219
The roles of hyperoxia and mechanical deformation in alveolar epithelial injury and repair Stuart Robert McKechnie Doctor of Philosophy The University of Edinburgh 2007

McKechnie SR PhD thesis 08.pdf - Edinburgh Research Archive

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

The roles of hyperoxia and mechanical deformation

in alveolar epithelial injury and repair

Stuart Robert McKechnie

Doctor of Philosophy

The University of Edinburgh

2007

ii

Declaration of Authorship

The work contained in this thesis is my own except where stated. This work has not been

submitted for any other degree or professional qualification.

Stuart Robert McKechnie

Edinburgh 2007

iii

Abstract

The alveolar epithelium is a key functional component of the air-blood barrier in the

lung. Comprised of two morphologically distinct cell types, alveolar epithelial type I

(ATI) and type II (ATII) cells, effective repair of the alveolar epithelial barrier following

injury appears to be an important determinant of clinical outcome. The prevailing view

suggests this repair is achieved by the proliferation of ATII cells and the

transdifferentiation of ATII cells into ATI cells.

Supplemental oxygen and mechanical ventilation are key therapeutic interventions in the

supportive treatment of respiratory failure following lung injury, but the effects of

hyperoxia and mechanical deformation in the injured lung, and on alveolar epithelial

repair in particular, are largely unknown. The clinical impression however, is that poor

outcome is associated with exposure of injured (repairing) epithelium to such iatrogenic

‘hits’. This thesis describes studies investigating the hypothesis that hyperoxia &

mechanical deformation inhibit normal epithelial repair.

The in vitro data presented demonstrate that hyperoxia reversibly inhibits the

transdifferentiation of ATII-like cells into ATI-like cells with time in culture. Whilst

confirming that hyperoxia is injurious to alveolar epithelial cells, these data further

suggest the ATII cell population harbours a subpopulation of cells resistant to

hyperoxia-induced injury. This subpopulation of cells appears to generate fewer reactive

oxygen species and express lower levels of the zonula adherens protein E-cadherin.

Using a panel of antibodies to ATI (RTI40) and ATII (MMC4 & RTII70) cell-selective

proteins, the effect of hyperoxia on the phenotype of the alveolar epithelium in a rat

model of resolving S. aureus-induced lung injury was investigated. These in vivo studies

support the view that, under normoxic conditions, alveolar epithelial repair occurs

through ATII cell proliferation & transdifferentiation of ATII cells into ATI cells, with

transdifferentiation occurring via a novel intermediate (MMC4/RTI40-coexpressing)

immunophenotype. However, in S. aureus-injured lungs exposed to hyperoxia, the

resolution of ATII cell hyperplasia was impaired, with an increase in ATII cell-staining

iv

membrane and a reduction in intermediate cell-staining membrane compared to injured

lungs exposed to normoxia alone. As hyperoxia is pro-apoptotic and known to inhibit

ATII cell proliferation, these data support the hypothesis that hyperoxia impairs normal

epithelial repair by inhibiting the transdifferentiation of ATII cells into ATI cells in vivo.

The effect of mechanical deformation on alveolar epithelial cells in culture was

investigated by examining changes in cell viability following exposure of epithelial cell

monolayers to quantified levels of cyclic equibiaxial mechanical strain. In the central

region of monolayers, deformation-induced injury was a non-linear function of

deformation magnitude, with significant injury occurring only following exposure to

strains greater than those associated with inflation of the intact lung to total lung

capacity. However, these studies demonstrate for the first time that different epithelial

cell phenotypes within the same culture system have different sensitivities to

deformation-induced injury, with spreading RTI40-expressing cells in the peripheral

region of epithelial cell monolayers and in the region of ‘repairing’ wounds being

injured even at physiological levels of mechanical strain. These findings are consistent

with the hypothesis that alveolar epithelial cells in regions of epithelial repair are highly

susceptible to deformation-induced injury.

v

Acknowledgements

I would like to express my sincere thanks to my supervisors Dr Mary McElroy and

Professor David Harrison without whose personal investment, advice and kindly support

the work in this thesis would not have been possible. Thanks also to Professors Tim

Walsh, Chris Haslett and Donald Salter who have all been very supportive and whose

help was instrumental in getting this project off the ground.

Bob Morris, Susan McIntyre and Kate Harris provided constant technical and logistic

support, while Kirsty Tyrell and Linda Franklin deserve special thanks for all their

patient help in the lab. Thanks to Spike Clay and Sharon Rossiter for their advice and

practical help in setting up the haemorrhagic shock model, Linda Wilson for assistance

with confocal microscopy and Susan Harvey for cutting the lung tissue sections. Chris

Baig kindly provided the foetal lung tissue sections used.

The work presented in this thesis describing the establishment of a haemorrhagic shock-

induced model of acute lung injury and the identification of MMC4/ RTI40–expressing

alveolar epithelial cells was performed in collaboration with Gareth Clegg. I would like

to thank Gareth for his help, good company, moral support and friendship throughout.

I am grateful to Professor Leland Dobbs for kindly providing anti-RTI40 and anti-RTII70

antibodies, Professor Michael Beers for anti-pro-SP-C antibody and Dr Tim Foster for S.

aureus strain 8325-4.

Finally, heartfelt thanks are due to my family and friends. To Jo, who gives so much to

everyone, for all her ‘wee ways’, and to Mum and Dad for just being there. Thank you.

This work was supported by a Wellcome Trust Clinical Research Training Fellowship.

vi

Table of contents

Declaration of Authorship .......................................................................................................ii

Abstract .................................................................................................................................... iii

Acknowledgements ...................................................................................................................v

Table of contents ......................................................................................................................vi

Figure List ................................................................................................................................xii

Abbreviations..........................................................................................................................xiv

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

1.1 Structure and function of the alveolus ......................................................................................1

1.1.1 Structure of the air-blood barrier...............................................................................................2

1.1.2 Pulmonary endothelium ..............................................................................................................2

1.1.3 Alveolar epithelium......................................................................................................................3

1.1.3.1 Alveolar epithelial type I (ATI) cells .......................................................................................3

1.1.3.2 Alveolar epithelial type II (ATII) cells ....................................................................................4

1.1.4 Alveolar epithelial cell junctions ................................................................................................4

1.1.4.1 Tight junctions ..........................................................................................................................5

1.1.4.2 Gap junctions ............................................................................................................................5

1.1.5 Extracellular matrix and structural features of the alveolar wall............................................6

1.1.5.1 Alveolar basement membranes ................................................................................................6

1.1.5.2 Alveolar interstitial connective tissue .....................................................................................6

1.1.5.3 Structural features of the alveolar wall...................................................................................7

1.1.6 Alveolar epithelial transport .......................................................................................................7

1.1.6.1 The role of sodium transport....................................................................................................8

1.1.6.2 The role of aquaporin water channels ....................................................................................8

1.1.7 Air-blood barrier function...........................................................................................................9

1.2 The role of the alveolar epithelium in disease pathogenesis ................................................11

1.2.1 The consequences of alveolar epithelial injury .......................................................................11

1.2.2 Acute respiratory distress syndrome (ARDS) & acute lung injury (ALI) ..............................12

1.2.2.1 Historical perspective and definitions...................................................................................12

1.2.2.2 Epidemiology of acute lung injury.........................................................................................13

1.2.2.3 Pathogenesis of acute lung injury .........................................................................................14

1.2.2.4 Treatment of acute lung injury...............................................................................................16

1.2.2.5 The significance of the alveolar epithelium in acute lung injury ........................................17

1.2.3 Bacterial pneumonia..................................................................................................................17

vii

1.2.3.1 Definition, aetiology and clinical presentation ....................................................................17

1.2.3.2 Epidemiology of bacterial pneumonia ..................................................................................19

1.2.3.3 Pathogenesis of bacterial pneumonia ...................................................................................19

1.2.3.4 The significance of the alveolar epithelium in bacterial pneumonia ..................................20

1.2.4 Idiopathic Pulmonary fibrosis ..................................................................................................21

1.2.4.1 Definition.................................................................................................................................21

1.2.4.2 Epidemiology of idiopathic pulmonary fibrosis ...................................................................22

1.2.4.3 Pathogenesis & the role of the alveolar epithelium in IPF .................................................23

1.3 Alveolar epithelial repair...........................................................................................................24

1.3.1 Historical perspective of cell cycle kinetics in the alveolar epithelium.................................24

1.3.2 Current model of alveolar epithelial repair.............................................................................25

1.3.3 Stem cells responsible for alveolar epithelial repair ..............................................................26

1.3.3.1 Alveolar stem cells..................................................................................................................26

1.3.3.2 Extra-alveolar stem cells........................................................................................................27

1.3.4 Alveolar epithelial repair and lung fibrosis.............................................................................28

1.4 The use of cell-selective markers in lung injury and repair.................................................30

1.4.1 Biomarkers and immunotargeting ............................................................................................30

1.4.2 Alveolar epithelial cell-selective markers ................................................................................31

1.4.2.1 ATI cell-specific and cell-selective proteins .........................................................................31

1.4.2.2 ATII cell-specific and cell-selective proteins........................................................................32

1.4.3 Alveolar epithelial cell-selective markers in the investigation of lung injury .......................33

1.4.3.1 Cell-selective markers in quantifying epithelial cell numbers and/or injury .....................33

1.4.3.2 Specific regulation of cell selective markers in lung injury.................................................35

1.4.3.3 Cell-selective markers and the phenotype of repairing epithelium.....................................35

1.5 Hyperoxia and the alveolar epithelium ...................................................................................37

1.5.1 Oxygen homeostasis ..................................................................................................................37

1.5.2 Hyperoxia ...................................................................................................................................38

1.5.3 Hyperoxia-induced lung injury .................................................................................................39

1.5.3.1 The effects of hyperoxia on the alveolar epithelium.............................................................40

1.6 Mechanical forces and the alveolar epithelium......................................................................43

1.6.1 Quantification of mechanical forces in situ .............................................................................43

1.6.2 The role of mechanical forces in lung development ................................................................44

1.6.2.1 The effect of mechanical forces on lung growth...................................................................44

1.6.2.2 Alveolar epithelial phenotype & differentiation...................................................................44

1.6.3 The effect of mechanical forces on the alveolar epithelium....................................................45

1.6.3.1 Surfactant secretion................................................................................................................45

viii

1.6.3.2 Phenotype................................................................................................................................46

1.6.3.3 Cytokine production ...............................................................................................................46

1.6.3.4 Apoptosis .................................................................................................................................47

1.6.3.5 Deformation-induced injury...................................................................................................47

1.6.4 Mechanical ventilation & lung injury: ventilator induced lung injury (VILI).......................48

1.6.4.1 Mechanisms of ventilator-induced lung injury .....................................................................49

1.6.4.2 The role of the alveolar epithelium in VILI...........................................................................52

1.7 Summary, thesis hypotheses and aims ....................................................................................53

1.7.1 Summary.....................................................................................................................................53

1.7.2 Hypotheses .................................................................................................................................54

1.7.3 Aims ............................................................................................................................................54

2. Materials and methods.......................................................................................................56

2.1 Primary alveolar epithelial type II cell isolation & culture.......................................................56

2.1.1 Alveolar epithelial type II cell isolation...................................................................................56

2.1.2 Modified Papanicolau staining & assessment of ATII purity.................................................57

2.1.3 Standard ATII cell culture.........................................................................................................58

2.1.4 ATII cell culture under hyperoxic conditions ..........................................................................59

2.1.5 ATII cell culture using the Bioflex! cell culture system..........................................................59

2.2 Protein quantification assay ........................................................................................................59

2.3 Measurement of cell viability in adherent cells ..........................................................................60

2.4 Lactate dehydrogenase (LDH)-cytotoxicity assay ......................................................................60

2.5 Feulgen staining & assessment of apoptosis ..............................................................................61

2.6 Estimation of reactive oxygen species (ROS) generation ..........................................................62

2.7 Estimation of ROS generation in response to oxidative stress ..................................................62

2.8 Quantification of antigens by ELISA-based dot blot assay........................................................63

2.9 Immunohistochemistry for fluorescence microscopy..................................................................64

2.9.1 General protocol for staining frozen sections .........................................................................65

2.9.2 General protocol for staining cultured ATII cells ...................................................................66

2.9.3 Bromo-deoxyuridine (BrdU) immunhistochemistry ................................................................66

2.9.4 Image acquisition.......................................................................................................................66

2.9.5 z-plane scanning and three-dimensional image reconstruction .............................................67

2.9.6 Image analysis ...........................................................................................................................68

2.9.6.1 Image density slicing ..............................................................................................................68

2.9.6.2 Quantification of epithelial surface staining ........................................................................68

2.9.6.3 Cell counts...............................................................................................................................68

2.9.6.4 Cell surface area.....................................................................................................................69

ix

2.10 Rat model of staphylococcus aureus pneumonia......................................................................69

2.10.1 Preparation of bacterial instillate ..........................................................................................69

2.10.2 Bacterial instillation................................................................................................................70

2.10.3 Initial recovery and randomisation ........................................................................................70

2.10.4 Housing under normoxic and hyperoxic conditions..............................................................70

2.10.5 Harvesting of blood, bronchoalveolar lavage (BAL) fluid and lung tissue.........................71

2.10.6 Bronchoalveolar lavage fluid processing ..............................................................................71

2.10.7 Lung tissue processing for biochemical analysis ..................................................................72

2.10.8 Lung tissue processing for morphological analysis ..............................................................72

2.11 Rat model of haemorrhagic shock-induced acute lung injury .................................................73

2.11.1 Animal preparation..................................................................................................................73

2.11.2 Experimental protocol.............................................................................................................74

2.12 Statistics.......................................................................................................................................74

3. The effect of hyperoxia on alveolar epithelial cells in culture .....................................76

3.1 Introduction & objectives ............................................................................................................76

3.2 Results ...........................................................................................................................................77

3.2.1 Characterisation of freshly isolated ATII cells........................................................................77

3.2.2 Change of morphology and phenotype of ATII cells in culture..............................................77

3.2.3 Identification of a novel intermediate cell phenotype .............................................................78

3.2.4 The immunoselectivity of the monoclonal antibody MMC6....................................................78

3.2.5 The effect of hyperoxia on ATII cells in culture.......................................................................80

3.2.6 The reversibility of hyperoxia-induced effects on ATII cells in culture .................................84

3.3 Discussion .....................................................................................................................................84

3.3.1 ATII cells transdifferentiate into ATI-like cells in culture ......................................................84

3.3.2 Identification of a novel intermediate cell phenotype .............................................................85

3.3.3 The MMC6 antigen appears to be ATI-selective .....................................................................85

3.3.4 Hyperoxia reversibly inhibits transdifferentiation in vitro .....................................................86

3.3.5 Hyperoxia is cytotoxic to alveolar epithelial cells ..................................................................87

3.3.6 A subpopulation of cells appears resistant to hyperoxia-induced injury...............................87

3.3.7 E-cadherin negative cells appear resistant to hyperoxia-induced injury ..............................88

3.3.8 Hyperoxia-resistant cells generate fewer ROS........................................................................89

3.3.9 Hyperoxia reversibly inhibits alveolar epithelial cell proliferation.......................................89

3.4 Summary .......................................................................................................................................90

4. The effect of hyperoxia on alveolar epithelial repair in vivo........................................91

4.1 Introduction & objectives ............................................................................................................91

4.2 Results ...........................................................................................................................................92

x

4.2.1 S. aureus-induced lung injury and the effect of hyperoxia......................................................92

4.2.2 The phenotype of the alveolar epithelium in control lung ......................................................93

4.2.3 The effect of hyperoxia on the proportion of ATI, ATII and intermediate cell-staining

membrane following S. aureus-induced lung injury .........................................................................94

4.2.4 The effect of hyperoxia on lung RTI40 concn following S.aureus-induced lung injury .........95

4.3 Discussion .....................................................................................................................................96

4.3.1 The effect of hyperoxic exposure on the resolution of lung injury .........................................96

4.3.2 The phenotype of the alveolar epithelium in control lungs.....................................................97

4.3.3 The phenotype of the alveolar epithelium in resolving S. aureus-induced lung injury.........97

4.3.4 The effect of hyperoxia on phenotype of the alveolar epithelium in resolving S. aureus-

induced lung injury .............................................................................................................................99

4.3.5 General discussion...................................................................................................................100

4.4 Summary .....................................................................................................................................102

5. Mechanical deformation-induced injury of alveolar epithelial cells ........................104

5.1 Introduction & objectives ..........................................................................................................104

5.2 Results .........................................................................................................................................105

5.2.1 Culture of alveolar epithelial cells in a mechanically active environment..........................105

5.2.2 Deformation-induced injury and magnitude of mechanical strain ......................................106

5.2.3 Deformation-induced injury and cell phenotype ...................................................................106

5.2.4 Deformation-induced injury in repairing wound...................................................................106

5.3 Discussion ...................................................................................................................................107

5.3.1 Deformation-induced injury is a non-linear function of deformation magnitude ...............107

5.3.2 Spreading RTI40-expressing cells in the peripheral region of epithelial cell monolayers are

susceptible to deformation-induced injury ......................................................................................108

5.3.3 Epithelial cells in the region of repairing epithelial monolayers appear susceptible to

deformation-induced injury ..............................................................................................................109

5.3.4 General discussion...................................................................................................................110

5.4 Summary .....................................................................................................................................112

6. Epithelial injury in a model of haemorrhagic shock-induced acute lung injury....113

6.1 Introduction ................................................................................................................................113

6.2 Results .........................................................................................................................................114

6.2.1 Characterisation of haemorrhagic shock-induced acute lung injury...................................114

6.2.2 Alveolar epithelial cell injury following haemorrhagic shock .............................................115

6.3 Discussion ...................................................................................................................................115

6.3.1 Haemorrhagic shock results in acute lung injury..................................................................115

6.3.2 Haemorrhagic shock is associated with ATI cell injury........................................................116

xi

6.4 Summary .....................................................................................................................................117

7. Summary ............................................................................................................................119

7.1 Hyperoxia inhibits the transdifferentiation of ATII cells into ATI cells..................................119

7.2 Spreading RTI40-expressing cells are susceptible to deformation-induced injury .................121

7.3 The multiple-hit hypothesis ........................................................................................................122

8. Future work .......................................................................................................................123

Figures ....................................................................................................................................125

Appendix I ..............................................................................................................................169

(A) Alveolar epithelial type I cell markers in adult lung ................................................................169

(B) Alveolar epithelial type II cell markers in adult lung...............................................................170

Appendix II Manufacturers and Suppliers.......................................................................171

Appendix III Buffers and Solutions ...................................................................................173

