Rev Respir Dis 118: 617-621, 1978.

47. Celedon JC, Lange C, Raby BA, Litonjua AA, Palmer LJ, DeMeo DL, Reilly

JJ, Kwiatkowski DJ, Chapman HA, Laird N, Sylvia JS, Hernandez M, et al. The

transforming growth factor-beta1 (TGFB1) gene is associated with chronic

obstructive pulmonary disease (COPD). Hum Mol Genet 13: 1649-1656, 2004.

48. Celli BR, Halbert RJ, Nordyke RJ, and Schau B. Airway obstruction in never

smokers: results from the Third National Health and Nutrition Examination Survey.

Am J Med 118: 1364-1372, 2005.

49. Centers for Disease C, and Prevention. Chronic obstructive pulmonary disease

among adults--United States, 2011. MMWR Morb Mortal Wkly Rep 61: 938-943,

2012.

50. Chambers DC, Enever D, Ilic N, Sparks L, Whitelaw K, Ayres J, Yerkovich

ST, Khalil D, Atkinson KM, and Hopkins PM. A phase 1b study of

placenta-derived mesenchymal stromal cells in patients with idiopathic pulmonary

fibrosis. Respirology 19: 1013-1018, 2014.

Page 218: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

199

51. Chan KH, Chan SC, Yeung SC, Man RY, Ip MS, and Mak JC. Inhibitory

effect of Chinese green tea on cigarette smoke-induced up-regulation of airway

neutrophil elastase and matrix metalloproteinase-12 via antioxidant activity. Free

Radic Res 46: 1123-1129, 2012.

52. Chan KH, Ho SP, Yeung SC, So WH, Cho CH, Koo MW, Lam WK, Ip MS,

Man RY, and Mak JC. Chinese green tea ameliorates lung injury in cigarette

smoke-exposed rats. Respir Med 103: 1746-1754, 2009.

53. Chelikani P, Fita I, and Loewen PC. Diversity of structures and properties

among catalases. Cell Mol Life Sci 61: 192-208, 2004.

54. Cho HY, Zhang LY, and Kleeberger SR. Ozone-induced lung inflammation and

hyperreactivity are mediated via tumor necrosis factor-alpha receptors. Am J Physiol

Lung Cell Mol Physiol 280: L537-546, 2001.

55. Cho YM, Kim JH, Kim M, Park SJ, Koh SH, Ahn HS, Kang GH, Lee JB,

Park KS, and Lee HK. Mesenchymal stem cells transfer mitochondria to the cells

with virtually no mitochondrial function but not with pathogenic mtDNA mutations.

PLoS One 7: e32778, 2012.

56. Chung KF. The role of airway smooth muscle in the pathogenesis of airway wall

remodeling in chronic obstructive pulmonary disease. Proc Am Thorac Soc 2:

347-354; discussion 371-342, 2005.

57. Chung KF, and Adcock IM. Multifaceted mechanisms in COPD: inflammation,

immunity, and tissue repair and destruction. Eur Respir J 31: 1334-1356, 2008.

58. Church DF, and Pryor WA. Free-radical chemistry of cigarette smoke and its

toxicological implications. Environ Health Perspect 64: 111-126, 1985.

59. Cosio MG, Hale KA, and Niewoehner DE. Morphologic and morphometric

effects of prolonged cigarette smoking on the small airways. Am Rev Respir Dis 122:

265-221, 1980.

60. Cosio MG, Saetta M, and Agusti A. Immunologic aspects of chronic obstructive

pulmonary disease. N Engl J Med 360: 2445-2454, 2009.

61. Crestani B. Bronchoalveolar lavage brings mesenchymal stem cells to the light.

Am J Respir Crit Care Med 185: 7-8, 2012.

Page 219: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

200

62. D'Hulst A I, Vermaelen KY, Brusselle GG, Joos GF, and Pauwels RA. Time

course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J 26:

204-213, 2005.

63. D'Souza N, Rossignoli F, Golinelli G, Grisendi G, Spano C, Candini O,

Osturu S, Catani F, Paolucci P, Horwitz EM, and Dominici M. Mesenchymal

stem/stromal cells as a delivery platform in cell and gene therapies. BMC Med 13: 186,

2015.

64. de Boer WI, Sont JK, van Schadewijk A, Stolk J, van Krieken JH, and

Hiemstra PS. Monocyte chemoattractant protein 1, interleukin 8, and chronic airways

inflammation in COPD. J Pathol 190: 619-626, 2000.

65. Decramer M, Janssens W, and Miravitlles M. Chronic obstructive pulmonary

disease. Lancet 379: 1341-1351, 2012.

66. Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D, Verleden GM,

Vermassen FE, Joos GF, and Brusselle GG. Accumulation of dendritic cells and

increased CCL20 levels in the airways of patients with chronic obstructive pulmonary

disease. Am J Respir Crit Care Med 175: 998-1005, 2007.

67. Demedts IK, Demoor T, Bracke KR, Joos GF, and Brusselle GG. Role of

apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res 7: 53,

2006.

68. Demedts IK, Morel-Montero A, Lebecque S, Pacheco Y, Cataldo D, Joos GF,

Pauwels RA, and Brusselle GG. Elevated MMP-12 protein levels in induced sputum

from patients with COPD. Thorax 61: 196-201, 2006.

69. Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, and Prockop DJ.

Propagation and senescence of human marrow stromal cells in culture: a simple

colony-forming assay identifies samples with the greatest potential to propagate and

differentiate. Br J Haematol 107: 275-281, 1999.

70. Ding X, and Kaminsky LS. Human extrahepatic cytochromes P450: function in

xenobiotic metabolism and tissue-selective chemical toxicity in the respiratory and

gastrointestinal tracts. Annu Rev Pharmacol Toxicol 43: 149-173, 2003.

71. Doll H, and Miravitlles M. Health-related QOL in acute exacerbations of

Page 220: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

201

chronic bronchitis and chronic obstructive pulmonary disease: a review of the

literature. Pharmacoeconomics 23: 345-363, 2005.

72. Donnelly LE, and Barnes PJ. Defective phagocytosis in airways disease. Chest

141: 1055-1062, 2012.

73. Eisner MD, Balmes J, Katz PP, Trupin L, Yelin EH, and Blanc PD. Lifetime

environmental tobacco smoke exposure and the risk of chronic obstructive pulmonary

disease. Environ Health 4: 7, 2005.

74. Eiswerth ME, Douglass Shaw W, and Yen ST. Impacts of ozone on the

activities of asthmatics: revisiting the data. J Environ Manage 77: 56-63, 2005.

75. Ezzati M. Indoor air pollution and health in developing countries. Lancet 366:

104-106, 2005.

76. Finlay GA, O'Driscoll LR, Russell KJ, D'Arcy EM, Masterson JB,

FitzGerald MX, and O'Connor CM. Matrix metalloproteinase expression and

production by alveolar macrophages in emphysema. Am J Respir Crit Care Med 156:

240-247, 1997.

77. Finlay GA, Russell KJ, McMahon KJ, D'Arcy E M, Masterson JB,

FitzGerald MX, and O'Connor CM. Elevated levels of matrix metalloproteinases in

bronchoalveolar lavage fluid of emphysematous patients. Thorax 52: 502-506, 1997.

78. Fitts DA. Ethics and animal numbers: informal analyses, uncertain sample sizes,

inefficient replications, and type I errors. J Am Assoc Lab Anim Sci 50: 445-453, 2011.

79. Fletcher C, and Peto R. The natural history of chronic airflow obstruction. Br

Med J 1: 1645-1648, 1977.

80. Francois M, and Galipeau J. New insights on translational development of

mesenchymal stromal cells for suppressor therapy. J Cell Physiol 227: 3535-3538,

2012.

81. Francois M, Romieu-Mourez R, Li M, and Galipeau J. Human MSC

suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and

bystander M2 macrophage differentiation. Mol Ther 20: 187-195, 2012.