Bibliography .........................................................................................................................174

xii

Figure List

1. Schematic representation of alveolar epithelial repair

2. Primary ATII cell isolation

3. The immunophenotype of alveolar epithelial cells in culture

4. MMC4 & RTI40 coexpression in intermediate cells

5. Control lung stained with MMC6, RTI40 & TO-PRO-3

6. Control lung stained with MMC6, RTI40 & MMC4

7. 3D localisation of MMC6 in control lung

8. E16 lung stained with MMC6, RTI40 & TO-PRO-3

9. E17 lung stained with MMC6, RTI40 & TO-PRO-3

10. E20 lung stained with MMC6 & RTII70

11. E20 lung stained with MMC6, RTI40 & TO-PRO-3

12. MMC6 expression in alveolar epithelial cells in culture

13. 3D localisation of MMC6 in ATI-like cells

14. The effect of hyperoxia on adherent cell viability

15. The effect of hyperoxia on cell spreading

16. Hyperoxia & ATI cell-selective protein expression - immunofluoresence

17. Hyperoxia & ATI cell-selective protein expression - ELISA

18. Hyperoxia & ATII cell-selective protein expression - immunofluoresence

19. Hyperoxia & ATII cell-selective MMC4 expression - ELISA

20. E-cadherin expression in normoxia- & hyperoxia-exposed cells

21. The effect of hyperoxia on ROS generation

22. The effect of hyperoxia on alveolar epithelial cell proliferation

23. Hyperoxia-induced cell cytotoxicity

24. Hyperoxia-induced apoptosis in alveolar epithelial cells

25. Experimental protocol for S. aureus pneumonia model

26. BALF protein content in S. aureus pneumonia model

27. BALF cell counts in S. aureus pneumonia model

28. BALF RTI40 in S. aureus pneumonia model

29. Lung homogenate RTI40 in S. aureus pneumonia model

30. Quantification of alveolar epithelial surface staining

31. Expression of ATI & ATII cell-selective markers in control lung

xiii

32. Tiled image of normoxia-exposed S. aureus pneumonia

33. Epithelial repair in normoxia-exposed S. aureus pneumonia

34. Tiled image of hyperoxia-exposed S. aureus pneumonia

35. Epithelial repair in hyperoxia-exposed S. aureus pneumonia

36. ATI & ATII-staining membrane in S. aureus pneumonia model

37. Frequency distribution of ATII cell-staining membrane lengths

38. RTI40/MMC4 coexpressing membrane in S. aureus pneumonia model

39. The Bioflex! cell culture system & Flexercell FX-4000T strain unit"

40. Deformation-induced injury – Live/Dead! staining

41. Effect of deformation magnitude on deformation-induced injury

42. Relationship between lung volume and # epithelial BM surface area

43. Deformation-induced injury in central and peripheral monolayer regions

44. Cell viability data in central and peripheral monolayer regions

45. Phalloidin staining of central and peripheral monolayer regions

46. Immunophenotype of central and peripheral monolayer regions

47. Deformation-induced injury in repairing monolayer wounds

48. Cell viability data in repairing epithelial monolayer wounds

49. Protocol for haemorrhagic shock-induced acute lung injury model

50. Blood pressure profile of haemorrhagic shock and resuscitation

51. BALF protein content in haemorrhagic shock model

52. BALF cell counts in haemorrhagic shock model

53. BALF ATI cell-selective proteins in haemorrhagic shock model

xiv

Abbreviations

ALI acute lung injury

ANTU $-naphthylthiourea

APN aminopeptidase N

AQP aquaporin

ARDS acute respiratory distress syndrome

ARDSnet acute respiratory distress syndrome network

ATI alveolar epithelial type I

ATII alveolar epithelial type II

ATP adenosine triphosphate

BAB blood agar base

BALF bronchoalveolar lavage fluid

Bax Bcl-associated x protein

BB blocking buffer

BBT blocking buffer with triton

Bcl-2 B-cell lymphoma-2

BM basement membrane

BrdU 5-bromo-2’-deoxyuridine

BTS British Thoracic Society

Calcein AM calcein acetoxymethylester

CAP community acquired pneumonia

CC10 Clara cell-specific protein

CCSP Clara cell secretory protein

CMH2DCFDA 5-(6-)-chloromethyl-2’,7’-dichlorodihydrofluorescein diacetate

CO2 carbon dioxide

CRP C-reactive protein

CT computed tomographic

Cx connexin

d day/s

xv

DDW double distilled water

DIC differential interference contrast

DMEM Dulbecco’s modified eagle’s medium

DNA deoxyribonucleic acid

du densitometry units

EBMSA epithelial basement membrane surface area

ECM extracellular matrix

EDTA ethylenediaminetetraacetic acid

EGF epidermal growth factor

EGTA ethylene glycol tetraacetic acid

ELISA enzyme-linked immunosorbent assay

ENaC amiloride sensitive ion channel

EthD-1 ethidium homodimer-1

FBS foetal bovine serum

FGF fibroblast growth factor

FRC functional residual capacity

fu fluorescent units

GFP green fluorescent protein

GSF granulocyte colony-stimulating factor

GSH glutathione

h hour/s

HBSS Hank’s balanced salt solution

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HGF hepatocyte growth factor

HIF-1$ hypoxia-inducible factor-1alpha

3HT tritiated thymidine

HTI56 human alveolar epithelial type I cell 56 kDa protein

ICAM-1 intracellular adhesion molecule-1

ICU intensive care unit

IFN-% interferon-gamma

xvi

IgG immunoglobulin G

IGF insulin-like growth factor

IL interleukin

IL-1& interleukin-1 beta

IL-ra interleukin-receptor antagonist

INT iodotetrazolium chloride

IPF idiopathic pulmonary fibrosis

KGF keratinocyte growth factor

LDH lactate dehydrogenase

LEL lectin lycoperiscon esculentum

LPS lipopolysaccharide

LSCM laser scanning confocal microscope

mRNA messenger ribonucleic acid

Mab monoclonal antibody

MIF macrophage migration inhibitory factor

MIP macrophage inflammatory protein

MMC Mary McElroy

MODS multiple organ dysfunction syndrome

MOF multiple organ failure

NAD &-nicotinamide adenine dinucleotide, oxidised form

NADH &-nicotinamide adenine dinucleotide, reduced form

Na+/K

+-ATPase sodium-potassium adenosine triphophatase

NF-'B nuclear factor-kappa B

NIH National Institute of Health

NO nitric oxide

NO2 nitrogen dioxide

O2 oxygen

OCT optimal cutting temperature compound

PAMP pathogen-associated molecular pattern

Pap Papanicolau

xvii

PBS phosphate buffered saline

PDGF platelet-derived growth factor

PEEP positive end-expiratory pressure

PKC protein kinase C

PMS phenazine methosulphate

pO2 partial pressure of oxygen

Pro-SP-C pro-surfactant protein-C

PRR pattern recognition receptor

PVDF polyvinylidene difluoride

RGB red green blue

RNS reactive nitrogen species

ROS reactive oxygen species

RTI40 rat alveolar epithelial cell type I 40 kDa protein

RTII70 rat alveolar epithelial cell type II 70 kDa protein

S. aureus Staphylococcus aureus

SA surface area

#SA change in surface area

SOD superoxide dismutase

SP-A/B/C/D surfactant protein-A/B/C/D

SSA sulfasalazine

T1$ (alveolar) type I cell alpha protein

TBS tris(hydroxymethyl)aminomethane-buffered saline

TGF-& transforming growth factor-beta

TIFF tagged image file format

TJ tight junction

TLC total lung capacity

TLR toll-like receptor

TNF-$ tumour necrosis factor-alpha

TRIS tris(hydroxymethyl)aminomethane

UK United Kingdom of Great Britain and Northern Ireland

xviii

US/USA United States/United States of America

VAP ventilator-associated pneumonia

VILI ventilator-induced lung injury

ZO zonna occludens

1

1. Introduction

Injury to the lung, resulting in impaired gas exchange and acute respiratory failure, is an

important cause of morbidity and mortality (Lewandowski et al. 1995; Behrendt 2000).

The alveolar epithelium plays a central role in the pathogenesis of this injury in a range

of common clinical conditions (Berthiaume et al. 1999; Newman et al. 2000; Ware and

Matthay 2000; Hippenstiel et al. 2006), with effective repair of the alveolar epithelial

barrier following injury an important determinant of clinical outcome (Adamson et al.

1988; Matthay and Wiener-Kronish 1990; Ware and Matthay 2001). Supplemental

oxygen and mechanical ventilation are key components of the supportive treatment of

respiratory failure but the effects of these therapeutic interventions in the injured lung

and on alveolar epithelial repair following injury are largely unknown. The clinical

impression however, is that poor outcome is associated with exposure of injured

(repairing) epithelium to such iatrogenic ‘hits’ (Ranieri et al. 1999; ARDSNet 2000;

Frank et al. 2002).

This thesis describes studies investigating the hypothesis that hyperoxia & mechanical

deformation inhibit normal epithelial repair following injury. In order to put these

studies into context, the introduction outlines the structure and function of the normal

alveolus and discusses the role of the alveolar epithelium in the pathogenesis of disease.

The process of alveolar epithelial repair is then discussed followed by a review of the

effects of hyperoxia and mechanical deformation in the lung.

1.1 Structure and function of the alveolus

The primary function of the lung is gas exchange. Inspired gases pass into the trachea

and through a series of branching conducting airways until they reach the respiratory

bronchioles, which have occasional alveoli budding from their walls, and finally the

alveolar ducts which are completely lined with alveoli. This alveoli-containing region of

2

the lung is known as the respiratory zone, and is where gas exchange occurs. In the

human lung, this region contains 300 million or so alveoli, with a total surface area in

the region of 50-100m2 (West, Weibel & Gomez 1962).

1.1.1 Structure of the air-blood barrier

Oxygen (O2) moves from air within alveoli into venous blood, and carbon dioxide (CO2)

from venous blood into alveoli. This process occurs by passive diffusion across the air-

blood barrier. This is a three-layered structure comprising pulmonary capillary

endothelium, extracellular matrix (ECM) and alveolar epithelium.

1.1.2 Pulmonary endothelium

The pulmonary vessels are lined by a squamous unistratified epithelium of mesodermal

origin, the endothelium. Alveolar capillary endothelial cells are large attenuated cells

(surface area (1000-1300µm2), with their nuclei usually oriented in the direction of

blood flow. Ultrastructurally, endothelial cells demonstrate a relative paucity of

intracellular organelles but have a large number of plasmalemmal transport vesicles,

thought to reflect adaptation of the vascular endothelium to its role in the transport of

macromolecules (Simionescu 1997). Joined by tight junctions, alveolar capillary

endothelial cells form a continuous (non-fenestratred) polarised monolayer with the

luminal surface of the cells in contact with blood, and the abluminal surface resting on

basement membrane.

In addition to its structural role as part of the air-blood barrier, the pulmonary

endothelium plays a pivotal role in regulating blood flow, coagulation and fibrinolysis

(Fishman 1982; Cines et al. 1998). Of central importance, is the role the endothelium

plays in controlling leucocyte migration from the blood, through the alveolar

interstitium, and into the alveolar airspace.

Leucocyte migration across the endothelial barrier occurs in many tissues as part of

normal immunosurveillance. However, the level of this cell migration can be increased

3

dramatically following endothelial cell activation - a term used to describe a change in

chemokine production and adhesion molecule expression on the luminal surface of the

endothelium in response to stimuli such as injury or infection (Cines et al. 1998).

Human lung microvascular endothelial cell activation has been demonstrated in vitro

following stimulation with tumour necrosis factor-$ (TNF-$) and interferon-% (IFN-%)

(Hillyer et al. 2003), and results in increased chemokine production and increased

expression of the adhesion molecules E-selectin and intercellular adhesion molecule-1

(ICAM-1). E-selectin, a member of the selectin family of transmembrane glycoproteins,

initiates the tethering and rolling of leucocytes on the endothelial cell surface (McEver et

al. 1995). Subsequent leucocyte activation enables &2 integrins on leucocytes to bind to

ICAM-1 (Springer 1995), and this is followed by migration of leucocytes between

endothelial cells into the tissues. These processes are thought to be of key importance in

the pathogenesis of acute lung injury (section 1.2.2.3) (Zimmerman et al. 1999).

1.1.3 Alveolar epithelium

The alveoli are lined by a continuous layer of epithelium comprised of two

morphologically distinct epithelial cell types: alveolar epithelial type I (ATI) and

alveolar epithelial type II (ATII) cells (Low 1952). Morphometric studies have

demonstrated that the distribution and morphology of these cell types is remarkably

similar across mammalian species including the rat and human (Crapo et al. 1983; Stone

et al. 1992).

1.1.3.1 Alveolar epithelial type I (ATI) cells

ATI cells comprise only (8-9% of the parenchymal cell population within the lung, but

cover (95% of the alveolar surface (Haies et al. 1981; Crapo et al. 1982).

Morphologically, ATI cells are large (luminal surface (5100µm2), flat (thickness

(0.2µm) and highly branched, with their thin attenuated cytoplasm facilitating passive

diffusion of gases across the cell (Weibel and Gomez 1962; Haies et al. 1981; Weibel

4

1984). Ultrastructurally, ATI cells appear relatively simple, with a small nucleus, a few

small mitochondria and inconspicuous endoplasmic reticulum and Golgi apparatus

(Schneeberger 1991). This paucity of organelles was thought to suggest little functional

role for ATI cells other than the passive facilitation of gas exchange, but more recently

ATI cells have been shown to have an important role in regulating alveolar fluid balance

(section 1.1.6) and, through mechanotransduction, the control of pulmonary surfactant

secretion (Dobbs et al. 1998; Johnson et al. 2002; Williams 2003).

1.1.3.2 Alveolar epithelial type II (ATII) cells

ATII cells comprise (15% of lung parenchymal cells, but cover only (3-5% of the

alveolar surface (Haies et al. 1981; Crapo et al. 1982). Unlike ATI cells, they are

cuboidal in shape (luminal surface (60µm2, thickness (3µm) with apical microvilli and

characteristic surfactant-containing organelles known as lamellar bodies (Crapo et al.

1983; Mason and Williams 1991). Typically located in the corners of alveoli, these cells

usually occur singly flanked by ATI cells. Known functions of ATII cells include the

synthesis, secretion and recycling of pulmonary surfactant, regulation of alveolar fluid

and electrolyte balance (section 1.1.6), host defence and immunomodulation (section

1.2.3.4) (Fehrenbach 2001). In addition to their synthetic and secretory capacities, ATII

cells function as the stem cell of the alveolar epithelium and have a central role in

alveolar epithelial repair (section 1.3.3.1).

1.1.4 Alveolar epithelial cell junctions

ATI and ATII cells are joined by both tight junctions (TJ) and gap junctions

(Schneeberger 1991).

5

1.1.4.1 Tight junctions

Tight junctions are gasket-like strands of protein connecting the apical portion of

alveolar epithelial cells and separating the apical and basolateral surfaces of the

epithelium (Schneeberger and Lynch 1992). They are thought to provide the primary

rate-limiting barrier to paracellular transport, with the majority of the barrier function

being provided by the integral membrane protein occludin (Saitou et al. 1997) and a

family of TJ proteins, the claudins (Wang et al. 2003). Tight junction-associated proteins

zona occludens (ZO)-1, -2 and -3, are intracellular proteins thought to act as a link

between the TJ and cytoskeletal proteins, although the functional significance of these

interactions is not known (Denker and Nigam 1998; Goodenough 1999).

Alveolar epithelial TJs are not static structures and appear to be highly regulated, with

the cytoskeleton playing a central role (Schneeberger and Lynch 1992). For example,

low levels of extracellular calcium and low levels of intracellular ATP (ATPi) have both

been shown to result in TJ dissociation, whilst activation of protein kinase C (PKC)

decreases TJ permeability (Gonzalez-Mariscal et al. 1985; Bacallao et al. 1994; Denker

and Nigam 1998). TJ dissociation, with reduced resistance of the paracellular pathway,

occurs in both hydrostatic and injury-induced pulmonary oedema and has been shown to

occur in response to inflammatory mediators in mice (Gon et al. 2005), hypoxia (Bouvry

et al. 2006) and pathological levels of mechanical strain in primary rat alveolar epithelial

cells (Cavanaugh et al. 2001).

1.1.4.2 Gap junctions

Gap junctions are transcellular channels that allow the passive diffusion of small

molecules and ions between adjacent cells. In vertebrates, gap junctions are composed of

hexamers of a family of gap junction proteins, the connexins (Cx) (Kumar and Gilula

1996; Alberts 2002). At least six different connexins are expressed in alveolar epithelial

cells, with ATI and ATII cells having different expression profiles (Abraham et al. 1999;

Isakson et al. 2003). The differential expression and post-translational modification of

6

these proteins is thought to influence gap junction permeability and function, and

appears to be highly regulated, although the functional significance of this remains

unclear (Isakson et al. 2003).

1.1.5 Extracellular matrix and structural features of the alveolar wall

Extracellular matrix (ECM) may be divided into two categories: basement membrane

(BM) and interstitial connective tissue (Kumar et al. 2004).

1.1.5.1 Alveolar basement membranes

Basement membranes are condensed sheet-like structures separating endothelial and

epithelial cells from interstitial connective tissue. In both the rat and human lung, the

alveolar epithelial BM and alveolar capillary BM has been shown to consist of highly

cross-linked type IV collagen, laminin and fibronectin (Gil and Martinez-Hernandez

1984; Kumar et al. 2004). The composition of the BM is thought to play a significant

role in preserving normal alveolar structure and function. In vitro, the ECM on which

alveolar epithelial cells are cultured has been shown to have a significant effect on cell

phenotype, with laminin-rich surfaces favouring retention of the differentiated ATII-like

phenotype and fibronectin-rich surfaces accelerating loss of the ATII phenotype and

acquisition of an ATI-like phenotype (Rannels and Rannels 1989; Olsen et al. 2005).

Fibronectin is believed to be involved in epithelial cell regeneration in many tissues

(Fujikawa et al. 1981; Clark et al. 1982), and comparison of the level and distribution of

fibronectin expression in normal and fibrotic lungs supports the hypothesis that

fibronectin plays an important role in alveolar epithelial cell repair (Torikata et al. 1985).

1.1.5.2 Alveolar interstitial connective tissue

The major components of alveolar interstitial connective tissue are types I collagen, type

7

III collagen and fibronectin (Gil and Martinez-Hernandez 1984; Kumar et al. 2004).

1.1.5.3 Structural features of the alveolar wall

In the rat lung, the alveolar wall has been shown to consist of thin areas, where capillary

endothelium and alveolar epithelium are separated by only a single fused alveolar and

capillary basement membrane, and thick areas, where endothelium and epithelium are

separated by their respective basement membranes and connective tissue of the

interstitial space (Vaccaro and Brody 1981). These differences suggest that the alveolar

basement membrane may contain functional and structural microdomains that regulate

alveolar epithelial cell phenotype: the single basement membrane separating capillary

blood from alveolar air in areas of thin alveolar wall provides the thinnest possible

barrier to gaseous diffusion, and during normal development and repair these areas

appear to favour the acquisition of an ATI phenotype. In contrast, areas of thick alveolar

wall, typically situated along alveolar septa and in the corners of alveoli, favour the

retention of an ATII phenotype (Sannes 1984; Lwebuga-Mukasa 1991). This is thought

to represent a plausible hypothesis to explain the relatively constant numerical ratio of

ATI and ATII cells and the non-random localisation of ATII cells within the lung

(Lwebuga-Mukasa 1991). Further evidence supporting the concept of basement

membrane-associated microdomains is the finding that the BM underlying ATI and ATII

cells differs in the distribution of cytochemically detected sulphate esters (Sannes 1984).

1.1.6 Alveolar epithelial transport

Current evidence suggests that the alveolar epithelium is at least an order of magnitude

less permeable to protein and small solutes than the pulmonary endothelium, and hence

is the main regulator of fluid and solute exchange across the air-blood barrier (Taylor

and Gaar 1970; Normand et al. 1971). The alveolar epithelium is more than a passive

barrier however, and plays an active role both in fluid homeostasis in the normal

alveolus and critically, in the reabsorption of fluid that enters the alveolus following

8

injury (Carden et al. 1998; Crandall and Matthay 2001).

1.1.6.1 The role of sodium transport

Active sodium transport appears to be the primary mechanism driving fluid reabsorption

within the alveolus. Sodium ions enter the apical membrane of alveolar epithelial cells,

at least in part, through amiloride-sensitive ion channels (ENaC) and are actively

transported across the basolateral membrane by Na+/K

+-ATPase. Chloride and water

follow the transport of sodium through both transcellular and paracellular pathways to

maintain electrochemical and osmotic neutrality across the alveolar epithelial membrane

(Crandall and Matthay 2001).

Initial in situ immunocytochemical studies demonstrated expression of Na+/K

+-ATPase

in ATII cells but not ATI cells, leading to the inference that alveolar fluid transport was

regulated largely by ATII cells, with ATI cells playing only a passive role (Schneeberger

and McCarthy 1986). However, subsequent in vitro studies have shown that ATI cells

not only have the highest known water permeability of any mammalian cell type, but

also express Na+/K

+-ATPase, suggesting that ATI cells may also play a significant role

in vectorial ion and water transport (Dobbs et al. 1998; Johnson et al. 2002).

1.1.6.2 The role of aquaporin water channels

Water transport down the osmotic gradient generated by active sodium transport is

thought to occur predominantly through aquaporin (AQP) water channels. The

aquaporins are a family of hydrophobic integral membrane proteins, four members of

which have been shown to be expressed in the lung (Verkman et al. 2000). AQP1 has

been immunolocalised to the microvascular endothelium and to a limited extent ATII

cells (Effros et al. 1997), AQP3 has been shown to be expressed on the basolateral

membrane of ATII cells (Kreda et al. 2001), whilst both AQP4 and AQP5 appear to be

expressed on the apical surface of ATI cells (Kreda et al. 2001; Williams 2003).

9

The precise role of aquaporins in the maintenance of alveolar fluid homeostasis remains

unclear. Deletions of AQP1 and AQP5 produce a dramatic decrease in osmotically

driven water transport, although not in isosmolar fluid transport (Bai et al. 1999; Ma et

al. 2000), and although AQP4 is thought to be the main aquaporin responsible for

removal of fluid from the neonatal alveolus, AQP1, AQP4 and AQP5 knock out mice

appear to have the same ability to clear lung fluid in the perinatal period as wild type

mice (Song et al. 2000). In addition, deletion of AQP1, AQP4 and AQP5 does not effect

the formation or clearance of lung oedema following acid- or hyperoxia-induced lung

injury (Song et al. 2000).

1.1.7 Air-blood barrier function

Because the epithelial barrier is much less permeable than the endothelial barrier,

epithelial injury removes a significant physical barrier to the passage of fluid into the

alveolus (Wiener-Kronish et al. 1991). In addition, loss of alveolar epithelial integrity

significantly disrupts normal epithelial fluid transport, impairing alveolar fluid clearance

capacity (Ware and Matthay 2001; Matthay et al. 2002; Matthay et al. 2005). It is

therefore unsuprising that preservation of alveolar epithelial barrier integrity appears to

have significant prognostic value in acute lung injury (ALI), with patients who maintain

a functional epithelial barrier having a better chance of survival than those with loss of

epithelial barrier integrity (Matthay and Wiener-Kronish 1990; Carden et al. 1998).

The majority of patients who die from acute lung injury do not do so from respiratory

failure, but from multiple organ failure (MOF)(Montgomery et al. 1985; Wyncoll and

Evans 1999). One mechanism postulated for this finding is that disruption of the air-

blood barrier allows the release of pro-inflammatory cytokines produced in the lung into

the pulmonary circulation with the subsequent development of systemic inflammation

and multiple organ dysfunction syndrome (MODS) (Douzinas et al. 1997; Slutsky and

Tremblay 1998). Loss of compartmentalisation of alveolar TNF-$ has been

demonstrated following $-naphthylthiourea (ANTU)-induced lung injury in rats (Tutor

10

et al. 1994), and also after Pseudomonas aeruginosa pneumonia in rabbits (Kurahashi et

al. 1999). In humans with acute lung injury, conventional mechanical ventilation is

associated with both a pulmonary and systemic cytokine response. Both of these

responses are attenuated by lung-protective ventilation designed to reduce further injury

to the air-blood barrier, supporting the concept that release of pro-inflammatory

cytokines from the lung into the systemic circulation may be related to integrity of the

air-blood barrier (section 1.6.4.1.1) (Ranieri et al. 1999).

Loss of alveolar compartmentalisation following disruption of the air-blood barrier has

also been shown to facilitate bacterial translocation from the alveoli into the systemic

circulation in both hamsters and dogs (Johanson et al. 1985; Nahum et al. 1997).

Changes in the expression of adhesion molecules following activation or injury of the

alveolar epithelium may play an important part in the further recruitment and migration

of inflammatory cells across the epithelial barrier. In support of this concept, acid

induced-injury, hyperoxia, and the inflammatory cytokines interleukin-1 (IL-1) and

TNF-$ have all been shown to upregulate alveolar epithelial cell expression of ICAM-1

(Tosi et al. 1992; Ohwada et al. 2003). The role of the endothelium in controlling

leucocyte migration from the blood, through the alveolar interstitium, and into the

alveolar air space is discussed above (section 1.1.2).

11

1.2 The role of the alveolar epithelium in disease pathogenesis

1.2.1 The consequences of alveolar epithelial injury

The alveolar epithelium has key structural and functional roles in maintaining normal

lung function. Alveolar epithelial injury, with disruption of the integrity of the air-blood

barrier, has profound consequences:

i. Loss of epithelial barrier function: Epithelial cell injury and tight junction disruption

removes the most significant physical barrier to the passage of fluid into the alveolus

and alveolar flooding results. Upregulation of adhesion molecules following injury

also facilitates inflammatory cell recruitment and trafficking across the air-blood

barrier (section 1.1.7).

ii. Impaired alveolar fluid clearance: Disruption of normal epithelial fluid transport

impairs the removal of oedema fluid from the alveolar air spaces (sections 1.1.6 &

1.1.7).

iii. Surfactant abnormalities: ATII cell injury impairs normal production, secretion and

recycling of pulmonary surfactant. This leads to loss of surface tension-reducing

phospholipids, alveolar instability and collapse (Lewis and Jobe 1993; Greene et al.

1999).

iv. Impaired innate immunity: The alveolar epithelium forms a significant mechanical

barrier to microbial invasion, and plays a central role in pathogen recognition and the

initiation of the innate immune response. Injury, together with surfactant depletion,

impairs this response (section 1.2.3.4).

v. Loss of alveolar compartmentalisation: Physical disruption of the epithelial barrier

may result in loss of compartmentalisation within the lung allowing the release of

bacteria and inflammatory mediators into the systemic circulation (section 1.1.7).

vi. Pulmonary fibrosis: In severe epithelial injury, disorganised or inadequate epithelial

repair may lead to fibrosis (section 1.3.4)(Adamson et al. 1988; Bitterman 1992).

It is unsurprising given the extent of these consequences that alveolar epithelial injury,

12

and effective repair of the alveolar epithelium following injury, play a central role in the

pathogenesis and resolution of a wide range of clinical conditions. Three such

conditions, with relevance to the work presented in this thesis are acute lung injury,

bacterial pneumonia and & idiopathic pulmonary fibrosis.

1.2.2 Acute respiratory distress syndrome (ARDS) & acute lung injury (ALI)

1.2.2.1 Historical perspective and definitions

The first description of respiratory distress syndrome was published in 1967, when

Ashbaugh and colleagues described patients in intensive care with acute respiratory

distress, hypoxaemia refractory to oxygen therapy, decreased lung compliance, and

diffuse alveolar infiltrates on chest radiograph (Ashbaugh et al. 1967). Initially called

the adult respiratory distress syndrome (Petty and Ashbaugh 1971), the term acute

respiratory distress syndrome (ARDS) was subsequently adopted, as the condition is

also known to occur in children. In 1994, the American-European Consensus

Conference Committee on ARDS recommended a new, and now widely accepted

definition, in which the severity of clinical lung injury was recognised. Based upon this

definition, patients with less severe hypoxaemia (defined by a ratio of the partial

pressure of arterial oxygen to the fraction of inspired oxygen) are considered to have

acute lung injury (ALI), whilst those with more severe hypoxaemia are considered to

have the acute respiratory distress syndrome (table 1) (Bernard et al. 1994). In addition,

the American-European Consensus Conference definition recognised that ALI is

associated with a precipitating or predisposing event. These events can be separated into

those associated with direct injury to the lung, such as pneumonia, and those causing

indirect injury in the setting of a systemic process, such as sepsis or haemorrhage (table

2).

13

Onset Oxygenation Chest radiograph Pulmonary artery occlusion pressure

ALI Acute paO2/FiO2 ! 300 Bilateral infiltrates ! 18mmHg / no left atrial hypertension

ARDS Acute paO2/FiO2 ! 200 Bilateral infiltrates ! 18mmHg / no left atrial hypertension

Table 1 Definitions of acute lung injury (ALI) & the acute respiratory distress syndrome

(ARDS) from American-European Consensus Conference 1994 (Bernard, Artigas et al. 1994).

paO2 in mmHg.

Direct injury Indirect injury

Pneumonia

Aspiration

Lung contusion

Near drowning

Toxic inhalation

Sepsis

Transfusion-related

Pancreatitis

Severe non-thoracic trauma

Cardiopulmonary bypass

Table 2 Clinical disorders associated with development of ALI & ARDS, as identified by the

American-European Consensus Conference Committee 1994 (Bernard, Artigas et al. 1994).

1.2.2.2 Epidemiology of acute lung injury

Incidence: Accurate estimation of the incidence of ALI has been hindered historically by

lack of uniform diagnostic criteria coupled with the heterogenicity of the causes and

clinical manifestations of the disease (Ware and Matthay 2000). Because of this, the

reported incidence has varied widely from 2 to 75 cases per 100,000 person-years (NIH

1977; Webster et al. 1988; Luhr et al. 1999). However, a large prospective population-

based cohort study using the 1994 consensus definitions recently estimated the age-

adjusted incidence of ALI in the United States as 86 per 100,000 person-years, and of

ARDS as 64 per 100,000 person-years (Rubenfeld et al. 2005). The associated in-

14

hospital mortality in this study was 39%, in keeping with that reported in previous

studies of 40-60% (Ware and Matthay 2000). These data suggest that there are 200,000

cases of ALI each year in the United States, associated with 74,500 deaths.

Outcome: ARDS has a mortality of 30-40% in the UK (Abel et al. 1998). Of the patients

who die, the majority of deaths are attributed to sepsis or multiple organ failure rather

than primary respiratory failure (Montgomery et al. 1985; Wyncoll and Evans 1999). It

is likely, however, that pulmonary dysfunction contributes significantly to these

processes. The respiratory failure associated with ALI often necessitates mechanical

ventilatory support, and the recent therapeutic success of ventilation with low tidal

volumes appears to suggest that in some cases death may be directly related to a

combination of existing and iatrogenic lung injury (ARDSNet 2000).

Patients who survive ALI have been shown to have a reduced health-related quality of

life (Weinert et al. 1997) and persistent functional limitation, largely as a result of

muscle wasting and weakness (Herridge et al. 2003). Although mild restrictive patterns

on lung-function testing, and mild-moderate reductions in carbon monoxide diffusion

capacity have been demonstrated, these abnormalities are usually asymptomatic

(Herridge et al. 2003), and in most patients, pulmonary function returns to normal within

6-12 months (McHugh et al. 1994).

1.2.2.3 Pathogenesis of acute lung injury

The acute phase of ALI, corresponding to the first few days following the onset of

injury, is characterised by the influx of protein-rich oedema fluid and inflammatory cells

into the alveolar air spaces with the formation of characteristic hyaline membranes. This

has been termed the exudative phase of ALI, and arises as a consequence of both

endothelial and epithelial cell injury with increased permeability of the alveolar-

capillary barrier (Pugin et al. 1999; Ware and Matthay 2000; Tomashefski 2003). The

importance of endothelial injury and increased vascular permeability in the development

of pulmonary oedema has been well established, but the most conspicuous structure

15

damaged in the acute phase of ALI appears to be the alveolar epithelium, particularly

ATI cells (Bachofen and Weibel 1982; Berthiaume et al. 1999; Ware and Matthay

2000). The significance of epithelial injury is discussed below (section 1.2.2.5).

The second histopathological stage in the natural history of ALI, known as the

proliferative phase, is defined by the proliferation of ATII cells along alveolar septa and

the onset of epithelial regeneration (Tomashefski 2003). In uncomplicated cases,

denuded basement membrane is reepithelialised with restoration of normal alveolar

architecture and epithelial fluid transport capacity and rapid reabsorption of alveolar

oedema. However, in some patients alveolar oedema persists, mesenchymal cells fill the

alveolar spaces with subsequent organisation and progression to fibrotic lung injury

(Bachofen and Weibel 1982; Ware and Matthay 2000).

1.2.2.3.1 Inflammatory mechanisms in the pathogenesis of acute lung injury

Cytokines: A complex balance of pro- and anti-inflammatory cytokines appears to

initiate and regulate the inflammatory response in acute lung injury (Ware and Matthay

2000; Park et al. 2001). Macrophage migration inhibitory factor (MIF) is released from

the anterior pituitary and alveolar macrophages in response to a range of known

precipitants of lung injury such as trauma and sepsis (Joshi et al. 2000; Lehmann et al.

2001). MIF not only inhibits the anti-inflammatory effects of glucocorticoids (Calandra

et al. 1995), but also stimulates the release of the major neutrophil chemoattractant,

interleukin-8 (IL-8), and the pro-inflammatory mediators TNF-$, interleukin-1 (IL-1)

and interleukin-6 (IL-6) from alveolar macrophages (Donnelly et al. 1997; Abraham et

al. 2000).

Specific inhibitors of the proinflammatory cytokines, including IL-1-receptor antagonist

(IL-1ra), soluble TNF receptor (sTNF-R) and autoantibodies against IL-8 have been

described. In conjunction with the non-specific anti-inflammatory cytokines interleukin-

10 (IL-10) and interleukin-11 (IL-11), these mediators are thought to modulate

inflammatory lung injury, a hypothesis supported by the finding that low concentrations

16

of IL-10 and IL-1ra in the bronchoalveoalr fluid of patients with ARDS appears to be

associated with increased mortality (Donnelly et al. 1996; Pittet et al. 1997; Ware and

Matthay 2000; Park et al. 2001).

The role of the neutrophil: Histological studies have consistently shown a marked

accumulation of polymorphonuclear neutrophils in the acute phase of ALI, suggesting

that neutrophils play an important role in the pathogenesis of lung injury (Bachofen and

Weibel 1982; Abraham et al. 2000; Lee and Downey 2001; Martin 2002). Many animal

models of ALI appear to be neutrophil-dependent, and inhibiting neutrophil recruitment

by blocking IL-8 appears protective against acid-induced lung injury (Folkesson et al.

1995). However whether the neutrophil is the cause or result of lung injury remains

unclear. Some animal models appear neutrophil-independent, and patients with profound

neutropenia may still develop ALI (Laufe et al. 1986; Wiener-Kronish et al. 1991). In

addition, studies using granulocyte colony-stimulating factor (G-CSF) in humans with

severe pneumonia have demonstrated that increasing the number of circulating

neutrophils is not associated with an increase in either the incidence or severity of lung

injury (Nelson et al. 1998).

1.2.2.4 Treatment of acute lung injury

For many years, treatment of ALI was limited to the supportive treatment of organ

failure and treatment of the underlying cause of lung injury. Improvements in this

supportive treatment, rather than the success of a particular therapeutic intervention, are

thought to account for the recently observed decline in mortality from (40-60% to (30-

40% (Abel et al. 1998; Ware and Matthay 2000).

Despite significant advances in the understanding of the pathogenesis of ALI, specific

pharmacological interventions, including surfactant therapy, inhaled nitric oxide,

glucocorticoids and other anti-inflammatory agents, have shown little benefit (Adhikari

et al. 2004). However, a study by the ARDS Network (ARDSNet) comparing

conventional mechanical ventilation with a lung protective strategy of ventilation

17

reported a 22% reduction in mortality in patients ventilated using protective ventilation

(section 1.6.4.1.2) (ARDSNet 2000).

1.2.2.5 The significance of the alveolar epithelium in acute lung injury

The importance of epithelial injury in the development of ALI is now widely recognised

(Bachofen and Weibel 1982; Wiener-Kronish et al. 1991; Berthiaume et al. 1999; Ware

and Matthay 2000). In human studies, acute lung injury is associated with a significant

rise in both plasma and bronchoalveolar lavage fluid levels of the human ATI cell-

specific apical plasma membrane protein, HTI56, and the extent of alveolar epithelial

injury, as determined by the degree of impairment of alveolar fluid clearance, appears to

be an important predictor of outcome (Newman et al. 2000; Ware and Matthay 2001).

Alveolar epithelial recovery appears to be a key feature in surviving ALI, with

restoration of intact epithelial barrier function and normal alveolar fluid transport, being

associated with improved outcome (Matthay and Wiener-Kronish 1990; Ware and

Matthay 2001). However, abnormal alveolar epithelial repair appears to be a key

pathogenic mechanism in the development of fibrosis (Adamson et al. 1988).

Protective lung ventilation is the only therapeutic intervention of any proven benefit in

the treatment of ARDS (ARDSNet 2000), and the knowledge that the alveolar

epithelium plays a central role in the pathogenesis of ventilator-induced lung injury

(section 1.6.4.2), further highlights the central role of the alveolar epithelium in the

pathogenesis and recovery from acute lung injury.

1.2.3 Bacterial pneumonia

1.2.3.1 Definition, aetiology and clinical presentation

Pneumonia is defined as an inflammation of the lung parenchyma. It is typically caused

18

by microbial pathogens, including bacteria, viruses, fungi and parasites. A number of

modifying terms are used to classify pneumonia: acute or chronic dependent on the

duration of symptoms; lobar or bronchopneumonia dependent on chest radiograph

findings; community-acquired or hospital-acquired (also called nosocomial pneumonia)

dependent on the place of acquisition (Weatherall et al. 2003). Ventilator-associated

pneumonia (VAP), is a common form of hospital-acquired pneumonia, and describes

pneumonia occurring in patients who have required mechanical ventilation for at least

48 hours. The common aetiological agents causing pneumonia are listed in tables 3 & 4

(Bartlett et al. 1986; BTS 2001).

Aetiological organism %

Streptococcus pneumoniae

Haemophilus influenzae

Legionella spp

Staphylococcus aureus

Gram negative bacilli

Mycoplasma pneumoniae

Chlamydia psittaci

Influenza A & B

No pathogen isolated

36.0

10.2

0.4

0.8

1.3

1.3

1.3

8.1

45.3

Table 3 Causes of community-acquired pneumonia (CAP) (BTS 2001).

Aetiological organism %

Gram negative bacilli

Anaerobic bacteria

Staphylococcus aureus

Streptococcus pneumoniae

47

35

31

26

Table 4 Causes of hospital-acquired pneumonia (Bartlett, O’Keefe et al. 1986).