82. Fromer L, and Cooper CB. A review of the GOLD guidelines for the diagnosis

and treatment of patients with COPD. Int J Clin Pract 62: 1219-1236, 2008.

Page 221: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

202

83. Fulda S, Galluzzi L, and Kroemer G. Targeting mitochondria for cancer therapy.

Nat Rev Drug Discov 9: 447-464, 2010.

84. Galan I, Tobias A, Banegas JR, and Aranguez E. Short-term effects of air

pollution on daily asthma emergency room admissions. Eur Respir J 22: 802-808,

2003.

85. Garcia O, Carraro G, Navarro S, Bertoncello I, McQualter J, Driscoll B,

Jesudason E, and Warburton D. Cell-based therapies for lung disease. Br Med Bull

101: 147-161, 2012.

86. Ghaedi M, Calle EA, Mendez JJ, Gard AL, Balestrini J, Booth A, Bove PF,

Gui L, White ES, and Niklason LE. Human iPS cell-derived alveolar epithelium

repopulates lung extracellular matrix. J Clin Invest 123: 4950-4962, 2013.

87. Giuliani M, Oudrhiri N, Noman ZM, Vernochet A, Chouaib S, Azzarone B,

Durrbach A, and Bennaceur-Griscelli A. Human mesenchymal stem cells derived

from induced pluripotent stem cells down-regulate NK-cell cytolytic machinery.

Blood 118: 3254-3262, 2011.

88. Gould NS, Min E, Gauthier S, Chu HW, Martin R, and Day BJ. Aging

adversely affects the cigarette smoke-induced glutathione adaptive response in the

lung. Am J Respir Crit Care Med 182: 1114-1122, 2010.

89. Granados-Castro LF, Rodriguez-Rangel DS, Montano M, Ramos C, and

Pedraza-Chaverri J. Wood smoke exposure induces a decrease in respiration

parameters and in the activity of respiratory complexes I and IV in lung mitochondria

from guinea pigs. Environ Toxicol 30: 461-471, 2015.

90. Green DR, and Reed JC. Mitochondria and apoptosis. Science 281: 1309-1312,

1998.

91. Green MD, Chen A, Nostro MC, d'Souza SL, Schaniel C, Lemischka IR,

Gouon-Evans V, Keller G, and Snoeck HW. Generation of anterior foregut

endoderm from human embryonic and induced pluripotent stem cells. Nat Biotechnol

29: 267-272, 2011.

92. Gu D, Wu X, Reynolds K, Duan X, Xin X, Reynolds RF, Whelton PK, and

He J. Cigarette smoking and exposure to environmental tobacco smoke in China: the

Page 222: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

203

international collaborative study of cardiovascular disease in Asia. Am J Public

Health 94: 1972-1976, 2004.

93. Gu W, Song L, Li XM, Wang D, Guo XJ, and Xu WG. Mesenchymal stem

cells alleviate airway inflammation and emphysema in COPD through

down-regulation of cyclooxygenase-2 via p38 and ERK MAPK pathways. Sci Rep 5:

8733, 2015.

94. Guan XJ, Song L, Han FF, Cui ZL, Chen X, Guo XJ, and Xu WG.

Mesenchymal stem cells protect cigarette smoke-damaged lung and pulmonary

function partly via VEGF-VEGF receptors. J Cell Biochem 114: 323-335, 2013.

95. Hajnoczky G, Csordas G, and Yi M. Old players in a new role:

mitochondria-associated membranes, VDAC, and ryanodine receptors as contributors

to calcium signal propagation from endoplasmic reticulum to the mitochondria. Cell

Calcium 32: 363-377, 2002.

96. Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, and Mannino DM.

Global burden of COPD: systematic review and meta-analysis. Eur Respir J 28:

523-532, 2006.

97. Han D, Canali R, Rettori D, and Kaplowitz N. Effect of glutathione depletion

on sites and topology of superoxide and hydrogen peroxide production in

mitochondria. Mol Pharmacol 64: 1136-1144, 2003.

98. Hardaker EL, Bacon AM, Carlson K, Roshak AK, Foley JJ, Schmidt DB,

Buckley PT, Comegys M, Panettieri RA, Jr., Sarau HM, and Belmonte KE.

Regulation of TNF-alpha- and IFN-gamma-induced CXCL10 expression:

participation of the airway smooth muscle in the pulmonary inflammatory response in

chronic obstructive pulmonary disease. FASEB J 18: 191-193, 2004.

99. Harman D. Free radical theory of aging: an update: increasing the functional life

span. Ann N Y Acad Sci 1067: 10-21, 2006.

100. Heeschen C, Lehmann R, Honold J, Assmus B, Aicher A, Walter DH,

Martin H, Zeiher AM, and Dimmeler S. Profoundly reduced neovascularization

capacity of bone marrow mononuclear cells derived from patients with chronic

ischemic heart disease. Circulation 109: 1615-1622, 2004.

Page 223: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

204

101. Heguy A, O'Connor TP, Luettich K, Worgall S, Cieciuch A, Harvey BG,

Hackett NR, and Crystal RG. Gene expression profiling of human alveolar

macrophages of phenotypically normal smokers and nonsmokers reveals a previously

unrecognized subset of genes modulated by cigarette smoking. J Mol Med (Berl) 84:

318-328, 2006.

102. Heijink IH, Brandenburg SM, Postma DS, and van Oosterhout AJ. Cigarette

smoke impairs airway epithelial barrier function and cell-cell contact recovery. Eur

Respir J 39: 419-428, 2012.

103. Hodge S, Hodge G, Holmes M, and Reynolds PN. Increased airway epithelial

and T-cell apoptosis in COPD remains despite smoking cessation. Eur Respir J 25:

447-454, 2005.

104. Hoffman AM, Paxson JA, Mazan MR, Davis AM, Tyagi S, Murthy S, and

Ingenito EP. Lung-derived mesenchymal stromal cell post-transplantation survival,

persistence, paracrine expression, and repair of elastase-injured lung. Stem Cells Dev

20: 1779-1792, 2011.

105. Hoffmann RF, Zarrintan S, Brandenburg SM, Kol A, de Bruin HG, Jafari S,

Dijk F, Kalicharan D, Kelders M, Gosker HR, Ten Hacken NH, van der Want JJ,

et al. Prolonged cigarette smoke exposure alters mitochondrial structure and function

in airway epithelial cells. Respir Res 14: 97, 2013.

106. Hogan BL, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CC,

Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, et al. Repair and

regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung

stem cell function. Cell Stem Cell 15: 123-138, 2014.

107. Hogg JC. Pathophysiology of airflow limitation in chronic obstructive

pulmonary disease. Lancet 364: 709-721, 2004.

108. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack

RM, Rogers RM, Sciurba FC, Coxson HO, and Pare PD. The nature of

small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350:

2645-2653, 2004.

109. Holloway RA, and Donnelly LE. Immunopathogenesis of chronic obstructive

Page 224: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

205

pulmonary disease. Curr Opin Pulm Med 19: 95-102, 2013.

110. Houben JM, Mercken EM, Ketelslegers HB, Bast A, Wouters EF, Hageman

GJ, and Schols AM. Telomere shortening in chronic obstructive pulmonary disease.

Respir Med 103: 230-236, 2009.

111. Huang SX, Green MD, de Carvalho AT, Mumau M, Chen YW, D'Souza SL,

and Snoeck HW. The in vitro generation of lung and airway progenitor cells from

human pluripotent stem cells. Nat Protoc 10: 413-425, 2015.

112. Huh JW, Kim SY, Lee JH, Lee JS, Van Ta Q, Kim M, Oh YM, Lee YS, and

Lee SD. Bone marrow cells repair cigarette smoke-induced emphysema in rats. Am J

Physiol Lung Cell Mol Physiol 301: L255-266, 2011.

113. Hunninghake GM, Cho MH, Tesfaigzi Y, Soto-Quiros ME, Avila L, Lasky-Su

J, Stidley C, Melen E, Soderhall C, Hallberg J, Kull I, Kere J, et al. MMP12, lung

function, and COPD in high-risk populations. N Engl J Med 361: 2599-2608, 2009.

114. Hvidsten SC, Storesund L, Wentzel-Larsen T, Gulsvik A, and Lehmann S.

Prevalence and predictors of undiagnosed chronic obstructive pulmonary disease in a

Norwegian adult general population. Clin Respir J 4: 13-21, 2010.

115. Ichinose M, Sugiura H, Yamagata S, Koarai A, and Shirato K. Increase in

reactive nitrogen species production in chronic obstructive pulmonary disease airways.

Am J Respir Crit Care Med 162: 701-706, 2000.

116. Imai K, Mercer BA, Schulman LL, Sonett JR, and D'Armiento JM.

Correlation of lung surface area to apoptosis and proliferation in human emphysema.

Eur Respir J 25: 250-258, 2005.

117. Ingenito EP, Tsai L, Murthy S, Tyagi S, Mazan M, and Hoffman A.

Autologous lung-derived mesenchymal stem cell transplantation in experimental

emphysema. Cell Transplant 21: 175-189, 2012.

118. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ,

Quadri SK, Bhattacharya S, and Bhattacharya J. Mitochondrial transfer from

bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung

injury. Nat Med 18: 759-765, 2012.

119. Ito K, and Barnes PJ. COPD as a disease of accelerated lung aging. Chest 135:

Page 225: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

206

173-180, 2009.

120. Janssens JP, Pache JC, and Nicod LP. Physiological changes in respiratory

function associated with ageing. Eur Respir J 13: 197-205, 1999.

121. John M, Au BT, Jose PJ, Lim S, Saunders M, Barnes PJ, Mitchell JA,

Belvisi MG, and Chung KF. Expression and release of interleukin-8 by human

airway smooth muscle cells: inhibition by Th-2 cytokines and corticosteroids. Am J

Respir Cell Mol Biol 18: 84-90, 1998.

122. Jung Y, Bauer G, and Nolta JA. Concise review: Induced pluripotent stem

cell-derived mesenchymal stem cells: progress toward safe clinical products. Stem

Cells 30: 42-47, 2012.

123. Kaasik A, Safiulina D, Zharkovsky A, and Veksler V. Regulation of

mitochondrial matrix volume. Am J Physiol Cell Physiol 292: C157-163, 2007.

124. Kagan VE, Borisenko GG, Tyurina YY, Tyurin VA, Jiang J, Potapovich AI,

Kini V, Amoscato AA, and Fujii Y. Oxidative lipidomics of apoptosis: redox

catalytic interactions of cytochrome c with cardiolipin and phosphatidylserine. Free

Radic Biol Med 37: 1963-1985, 2004.

125. Kanazawa H, and Yoshikawa J. Elevated oxidative stress and reciprocal

reduction of vascular endothelial growth factor levels with severity of COPD. Chest

128: 3191-3197, 2005.

126. Karrasch S, Holz O, and Jorres RA. Aging and induced senescence as factors

in the pathogenesis of lung emphysema. Respir Med 102: 1215-1230, 2008.

127. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, and Voelkel NF.

Endothelial cell death and decreased expression of vascular endothelial growth factor

and vascular endothelial growth factor receptor 2 in emphysema. Am J Respir Crit

Care Med 163: 737-744, 2001.

128. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S,

Hirth PK, Waltenberger J, and Voelkel NF. Inhibition of VEGF receptors causes

lung cell apoptosis and emphysema. J Clin Invest 106: 1311-1319, 2000.

129. Kassmer SH, and Krause DS. Detection of bone marrow-derived lung

epithelial cells. Exp Hematol 38: 564-573, 2010.

Page 226: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

207

130. Katsha AM, Ohkouchi S, Xin H, Kanehira M, Sun R, Nukiwa T, and Saijo Y.

Paracrine factors of multipotent stromal cells ameliorate lung injury in an

elastase-induced emphysema model. Mol Ther 19: 196-203, 2011.

131. Kauffman SL. Cell proliferation in the mammalian lung. Int Rev Exp Pathol 22:

131-191, 1980.

132. Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 10:

709-716, 2012.

133. Kirkham P, and Rahman I. Oxidative stress in asthma and COPD:

Antioxidants as a therapeutic strategy. Pharmacology &amp; Therapeutics 111:

476-494, 2006.

134. Kirkham PA, and Barnes PJ. Oxidative stress in COPD. Chest 144: 266-273,

2013.

135. Kluchova Z, Petrasova D, Joppa P, Dorkova Z, and Tkacova R. The

association between oxidative stress and obstructive lung impairment in patients with

COPD. Physiol Res 56: 51-56, 2007.

136. Kohansal R, Martinez-Camblor P, Agusti A, Buist AS, Mannino DM, and

Soriano JB. The natural history of chronic airflow obstruction revisited: an analysis

of the Framingham offspring cohort. Am J Respir Crit Care Med 180: 3-10, 2009.

137. Kojima S, Sakakibara H, Motani S, Hirose K, Mizuno F, Ochiai M, and

Hashimoto S. Incidence of chronic obstructive pulmonary disease, and the

relationship between age and smoking in a Japanese population. J Epidemiol 17:

54-60, 2007.

138. Kumar PA, Hu Y, Yamamoto Y, Hoe NB, Wei TS, Mu D, Sun Y, Joo LS,

Dagher R, Zielonka EM, Wang de Y, Lim B, et al. Distal airway stem cells yield

alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell

147: 525-538, 2011.

139. Kwon YW, Heo SC, Jeong GO, Yoon JW, Mo WM, Lee MJ, Jang IH, Kwon

SM, Lee JS, and Kim JH. Tumor necrosis factor-alpha-activated mesenchymal stem

cells promote endothelial progenitor cell homing and angiogenesis. Biochim Biophys

Acta 1832: 2136-2144, 2013.

Page 227: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

208

140. Lacoste JY, Bousquet J, Chanez P, Van Vyve T, Simony-Lafontaine J,

Lequeu N, Vic P, Enander I, Godard P, and Michel FB. Eosinophilic and

neutrophilic inflammation in asthma, chronic bronchitis, and chronic obstructive

pulmonary disease. J Allergy Clin Immunol 92: 537-548, 1993.

141. Lamprecht B, McBurnie MA, Vollmer WM, Gudmundsson G, Welte T,

Nizankowska-Mogilnicka E, Studnicka M, Bateman E, Anto JM, Burney P,

Mannino DM, Buist SA, et al. COPD in never smokers: results from the

population-based burden of obstructive lung disease study. Chest 139: 752-763, 2011.

142. Lams BE, Sousa AR, Rees PJ, and Lee TH. Immunopathology of the

small-airway submucosa in smokers with and without chronic obstructive pulmonary

disease. Am J Respir Crit Care Med 158: 1518-1523, 1998.

143. Lau WK, Chan SC, Law AC, Ip MS, and Mak JC. The role of MAPK and

Nrf2 pathways in ketanserin-elicited attenuation of cigarette smoke-induced IL-8

production in human bronchial epithelial cells. Toxicol Sci 125: 569-577, 2012.

144. Le Blanc K, and Mougiakakos D. Multipotent mesenchymal stromal cells and

the innate immune system. Nat Rev Immunol 12: 383-396, 2012.

145. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL,

Semprun-Prieto L, Delafontaine P, and Prockop DJ. Intravenous hMSCs improve

myocardial infarction in mice because cells embolized in lung are activated to secrete

the anti-inflammatory protein TSG-6. Cell Stem Cell 5: 54-63, 2009.

146. Lennartsson J, and Ronnstrand L. Stem cell factor receptor/c-Kit: from basic

science to clinical implications. Physiol Rev 92: 1619-1649, 2012.