The clinical presentation of pneumonia varies according to the aetiological agent, but

typical features include cough and fever with the variable presence of sputum, dyspnoea

and pleuritic chest pain. Haematological investigations typically demonstrate a raised

white cell count and C reactive protein (CRP). Although radiological changes may lag

19

behind the clinical course, a chest radiograph confirming areas of consolidation remains

the most reliable clinical investigation in the diagnosis. Confirmation of the microbial

cause of pneumonia typically depends on sputum culture, blood culture and serology.

However, the aetiological agent remains unidentified in (45% of cases (BTS 1992; BTS

2001; Weatherall et al. 2003).

1.2.3.2 Epidemiology of bacterial pneumonia

Pneumonia is the third leading cause of death worldwide and the leading cause of death

due to infectious disease in industrialised countries (Hippenstiel et al. 2006). The annual

incidence of community acquired pneumonia (CAP) in the UK is thought to be 5-11 per

1000, with (20-40% of these cases requiring hospital admission (Woodhead et al. 1987;

Guest and Morris 1997). The mortality rate varies widely, but appears to be (5-12% in

those requiring hospitalisation, rising to more than 50% in patients requiring admission

to an intensive care unit (ICU) (Woodhead et al. 1985; BTS 1992; BTS and Service

1992; Hirani and Macfarlane 1997). Hospital-acquired pneumonia occurs in up to 10%

of patients admitted to hospital, with ventilator-associated pneumonia having an

incidence of (8-28% and an associated mortality of (24-50% (Chastre and Fagon 2002).

1.2.3.3 Pathogenesis of bacterial pneumonia

In animal models, microbial pathogens trigger an inflammatory response resulting in

both epithelial and endothelial cell injury (Seeger et al. 1990; McElroy et al. 1995;

Clegg et al. 2005). In bacterial pneumonia, the primary inflammatory trigger for this

appears to be the bacterial cell wall, with pepdtidoglycan binding to alveolar

macrophage cell surface receptors initiating a pro-inflammatory cytokine cascade

(McCullers and Tuomanen 2001). This leads to the release of the neutrophil

chemoattractants macrophage inflammatory protein-2 (MIP-2) and IL-8, activation of

the transcription factor nuclear factor kappa B (NF-'B) and release of the pro-

20

inflammatory cytokines TNF-$, IL-1 and IL-6 (Spellerberg et al. 1996; Delclaux and

Azoulay 2003). Direct activation of the alternative complement pathway by bacterial

cell wall components, such as teichoic acid, provides a further chemotactic signal for

leucocytes (Winkelstein and Tomasz 1978).

Disruption of the air-blood barrier results in flooding of the alveolar air space with a

protein-rich exudate containing bacteria, macrophages, neutrophils and erythrocytes. In

areas of confluent injury, this leads to lung consolidation known as red hepatisation

(Kumar et al. 2004). As erythrocytes disintegrate, the influx of leucocytes continues and

bacteria are cleared slowly by inefficient complement-mediated opsonisation and

phagocytosis. The pathological appearance changes from red to grey hepatisation during

this time.

Reduction of inflammation and resolution of pneumonia is dependent upon the

appearance of specific anti-microbial antibodies enabling efficient opsonisation and

phagocytosis by alveolar macrophages. Bacterial clearance is accompanied by

enzymatic and cellular degradation of consolidated alveolar exudate, with restoration of

normal alveolar structure and function (McCullers and Tuomanen 2001; Kumar et al.

2004).

1.2.3.4 The significance of the alveolar epithelium in bacterial pneumonia

Localised at the interface between the environment and the host, the pulmonary

epithelium not only forms a significant mechanical barrier to microbial invasion, but

also appears to play a central role in pathogen recognition and the initiation of the innate

immune response (Hippenstiel et al. 2006).

Transmembrane toll-like receptors (TLRs) (Akira and Takeda 2004; Armstrong et al.

2004), together with a family of cytosolic receptors known as the NLR family (Martinon

and Tschopp 2005), act as pattern recognition receptors (PRRs), detecting a variety of

highly conserved pathogen-associated molecular patterns (PAMPs) (Janeway and

Medzhitov 2002). The recognition of PAMPs by PRRs activates a network of signal

21

transduction pathways, a central aspect of which appears to be the upregulation of NF-

'B with resultant cytokine and chemokine production and neutrophilic inflammation

(Sadikot et al. 2003; Hippenstiel et al. 2006). This pathogen-related activation of airway

epithelial cells results in auto- and paracrine stimulation of further inflammatory

mediators (Cooper et al. 2001) and upregulation of adhesion molecules (Cooper et al.

2001; Neff et al. 2006), complement component C3 (Cortes et al. 2002) and

antimicrobial peptides such as the defensins, cathelicidins and lactoferrin (Bals and

Hiemstra 2004).

Many clinically relevant pathogens secrete cytotoxic virulence factors, such as

pneumolysin (Streptococcus pneumoniae), alpha toxin (Staphylococcus aureus) and the

type III secretion system-associated toxins (Pseudomonas aeruginosa). In combination

with the inflammatory mediators released as part of the host inflammatory response,

these virulence factors lead to significant alveolar epithelial cell injury (Duane et al.

1993; Rubins et al. 1993; McElroy et al. 1995; McElroy et al. 1999). In a model of

Pseudomonas aeruginosa pneumonia, McElroy et al. demonstrated that the alveolar

fluid content of the ATI cell-specific protein RTI40 correlated with both increased

permeability of the air-blood barrier and morphological assessment of epithelial injury,

suggesting that ATI cell injury is a central feature in the pathogenesis of Pseudomonas

aeruginosa pneumonia (McElroy et al. 1995). Significant epithelial injury has also been

demonstrated in models of Streptococcus pneumoniae and Staphylococcus aureus

pneumonia (McElroy et al. 1999; Clegg et al. 2005).

1.2.4 Idiopathic Pulmonary fibrosis

1.2.4.1 Definition

Idiopathic pulmonary fibrosis (IPF), also known as cryptogenic fibrosing alveolitis, is a

progressive and devastating interstitial lung disease. The disorder may be defined as a

clinicopathological syndrome characterised by cough, exertional dyspnoea, bilateral

22

inspiratory crepitations on auscultation, a restrictive defect on pulmonary function

testing, honeycombing on computed tomographic (CT) scan and a histological diagnosis

of usual interstitial pneumonia on lung biopsy (table 5) (ATS 2000; Selman et al. 2004).

Cardinal feature Ancillary features

Heterogeneous involvement

Predilection for subpleural & basilar regions

Bilateral involvement

Fibroblastic foci

Honeycomb cysts

Thickened, distorted alveolar walls

Excessive collagen & extracellular matrix

Scattered inflammatory cells

Bronchiolectasis ± bronchiectasis

Smooth muscle hypertrophy

Hyper- & metaplasia of ATII cells

Mucostasis

Secondary pulmonary hypertensive changes

Absence of granulomas or vasculitis

Absence of microorganisms or minerals

Table 5 Histopathological features of idiopathic pulmonary fibrosis (Selmann, Thannickal et

al. 2004).

1.2.4.2 Epidemiology of idiopathic pulmonary fibrosis

IPF is rare, with a prevalence of 3-20 cases per 100,000 people. The disease occurs

predominantly from middle age onwards, and is more prevalent in males. A familial

variant of the disease has been recognised but, other than the age of onset, is clinically

indistinguishable from non-familial cases.

Following presentation, clinical symptoms worsen over months to years with

progressive fibrosis and destruction of lung parenchyma. Most patients die from

respiratory failure within 3-8 years of the onset of symptoms. Current treatments,

typically using anti-inflammatory or immunosuppressive agents, remain of unproven

benefit and there is no evidence that any therapy alters the natural history of the disease

(Selman et al. 2004).

23

1.2.4.3 Pathogenesis & the role of the alveolar epithelium in IPF

The aetiology and pathogenesis of IPF remain unknown. Early hypotheses focused on an

unidentified insult to the lung initiating a cycle of chronic alveolar inflammation leading

to fibrosis and destruction of normal lung architecture (Crystal et al. 1976). Based upon

this hypothesis, the alveolar epithelial alterations characteristic of IPF - namely, loss of

ATI cells and proliferation of ATII cells - are caused by an unresolved inflammatory

process in the surrounding microenvironment.

More recent hypotheses, however, suggest that IPF, rather than being a chronic

inflammatory disorder, results from an aberrant host response to wound healing

orchestrated by epithelial cell activation and abnormal epithelial-mesenchymal

interactions (Geiser 2003; Selman et al. 2004; Selman and Pardo 2006). Alveolar

epithelial hyperplasia and the presence of several altered cell phenotypes is a striking

feature of IPF, suggesting that alveolar reepithelialisation is severely disturbed (section

1.3.4). This is thought to disrupt normal epithelial-mesenchymal interactions generating

a profibrotic environment leading to the migration, proliferation and activation of

mesenchymal cells with the formation of active fibroblastic/myofibroblastic foci and

excessive accumulation of extracellular matrix (Selman and Pardo 2006).

24

1.3 Alveolar epithelial repair

1.3.1 Historical perspective of cell cycle kinetics in the alveolar epithelium

The concept of the ATII cell as a stem cell was first raised by Kapanci and colleagues

following study of the histological changes in the lungs of monkeys exposed to 100%

oxygen (Kapanci et al. 1969). These studies demonstrated that ATI epithelium,

destroyed after 4 days exposure to 100% oxygen, was initially replaced by cuboidal

ATII epithelium followed by the recovery of histologically normal lung upon re-

exposure of the animals to normal room air.

However, it is studies by Evans and colleagues (Evans et al. 1972; Evans et al. 1975),

combining the use of thymidine labelling and electron microscopy, which provide the

foundations for the current understanding of the response of the alveolar epithelium to

injury (Uhal 1997). They examined the ultrastructural changes occurring in the alveolar

epithelium following 48 h exposure to the oxidant gas nitrogen dioxide. The exposure

was followed immediately by an intraperitoneal injection of tritiated thymidine (3HT) to

achieve a pulse of label available for incorporation into newly synthesised DNA during

the S phase of the cell cycle. In lung sections obtained 1 h after labelling, virtually all

labelled cells were ATII cells. By 24 h after labelling, labelled ATI cells began to

appear, and, by 48 h the percentage of ATI cells had increased 10-fold compared to that

observed at 1 h. Over the same time period, the percentage of ATII cells had fallen to a

third of the original. In addition, a population of 3HT labelled ‘transitional’ cells with

morphological characteristics of both ATII and ATI cells could be identified. These data

suggested that alveolar epithelial repair following injury was achieved by ATII cell

division and the transition of ATII cells into ATI cells.

Similar studies by Adamson and colleagues (Adamson and Bowden 1974; Adamson and

Bowden 1975), in which mice were exposed to 90% oxygen for 6 days followed by

recovery in room air, examined the cell kinetics of alveolar epithelial repair using a

combination of 3HT labelling and colchicine-induced mitotic arrest. They found that

25

alveolar epithelial injury appeared to be limited to ATI cells, with relative sparing of

ATII cells. 3HT labelling of the alveolar epithelium became maximal 2 days following

termination of exposure to 90% oxygen, at which time-point mitoses were observed in

the ATII cell population. At this same time-point, neither 3HT labelling nor mitoses were

found in ATI cells, supporting the hypothesis that proliferation during recovery was

confined to the ATII cell population. By 3 days after exposure, labelling was observed in

the initially unlabelled ATI cell population with a concomitant decrease in the number of

labelled ATII cells. Analysis of bronchoalveolar lavage fluid (BALF) showed that ATII

cells were not being sloughed into the airspaces. Together with the observation of

‘intermediate’ cells exhibiting morphological characteristics of both ATII and ATI cells,

these data supported the notion of a direct transition of ATII cells to ATI cells during

alveolar epithelial repair.

Further evidence supporting the hypothesis that ATI cells must be derived from ATII

cells was the demonstration that ATI cells appear incapable of cell division, with

Adamson and colleagues unable to find evidence of ATI cell mitoses despite the

presence of abundant mitoses in neighbouring ATII cells following oxygen-induced

injury (Adamson and Bowden 1974; Adamson and Bowden 1975).

Support for this concept has been provided by in vitro studies of primary ATII cell

isolates, in which the ATII-like morphology of cells observed immediately after

isolation is replaced over a period of several days in culture by cells with an ATI-like

morphology (Kikkawa and Yoneda 1974). Immunohistochemistry has been used to

substantiate this observation, showing that primary ATII cells lose the expression of

ATII-selective proteins and gain the expression of ATI-selective proteins with time in

culture (Dobbs et al. 1985; Dobbs et al. 1988; Fehrenbach 2001).

1.3.2 Current model of alveolar epithelial repair

Based on these historical studies, it has been widely accepted for many years that an

important function of the ATII cell compartment is re-epithelialisation of denuded

26

basement membrane following alveolar injury, with damaged ATI cells being replaced

by the proliferation of ATII cells and the subsequent differentiation of a subpopulation

of daughter cells into ATI cells (figure 1A) (Uhal 1997).

However, more recent studies have refined the role of ATII cells in alveolar epithelial

repair (section 1.3.3.1), and also raised alternative mechanisms of ATI cell repair

(figure 1B) (McElroy and Kasper 2004).

It appears injured ATI cells may also be replaced by non-ciliated bronchiolar epithelial

progenitor cells and bone marrow cells (section 1.3.3.2). The concept that ATI cells are

terminally differentiated has also been challenged by studies examining the role of

mechanical load in the foetal lung, which suggest that ATI cells have the potential to

transdifferentiate into ATII cells (Flecknoe et al. 2002). The role this process plays in

epithelial repair remains unclear, and it is not known whether ATI cells are capable of

replacing ATII cells in the adult lung. Whilst the transformation of ATII cells into ATI

cells may be reversible, re-entry into the cell cycle appears to require reversion to the

ATII phenotype (Uhal 1997).

1.3.3 Stem cells responsible for alveolar epithelial repair

1.3.3.1 Alveolar stem cells

It is generally accepted that during homeostasis, and in response to injury, ATII cells

serve as progenitor cells within the alveolar compartment (Otto 2002). However, there

may be subpopulations of ATII cells with different susceptibilities to injury and different

proliferative capabilities following injury. Reddy and colleagues have suggested that

ATII cells isolated from hyperoxia-treated rat lungs can be segregated based on their

levels of E-cadherin expression (Reddy et al. 2004). E-cadherin negative cells are

damage resistant, proliferative and exhibit high levels of telomerase activity, suggesting

that they may represent a transiently amplifying progenitor subpopulation of the ATII

27

cell compartment (Reddy et al. 2004).

1.3.3.2 Extra-alveolar stem cells

Following severe injury to the alveolus, it is possible that repair mechanisms may

involve progenitor cells originating from outwith the alveolus (Otto 2002).

Tissue-specific stem cells: Examination of distal airway epithelium in mice following

bleomycin-induced lung injury revealed a subpopulation of cells containing Clara cell

specific protein (CC10) mRNA in alveolar-like structures (Daly et al. 1998). This was

thought to be the result of Clara cell migration or outpocketing, and the inference drawn

by Daly and colleagues was that alveolar epithelial progenitors could arise from

bronchiolar epithelium. This hypothesis is supported by the finding of pollutant-resistant

Clara cell secretory protein (CCSP)-expressing cells localised to the bronchoalveolar

duct junction that retain multipotent differentiation potential (Giangreco et al. 2002;

Reynolds et al. 2004).

Non-tissue-specific stem cells: Recent studies in experimental models and in humans

following lung transplant suggest that circulating bone marrow-derived cells may

participate in alveolar epithelial repair. Engraftment by circulating pluripotent cells

seems to occur in the lung parenchyma following injury (Ishizawa et al. 2004; Yamada

et al. 2004), and studies of bone marrow graft recipients have demonstrated numerous

donor-derived epithelial and endothelial cells in the lungs (Kleeberger et al. 2003; Suratt

et al. 2003).

However, the contribution of circulating bone marrow-derived stem cells to epithelial

repair remains controversial. Using a lineage-specific reporter system based on

transgenic mice that express green fluoresecent protein (GFP) only in ATII cells

(surfactant protein-C (SP-C)-GFP), Kotton and colleagues demonstrated that there was

no detectable reconstitution of alveolar epithelial cells by unfractionated bone marrow or

purified haematopoietic stem cells (Kotton et al. 2005).

28

1.3.4 Alveolar epithelial repair and lung fibrosis

Abnormal alveolar epithelial repair following injury appears to be a key pathogenic

mechanism in the development of fibrosis (Adamson et al. 1988). Normal alveolar

epithelial repair, on the other hand, appears to inhibit the development of pulmonary

fibrosis. Keratinocyte growth factor (KGF), a fibroblast-derived mitogen, has been

shown to stimulate the proliferation of ATII cells in normal rat lungs (Ulich et al. 1994),

and pre-treatment with KGF prior to bleomycin-induced injury has been shown to

facilitate epithelial repair and prevent fibrosis (Deterding et al. 1997; Guo et al. 1998).

Facilitation of epithelial repair by inhibiting ATII cell apoptosis has also been shown to

inhibit fibrosis in rats following bleomycin-induced injury (Wang et al. 2000).

In a comparison of models of normal repair (following hyperoxia-induced injury) and

abnormal repair with fibrosis (following bleomycin-induced injury) in the mouse,

Adamson and colleagues observed that, following ATI cell necrosis, normal repair was

associated with a significant, but short-lived, peak in ATII cell division. This was not

associated with fibrosis. However, abnormal repair was associated with an extended

period of ATII proliferation accompanied by fibroblast proliferation and collagen

deposition (Adamson et al. 1988). The number of epithelial-interstitial cell contacts in

these experiments appeared to be inversely related to the rate of ATII cell division,

suggesting that disruption of normal epithelial-fibroblast interactions may be a key event

in the progression to abnormal epithelial repair with fibrosis (Adamson et al. 1990).

The concept that abnormal repair is orchestrated by aberrant epithelial-mesenchymal

interactions is now widely accepted (Selman and Pardo 2002). Epithelial and

mesenchymal cells, functioning as a notional 'epithelial-mesenchymal trophic unit’,

signal each other dynamically and bidirectionally either by the release of soluble

mediators in a paracrine fashion or by direct cell-cell contact through spanning apertures

in the basement membrane (Evans et al. 1999; Fang 2000; Sirianni et al. 2003).

Epithelial cells are known to release transforming growth factor TGF-& and platelet-

derived growth factor (PDGF), both of which regulate fibroblast migration, proliferation,

matrix deposition and apoptosis. In turn, fibroblasts influence epithelial cell function by

29

releasing epidermal growth factor (EGF), keratinocyte growth factor (KGF) and

hepatocyte growth factor (HGF), all of which regulate ATII migration, proliferation,

differentiation and deposition of matrix proteins (Fang 2000).

It seems clear that normal repair following lung injury requires ordered and timely

reepithelialisation to restore alveolar epithelial integrity and normalise epithelial-

mesenchymal interactions. Failure of these processes appears to generate profibrotic

microenvironments within the lung leading to the migration, proliferation and activation

of mesenchymal cells and the formation of active fibroblastic foci (Bitterman 1992;

Fang 2000; Selman and Pardo 2006).

30

1.4 The use of cell-selective markers in lung injury and repair

Studies of changes in gas exchange, lung mechanics, pulmonary oedema formation,

alveolar protein leak and alveolar fluid clearance provide important information

regarding the functional consequences of lung injury. However, these studies provide

little information of disease process at the cellular level (Parker and Townsley 2004).