147. Li F, Zhang M, Hussain F, Triantaphyllopoulos K, Clark AR, Bhavsar PK,

Zhou X, and Chung KF. Inhibition of p38 MAPK-dependent bronchial contraction

after ozone by corticosteroids. Eur Respir J 37: 933-942, 2011.

148. Li H, Fan X, Kovi RC, Jo Y, Moquin B, Konz R, Stoicov C, Kurt-Jones E,

Grossman SR, Lyle S, Rogers AB, Montrose M, et al. Spontaneous expression of

embryonic factors and p53 point mutations in aged mesenchymal stem cells: a model

of age-related tumorigenesis in mice. Cancer Res 67: 10889-10898, 2007.

149. Li X, Fang P, Mai J, Choi ET, Wang H, and Yang XF. Targeting

Page 228: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

209

mitochondrial reactive oxygen species as novel therapy for inflammatory diseases and

cancers. J Hematol Oncol 6: 19, 2013.

150. Li X, Zhang Y, Yeung SC, Liang Y, Liang X, Ding Y, Ip MS, Tse HF, Mak JC,

and Lian Q. Mitochondrial transfer of induced pluripotent stem cell-derived

mesenchymal stem cells to airway epithelial cells attenuates cigarette smoke-induced

damage. Am J Respir Cell Mol Biol 51: 455-465, 2014.

151. Li Y, Gu C, Xu W, Yan J, Xia Y, Ma Y, Chen C, He X, and Tao H.

Therapeutic effects of amniotic fluid-derived mesenchymal stromal cells on lung

injury in rats with emphysema. Respir Res 15: 120, 2014.

152. Lian Q, Lye E, Suan Yeo K, Khia Way Tan E, Salto-Tellez M, Liu TM,

Palanisamy N, El Oakley RM, Lee EH, Lim B, and Lim SK. Derivation of

clinically compliant MSCs from CD105+, CD24- differentiated human ESCs. Stem

Cells 25: 425-436, 2007.

153. Lian Q, Zhang Y, Zhang J, Zhang HK, Wu X, Lam FF, Kang S, Xia JC, Lai

WH, Au KW, Chow YY, Siu CW, et al. Functional mesenchymal stem cells derived

from human induced pluripotent stem cells attenuate limb ischemia in mice.

Circulation 121: 1113-1123, 2010.

154. Lim S, Roche N, Oliver BG, Mattos W, Barnes PJ, and Chung KF. Balance

of matrix metalloprotease-9 and tissue inhibitor of metalloprotease-1 from alveolar

macrophages in cigarette smokers. Regulation by interleukin-10. Am J Respir Crit

Care Med 162: 1355-1360, 2000.

155. Lin MY, and Sheng ZH. Regulation of mitochondrial transport in neurons. Exp

Cell Res 334: 35-44, 2015.

156. Linden M, Rasmussen JB, Piitulainen E, Tunek A, Larson M, Tegner H,

Venge P, Laitinen LA, and Brattsand R. Airway inflammation in smokers with

nonobstructive and obstructive chronic bronchitis. Am Rev Respir Dis 148: 1226-1232,

1993.

157. Lindsey JY, Ganguly K, Brass DM, Li Z, Potts EN, Degan S, Chen H,

Brockway B, Abraham SN, Berndt A, Stripp BR, Foster WM, et al. c-Kit is

essential for alveolar maintenance and protection from emphysema-like disease in

Page 229: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

210

mice. Am J Respir Crit Care Med 183: 1644-1652, 2011.

158. Liu AN, Mohammed AZ, Rice WR, Fiedeldey DT, Liebermann JS, Whitsett

JA, Braciale TJ, and Enelow RI. Perforin-independent CD8(+) T-cell-mediated

cytotoxicity of alveolar epithelial cells is preferentially mediated by tumor necrosis

factor-alpha: relative insensitivity to Fas ligand. Am J Respir Cell Mol Biol 20:

849-858, 1999.

159. Longmire TA, Ikonomou L, Hawkins F, Christodoulou C, Cao Y, Jean JC,

Kwok LW, Mou H, Rajagopal J, Shen SS, Dowton AA, Serra M, et al. Efficient

derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell

Stem Cell 10: 398-411, 2012.

160. Louhelainen N, Rytila P, Haahtela T, Kinnula VL, and Djukanovic R.

Persistence of oxidant and protease burden in the airways after smoking cessation.

BMC Pulm Med 9: 25, 2009.

161. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham

J, Adair T, Aggarwal R, Ahn SY, Alvarado M, Anderson HR, et al. Global and

regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a

systematic analysis for the Global Burden of Disease Study 2010. Lancet 380:

2095-2128, 2012.

162. Ma N, Gai H, Mei J, Ding FB, Bao CR, Nguyen DM, and Zhong H. Bone

marrow mesenchymal stem cells can differentiate into type II alveolar epithelial cells

in vitro. Cell Biol Int 35: 1261-1266, 2011.

163. MacNee W. Accelerated lung aging: a novel pathogenic mechanism of chronic

obstructive pulmonary disease (COPD). Biochem Soc Trans 37: 819-823, 2009.

164. Macnee W. Pathogenesis of chronic obstructive pulmonary disease. Clin Chest

Med 28: 479-513, v, 2007.

165. Madamanchi NR, and Runge MS. Mitochondrial dysfunction in

atherosclerosis. Circ Res 100: 460-473, 2007.

166. Majo J, Ghezzo H, and Cosio MG. Lymphocyte population and apoptosis in

the lungs of smokers and their relation to emphysema. Eur Respir J 17: 946-953,

2001.

Page 230: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

211

167. Mak JC. Pathogenesis of COPD. Part II. Oxidative-antioxidative imbalance. Int

J Tuberc Lung Dis 12: 368-374, 2008.

168. Mannino DM, and Buist AS. Global burden of COPD: risk factors, prevalence,

and future trends. Lancet 370: 765-773, 2007.

169. Mannino DM, and Buist AS. Global burden of COPD: risk factors, prevalence,

and future trends. The Lancet 370: 765-773, 2007.

170. Marchi S, Giorgi C, Suski JM, Agnoletto C, Bononi A, Bonora M, De

Marchi E, Missiroli S, Patergnani S, Poletti F, Rimessi A, Duszynski J, et al.

Mitochondria-ros crosstalk in the control of cell death and aging. J Signal Transduct

2012: 329635, 2012.

171. Maria OM, and Tran SD. Human mesenchymal stem cells cultured with

salivary gland biopsies adopt an epithelial phenotype. Stem Cells Dev 20: 959-967,

2011.

172. Mathers CD, and Loncar D. Projections of global mortality and burden of

disease from 2002 to 2030. PLoS Med 3: e442, 2006.

173. Medina-Ramon M, Zanobetti A, and Schwartz J. The effect of ozone and

PM10 on hospital admissions for pneumonia and chronic obstructive pulmonary

disease: a national multicity study. Am J Epidemiol 163: 579-588, 2006.

174. Michaeloudes C, Sukkar MB, Khorasani NM, Bhavsar PK, and Chung KF.

TGF-beta regulates Nox4, MnSOD and catalase expression, and IL-6 release in

airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 300: L295-304,

2011.

175. Mikko M, Forsslund H, Cui L, Grunewald J, Wheelock AM, Wahlstrom J,

and Skold CM. Increased intraepithelial (CD103+) CD8+ T cells in the airways of

smokers with and without chronic obstructive pulmonary disease. Immunobiology 218:

225-231, 2013.

176. Montuschi P. Exhaled breath condensate analysis in patients with COPD. Clin

Chim Acta 356: 22-34, 2005.

177. Moriyama C, Betsuyaku T, Ito Y, Hamamura I, Hata J, Takahashi H,

Nasuhara Y, and Nishimura M. Aging enhances susceptibility to cigarette

Page 231: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

212

smoke-induced inflammation through bronchiolar chemokines. Am J Respir Cell Mol

Biol 42: 304-311, 2010.