Light microscopy can provide qualitative evidence of lung injury, but accurate cellular

identification, particularly of attenuated ATI cells, is difficult at the level of resolution of

the light microscope. Because of this, most historical studies of alveolar epithelial injury

have relied on the use of electron microscopy (Evans et al. 1972; Evans et al. 1975;

McElroy and Kasper 2004).

Whilst use of the electron microscope to identify characteristic ultrastructural markers

does allow accurate identification of alveolar epithelial cells in both normal and

repairing lungs (Evans et al. 1975), it is only practicable to study very small areas of

lung using this approach, making representative sampling and accurate quantification

difficult. Given the heterogeneity of many forms of lung injury (Maunder et al. 1986;

Rouby et al. 2003), this presents obvious problems, and combining adequate numerical

analysis with precise cellular identification has presented significant difficulty in

studying the behaviour of the alveolar epithelium (Adamson and Bowden 1974).

1.4.1 Biomarkers and immunotargeting

The use of monoclonal antibodies as biomarkers to identify antigens specific to cells of a

particular lineage or stage of differentiation, has been applied to a variety of cell types

and organ systems including the alveolar epithelium (Funkhouser and Peterson 1989;

Williams and Dobbs 1990). Funkhouser and colleagues described this approach as

‘immunotargeting’, and when combined with immunofluoresence and laser scanning

confocal microscopy, this provides a powerful tool for characterising the alveolar

31

epithelium both in control lungs and in repairing lungs following injury, during lung

development and in primary cell culture.

1.4.2 Alveolar epithelial cell-selective markers

A number of proteins expressed by only one alveolar epithelial cell type have been

identified (Appendix I). These have been described as either cell-specific, when

expressed by the target cell only in the lung, or cell-selective, when expressed by the

target cell and other lung cells but not other alveolar epithelial cells (McElroy and

Kasper 2004). The cell-specific and cell-selective proteins used in the studies in this

thesis are described below.

1.4.2.1 ATI cell-specific and cell-selective proteins

Rat Type I 40-kDa protein (RTI40) or Type I cell alpha protein (T1"): RTI40 is an apical

membrane sialoglycoprotein expressed specifically by ATI cells in the lung. It is also

expressed in the choroid plexus and ciliary epithelium (Williams et al. 1996). The

function of RTI40 is unknown, but mice with targeted mutations of the T1$ locus die at

birth with respiratory failure, and histological examination of the lungs of these animals

demonstrate reduced numbers of ATI cells and defective alveolar formation (Ramirez et

al. 2003). Over expression of T1$ in endothelial cells and the homologous protein

PA2.26 in keratinocytes promotes the formation of long thin cell extensions and

increases migration rate in wounding experiments (Schacht et al. 2003). Based on these

data it has been speculated that RTI40 may regulate distal airway cell proliferation during

lung development and, in the adult lung, may be important in initiating and maintaining

the characteristic extended and flattened ATI phenotype (Williams 2003; McElroy and

Kasper 2004).

Aquaporin 5 (AQP5): AQP5 is a water channel protein located in the apical membrane

of ATI cells and bronchiolar epithelial cells in the lung (King and Agre 1996; Nielsen et

32

al. 1997). Studies using AQP5-deficient mice demonstrate that AQP5 provides the

principal pathway for the passive osmotically-driven movement of water across the

apical surface of the alveolar epithelium (Ma et al. 2000), although deletion appears to

have no significant effect on active alveolar fluid clearance out of the airspaces or the

accumulation of extravascular fluid arising from increases in hydrostatic pressure (Ma et

al. 2000).

MMC6 Antigen: MMC6 is a monoclonal antibody that was generated as a product of

work to develop novel antibodies recognising antigens expressed on the surface of rat

ATII cells (Boylan et al. 2001). The function of the MMC6 antigen is unknown, but

initial studies suggest that it is a lung-specific novel apical membrane protein expressed

by ATI cells (unpublished data McElroy & Franklin). Studies of the immunoselectivity

of MMC6 both in vitro and ex vivo form part of the work described in this thesis

(chapter 3). These studies confirm that the MMC6 antigen is ATI-specific.

1.4.2.2 ATII cell-specific and cell-selective proteins

Rat Type II 70-kDa protein (RTII70): Anti-RTII70 is a monoclonal antibody recognising

the rat ATII cell-specific apical membrane protein RTII70 (Dobbs et al. 1997). The

function of RTII70 is unknown.

MMC4 antigen or aminopeptidase N (APN) or p146: Aminopeptidase N (APN),

previously known as p146, is an apical membrane protein expressed in ATII and Clara

cells in the lung (Funkhouser et al. 1991). Boylan and colleagues described a novel

monoclonal antibody, MMC4, which selectively recognised an integral membrane

protein on the apical surface of rat ATII and Clara cells in the lung, proximal tubular

epithelial cells in the kidney and epithelial cells in the small intestine. Subsequent

characterisation has shown that the MMC4 antigen is APN (Franklin et al. 2004). The

function of MMC4 or APN in the lung remains unclear, although its main function in

other organs appears to be post-translational modification of proteins.

Pro-surfactant protein C (pro-SP-C): Surfactant protein-C (SP-C) is a lung specific

33

hydrophobic protein accounting for (4% of the protein content of pulmonary surfactant.

Beyond its role in reducing alveolar surface tension however, the precise role of

surfactant protein-C remains unknown, and difficulty purifying the protein together with

its tendency to denature and aggregate have made studying the protein difficult (Ikegami

and Jobe 1998). Alveolar SP-C (3.5 kD) is secreted by rat ATII cells, both in vivo and in

vitro, following synthesis and proteolytic cleavage of the precursor molecule pro-SP-C

(21 kD) (Beers and Fisher 1992; Beers and Lomax 1995). Anti-pro-SP-C is a cell-

specific polyclonal antibody recognising intracellular pro-SP-C within ATII cells (Beers

et al. 1992).

1.4.3 Alveolar epithelial cell-selective markers in the investigation of lung injury

1.4.3.1 Cell-selective markers in quantifying epithelial cell numbers and/or injury

ATI markers: Ultrastructural studies in injured lungs have confirmed that necrotic ATI

cells are sloughed from the basement membrane into the airspaces, and several studies

suggest that the quantity of ATI cell-specific protein recovered from bronchoalveolar

lavage fluid (BALF) can be used to quantify the extent of ATI cell necrosis following

injury (McElroy and Kasper 2004). McElroy and colleagues have shown that in a model

of severe lung injury caused by Pseudomonas aeruginosa, BALF RTI40 levels were

elevated 80-fold above controls. This correlated with widespread ATI cell necrosis on

morphological analysis by EM and complete abolition of alveolar fluid clearance, a

functional marker of epithelial integrity (McElroy et al. 1995). In contrast, in a model of

focal ATI cell injury using nitrogen dioxide, BALF RTI40 levels were elevated only 2-

fold above controls and alveolar fluid clearance was stimulated (McElroy et al. 1997).

Plasma levels of RTI40 have also been shown to correlate with the degree of alveolar

epithelial injury following acid-induced lung injury (Frank et al. 2002).

HTI56 is a human ATI-specific integral membrane protein that has been shown to be

significantly elevated in the alveolar oedema fluid of patients with pulmonary oedema

34

associated with epithelial cell injury (ARDS), but not in patients with pulmonary

oedema occurring in the absence of epithelial injury (hydrostatic pulmonary oedema).

This suggests that HTI56 could be used as a biochemical marker of ATI injury in humans

(Newman et al. 2000).

ATII markers: The use of ATII-selective surfactant proteins as markers of cell injury has

been particularly studied in the context of acute lung injury (Hermans and Bernard

1999). Both SP-A and SP-B BALF levels have been shown to decrease in patients with

ARDS (Gregory et al. 1991; Gunther et al. 1996), and in the case of SP-A, in those at

risk of developing ARDS (Gregory et al. 1991). By contrast no alteration was observed

in BALF levels of SP-A and SP-B in patients with cardiogenic pulmonary oedema

(Gunther et al. 1996). In traumatic lung injury, a transient decrease in BALF SP-A has

been demonstrated, with levels returning to normal over a time course dependent on the

severity of injury (Pison et al. 1992). In addition to the reduction of SP-A and SP-B in

BALF suggestive of ATII cell damage, both SP-A and SP-B levels have been shown to

increase in the serum of patients with ARDS (Doyle et al. 1995; Doyle et al. 1997). The

elevation of SP-A and SP-B in the serum appears to correlate negatively with both

oxygenation and clinical outcome, and is thought to reflect disruption of the air-blood

barrier (Doyle et al. 1997).

KL-6 is a human circulating high molecular weight glycoprotein. The gene for KL-6 has

been classified as belonging to the MUC-1 gene family which encodes membrane-

associated mucins (Hermans and Bernard 1999). In the lung, KL-6 is ATII cell-selective

(Kohno et al. 1988). KL-6 is elevated in both sera and BALF in patients with idiopathic

pulmonary fibrosis and pulmonary alveolar proteinosis and this elevation appears to

correlate with the clinical activity of disease (Kobayashi and Kitamura 1995). ATII cell

hyperplasia is one of the major histopathological findings in these conditions and

immunohistochemical studies suggest that the origin of the elevated levels KL-6 is

proliferating ATII cells (Takahashi et al. 1998).

35

1.4.3.2 Specific regulation of cell-selective markers in lung injury

Changes in levels of cell-selective proteins may not simply reflect cell damage or a

change in cell number, but rather specific regulation.

Hyperoxia has frequently been used as a model of lung injury, and is known to cause

ATI damage (Crapo 1986). However, Cao and colleagues investigating the levels of the

ATI-selective proteins in the lung following hyperoxic exposure found that both RTI40

and AQP5 levels were significantly increased despite ATI cell injury, suggesting

upregulation of these proteins by ATI cells in response to hyperoxic stress (Cao et al.

2003).

In contrast, in a viral model of pneumonia, Towne and colleagues demonstrated that

levels of AQP5 and its mRNA transcript were decreased in the absence of significant

ATI cell necrosis, suggesting downregulation of AQP5 gene expression following viral

pneumonia (Towne et al. 2000). AQP5 also appears to be downregulated following

lipopolysaccharide (LPS)-induced lung injury (Jiao et al. 2002).

ICAM-1 is ATI cell selective in the lung, but has been shown to be expressed by ATII

cells in culture as they become ATI-like (Christensen et al. 1993). ICAM-1 expression

also dramatically increases in ATII cells following both pneumonia and hyperoxia-

induced lung injury, and it has been suggested that this upregulation of ICAM-1 by ATII

cells in-vivo could represent an early differentiation event (McElroy and Kasper 2004).

p172 is a rat ATII- and Clara cell-selective apical membrane protein recognised by the

monoclonal antibody 3F9. Although the function of p172 is unknown, Girod and

colleagues have demonstrated that this protein is upregulated just prior to birth and

following hyperoxia-induced injury in the adult lung suggesting an adaptive function

(Girod et al. 1996).

1.4.3.3 Cell-selective markers and the phenotype of repairing epithelium

Following injury to the lung, the alveolar wall is populated by proliferating ATII cells,

36

ATI cells and a population of alveolar epithelial intermediate cells (McElroy and Kasper

2004). Although little is known about the proteins expressed by these cells, the use of

cell-selective proteins has been used in an attempt to quantify the relative numbers of the

different cell populations and characterise their interactions at sites of active injury

repair.

Using antibodies to the ATII selective SP-D and the ATI-selective lectin lycoperiscon

esculentum agglutinin (LEL), Fehrenbach and colleagues studied the epithelial

remodelling that occurs following (KGF)-induced ATII cell hyperplasia. They found

that the number of ATII cells was maximal at 2-3 days following KGF instillation, with

ATII cells covering (45% of the alveolar surface compared to (4% in control lungs. By

day 3 following KGF instillation, a population of intermediate cells expressing both SP-

D and LEL was apparent. These intermediate cells increased in number to cover 7% of

the alveolar surface by day 7 post-KGF instillation, compared to (0.1-0.8% in control

lungs, at which time alveolar architecture was almost normal with the majority of the

alveolar surface covered by ATI cells. Alveolar epithelial cell apoptosis was increased

from day 2, and appeared maximal at 7 days post-KGF instillation. These data suggest

that the epithelial remodelling following KGF-induced ATII cell hyperplasia was

achieved in vivo by both the differentiation of hyperplastic ATII cells into ATI cells and

ATII cell apoptosis (Fehrenbach et al. 1999).

Examination of repairing alveolar epithelium following Staphylococcus aureus-induced

lung injury by Clegg and colleagues, revealed that at day 3 post-inoculation the alveolar

wall was thickened secondary to ATII cell hyperplasia with a 5-fold increase in the

fractional surface area of the alveolar wall expressing the ATII-selective proteins RTII70

and MMC4 and a decrease in the area expressing the ATI-selective RTI40. S.aureus

treated lungs also contained RTI40/MMC4/RTII70 and RTI40/MMC4 coexpressing cells

not observed in control lungs. These cells were thought to be intermediates in the

transition of ATII cells into ATI cells (Clegg et al. 2005).

37

1.5 Hyperoxia and the alveolar epithelium

1.5.1 Oxygen homeostasis

The sequential reduction of molecular oxygen within the mitochondria during oxidative

phosphorylation generates a group of molecules and free radicals known collectively as

reactive oxygen species (ROS). ROS include the superoxide anion (O2-.), hydrogen

peroxide (H2O2) and the hydroxyl radical (OH.). ROS may react with other radicals

including nitric oxide (NO.) to generate further potent oxidants, such as peroxynitrite

(ONOO-). The products derived from NO

. are known collectively as reactive nitrogen

species (RNS). Small fluctuations in the steady-state concentrations of ROS appear to

play a role in intracellular signalling (Suzuki et al. 1997), but large increases lead to free

radical chain reactions that indiscriminately target cellular proteins, lipids,

polysaccharides and DNA, often leading to cell death (Thannickal and Fanburg 2000;

Turrens 2003).

All aerobic organisms face the threat of oxidative injury and have developed antioxidant

systems and gene-regulated adaptive responses to ensure oxygen homeostasis (Haddad

2002). The term ‘oxidative stress’ is frequently used to describe the deleterious

processes resulting from imbalances between ROS/RNS generation and these defence

mechanisms (Turrens 2003).

The principal antioxidant systems include the superoxide dismutases (SOD), a family of

metalloenzymes which enzymatically convert the superoxide anion to hydrogen

peroxide, and glutathione (GSH), a ubiquitous essential tripeptide (L-%-glutamyl-L-

cysteine-glycine) able to act as a substrate in the reduction of many free radicals and

ROS. Glutathione-associated metabolism in particular, provides a crucial equilibrium

interface between oxidative stress and the adaptive responses arising from changes in

gene expression (Haddad 2002; Turrens 2003).

Variations in the partial pressure of oxygen appear to regulate the pattern of gene

38

expression within cells primarily through the activation of the redox-sensitive

transcription factors hypoxia-inducible factor-1$ (HIF-1$) and nuclear factor-'B (NF-

'B). These are differentially regulated during oxidative stress, with HIF-1$ activity

increased under hypoxic conditions and NF-'B under hyperoxic conditions (D'Angio

and Finkelstein 2000; Haddad 2002; Michiels et al. 2002).

1.5.2 Hyperoxia

Hyperoxia can be defined as exposure of body tissues to higher than normal partial

pressures of oxygen. Although the brain and the preterm retina are susceptible to

hyperoxic injury, it is the lung which is normally exposed to the highest concentrations

of oxygen and therefore at greatest risk of oxidative injury.

The percentage of oxygen in air is a relatively constant 21%, and at normal atmospheric

pressure, the partial pressure of oxygen (pO2) in dry inspired air is therefore (21 kPa. As

air is transported into the lungs, through the conducting airways and into the alveoli, the

pO2 falls as a consequence of humidification of inspired gas, the uptake of O2 and the

admixture of carbon dioxide. By the time the inspired gas reaches the alveolus, the

normal alveolar partial pressure of oxygen (normoxia) is (13-14 kPa (West 2004).

Relative hyperoxia occurs in the neonate during the rapid transition from an intrauterine

pO2 of (3-4 kPa to an alveolar pO2 of (13-14 kPa following birth. Key developmental

changes in late gestation prepare for this ontogenic event by increasing antioxidant

defences in the lung, and the absence of these protective changes is thought to explain

the relative sensitivity of the preterm lung to oxidant injury and the development of

bronchopulmonary dysplasia (BPD) (O'Donovan and Fernandes 2004).

In adults, hyperoxia is invariably a consequence of the clinical use of therapeutic oxygen

to prevent hypoxaemia, and in severe respiratory failure, the prolonged use of high

fractions of inspired oxygen can result in an alveolar pO2 >90 kPa, with the alveolar

epithelium exposed to significant hyperoxia and oxidative stress as a result.

39

1.5.3 Hyperoxia-induced lung injury

Continuous exposure of normal adult rats to 100% oxygen causes characteristic

progressive lung injury, respiratory failure and eventually death (Crapo 1986; Clerch

and Massaro 1993). Atelectasis occurs within the first few hours of exposure due to

absence of nitrogen within the alveoli, and although there is little clinical evidence of

pulmonary oedema or inflammatory cell influx within the first 48 h, significant alveolar

protein leak has been demonstrated in the human lung within 24 h of exposure (Davis et

al. 1983). Between 48 and 60 h of exposure, pulmonary oedema and pleural effusions

develop, with animals beginning to die at 60 h. By 72 h, the majority of animals are dead

(Clerch and Massaro 1993). Examination of the lungs in these animals reveals a pattern

of injury similar to ARDS, with endothelial damage, extensive ATI cell necrosis,

alveolar-capillary oedema, lung protein leak, neutrophil infiltration and hyaline

membrane formation (Crapo 1986; Fracica et al. 1991).

Hyperoxia-induced lung injury appears to be mediated by a combination of the cytotoxic

effects of ROS and activation of redox-sensitive transcription factors, most notably NF-

'B, with subsequent upregulation of stress-response genes and changes in cell fate

signalling pathways. Upregulation of the pro-inflammatory cytokines TNF-$, IL-1&, IL-

6 and IL-8 occurs as part of this response, reinforcing the concept that there is a link

between oxidative stress and the evolution of an inflammatory response in the lung

(D'Angio and Finkelstein 2000; Haddad 2002; Michiels et al. 2002; Sue et al. 2004).

The rate of mitochondrial ROS generation increases linearly with oxygen concentration.

However, the rate of release of ROS from mitochondria appears to be biphasic,

increasing dramatically when cells are exposed to concentrations of oxygen exceeding

60% (Turrens 2003). The slower release of ROS at lower pO2 suggests that antioxidant

defences are able to compensate for moderate hyperoxia, but become overwhelmed at

higher pO2. This concept is supported by the finding that pre-exposure to 85% O2

confers resistance to hyperoxia-induced injury through an adaptive increase in lung

40

antioxidant content (Haddad 2002), and may explain the observation that prolonged

exposure of normal lungs to oxygen concentrations )60% appears to be injurious, whilst

concentrations *50% do not (Hayatdavoudi et al. 1981; Crapo et al. 1983).

1.5.3.1 The effects of hyperoxia on the alveolar epithelium

The alveolar surface is a key target for oxidant injury, and alveolar epithelial injury is a

characteristic feature of hyperoxia-induced lung injury (Crapo et al. 1980; Crapo 1986).

Continuous exposure to 100% O2 has been shown to cause complete destruction of the

alveolar epithelium in the lungs of normal monkeys (Kapanci et al. 1969; Crapo 1986),

and sublethal (<85%) exposure to cause significant ATI cell necrosis in rodents

(Adamson and Bowden 1974; Crapo et al. 1980).

1.5.3.1.1 Hyperoxia and ATII cell proliferation

Repair of damaged epithelium following hyperoxia-induced injury is dependent on the

initiation of ATII cell proliferation and the differentiation of ATII cells into ATI cells

(Adamson and Bowden 1974; Thet et al. 1986). ATII cell proliferation has been clearly

demonstrated in animals exposed to 100% O2 and then recovered in room air (Adamson

and Bowden 1974; Thet et al. 1986), and stimulation of ATII cell proliferation by the

ATII cell mitogen KGF has been shown to reduce both mortality and severity of lung

injury following hyperoxic exposure in rodents (Panos et al. 1995; Barazzone et al.

1999). However, exposure to 100% O2 itself appears to inhibit ATII proliferation (Crapo

1986).

The mechanisms regulating ATII cell proliferation during and following hyperoxia have

been difficult to study in vitro, because isolated primary ATII cells only proliferate to a

limited degree and rapidly lose their differentiated characteristics in culture (Dobbs

1990). However, in the transformed (Simian virus) SV40 ATII cell line, hyperoxia has

been shown to inhibit proliferation by post-transcriptional regulation of the late cell

41

cycle genes histone and thymidine kinase and to induce G1 cell cycle arrest through

transforming growth factor-& (TGF-&) signalling and increased expression of p21

(Clement et al. 1992; Corroyer et al. 1996). Further studies in mice have demonstrated

that hyperoxia causes nuclear DNA damage and increases the alveolar epithelial

expression of both p21 and p53 in vivo, supporting the hypothesis that hyperoxia inhibits

ATII cell proliferation through cell cycle checkpoint activation (O'Reilly et al. 1998;

O'Reilly et al. 1998).

Transcriptional regulation of the insulin growth factor (IGF) system also appears to play

a role in the arrest of ATII cell proliferation induced by hyperoxia. IGF-I, IGF-II and

IGF-IR have been shown to be upregulated by hyperoxia in vivo (Narasaraju et al.

2006), and in vitro the expression of IGF-II, IGF-IIR and IGF-binding protein-2

(IGFBP-2) increases in cells whose proliferation has been arrested by hyperoxia, falling

rapidly to control levels when cells are allowed to resume proliferation in normoxia

(Cazals et al. 1994).

1.5.3.1.2 Hyperoxia-induced alveolar epithelial apoptosis and necrosis

Alveolar epithelial cell injury is an important feature of hyperoxia-induced lung injury,

and although necrosis is known to occur following hyperoxic exposure, apoptosis

appears to be the principal mode of cell death in vivo (Kazzaz et al. 1996; Barazzone et

al. 1998).

Several studies suggest that the differential expression of signalling factors determining

cell fate in the alveolar epithelium is redox-sensitive (Haddad and Land 2000; Ward et

al. 2000; Buccellato et al. 2004), and that ROS act as (pro-apoptotic) upstream signalling

molecules in determining this fate (Lee and Choi 2003).

In support of this hypothesis, activation of the pro-apoptotic Bcl-associated x protein

(Bax) and expression of the pro-apoptotic cell cycle regulator p53 have both been shown

to increase in alveolar epithelial cells in parallel with increasing pO2 (Haddad and Land

2000), and hyperoxia-induced Bax activation has been shown to require the generation

42

of ROS (Buccellato et al. 2004). In a study using IL-6 over-expressing transgenic mice,

Ward and colleagues highlighted the significance of these findings by demonstrating that

IL-6 markedly diminished hyperoxia-induced lung injury and cell death and this

protective effect was associated with increased expression of the anti-apoptotic Bcl-2 but

not the pro-apoptotic Bax (Ward et al. 2000).

The pattern of hyperoxia-induced cell death in vitro is less clear. ATII cells isolated

from the lungs of rats exposed to sublethal hyperoxia prior to isolation demonstrate

DNA damage and increased levels of the pro-apoptotic proteins p21, p53 and Bax

(Buckley et al. 1998). However, this may simply reflect the fact that sublethal exposure

of ATII cells in vivo is a pro-apoptotic stimulus, damaging the cells sufficiently to result

in apoptosis in ex vivo culture. Other in vitro studies suggest that hyperoxia causes

necrosis or a cellular death pathway sharing features of both apoptosis and necrosis

(Kazzaz et al. 1996; Wang et al. 2003; De Paepe et al. 2005).

Interestingly, it appears that the alveolar epithelium may harbour a subpopulation of

cells resistant to hyperoxia-induced cell injury. In a study examining the phenotype of

ATII cells isolated from hyperoxia-treated rats, Reddy and colleagues found that the

isolated ATII cell population could be functionally segregated based on their expression

of E-cadherin (Reddy et al. 2004). The E-cadherin positive subpopulation contained

damaged cells and was non-proliferative with low levels of telomerase activity, whilst in

contrast, the E-cadherin negative subpopulation was undamaged, proliferative and

expressed high levels of telomerase activity. These data suggest that the ATII cell

population isolated from hyperoxia-treated rats is heterogeneous, with E-cadherin

expressing cells more sensitive to hyperoxia-induced injury than cells with low levels of

E-cadherin expression. The functional significance of these findings remains unclear,

but Reddy and colleagues have speculated that the E-cadherin negative subpopulation

may be the source of the transiently amplifying cells responsible for epithelial repair

following injury.

43

1.6 Mechanical forces and the alveolar epithelium

The lung is a dynamic organ that is subjected to mechanical forces throughout

development and adult life, and mechanical deformation appears to play an important

role in regulating both normal and pathological responses of the lung. The alveolar

epithelium appears to play a key role in regulating these responses (Riley et al. 1990;

Wirtz and Dobbs 2000; Edwards 2001).

1.6.1 Quantification of mechanical forces in situ

Several approaches have been used in an attempt to quantify the mechanical

deformations experienced by alveolar epithelial cells in situ. These include measurement

of changes in the surface area of the air-tissue interface using alveolar three-dimensional

reconstruction in serially sectioned rat lungs (Mercer et al. 1987) and measurement of

changes in epithelial basement membrane surface area (EBMSA) at different points on

the pressure-volume curve using morphometric analysis of electron micrographs

(Tschumperlin and Margulies 1999). These studies suggest that when the rat lung is

inflated from functional residual capacity (FRC) changes in epithelial cell basal surface

area are relatively small at low lung volumes increasing to between 16% (Bachofen et al.

1987) and 34% (Mercer and Crapo 1990; Tschumperlin and Margulies 1999) at total

lung capacity (TLC). Support for the magnitude of these changes in EBMSA with lung

volume comes from morphological studies of collagen fibril geometry in the alveolar

septa of lungs fixed at functional residual capacity (Mercer and Crapo 1990). These

studies found that collagen fibrils could theoretically be extended by (16% before the

wavelike structure of the fibrils straighten limiting further extension. This change in

fibril length translates to an increase in surface area of (32% (Mercer and Crapo 1990;

Tschumperlin and Margulies 1999).

44

1.6.2 The role of mechanical forces in lung development

1.6.2.1 The effect of mechanical forces on lung growth

During intrauterine life, the foetus makes foetal breathing movements generating

intrathoracic pressure changes similar to those generated in postnatal breathing

(Kitterman 1996). In addition, fluid is actively secreted into the lumen of the developing

lung generating a constant distending pressure in the potential air spaces (Kitterman

1996). It appears that both of these forces regulate normal growth and maturation of the

lung. Increasing foetal lung distension in sheep by chronic tracheal ligation has been

shown to both increase and accelerate lung growth (Alcorn et al. 1977; Hooper et al.

1993; Joe et al. 1997), whilst reducing foetal lung distension by chronic tracheal

drainage inhibits normal growth (Hooper et al. 1993). Abolition of foetal breathing

movements by cervical spinal cord transection also appears to inhibit lung growth

(Liggins et al. 1981; Harding et al. 1993).

Mechanical forces are also thought to play a role in the compensatory lung growth that

occurs following pneumonectomy in humans (Hsia 2004).

1.6.2.2 Alveolar epithelial phenotype & differentiation

Development of a normal alveolar epithelium is dependent on the regulated proliferation

and subsequent differentiation of alveolar epithelial cells into separate ATI and ATII cell

populations. Mechanical forces are thought to play a key role in the regulation of these

processes (Wirtz and Dobbs 2000). Increasing foetal lung distension by tracheal ligation

stimulates ATII cell proliferation (Hooper et al. 1993; De Paepe et al. 1999) but

qualitatively decreases the proportion of ATII cells in the foetal lung, whilst tracheal

drainage appears to increase the proportion of ATII cells (Alcorn et al. 1977; Joe et al.

1997). Electron microscopy reveals that the reduction in the number of ATII cells

following tracheal ligation in sheep is accompanied by a significant increase in the

45

number of cells of an intermediate cell phenotype, displaying characteristics of both

ATII and ATI cells, and this is followed, in a time-dependent fashion, by a fall in the

number of intermediate cells and a significant increase in the number of ATI cells. These

findings suggest that foetal lung distension stimulates the differentiation of ATII cells

into ATI cells via an intermediate cell type (Flecknoe et al. 2000).

Further evidence from animal models supporting the hypothesis that mechanical

distension stimulates the expression of the ATI cell phenotype and inhibits the ATII cell

phenotype is provided by the finding that mechanical distension of foetal lung decreases

the mRNA of the ATII cell-specific surfactant proteins SP-A, SP-B and SP-C whilst

increasing the mRNA of the ATI cell-specific protein RTI40 (Gutierrez et al. 1999; Lines

et al. 1999).

Intriguingly, Flecknoe and colleagues have also shown that reducing foetal lung

expansion in sheep stimulates the transdifferentiation of ATI cells into ATII cells,

challenging the long-accepted concept that ATI cells are terminally differentiated

(section 1.3.2) (Flecknoe et al. 2002).

1.6.3 The effect of mechanical forces on the alveolar epithelium

1.6.3.1 Surfactant secretion

The predominant physiological trigger for surfactant release appears to be the

mechanical forces occurring during breathing. Hyperpnoea during exercise, and the

application of a single deep inflation to isolated perfused rat lungs have both been shown

to cause a significant increase in surfactant release and a reduction in the volume density

of ATII cell lamellar bodies (Nicholas et al. 1982; Nicholas et al. 1982; Massaro and

Massaro 1983). Mechanical distension of primary rat ATII cells also stimulates

surfactant release, with stretch causing a transient increase in intracellular calcium and

sustained release of phosphatidylcholine (Wirtz and Dobbs 1990; Edwards et al. 1999).

46

1.6.3.2 Phenotype

Mechanical distension of foetal ovine alveolar epithelial cells in utero appears to favour

expression of the ATI cell phenotype and inhibit expression of the ATII cell phenotype

(section 1.6.2.2). The hypothesis that strain induces differentiation of ATII into ATI

cells is also supported by several in vitro studies. Sustained tonic distension of primary

rat ATII cells significantly increases the expression of the ATI cell-specific protein

RTI40 whilst reducing the expression of the ATII cell-specific proteins SP-B and SP-C

(Gutierrez et al. 1998). Sustained tonic contraction appears to have the reverse effect,

favouring expression of the ATII phenotype with reduced RTI40 expression and

increased expression of SP-A, SP-B and SP-C (Gutierrez et al. 2003). These effects

appear to be mediated at the transcriptional level (Gutierrez et al. 1999).

1.6.3.3 Cytokine production

Mechanical strain of the alveolar epithelium may contribute to the pathogenesis of

ventilator-induced lung injury (VILI) (section 1.6.4) by stimulating the release of pro-

inflammatory cytokines. Cyclical mechanical strain has been shown to upregulate the

expression of IL-8 in human lung carcinoma-derived A549 (ATII-like) cells (Vlahakis et

al. 1999; Li et al. 2003; Jafari et al. 2004), with this effect inhibited by the antioxidant

glutathione (Jafari et al. 2004).

However, in the majority of studies, stretch alone does not appear to significantly effect

cytokine expression, and DNA microarray analysis of changes in gene expression

following exposure of human alveolar epithelial cells to mechanical strain appears to

confirm this, with the expression of the commonly studied cytokines either not

significantly effected or decreased (Waters et al. 2002; dos Santos et al. 2004).

47

1.6.3.4 Apoptosis

ATII cells undergo apoptosis as part of normal cell turnover in the lung. ATII cell

apoptosis also appears to be a key mechanism in controlling the number of ATII cells in

the developing lung (Schittny et al. 1998), and in the restoration of normal alveolar

epithelium following ATII cell proliferation in repairing epithelium (Bardales et al.

1996; Fehrenbach et al. 1999; Fehrenbach et al. 2000; Fine et al. 2000). Mechanical

forces are thought to play a key role regulating this process (Edwards 2001).

Tracheal occlusion, with sustained foetal lung distension, has been shown to

significantly increase ATII cell apoptosis (De Paepe et al. 2004), and this, together with

the transdifferentiation of ATII cells into ATI cells (Flecknoe et al. 2000), is thought to

regulate the number of ATII and ATI cells in the developing alveolar epithelium. Cyclic

mechanical strain of primary rat ATII cells, and transformed human ATII-like (A549)

cells has also been shown to induce ATII cell apoptosis (Edwards et al. 1999;

Hammerschmidt et al. 2004). This effect appears to be attenuated by nitric oxide (NO)

released from co-cultured alveolar macrophages suggesting a physiological role of

alveolar macrophages in protecting ATII cells from stretch-induced apoptosis (Edwards

et al. 2000).

1.6.3.5 Deformation-induced injury

In addition to inducing apoptosis, mechanical deformation of the alveolar epithelial

basement membrane has been shown to induce necrotic cell death (Tschumperlin and

Margulies 1998; Hammerschmidt et al. 2004). This deformation-induced injury has been

implicated in the disruption of the air-blood barrier which occurs in animal models

during the development of ventilator-induced lung injury (Egan 1982; Tschumperlin and

Margulies 1998).

Tschumperlin and colleagues assessed the vulnerability of primary rat ATII cells in

culture to mechanical deformation-induced injury by subjecting cells to equibiaxial

deformations of 12, 24, 37 and 50%, representing lung inflation to ~60, 80, 100 and

48

>100% of total lung capacity respectively. Deformation-induced injury increased with

deformation magnitude and frequency in these studies, increasing rapidly with

deformations exceeding those associated with inflation to total lung capacity. This

appears to support the hypothesis that the alveolar epithelium undergoes irreversible cell

damage at high lung volumes (Tschumperlin and Margulies 1998; Tschumperlin et al.

2000). In addition, Tschumperlin and colleagues found that ATII-like cells (primary

ATII cells after 1 day in culture) appeared more sensitive to deformation-induced injury

than ATI-like cells (primary ATII cells after 5 days in culture), suggesting that the

morphological and phenotypic changes of ATII cells with time in culture also

significantly affect the vulnerability of alveolar epithelial cells to deformation

(Tschumperlin and Margulies 1998).

The mechanism underlying deformation-induced epithelial cell injury remains unclear,

although plasma membrane stress failure (Vlahakis and Hubmayr 2000), and oxidative

stress secondary to stretch-induced antioxidant depletion have both been implicated

(Jafari et al. 2004). A recent study by Chapman and colleagues, demonstrating increased

ROS generation in immortalised human alveolar epithelial (A549) cells in response to

mechanical deformation, supports the concept that oxidative stress may play a central

role in deformation-induced epithelial cell injury (Chapman et al. 2005).

1.6.4 Mechanical ventilation & lung injury: ventilator induced lung injury (VILI)

Mechanical ventilation is routinely used to provide respiratory support in critically ill

patients. However, both clinical and animal studies have shown that mechanical

ventilation can worsen existing lung injury and produce an iatrogenic injury known as

ventilator-induced lung injury (VILI) (Parker et al. 1993; Dreyfuss and Saumon 1998;

Slutsky 1999; Dos Santos and Slutsky 2000).

The clinical significance of VILI has been highlighted by several clinical trials

demonstrating that protective ventilatory strategies (section 1.6.4.1.2) are associated

with lower levels of both pulmonary and systemic inflammatory mediators, reduced

49

levels of organ dysfunction, and reduced mortality (Amato et al. 1998; Ranieri et al.

1999; ARDSNet 2000).

1.6.4.1 Mechanisms of ventilator-induced lung injury

For many years, ventilator-induced lung injury and barotrauma were thought to be

synonymous, with high airway pressures causing physical disruption of the alveolar

wall, air leaks and accumulation of extra-alveolar air (Dreyfuss and Saumon 1998;

Slutsky 1999). However, morphological examination of the lungs following ventilator-

induced injury reveals that more subtle changes occur following injurious ventilation,

with severe alveolar damage and changes in endothelial and epithelial permeability that

are indistinguishable from other forms of lung injury (Dreyfuss et al. 1985).

Both high pressure and high volume ventilation cause VILI in experimental models

(Dreyfuss and Saumon 1998; 1999), but alveolar overdistension (volutrauma) rather than

high airway pressures (barotrauma) appears to be the primary determinant of VILI

(Dreyfuss et al. 1988), with repeated alveolar collapse and reopening (atelectrauma)

contributing by the application of large shear forces (Mead et al. 1970; Webb and

Tierney 1974; Argiras et al. 1987).

These factors are of particular significance in acute lung injury, where patchy

parenchymal consolidation results in an uneven distribution of aerated, poorly aerated

and non-aerated lung (Maunder et al. 1986; Rouby et al. 2003). Positive pressure

ventilation in this context is thought to result in alveolar overdistension in uninjured

(aerated) alveoli with repeated alveolar opening and collapse secondary to cyclic

alveolar recruitment in poorly aerated regions. It is postulated that the resulting

ventilator-induced injury plays a key role in determining the poor outcome in patients

requiring mechanical ventilation through the development of a systemic inflammatory

response (Ranieri et al. 1999) and multiple organ failure (Slutsky and Tremblay 1998;

Tremblay and Slutsky 1998; Imai et al. 2003).

50

1.6.4.1.1 Inflammatory mediators in ventilator-induced injury: biotrauma

Barotrauma, volutrauma and atelectrauma are thought to be mechanical injuries caused

largely by stress failure of the plasma membrane and of the endothelial and epithelial

barriers (Slutsky 1999; Uhlig 2002). However, there is increasing evidence that

mechanical ventilation also leads to injury that is cell and inflammatory mediator based.

Slutsky and colleagues have coined the term biotrauma to describe this form of

ventilator-induced injury (Tremblay and Slutsky 1998).

Kawano and colleagues found that conventional ventilation of surfactant-depleted

rabbits resulted in lung injury characterised by the sequestration of large numbers of

neutrophils. This injury was attenuated by neutrophil depletion (Kawano et al. 1987).

Further evidence for the central role of the neutrophil in VILI has been provided by the

work of Zhang and colleagues, in which alveolar lavage fluid taken from ventilated

patients with ARDS was incubated with neutrophils from normal volunteers. Markers of

neutrophil activation were significantly higher in those ventilated conventionally

compared to those ventilated with a lung protective strategy (Zhang et al. 2002).

The alveolar macrophage may also play a role in the development of VILI. Mechanical

deformation of human alveolar macrophages in vitro, has been shown to increase the

expression of IL-8 and matrix-metalloproteinase-9 (Pugin et al. 1998), and in a rat model

of acute lung injury, Lentsch and colleagues demonstrated that alveolar macrophage

depletion suppressed NF-'B activation in the lung which appears to be an important step

in mediator release in response to overdistension (Lentsch et al. 1999; Held et al. 2001).