178. Morla M, Busquets X, Pons J, Sauleda J, MacNee W, and Agusti AG.

Telomere shortening in smokers with and without COPD. Eur Respir J 27: 525-528,

2006.

179. Mou H, Zhao R, Sherwood R, Ahfeldt T, Lapey A, Wain J, Sicilian L,

Izvolsky K, Musunuru K, Cowan C, and Rajagopal J. Generation of multipotent

lung and airway progenitors from mouse ESCs and patient-specific cystic fibrosis

iPSCs. Cell Stem Cell 10: 385-397, 2012.

180. Muller KC, Welker L, Paasch K, Feindt B, Erpenbeck VJ, Hohlfeld JM,

Krug N, Nakashima M, Branscheid D, Magnussen H, Jorres RA, and Holz O.

Lung fibroblasts from patients with emphysema show markers of senescence in vitro.

Respir Res 7: 32, 2006.

181. Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, Ezzati

M, Shibuya K, Salomon JA, Abdalla S, Aboyans V, Abraham J, et al.

Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions,

1990-2010: a systematic analysis for the Global Burden of Disease Study 2010.

Lancet 380: 2197-2223, 2012.

182. Nombela-Arrieta C, Ritz J, and Silberstein LE. The elusive nature and

function of mesenchymal stem cells. Nat Rev Mol Cell Biol 12: 126-131, 2011.

183. Nunnari J, and Suomalainen A. Mitochondria: in sickness and in health. Cell

148: 1145-1159, 2012.

184. Nur EKA, Gross SR, Pan Z, Balklava Z, Ma J, and Liu LF. Nuclear

translocation of cytochrome c during apoptosis. J Biol Chem 279: 24911-24914, 2004.

185. Nystedt J, Anderson H, Tikkanen J, Pietila M, Hirvonen T, Takalo R,

Heiskanen A, Satomaa T, Natunen S, Lehtonen S, Hakkarainen T, Korhonen M,

et al. Cell surface structures influence lung clearance rate of systemically infused

mesenchymal stromal cells. Stem Cells 31: 317-326, 2013.

186. O'Donnell DE. Hyperinflation, dyspnea, and exercise intolerance in chronic

obstructive pulmonary disease. Proc Am Thorac Soc 3: 180-184, 2006.

Page 232: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

213

187. O'Donnell RA, Peebles C, Ward JA, Daraker A, Angco G, Broberg P,

Pierrou S, Lund J, Holgate ST, Davies DE, Delany DJ, Wilson SJ, et al.

Relationship between peripheral airway dysfunction, airway obstruction, and

neutrophilic inflammation in COPD. Thorax 59: 837-842, 2004.

188. Ohashi M, Runge MS, Faraci FM, and Heistad DD. MnSOD deficiency

increases endothelial dysfunction in ApoE-deficient mice. Arterioscler Thromb Vasc

Biol 26: 2331-2336, 2006.

189. Ohnishi K, Takagi M, Kurokawa Y, Satomi S, and Konttinen YT. Matrix

metalloproteinase-mediated extracellular matrix protein degradation in human

pulmonary emphysema. Lab Invest 78: 1077-1087, 1998.

190. Opitz B, van Laak V, Eitel J, and Suttorp N. Innate immune recognition in

infectious and noninfectious diseases of the lung. Am J Respir Crit Care Med 181:

1294-1309, 2010.

191. Orozco-Levi M, Garcia-Aymerich J, Villar J, Ramirez-Sarmiento A, Anto

JM, and Gea J. Wood smoke exposure and risk of chronic obstructive pulmonary

disease. Eur Respir J 27: 542-546, 2006.

192. Otsu K, Das S, Houser SD, Quadri SK, Bhattacharya S, and Bhattacharya J.

Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells.

Blood 113: 4197-4205, 2009.

193. Ott M, Robertson JD, Gogvadze V, Zhivotovsky B, and Orrenius S.

Cytochrome c release from mitochondria proceeds by a two-step process. Proc Natl

Acad Sci U S A 99: 1259-1263, 2002.

194. Paredi P, Kharitonov SA, and Barnes PJ. Analysis of expired air for oxidation

products. Am J Respir Crit Care Med 166: S31-37, 2002.

195. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, and Barnes PJ.

Exhaled ethane, a marker of lipid peroxidation, is elevated in chronic obstructive

pulmonary disease. Am J Respir Crit Care Med 162: 369-373, 2000.

196. Pasquier J, Guerrouahen BS, Al Thawadi H, Ghiabi P, Maleki M,

Abu-Kaoud N, Jacob A, Mirshahi M, Galas L, Rafii S, Le Foll F, and Rafii A.

Preferential transfer of mitochondria from endothelial to cancer cells through

Page 233: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

214

tunneling nanotubes modulates chemoresistance. J Transl Med 11: 94, 2013.

197. Peel JL, Tolbert PE, Klein M, Metzger KB, Flanders WD, Todd K,

Mulholland JA, Ryan PB, and Frumkin H. Ambient air pollution and respiratory

emergency department visits. Epidemiology 16: 164-174, 2005.

198. Peto R, Speizer FE, Cochrane AL, Moore F, Fletcher CM, Tinker CM,

Higgins IT, Gray RG, Richards SM, Gilliland J, and Norman-Smith B. The

relevance in adults of air-flow obstruction, but not of mucus hypersecretion, to

mortality from chronic lung disease. Results from 20 years of prospective observation.

Am Rev Respir Dis 128: 491-500, 1983.

199. Petrache I, Natarajan V, Zhen L, Medler TR, Richter AT, Cho C, Hubbard

WC, Berdyshev EV, and Tuder RM. Ceramide upregulation causes pulmonary cell

apoptosis and emphysema-like disease in mice. Nat Med 11: 491-498, 2005.

200. Phinney DG. Functional heterogeneity of mesenchymal stem cells: implications

for cell therapy. J Cell Biochem 113: 2806-2812, 2012.

201. Pichavant M, Taront S, Jeannin P, Breuilh L, Charbonnier AS, Spriet C,

Fourneau C, Corvaia N, Heliot L, Brichet A, Tonnel AB, Delneste Y, et al. Impact

of bronchial epithelium on dendritic cell migration and function: modulation by the

bacterial motif KpOmpA. J Immunol 177: 5912-5919, 2006.

202. Pinamonti S, Leis M, Barbieri A, Leoni D, Muzzoli M, Sostero S, Chicca

MC, Carrieri A, Ravenna F, Fabbri LM, and Ciaccia A. Detection of xanthine

oxidase activity products by EPR and HPLC in bronchoalveolar lavage fluid from

patients with chronic obstructive pulmonary disease. Free Radic Biol Med 25:

771-779, 1998.

203. Pinart M, Zhang M, Li F, Hussain F, Zhu J, Wiegman C, Ryffel B, and

Chung KF. IL-17A modulates oxidant stress-induced airway hyperresponsiveness but

not emphysema. PLoS One 8: e58452, 2013.

204. Plotnikov EY, Khryapenkova TG, Vasileva AK, Marey MV, Galkina SI,

Isaev NK, Sheval EV, Polyakov VY, Sukhikh GT, and Zorov DB. Cell-to-cell

cross-talk between mesenchymal stem cells and cardiomyocytes in co-culture. J Cell

Mol Med 12: 1622-1631, 2008.

Page 234: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

215

205. Prokopczyk B, Hoffmann D, Bologna M, Cunningham AJ, Trushin N,

Akerkar S, Boyiri T, Amin S, Desai D, Colosimo S, Pittman B, Leder G, et al.

Identification of tobacco-derived compounds in human pancreatic juice. Chem Res

Toxicol 15: 677-685, 2002.

206. Rahman I, van Schadewijk AA, Crowther AJ, Hiemstra PS, Stolk J,

MacNee W, and De Boer WI. 4-Hydroxy-2-nonenal, a specific lipid peroxidation

product, is elevated in lungs of patients with chronic obstructive pulmonary disease.

Am J Respir Crit Care Med 166: 490-495, 2002.

207. Ramirez-Venegas A, Sansores RH, Quintana-Carrillo RH, Velazquez-Uncal

M, Hernandez-Zenteno RJ, Sanchez-Romero C, Velazquez-Montero A, and

Flores-Trujillo F. FEV1 decline in patients with chronic obstructive pulmonary

disease associated with biomass exposure. Am J Respir Crit Care Med 190: 996-1002,

2014.

208. Ranu H, Wilde M, and Madden B. Pulmonary function tests. Ulster Med J 80:

84-90, 2011.

209. Rennard SI, and Daughton DM. Smoking cessation. Chest 117: 360S-364S,

2000.

210. Ricciardi M, Malpeli G, Bifari F, Bassi G, Pacelli L, Nwabo Kamdje AH,

Chilosi M, and Krampera M. Comparison of epithelial differentiation and immune

regulatory properties of mesenchymal stromal cells derived from human lung and

bone marrow. PLoS One 7: e35639, 2012.

211. Rico de Souza A, Zago M, Pollock SJ, Sime PJ, Phipps RP, and Baglole CJ.

Genetic ablation of the aryl hydrocarbon receptor causes cigarette smoke-induced

mitochondrial dysfunction and apoptosis. J Biol Chem 286: 43214-43228, 2011.

212. Rizzuto R, and Pozzan T. Microdomains of intracellular Ca2+: molecular

determinants and functional consequences. Physiol Rev 86: 369-408, 2006.

213. Roos-Engstrand E, Ekstrand-Hammarstrom B, Pourazar J, Behndig AF,

Bucht A, and Blomberg A. Influence of smoking cessation on airway T lymphocyte

subsets in COPD. COPD 6: 112-120, 2009.

214. Russell RE, Culpitt SV, DeMatos C, Donnelly L, Smith M, Wiggins J, and

Page 235: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

216

Barnes PJ. Release and activity of matrix metalloproteinase-9 and tissue inhibitor of

metalloproteinase-1 by alveolar macrophages from patients with chronic obstructive

pulmonary disease. Am J Respir Cell Mol Biol 26: 602-609, 2002.

215. Rustom A, Saffrich R, Markovic I, Walther P, and Gerdes HH. Nanotubular

highways for intercellular organelle transport. Science 303: 1007-1010, 2004.

216. Saetta M, Baraldo S, Corbino L, Turato G, Braccioni F, Rea F, Cavallesco G,

Tropeano G, Mapp CE, Maestrelli P, Ciaccia A, and Fabbri LM. CD8+ve cells in

the lungs of smokers with chronic obstructive pulmonary disease. Am J Respir Crit

Care Med 160: 711-717, 1999.

217. Saetta M, Turato G, Facchini FM, Corbino L, Lucchini RE, Casoni G,

Maestrelli P, Mapp CE, Ciaccia A, and Fabbri LM. Inflammatory cells in the

bronchial glands of smokers with chronic bronchitis. Am J Respir Crit Care Med 156:

1633-1639, 1997.

218. Saretzki G, and Von Zglinicki T. Replicative aging, telomeres, and oxidative

stress. Ann N Y Acad Sci 959: 24-29, 2002.

219. Savale L, Chaouat A, Bastuji-Garin S, Marcos E, Boyer L, Maitre B, Sarni

M, Housset B, Weitzenblum E, Matrat M, Le Corvoisier P, Rideau D, et al.

Shortened telomeres in circulating leukocytes of patients with chronic obstructive

pulmonary disease. Am J Respir Crit Care Med 179: 566-571, 2009.

220. Schaberg T, Klein U, Rau M, Eller J, and Lode H. Subpopulations of alveolar

macrophages in smokers and nonsmokers: relation to the expression of CD11/CD18

molecules and superoxide anion production. Am J Respir Crit Care Med 151:

1551-1558, 1995.

221. Scholzen T, and Gerdes J. The Ki-67 protein: from the known and the unknown.

J Cell Physiol 182: 311-322, 2000.

222. Schweitzer KS, Johnstone BH, Garrison J, Rush NI, Cooper S, Traktuev DO,

Feng D, Adamowicz JJ, Van Demark M, Fisher AJ, Kamocki K, Brown MB, et al.

Adipose stem cell treatment in mice attenuates lung and systemic injury induced by

cigarette smoking. Am J Respir Crit Care Med 183: 215-225, 2011.

223. Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Becerril C, and Selman M.

Page 236: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

217

Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal

cell death in COPD. Chest 117: 684-694, 2000.

224. Sepe SM, and Clark RA. Oxidant membrane injury by the neutrophil

myeloperoxidase system. II. Injury by stimulated neutrophils and protection by

lipid-soluble antioxidants. J Immunol 134: 1896-1901, 1985.

225. Sexton P, Metcalf P, and Kolbe J. Respiratory effects of insulin sensitisation

with metformin: a prospective observational study. COPD 11: 133-142, 2014.

226. Shaykhiev R, Otaki F, Bonsu P, Dang DT, Teater M, Strulovici-Barel Y, Salit

J, Harvey BG, and Crystal RG. Cigarette smoking reprograms apical junctional

complex molecular architecture in the human airway epithelium in vivo. Cell Mol Life

Sci 68: 877-892, 2011.

227. Sheng ZH, and Cai Q. Mitochondrial transport in neurons: impact on synaptic

homeostasis and neurodegeneration. Nat Rev Neurosci 13: 77-93, 2012.

228. Shi MM, Iwamoto T, and Forman HJ. gamma-Glutamylcysteine synthetase

and GSH increase in quinone-induced oxidative stress in BPAEC. Am J Physiol 267:

L414-421, 1994.

229. Shigemura N, Okumura M, Mizuno S, Imanishi Y, Matsuyama A, Shiono H,

Nakamura T, and Sawa Y. Lung tissue engineering technique with adipose stromal

cells improves surgical outcome for pulmonary emphysema. Am J Respir Crit Care

Med 174: 1199-1205, 2006.

230. Shigemura N, Okumura M, Mizuno S, Imanishi Y, Nakamura T, and Sawa

Y. Autologous transplantation of adipose tissue-derived stromal cells ameliorates

pulmonary emphysema. Am J Transplant 6: 2592-2600, 2006.

231. Shimada T. Xenobiotic-metabolizing enzymes involved in activation and

detoxification of carcinogenic polycyclic aromatic hydrocarbons. Drug Metab

Pharmacokinet 21: 257-276, 2006.

232. Sies H. Glutathione and its role in cellular functions. Free Radic Biol Med 27:

916-921, 1999.

233. Silverman EK, Weiss ST, Drazen JM, Chapman HA, Carey V, Campbell EJ,

Denish P, Silverman RA, Celedon JC, Reilly JJ, Ginns LC, and Speizer FE.

Page 237: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

218

Gender-related differences in severe, early-onset chronic obstructive pulmonary

disease. Am J Respir Crit Care Med 162: 2152-2158, 2000.

234. Sin DD, Anthonisen NR, Soriano JB, and Agusti AG. Mortality in COPD:

Role of comorbidities. Eur Respir J 28: 1245-1257, 2006.

235. Singer NG, and Caplan AI. Mesenchymal stem cells: mechanisms of

inflammation. Annu Rev Pathol 6: 457-478, 2011.

236. Soler-Cataluna JJ, Martinez-Garcia MA, Roman Sanchez P, Salcedo E,

Navarro M, and Ochando R. Severe acute exacerbations and mortality in patients

with chronic obstructive pulmonary disease. Thorax 60: 925-931, 2005.

237. Song L, Guan XJ, Chen X, Cui ZL, Han FF, Guo XJ, and Xu WG.

Mesenchymal stem cells reduce cigarette smoke-induced inflammation and airflow

obstruction in rats via TGF-beta1 signaling. COPD 11: 582-590, 2014.