Work by Frank and colleagues also suggests macrophage activation is an early and

critical event in the initiation of VILI, with BALF from rats exposed to only 20 minutes

injurious ventilation capable of activating naïve primary alveolar macrophages in culture

and macrophage depletion decreasing ventilator-induced endothelial and epithelial

injury (Frank et al. 2006).

In animal studies, injurious ventilation of normal lungs has been shown to increase

concentrations of the pro-inflammatory cytokines TNF-$ and IL-6 in lavage fluid

(Tremblay et al. 1997; Veldhuizen et al. 2001; Tremblay et al. 2002) and in the perfusate

51

of isolated-perfused lungs (von Bethmann et al. 1998). In-situ hybridisation suggests that

the alveolar epithelium is a major source of these cytokines (Tremblay et al. 2002).

Clinical studies seem to support the concept that injurious ventilation promotes a

pulmonary inflammatory response and cytokine decompartmentalisation. Ranieri and

colleagues reported significant decreases in BALF and plasma concentrations of many

inflammatory mediators (TNF-$, IL-6 and IL-8) in patients ventilated with a lung

protective strategy (Ranieri et al. 1999), and in the ARDSnet trial, patients ventilated

with a lung protective strategy had a greater decrease in levels of plasma IL-6

(ARDSNet 2000).

1.6.4.1.2 The effect of protective ventilatory strategies

The avoidance of alveolar overdistension by limiting tidal volume and/or airway

pressures, with the addition of positive end inspiratory pressure (PEEP) to maintain

alveolar recruitment throughout the respiratory cycle, form the basis of lung protective

ventilatory strategies. Both clinical and animal studies have demonstrated the ability of

these manoeuvres to reduce VILI.

In a rat model of acid-induced lung injury, Frank and colleagues demonstrated that tidal

volume reduction, maintaining the same level of PEEP, significantly reduced both

endothelial and epithelial injury and this was associated with lower histological lung

injury severity scores (Frank et al. 2002).

Ranieri and colleagues reported significant decreases in the levels of both pulmonary

and systemic inflammatory mediators in patients ventilated using a lung protective

strategy (Ranieri et al. 1999), and in the ARDSnet trial, mortality was reduced by 22%

in patients ventilated with lower tidal volumes (6 ml/kg) compared to those ventilated

with traditional tidal volumes (12 ml/kg) (ARDSNet 2000). The protective effect of

PEEP has been highlighted in a study by Amato and colleagues, demonstrating that an

“open-lung” approach to mechanical ventilation, in which alveolar recruitment was

optimised with the use of high levels of PEEP, resulted in improved 28 day mortality

52

(Amato et al. 1998).

1.6.4.2 The role of the alveolar epithelium in VILI

The precise mechanisms of ventilator-induced lung injury remain unclear. It appears that

physical disruption with stress failure of plasma membranes and stress failure of the air-

blood barrier occurs in conjunction with cellular activation and the release of

inflammatory mediators (Slutsky 1999; Uhlig 2002). Neutrophils and alveolar

macrophages have both been implicated in the pathogenesis, but there is compelling

evidence that the alveolar epithelium plays a key regulatory role in the initiation and

regulation of the inflammatory response in VILI (Tremblay and Slutsky 1998; Slutsky

1999; Frank et al. 2006).

Localised at the air interface, the alveolar epithelial surface is clearly exposed to the

pressures generated during positive pressure ventilation, and morphometric analysis of

lungs exposed to injurious ventilation reveals significant epithelial injury, with

widespread ATI cell injury and ATII cell proliferation. The structural and functional

consequences of epithelial barrier disruption are discussed above (section 1.2.1).

Whether mechanical ventilation has a significant impact on epithelial repair following

injury remains unknown.

53

1.7 Summary, thesis hypotheses and aims

1.7.1 Summary

The alveolar surface of the lung is lined by the alveolar epithelium. This is comprised of

two morphologically distinct cell types: ATI and ATII cells. Joined by tight junctions,

these cells together constitute a key structural and functional component of the air-blood

barrier (section 1.1). Injury to the alveolar epithelium, with disruption of the integrity of

the air-blood barrier, is a central feature in the pathogenesis of a range of clinical

conditions (section 1.2).

ATI cells are more susceptible to injury than ATII cells but appear incapable of cell

division. The accepted paradigm of alveolar epithelial repair following injury posits that

damaged ATI cells are replaced by proliferation of ATII cells and the differentiation of a

subpopulation of daughter cells into ATI cells (section 1.3).

Timely and effective epithelial repair, with restoration of an intact epithelial barrier and

normal alveolar fluid transport, is a key determinant of clinical outcome following lung

injury. Abnormal epithelial repair on the other hand appears to be a key pathogenic

mechanism in progression to pulmonary fibrosis.

Oxygen and mechanical ventilation are key components in the supportive treatment of

respiratory failure and the prevention of hypoxaemia. However, both hyperoxia (section

1.5) and the lung deformations associated with mechanical ventilation (sections 1.6) are

potentially injurious to the lung. Localised at the air interface, the alveolar epithelial

surface is exposed to oxidative stress with the use of supplemental oxygen and

significant mechanical force during positive pressure ventilation. As such, it is a key

target for oxygen- and ventilation-induced iatrogenic injury.

Exposure of the normal lung to significant hyperoxia is known to cause ATI cell

necrosis and inhibit ATII cell proliferation. Its effect on the differentiation of ATII cells

into ATI cells has not been studied and is unknown. The effects of hyperoxic exposure

54

on the injured lung and on epithelial repair are also not known.

Similarly, although clinical experience suggests that mechanical ventilation is more

damaging to injured (repairing) lung than to normal lung, the effects of mechanical

deformation (± hyperoxia) on alveolar epithelial repair are not known.

The use of a panel of ATI and ATII cell-selective antibodies as biomarkers provides a

powerful tool for characterising the phenotype of alveolar epithelial cells in vitro and of

the alveolar epithelium ex vivo (section 1.4). It also enables the quantification of

individual cell phenotypes within the alveolar compartment. As normal epithelial repair

is associated with characteristic changes in epithelial cell number and cell phenotype

(ATII cell proliferation and differentiation of ATII cells into ATI cells), this

immunotargeting approach offers a novel means of extracting both qualitative and

quantitative data regarding the cell populations present in regions of alveolar epithelial

repair and of investigating the factors influencing alveolar epithelial repair in vivo.

1.7.2 Hypotheses

1. Hyperoxia inhibits normal alveolar epithelial repair by inhibiting the

transdifferentiation of ATII cells into ATI cells.

2. Alveolar epithelial cells in regions of epithelial repair are more susceptible to

mechanical deformation-induced injury than normal alveolar epithelial cells.

1.7.3 Aims

1. Establish the isolation and culture of primary alveolar epithelial cells (chapter 3).

2. Use a panel of cell-selective antibodies to characterise the immunophenotype of

alveolar epithelial cells in culture (chapter 3).

3. Investigate the immunoselectivity of a new monoclonal antibody (MMC6) in the

developing and adult lung and in alveolar epithelial cells in culture (chapter 3).

55

4. Determine the effect of hyperoxia on the transdifferentiation of alveolar epithelial

cells from an ATII- to an ATI-like phenotype in culture (chapter 3).

5. Further characterise an established model of resolving direct lung injury

(Staphylococcus aureus-induced pneumonia) to investigate exposure of the repairing

lung to hyperoxia (chapter 4).

6. Use a panel of cell-selective antibodies combined with immunofluoresence and laser

scanning confocal microscopy to quantify the proportion of the alveolar surface

associated with ATI, ATII and intermediate cell phenotypes in a model of resolving

lung injury and determine whether hyperoxia delays normal epithelial repair by

inhibiting the transdifferentiation of ATII cells into ATI cells in vivo (chapter 4).

7. Establish the primary cell culture of alveolar epithelial cells in a mechanically active

environment and determine the sensitivity of these cells to mechanical deformation-

induced injury (chapter 5).

8. Using an in vitro model, determine the susceptibility of cells in the ‘repairing’ region

of wounded primary alveolar epithelial cell monolayers to deformation-induced

injury (chapter 5).

9. Establish a comparative recovery model of indirect acute lung injury for investigation

of the effects of mechanical ventilation ± hyperoxia on alveolar epithelial injury and

repair (chapter 6).

56

2. Materials and methods

Reagents were obtained from Sigma-Aldrich Ltd unless otherwise indicated. Anaesthetic

agents and veterinary pharmaceuticals were supplied by Genusxpress. A list of

manufacturers and suppliers is given in Appendix II. Details of buffers, stock solutions

and culture media are given in Appendix III. Animal work was carried out in

accordance with the Animals Scientific Procedures Act 1986.

2.1 Primary alveolar epithelial type II cell isolation & culture

2.1.1 Alveolar epithelial type II cell isolation

ATII cells were isolated from the lungs of 150-250g male Sprague-Dawley specific

pathogen-free rats (Harlan) using a modified version of the method described by Dobbs

et al. (Dobbs et al. 1980; Dobbs et al. 1986; Dobbs 1990). Two rats were used per

isolation, with isolated cells being pooled prior to immunoglobulin G (IgG) panning.

Quantities stated are per rat. Isolation buffers and culture media were pre-warmed to

37+C, elastase (Roche) solution to 39+C.

Rats were anaesthetised by intraperitoneal injection of a mixture of midazolam (5

mg/kg) and ketamine (100 mg/kg). Heparin sulphate (500 iu) was administered by

separate intraperitoneal injection. Once anaesthetised, laparotomy was performed and

rats exsanguinated by abdominal aortotomy. The chest was opened, trachea cannulated

(18G Monoject blunt needle, Harvard Apparatus) and the lungs mechanically ventilated.

The left atrium was opened, and the lungs perfused until marble white by injection of

isolation buffer through the right ventricle. The heart and lungs were then explanted and

endobronchial lavage performed by inflation to total lung capacity (TLC) ten times with

Ca/Mg-free isolation buffer followed by four times with standard isolation buffer. The

57

lungs were then inflated to TLC with a solution of elastase (5mg in 20ml isolation

buffer) and incubated in saline at 39+C for 15 minutes. Remaining elastase solution was

then infused intratracheally over a further 15 minutes. Individual lobes of lung were

dissected from residual tissue, placed in DNAse solution (4mg in 2ml isolation buffer,

Roche) and elastase activity inhibited by addition of 4 ml foetal bovine serum (FBS).

The lungs were then chopped into (3 mm cubes, the resulting tissue suspension made up

to 40 ml with isolation buffer and hand shaken for three minutes, before sequential

filtration through 2 and 4-layer surgical gauze followed by 150, 20 and 10 µm nylon

mesh filters (Precision Textiles Ltd.). The resulting cell suspension was centrifuged

(257g, 6 min), resuspended in 20 ml serum-free Dulbecco’s modified eagle’s medium

(DMEM) and incubated at 37+C for 60 minutes on IgG coated bacteriological dishes

(Greiner Bio-One) to remove contaminant macrophages. The cells remaining unattached

at the end of this period were centrifuged (257g, 6 min) and resuspended in ATII culture

media.

Cell viability, assessed by exclusion of trypan blue, was determined by differential cell

count in a coverslipped Neubauer haemacytometer. Only isolations with a final cell

suspension cell viability > 90% were used in these studies.

Cell yield was assessed by cell count of crystal violet stained cells in a Neubauer

haemocytometer. The mean yield was 12.8 ± 3.1 x 106 cells per rat.

2.1.2 Modified Papanicolau staining & assessment of ATII purity

ATII cell purity was assessed by modified Papanicolau (Pap) staining of freshly isolated

cells (Dobbs 1990). (3x105 cells in 200µl media were cytospun (300 rpm, 3 min) onto

glass slides and air-dried. Slides were dipped in haematoxylin (Harris formulation) for

3mins, rinsed in DDW and dipped in saturated lithium carbonate (256mg Li2CO3 in 20

ml DDW) for 2min. After rinsing in DDW, slides were then sequentially dipped in 50%

ethanol for 90sec, 80% ethanol for 15sec, 95% ethanol for 15sec, 100% ethanol for

30sec, xylene:ethanol (1:1) for 30sec and finally 100% xylene for 1min. Cells were then

58

coverslipped using Pertex mounting medium.

Differential cell counts were performed at high magnification (100x oil immersion

objective), with ATII cells being identified as cells containing )3 purple-staining

inclusion (lamellar) bodies. Macrophages, neutrophils and lymphocytes were identified

by their characteristic cell morphology. A minimum of 500 cells was counted and

differential cell counts expressed as a percentage of totals.

2.1.3 Standard ATII cell culture

ATII cells were seeded at a density of 0.5 x 106 cells per cm

2 onto 13mm diameter

fibronectin-coated glass coverslips (Fisher Scientific) in ATII culture media and

incubated at 37°C in a humidified atmosphere of 95% air and 5% CO2 (normoxia) unless

otherwise stated. After 6 h, cells were washed 3x with serum free DMEM and the media

replaced with fresh ATII culture media. Media was subsequently changed every 48

hours throughout time in culture. The pO2, pCO2 and pH of culture media was measured

(Bayer RapidLab® 248 analyser) every 12 h during experiments under both normoxic

and hyperoxic conditions, with the results shown in table 6.

Normoxia Hyperoxia

pO2 (kPa) 16.3 ± 1.6 95.4 ± 4.3

pCO2 (kPa) 6.2 ± 0.7 6.4 ± 0.3

pH 7.50 ± 0.07 7.51 ± 0.02

Table 6 Culture media pH and partial pressures of oxygen and carbon dioxide

throughout time in culture under normoxic and hyperoxic conditions. Measurements

made using a Bayer RapidLab® 248 analyser (n=12).

59

2.1.4 ATII cell culture under hyperoxic conditions

ATII cells were seeded as for standard ATII cell culture. Cell culture plates were then

sealed in a humidified modular incubator chamber (MIC-101, Billups-Rothenberg inc.)

and the chamber flushed with filtered medical grade gas (95% O2, 5% CO2) from

cylinder supply (BOC Gases) at a flow rate of 20 l/min (Dual Flow Meter DFM 3002,

Billups-Rothenberg inc.) for 10 min. After 6 h, cells were washed 3x with serum free

DMEM and the media replaced with fresh ATII culture media. Media was subsequently

changed every 48 hours throughout time in culture. Media changes were carried out as

rapidly as possible under standard (normoxic) conditions. The pH and partial pressures

of O2 and CO2 measured in culture media during experiments under hyperoxic

conditions are outlined in table 6.

2.1.5 ATII cell culture using the Bioflex, cell culture system

Freshly isolated ATII cells were seeded onto the fibronectin-coated silastic membranes

of Bioflex, 6-well culture plates (Dunn Labortechnik GmbH) at a density of 0.5x106

cells/cm2. In all experiments, cell attachment was confined to the central portion of the

membrane by seeding within a removable silicon gasket (Grace Bio-Labs). Cells were

washed 6 h after seeding and silicon gaskets removed after 24 h in culture. In other

respects cell culture was identical to that for standard ATII cell culture.

Mechanical deformation of cells in culture was achieved using the Flexercell FX-

4000T" plates (Dunn Labortechnik GmbH) and is described in section 5.2.1.

2.2 Protein quantification assay

Protein concentrations in aqueous solution were determined using the Bradford (Bio-

Rad Laboratories) protein assay, a dye-binding assay based on the concentration-

dependent shift of the absorbance maximum of an acidic solution of Coomassie Brilliant

Blue G-250 from 465 nm to 595 nm upon binding protein (Bradford 1976).

60

Protein concentrations were obtained from the linear range of a standard curve

constructed by plotting absorbance against concentration (0-1.4 mg/ml) of bovine serum

albumin. Absorbance of samples was read at 630 nm using an ELISA plate reader (MRX

Microplater Reader, Revelation 3.04 software, Dynatech Laboratories). Each sample

was tested at two different dilutions with three repeats of each dilution.

2.3 Measurement of cell viability in adherent cells

The viability of adherent cells was assessed using fluorescent microscopy and a

LIVE/DEAD, viability/cytotoxicity kit (Molecular Probes, Invitrogen) comprising

ethidium homodimer-1 (EthD-1) and calcein acetoxymethyl ester (calcein AM). EthD-1

is excluded by the intact plasma membranes of live cells, but enters cells with damaged

membranes undergoing fluoresecent enhancement upon binding to nucleic acids and

producing red fluorescence (ex/em (495nm/(635nm). Cell permeant Calcein AM

undergoes enzymatic conversion by intracellular esterases present within live cells to the

polyanionic dye calcein. Calcein is well retained within live cells, producing a uniform

green fluorescence (ex/em (495nm/(515 nm). Adherent cells were washed 3x with

warmed serum-free DMEM containing 20mM 4-(2-hydroxyethyl)-1-

piperazineethanesulfonic acid (HEPES), and then incubated at 37 °C with 4µM EthD-1

and 2µM calcein AM in HEPES-containing DMEM for 60min. Cells were examined

with an inverted epifluorescnt microscope and images acquired from 3 random locations

using a digital-imaging system (Metamorph, Molecular Devices). The total number of

EthD-1 and calcein AM staining cells in each field was counted and data expressed as

percentage dead (EthD-1 positive) or viable (calcein AM positive) of total.

2.4 Lactate dehydrogenase (LDH)-cytotoxicity assay

Measurement of the increase in lactate dehydrogenase (LDH) activity following release

of cytoplasmic LDH from damaged cells is a widely accepted method of quantifying cell

61

cytotoxicity (Korzeniewski and Callewaert 1983). LDH activity may be determined

using a coupled enzymatic reaction in which the tetrazolium salt iodotetrazolium

chloride (INT) (yellow) is reduced to formazan (red) by NADH produced during the

LDH-catalysed conversion of lactate to pyruvate. Increases in LDH activity are directly

proportional to the amount of formazan produced in this coupled reaction, and this can

be determined colorimetrically.

After gentle agitation, cell supernatants were collected and remaining adherent cells

lysed by the addition of 100µl of 0.1% Triton X-100 solution to each culture well. After

15 min, culture wells were gently scraped with a cell scraper and the lysate removed.

Supernatant and lysate samples were stored at -70°C prior to analysis. LDH assays were

performed in triplicate on supernatant and paired lysate samples in 96 well culture

plates. 20µl Tris (hydroxymethyl)aminomethane buffer, 10µl sample and 50µl LDH

substrate were added to each well, mixed and incubated at 37°C for 5 min. 20µl of

freshly prepared chromagen solution was then added to each well, mixed and plates

incubated at 37°C for a further 5 min. 150µl 0.5M HCl was then added to each well,

mixed and formazan absorption at 540 nm read immediately (MRX Microplater Reader,

Revelation 3.04 software, Dynatech Laboratories). Samples were compared to control

wells in which sample was replaced with 10µl blank oxalate solution.

The percentage of cell death was calculated by determining the LDH activity in the

supernatant of each well as a percentage of total LDH activity (supernatant + lysate).

2.5 Feulgen staining & assessment of apoptosis

Apoptosis was assessed by nuclear morphology following Feulgen’s staining. Cells were

fixed in modified Bouin’s fixative at 4°C overnight then denatured by incubation with

5M HCl for 45 minutes at room temperature. After rinsing in tap water, cells were

incubated in Schiff’s reagent (BDH) for 1 hour and rinsed thoroughly ((15 min) in tap

water until red-purple DNA staining developed. Cytoplasm was counterstained with

0.1% light green, cells air-dried and mounted in cedarwood oil. Apoptotic cells were

62

distinguished by nuclear morphology using light microscopy (Bellamy et al. 1997). A

minimum of 500 cells was counted and results expressed as percentage of apoptosis.