238. Spees JL, Olson SD, Whitney MJ, and Prockop DJ. Mitochondrial transfer

between cells can rescue aerobic respiration. Proc Natl Acad Sci U S A 103:

1283-1288, 2006.

239. Stenfors N, Pourazar J, Blomberg A, Krishna MT, Mudway I, Helleday R,

Kelly FJ, Frew AJ, and Sandstrom T. Effect of ozone on bronchial mucosal

inflammation in asthmatic and healthy subjects. Respir Med 96: 352-358, 2002.

240. Stockley RA. Neutrophils and the pathogenesis of COPD. Chest 121:

151S-155S, 2002.

241. Stoller JK, and Aboussouan LS. Alpha1-antitrypsin deficiency. Lancet 365:

2225-2236, 2005.

242. Stringer KA, Tobias M, O'Neill HC, and Franklin CC. Cigarette smoke

extract-induced suppression of caspase-3-like activity impairs human neutrophil

phagocytosis. Am J Physiol Lung Cell Mol Physiol 292: L1572-1579, 2007.

243. Sueblinvong V, Loi R, Eisenhauer PL, Bernstein IM, Suratt BT, Spees JL,

and Weiss DJ. Derivation of lung epithelium from human cord blood-derived

mesenchymal stem cells. Am J Respir Crit Care Med 177: 701-711, 2008.

244. Sun YQ, Deng MX, He J, Zeng QX, Wen W, Wong DS, Tse HF, Xu G, Lian

Q, Shi J, and Fu QL. Human pluripotent stem cell-derived mesenchymal stem cells

Page 238: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

219

prevent allergic airway inflammation in mice. Stem Cells 30: 2692-2699, 2012.

245. Tager IB, Ngo L, and Hanrahan JP. Maternal smoking during pregnancy.

Effects on lung function during the first 18 months of life. Am J Respir Crit Care Med

152: 977-983, 1995.

246. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, and

Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by

defined factors. Cell 131: 861-872, 2007.

247. Takahashi K, and Yamanaka S. Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell 126: 663-676, 2006.

248. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, Okada Y,

Yamasawa F, Nakahara K, and Umeda A. Increased expression of transforming

growth factor-beta1 in small airway epithelium from tobacco smokers and patients

with chronic obstructive pulmonary disease (COPD). Am J Respir Crit Care Med 163:

1476-1483, 2001.

249. Tang K, Rossiter HB, Wagner PD, and Breen EC. Lung-targeted VEGF

inactivation leads to an emphysema phenotype in mice. J Appl Physiol (1985) 97:

1559-1566; discussion 1549, 2004.

250. Taylor AE, Finney-Hayward TK, Quint JK, Thomas CM, Tudhope SJ,

Wedzicha JA, Barnes PJ, and Donnelly LE. Defective macrophage phagocytosis of

bacteria in COPD. Eur Respir J 35: 1039-1047, 2010.

251. Teo AK, and Vallier L. Emerging use of stem cells in regenerative medicine.

Biochem J 428: 11-23, 2010.

252. Tibboel J, Keijzer R, Reiss I, de Jongste JC, and Post M. Intravenous and

intratracheal mesenchymal stromal cell injection in a mouse model of pulmonary

emphysema. COPD 11: 310-318, 2014.

253. Triantaphyllopoulos K, Hussain F, Pinart M, Zhang M, Li F, Adcock I,

Kirkham P, Zhu J, and Chung KF. A model of chronic inflammation and

pulmonary emphysema after multiple ozone exposures in mice. Am J Physiol Lung

Cell Mol Physiol 300: L691-700, 2011.

254. Tsuji T, Aoshiba K, and Nagai A. Alveolar cell senescence in patients with

Page 239: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

220

pulmonary emphysema. Am J Respir Crit Care Med 174: 886-893, 2006.

255. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y,

Salvemini D, Voelkel NF, and Flores SC. Oxidative stress and apoptosis interact and

cause emphysema due to vascular endothelial growth factor receptor blockade. Am J

Respir Cell Mol Biol 29: 88-97, 2003.

256. Tzouvelekis A, Paspaliaris V, Koliakos G, Ntolios P, Bouros E, Oikonomou A,

Zissimopoulos A, Boussios N, Dardzinski B, Gritzalis D, Antoniadis A,

Froudarakis M, et al. A prospective, non-randomized, no placebo-controlled, phase

Ib clinical trial to study the safety of the adipose derived stromal cells-stromal

vascular fraction in idiopathic pulmonary fibrosis. J Transl Med 11: 171, 2013.

257. Vagaggini B, Carnevali S, Macchioni P, Taccola M, Fornai E, Bacci E,

Bartoli ML, Cianchetti S, Dente FL, Di Franco A, Giannini D, and Paggiaro PL.

Airway inflammatory response to ozone in subjects with different asthma severity.

Eur Respir J 13: 274-280, 1999.

258. Vallabhaneni KC, Haller H, and Dumler I. Vascular smooth muscle cells

initiate proliferation of mesenchymal stem cells by mitochondrial transfer via

tunneling nanotubes. Stem Cells Dev 21: 3104-3113, 2012.

259. van den Boom G, van Schayck CP, van Mollen MP, Tirimanna PR, den

Otter JJ, van Grunsven PM, Buitendijk MJ, van Herwaarden CL, and van Weel

C. Active detection of chronic obstructive pulmonary disease and asthma in the

general population. Results and economic consequences of the DIMCA program. Am

J Respir Crit Care Med 158: 1730-1738, 1998.

260. van der Flier LG, and Clevers H. Stem cells, self-renewal, and differentiation

in the intestinal epithelium. Annu Rev Physiol 71: 241-260, 2009.

261. van der Vliet A, Eiserich JP, Shigenaga MK, and Cross CE. Reactive nitrogen

species and tyrosine nitration in the respiratory tract: epiphenomena or a

pathobiologic mechanism of disease? Am J Respir Crit Care Med 160: 1-9, 1999.

262. Verbeken EK, Cauberghs M, Mertens I, Clement J, Lauweryns JM, and

Van de Woestijne KP. The senile lung. Comparison with normal and emphysematous

lungs. 1. Structural aspects. Chest 101: 793-799, 1992.

Page 240: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

221

263. Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, Barnes

PJ, Fabbri LM, Martinez FJ, Nishimura M, Stockley RA, Sin DD, et al. Global

strategy for the diagnosis, management, and prevention of chronic obstructive

pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med 187:

347-365, 2013.

264. Walker N, Badri L, Wettlaufer S, Flint A, Sajjan U, Krebsbach PH,

Keshamouni VG, Peters-Golden M, and Lama VN. Resident tissue-specific

mesenchymal progenitor cells contribute to fibrogenesis in human lung allografts. Am

J Pathol 178: 2461-2469, 2011.

265. Wang CH, Verma S, Hsieh IC, Hung A, Cheng TT, Wang SY, Liu YC,

Stanford WL, Weisel RD, Li RK, and Cherng WJ. Stem cell factor attenuates

vascular smooth muscle apoptosis and increases intimal hyperplasia after vascular

injury. Arterioscler Thromb Vasc Biol 27: 540-547, 2007.

266. Waterman RS, Tomchuck SL, Henkle SL, and Betancourt AM. A new

mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1

or an Immunosuppressive MSC2 phenotype. PLoS One 5: e10088, 2010.

267. Wei H, Tan G, Manasi, Qiu S, Kong G, Yong P, Koh C, Ooi TH, Lim SY,

Wong P, Gan SU, and Shim W. One-step derivation of cardiomyocytes and

mesenchymal stem cells from human pluripotent stem cells. Stem Cell Res 9: 87-100,

2012.

268. Weiss DJ. Concise review: current status of stem cells and regenerative medicine

in lung biology and diseases. Stem Cells 32: 16-25, 2014.

269. Weiss DJ. Stem cells, cell therapies, and bioengineering in lung biology and

diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac

Soc 10: S45-97, 2013.

270. Weiss DJ, Bertoncello I, Borok Z, Kim C, Panoskaltsis-Mortari A, Reynolds

S, Rojas M, Stripp B, Warburton D, and Prockop DJ. Stem cells and cell therapies

in lung biology and lung diseases. Proc Am Thorac Soc 8: 223-272, 2011.

271. Weiss DJ, Casaburi R, Flannery R, LeRoux-Williams M, and Tashkin DP. A

placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest 143:

Page 241: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

222

1590-1598, 2013.

272. Wiegman CH, Li F, Clarke CJ, Jazrawi E, Kirkham P, Barnes PJ, Adcock

IM, and Chung KF. A comprehensive analysis of oxidative stress in the

ozone-induced lung inflammation mouse model. Clin Sci (Lond) 126: 425-440, 2014.

273. Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE,

Bao W, Pavlidis S, Barnes PJ, Kanerva J, Bittner A, Rao N, et al. Oxidative

stress-induced mitochondrial dysfunction drives inflammation and airway smooth

muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy

Clin Immunol 136: 769-780, 2015.

274. Williams AS, Eynott PR, Leung SY, Nath P, Jupp R, De Sanctis GT, Resnick

R, Adcock IM, and Chung KF. Role of cathepsin S in ozone-induced airway

hyperresponsiveness and inflammation. Pulm Pharmacol Ther 22: 27-32, 2009.

275. Williams AS, Issa R, Leung SY, Nath P, Ferguson GD, Bennett BL, Adcock

IM, and Chung KF. Attenuation of ozone-induced airway inflammation and

hyper-responsiveness by c-Jun NH2 terminal kinase inhibitor SP600125. J Pharmacol

Exp Ther 322: 351-359, 2007.

276. Wilson JG, Liu KD, Zhuo H, Caballero L, McMillan M, Fang X, Cosgrove

K, Vojnik R, Calfee CS, Lee JW, Rogers AJ, Levitt J, et al. Mesenchymal stem

(stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med 3:

24-32, 2015.

277. Wipfli H, and Samet JM. Global economic and health benefits of tobacco

control: part 1. Clin Pharmacol Ther 86: 263-271, 2009.

278. Wong AP, Bear CE, Chin S, Pasceri P, Thompson TO, Huan LJ, Ratjen F,

Ellis J, and Rossant J. Directed differentiation of human pluripotent stem cells into

mature airway epithelia expressing functional CFTR protein. Nat Biotechnol 30:

876-882, 2012.

279. Wong AP, Chin S, Xia S, Garner J, Bear CE, and Rossant J. Efficient

generation of functional CFTR-expressing airway epithelial cells from human

pluripotent stem cells. Nat Protoc 10: 363-381, 2015.

280. Xu F, Yin X, Shen H, Xu Y, Ware RS, and Owen N. Better understanding the

Page 242: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

223

influence of cigarette smoking and indoor air pollution on chronic obstructive

pulmonary disease: a case-control study in Mainland China. Respirology 12: 891-897,

2007.

281. Yakes FM, and Van Houten B. Mitochondrial DNA damage is more extensive

and persists longer than nuclear DNA damage in human cells following oxidative

stress. Proc Natl Acad Sci U S A 94: 514-519, 1997.

282. Yan C, Qu P, and Du H. Myeloid-specific expression of Stat3C results in

conversion of bone marrow mesenchymal stem cells into alveolar type II epithelial

cells in the lung. Sci China Life Sci 55: 576-590, 2012.

283. Yang E, Zha J, Jockel J, Boise LH, Thompson CB, and Korsmeyer SJ. Bad,

a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death.

Cell 80: 285-291, 1995.

284. Yang NC, Ho WM, Chen YH, and Hu ML. A convenient one-step extraction of

cellular ATP using boiling water for the luciferin-luciferase assay of ATP. Anal

Biochem 306: 323-327, 2002.

285. Yao H, and Rahman I. Current concepts on oxidative/carbonyl stress,

inflammation and epigenetics in pathogenesis of chronic obstructive pulmonary

disease. Toxicol Appl Pharmacol 254: 72-85, 2011.

286. Ylostalo JH, Bartosh TJ, Coble K, and Prockop DJ. Human mesenchymal

stem/stromal cells cultured as spheroids are self-activated to produce prostaglandin E2

that directs stimulated macrophages into an anti-inflammatory phenotype. Stem Cells

30: 2283-2296, 2012.

287. Yokohori N, Aoshiba K, Nagai A, and Respiratory Failure Research Group

in J. Increased levels of cell death and proliferation in alveolar wall cells in patients

with pulmonary emphysema. Chest 125: 626-632, 2004.

288. Yoon YG, Haug CL, and Koob MD. Interspecies mitochondrial fusion between

mouse and human mitochondria is rapid and efficient. Mitochondrion 7: 223-229,

2007.

289. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian

S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin, II, and Thomson JA.

Page 243: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

224

Induced pluripotent stem cell lines derived from human somatic cells. Science 318:

1917-1920, 2007.

290. Yuhgetsu H, Ohno Y, Funaguchi N, Asai T, Sawada M, Takemura G,

Minatoguchi S, Fujiwara H, and Fujiwara T. Beneficial effects of autologous bone

marrow mononuclear cell transplantation against elastase-induced emphysema in

rabbits. Exp Lung Res 32: 413-426, 2006.

291. Zanini A, Cherubino F, Zampogna E, Croce S, Pignatti P, and Spanevello A.

Bronchial hyperresponsiveness, airway inflammation, and reversibility in patients

with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 10:

1155-1161, 2015.

292. Zhang J, Ou JX, and Bai CX. Tobacco smoking in China: prevalence, disease

burden, challenges and future strategies. Respirology 16: 1165-1172, 2011.

293. Zhang Y, Liang X, Liao S, Wang W, Wang J, Li X, Ding Y, Liang Y, Gao F,

Yang M, Fu Q, Xu A, et al. Potent Paracrine Effects of human induced Pluripotent

Stem Cell-derived Mesenchymal Stem Cells Attenuate Doxorubicin-induced

Cardiomyopathy. Sci Rep 5: 11235, 2015.

294. Zhang Y, Liao S, Yang M, Liang X, Poon MW, Wong CY, Wang J, Zhou Z,

Cheong SK, Lee CN, Tse HF, and Lian Q. Improved cell survival and paracrine

capacity of human embryonic stem cell-derived mesenchymal stem cells promote

therapeutic potential for pulmonary arterial hypertension. Cell Transplant 21:

2225-2239, 2012.

295. Zhen G, Liu H, Gu N, Zhang H, Xu Y, and Zhang Z. Mesenchymal stem cells

transplantation protects against rat pulmonary emphysema. Front Biosci 13:

3415-3422, 2008.

296. Zhen G, Xue Z, Zhao J, Gu N, Tang Z, Xu Y, and Zhang Z. Mesenchymal

stem cell transplantation increases expression of vascular endothelial growth factor in

papain-induced emphysematous lungs and inhibits apoptosis of lung cells.

Cytotherapy 12: 605-614, 2010.

297. Zheng G, Huang L, Tong H, Shu Q, Hu Y, Ge M, Deng K, Zhang L, Zou B,

Cheng B, and Xu J. Treatment of acute respiratory distress syndrome with allogeneic

Page 244: Mitochondrial transfer from induced pluripotent stem cell ... · Mitochondrial transfer from induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial and

225

adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot

study. Respir Res 15: 39, 2014.

298. Zhu J, Qiu Y, Valobra M, Qiu S, Majumdar S, Matin D, De Rose V, and

Jeffery PK. Plasma cells and IL-4 in chronic bronchitis and chronic obstructive

pulmonary disease. Am J Respir Crit Care Med 175: 1125-1133, 2007.