2.6 Estimation of reactive oxygen species (ROS) generation

ROS generation was assessed by intracellular oxidation of 5-(and-6-)-chloromethyl-

2’,7’-dichlorodihydrofluorescein diacetate (CMH2DCFDA) (Molecular Probes,

Invitrogen). Cleavage of the lipophilic blocking groups of this cell-permeant probe by

intracellular esterases yields a charged form of the probe that is retained intracellularly.

Subsequent intracellular oxidation of nonfluorescent dichlorodihydrofluorescein to

(fluorescent) fluorescein can then be quantified spectrofluorometrically.

ATII cells on glass coverslips within 24 well plates (Greiner Bio-One) were washed 3x

with warmed phenol red-free Hank’s balanced salt solution (HBSS) (Invitrogen) and

500µl HBSS containing 10µM CMH2DCFDA added to each well. CMH2DCFDA was

omitted from otherwise identically treated control wells. Fluorescence was followed

continuously over 60min using a thermostated plate reading spectrofluorometer

(Fluoroskan Ascent, Thermo Scientific) at excitation 486 nm and emission 530 nm. The

protein content of each well was determined and the average rate of CMH2DCFDA

fluorescence corrected for variation in total protein between wells. Fluorescence

detected in control wells was subtracted from measurements obtained, and ROS

production reported as fluorescent units (fu)/mg protein/min.

2.7 Estimation of ROS generation in response to oxidative stress

ROS generation in response to oxidative stress was assessed by the oxidation of

CMH2DCFDA during exposure of ATII cells to hydrogen peroxide (H2O2). Controlled

exposure to H2O2 was produced by the addition of 50 milliunits of glucose oxidase

(catalysing the conversion of glucose within HBSS to D-glucono-1, 5-lactone and H2O2)

(Fluka and Riedel, Sigma-Aldrich) to each well containing 500µl HBSS and 10µM

63

CMH2DCFDA. ROS generation by ATII cells in response to H2O2 was then followed

continuously over 60min as detailed in section 2.6. Control wells contained HBSS and

CMH2DCFDA without glucose oxidase but were otherwise identical. Fluorescence

detected in control wells was subtracted from measurements obtained, and ROS

production reported as fu/mg protein/min.

2.8 Quantification of antigens by ELISA-based dot blot assay

The level of RTI40, MMC4 and MMC6 expression was determined semiquantitatively

using enzyme-linked immunoabsorbent assay (ELISA)-based dot blots. Samples were

immobilised by vacuum onto polyvinylidene difluoride (PVDF) Immobilon-P

membranes (Millipore) using a dot blot manifold (Schleicher & Schuell, Whatman

International). Immobilised samples were then blocked for 1.5 hours at room

temperature with 0.4% non-fat milk (casein) in tris(hydroxymethyl)aminomethane-

buffered saline (TBS, 154 mM, pH 8.2), incubated with primary antibody (1:500 RTI40

in DMEM; MMC4 hybridoma supernatant; MMC6 hybridoma supernatant) for 30

minutes, washed with TBS containing 0.05% Tween-20 (TBS-T) for 30 minutes and

then incubated with sheep anti-mouse secondary antibody conjugated to horseradish

peroxidase (1:3000, Amersham Pharmacia). Blots were subsequently washed again for

30 minutes with TBS-T before being developed with enhanced chemiluminescence

reagents (ECL, Amersham Biosciences) and developed to film (Kodak, X-omat AR

film, Sigma). Secondary only controls, in which incubation with primary antibody was

omitted, were performed as part of each assay. All samples for comparison were loaded

onto one membrane to allow for variation between blots. Semiquantitative data, reported

as densitometry or relative densitometry units per mg protein (du or rdu/mg protein),

were obtained from blots by determination of the absorbance of individual dots at 600

nm using a plate reader (MRX Microplate Reader, Revelation 3.04 software, Dynatech

Laboratories) normalised for sample protein concentration.

64

2.9 Immunohistochemistry for fluorescence microscopy

Details of the primary antibodies used for immunofluoresence are shown in tables 7 &

8. Alexa Fluor, goat anti-mouse isotype-specific secondary antibodies with conjugated

488 (green), 546 (red) or 647 (blue) fluorophores and anti-rabbit polyclonal secondary

antibody with conjugated 488 (green) fluorophore were used as secondary antibodies,

and were all obtained from Molecular Probes (Molecular Probes, Invitrogen).

Secondary-only controls and isotype-specific negative controls, using negative control

antibodies for mouse IgG1, IgG2a, IgG2b and IgG3 (Stratech Scientific), were performed

for each of the monoclonal primary antibodies used in these studies. Secondary-only

controls were performed for each of the polyclonal primary antibodies used. There was

no significant staining in controls unless otherwise stated. Frozen lung sections were

used as positive controls for ATI and ATII cell-selective primary antibodies. Positive

controls for the other primary antibodies used are detailed in table 8.

Primary antibody Antibody isotype Working dilution Source

MMC4 (hybridoma supernatant) Mouse IgG2a 50:100 Mary McElroy

Anti-RTII70 (hybridoma supernatant) Mouse IgG3 50:100 Leland Dobbs

Anti-Pro-SP-C (100 "g/ml) Rabbit polyclonal 10:100 Michael Beers

Anti-RTI40 (hybridoma supernatant) Mouse IgG1 2:100 Leland Dobbs

Anti-Aquaporin-5 (1.0 mg/ml) Rabbit polyclonal 1:100 Alpha Diagnostics

MMC6 (hybridoma supernatant) Mouse IgG2b 50:100 Mary McElroy

Table 7 ATI and ATII cell-specific/selective primary antibodies

65

Primary antibody Antibody isotype Dilution Positive control Source

Anti-BrdU (# 0.2 mg/ml) Mouse IgG1 5:100 A549 cell line Amersham

Anti-E-cadherin (250"g/ml) Mouse IgG2a 2:100 Lung tissue section BD Biosciences

Anti-active caspase-3 Rabbit polyclonal 2:100 Apoptotic hepatocytes BD Biosciences

Table 8 Primary antibodies used in immunohistochemistry

2.9.1 General protocol for staining frozen sections

Frozen tissue sections (~3"m thick) were removed from storage at –20°C and incubated

at room temperature for 15 minutes with blocking buffer (BB), or blocking buffer with

triton (BBT) if staining with an intracellular marker. After rinsing 5x with Ca/Mg-free

PBS, sections were then incubated for 20 minutes with 100µl of primary antibody

cocktail (working dilutions in BB detailed in tables 2 & 3), rinsed 5x with Ca/Mg-free

PBS, incubated for 15 minutes with 100µl of secondary antibody cocktail (1:100 in BB

for all secondaries) and again rinsed 5x with Ca/Mg-free PBS. If nuclear staining was

required, the sections were then incubated for 10 minutes with 100µl of TO-PRO,-3

(1:1000 in Ca/Mg-free PBS, Molecular Probes, Invitrogen) followed by a final rinse 5x

with Ca/Mg-free PBS. A coverslip was then mounted onto the section using Dako, anti-

fade fluorescence mounting medium (Dako). Once stained, sections were covered in foil

and stored at 4°C.

For staining of frozen lung sections with aquaporin 5, tissue sections were removed from

storage at –20°C and placed in methanol overnight prior to incubation with BB or BBT.

The foetal tissue sections used to investigate the immunoselectivity of MMC6 were

kindly provided by Chris Baig.

66

2.9.2 General protocol for staining cultured ATII cells

ATII cells cultured on glass coverslips in 24-well plates were fixed in 4%

paraformaldehyde or methanol (aquaporin 5 staining only) and stored at 4°C. Once

fixed, coverslips were removed from 24-well plates using fine forceps, rinsed 5x with

Ca/Mg-free PBS and stained in an identical fashion to that for frozen sections by placing

the coverslip cell-side down onto a drop of the required solution (BB, primary antibody

cocktail etc.) pre-positioned on a piece of Parafilm M. Rinsing between steps was

performed by gentle irrigation holding the coverslip in fine forceps.

ATII cells cultured in BioFlex, 6-well culture plates were fixed in 4% paraformaldehyde

and stored at 4°C. Cells were then stained in-situ within the 6-well culture plates. The

sequence followed was similar to that for frozen sections with the exception that 500µl

aliquots of primary and secondary antibody cocktails and TO-PRO,-3 were required to

incubate the cells within the culture wells. Once stained, cells were covered with Ca/Mg-

free PBS and imaged with a Zeiss 510 laser scanning confocal microscope equipped

with an upright Zeiss Axioskop microscope and dipping lens.

2.9.3 Bromo-deoxyuridine (BrdU) immunhistochemistry

ATII cells were incubated with 10µM BrdU (Amersham Biosciences) in ATII culture

medium for 6 hours and fixed in 4% paraformaldehyde. After rinsing 5x with Ca/Mg-

free PBS, cells were incubated for 45 mins at room temperature in 5M HCl, rinsed 5x

5min in Ca/Mg-free PBS, incubated in BBT for 15min and then 60min with mouse anti-

BrdU Alexa Fluor, 488 conjugate (1:20 in BBT, Molecular Probes). After rinsing 5x

with Ca/Mg-free PBS, nuclei were counter-stained with TO-PRO,-3 and mounted as

detailed above.

2.9.4 Image acquisition

Images were captured using a confocal laser scanning microscope (LSM) with Zeiss

67

LSM 510 scanning head and an Axiovert 100M microscope unless stated otherwise.

Three lasers were used for specimen excitation: argon/krypton (488nm), helium/neon

(543nm) and helium/neon (633nm). To eliminate spectral spill, scanning with each laser

was performed sequentially (multi-track imaging). Comparative images were obtained

with identical laser and detector settings and images used for quantification were not

further digitally enhanced. Secondary antibodies had emission spectra with peak

wavelengths at 488nm (Alexa 488, green), 546nm (Alexa 546, red), and 647nm (Alexa

647, blue). Alexa 488 emissions were filtered through a Band Pass 500-530nm filter,

Alexa 546 emissions through a Band Pass 565-615nm filter and Alexa 647 emissions

through a Low Pass 650nm filter. Images were captured as three channel (red, green, and

blue) 8-bit grey scale LSM files at a resolution of 1024x1024 pixels, and subsequently

exported in tagged image file format (TIFF). A differential interference contrast (DIC)

image was also captured to record morphology at the transmitted light level.

For quantification purposes, a minimum of 3 large contiguous areas of specimen (up to

585 µm2

or 4096x4096 pixels with the 63x objective) were imaged as 4x4 contiguous

fields of view using the image tiling facility of the LSM.

2.9.5 z-plane scanning and three-dimensional image reconstruction

After determining the focal plane of the top and bottom (maximum and minimum z-

coordinates) of a specimen, a series of sequential images was taken by scanning at

incrementally changing z-positions between the maximum and minimum z-coordinates

(xy-coordinates constant). The resulting optical sections, arranged to form an image

stack, were viewed as a three-dimensional image using Volocity 3.0 (Improvision).

To improve image resolution, particularly in the z-plane, image stacks were acquired

using Nyquist settings. The raw data obtained were deconvoluted using Huygens 2 (SVI)

software and reconstructed using Imaris 4.0 software (Bitplane AG).

68

2.9.6 Image analysis

Images used for quantification were acquired using identical LSM settings and were not

digitally enhanced. Openlab 3.5.2 (Improvision) software was used for quantification.

2.9.6.1 Image density slicing

Positive staining with fluorochromes was determined by density slicing using the

Density Slice… function in Openlab. RGB TIFF files (max 4000x4000 pixels) were split

into constituent red, green and blue channels (Layers>Select All>Split RGB). The lower

threshold of positive staining in each channel was determined by polling the pixel value

of a positive area using the HSI (Hue, Saturation, Intensity) Colorspy palette, and a

binary layer of positive staining generated using the Image>Density slice… function.

Binary layers from different channels were combined using the Boolean Operations…

function to produce binary layers of fluorochrome colocalisation.

2.9.6.2 Quantification of epithelial surface staining

Binary layers, representing positive staining with ATII and/or ATI-selective markers,

were generated using image density slicing as described in section 2.9.6.1. Quantitative

assessment of alveolar epithelial composition was then made by measurement of the

length of epithelial surface positively staining in these binary layers using the

Measurement… functions in Openlab. The contribution of individual cell phenotypes to

the alveolar epithelial surface was reported as a percentage of total alveolar surface

measured.

2.9.6.3 Cell counts

In vitro, fixed cells staining positively with fluorescent markers were counted after

image density slicing using the Measurement… functions of Openlab. Total cell number

69

was determined using the nuclear stain To-Pro-3. Counts of cell viability using the

Live/Dead, assay are described in section 2.3.

2.9.6.4 Cell surface area

Cell surface area was determined by tracing around the cell membrane of E-cadherin or

phalloidin-stained cells using the Region of Interest… measurement tool of Openlab.

2.10 Rat model of Staphylococcus aureus pneumonia

The Staphylococcus aureus strain used in these study was wild-type strain 8325-4. This

was kindly provided by Dr. Tim Foster, Department of Microbiology, Moyne Institute of

Preventive Medicine, Trinity College, Dublin, Ireland. The experimental protocol used

is outlined in figure 15, and was based upon the model previously established and

characterised by McElroy et al. (McElroy et al. 1999).

2.10.1 Preparation of bacterial instillate

Bacterial stocks were stored in peptone broth containing 25% glycerol at –70°C. 36 h

before planned bacterial instillation, a scraping of glycerol stock was spread onto blood

agar base (BAB) plates (BD Biosciences) and incubated at 37°C for 18 h. 3ml of Todd

Hewitt broth (BD Biosciences) was then inoculated with a single bacterial colony

scraped from the BAB plate and the resulting broth incubated and shaken (220rpm) at

37°C for 18 h. On the day of instillation, 50µl/rat ((108cfu/rat) of the resulting

suspension was aliquoted into a sterile Eppendorf, washed twice with sterile endotoxin-

free PBS with Ca/Mg, centrifuging (11300g, 3min) between washes, and the bacterial

pellet resuspended in 1.5ml sterile endotoxin-free PBS with Ca/Mg and kept on ice. The

dose of viable bacteria instilled, expressed as colony forming units (cfu), was

determined by plating 50µl of 10-3

and 10-6

dilutions of instillate onto BAB plates and

70

incubating at 37°C overnight.

2.10.2 Bacterial instillation

Specific pathogen-free male Sprague-Dawley rats (250-300g; Harlan) were

anaesthetised by intraperitoneal injection of a mixture of fentanyl citrate (1mg/kg),

fluanisone (33mg/kg) and midazolam hydrochloride (17mg/kg). Once anaesthetised, rats

were suspended on a horizontal wire by the upper incisors and an intubation wedge

(modified 2ml Eppendorf; Fisher Scientific) placed in the mouth. A cannula (9cm

length, 0.96mm OD, Portex Fine Bore Polythene Tubing, Smiths Medical), with distal

end mounted on a 21G microlance needle, was inserted into the trachea and 0.5ml

bacterial suspension (~108 cfu) instilled into the lungs using a 1ml syringe. Control

animals were instilled with 0.5ml of sterile PBS. Instillation of bacterial suspension (or

PBS only in controls) was followed by injection of 0.5ml of air to facilitate dispersion of

bacteria to distal airways. Successful tracheal cannulation was distinguished by passage

of the tracheal cannula over palpable cartilagenous rings, and successful instillation was

typically accompanied by a brief episode of apnoea followed by tachypnoea.

2.10.3 Initial recovery and randomisation

Rats were given a subcutaneous injection of 1-2ml warmed sterile saline and recovered

on a heating pad inside a class II safety cabinet for (6 h before transfer to standard

housing. 72 h following instillation, animals were transferred to custom-built housing

units and randomised to either normoxic (air) or hyperoxic (93±2% O2) conditions for a

further 48 h.

2.10.4 Housing under normoxic and hyperoxic conditions

Units used to house rats under normoxic and hyperoxic conditions were comprised of

71

inner and outer clear acrylic chambers. Rats were housed within the inner chamber, and

this placed within the outer chamber. Vent holes in the sides of the inner chamber

allowed gas flow between chambers. Fresh gas was supplied to the outer chamber from

cylinder supply (medical-grade air normoxia, 100% O2 hyperoxia, BOC Gases) at 3

l/min and circulated throughout both inner and outer chambers by means of a coaxial fan

positioned on the external surface of the inner chamber. The outer box was vented to

room air ensuring normobaric conditions. CO2 accumulation was prevented through

absorption by soda lime placed between the outer and inner chambers. Rats had free

access to food and water. Temperature, lighting and humidity were maintained within

usual limits throughout. Once placed within the chambers, rats were left undisturbed for

48 h. O2 and CO2 tensions within the chambers were monitored continuously using a

Datex Normocap 200 sensor.

2.10.5 Harvesting of blood, bronchoalveolar lavage (BAL) fluid and lung tissue

At the end of the 48 h period in normoxic or hyperoxic conditions (5 days post-bacterial

instillation), rats were terminally anaesthetised by intraperitoneal injection of sodium

pentobarbitone (45mg/kg) mixed with heparin sulphate (500iu/rat). Laparotomy was

performed, and blood collected from the abdominal aorta. The chest was opened and

trachea cannulated (18G Monoject blunt needle, Harvard Apparatus). The lungs were

lavaged with 2x10ml sterile endotoxin- and Ca/Mg-free PBS, the volume of BALF

recovered recorded, and the lungs removed. Lungs for morphological analysis were

fixed (without lavage) by inflation to TLC with 4% paraformaldehyde in Ca/Mg-free

PBS, the trachea clamped to retain lung inflation, and the lungs removed and placed in

4% paraformaldehyde.

2.10.6 Bronchoalveolar lavage fluid processing

1ml of BALF was centrifuged (5000g, 5min) and the resulting cell pellet resuspended in

100-1000µl crystal violet. Total cell count was then determined using a

72

haemocytometer. Remaining BALF was centrifuged (212g, 6min) and the supernatant

aliquoted and stored at –70°C. The residual cell pellet was resuspended in Ca/Mg-free

PBS and (3x105 cells in 200µl cytospun (300rpm, 3mins) onto glass slides, air-dried and

stained with Diff-Quick, a modified Wright-Giemsa stain. Differential cell counts were

performed under oil immersion (100x) using characteristic cell morphology. A minimum

of 500 cells was counted and differential cell counts expressed as percentage of totals.

BALF protein concentration was measured using a Bradford protein assay (section 2.2).

2.10.7 Lung tissue processing for biochemical analysis

The left upper lobe was used in controls. When harvesting lungs from animals instilled

with bacteria, inflamed and/or consolidated lung was identified macroscopically and

separated. After weighing, lung tissue was stored at –70°C. Lung tissue was thawed on

ice, added to 5x lung weight of homogenisation buffer and homogenised (3x10sec,

PowerGen 125 Homogeniser, Fisher Scientific). The resulting homogenate was

centrifuged (300rpm, 3mins) to remove larger tissue fragments, aliquoted and stored at –

70°C. Homogenised lung protein concentration was measured using a Bradford protein

assay.

2.10.8 Lung tissue processing for morphological analysis

Lungs were left in 4% paraformaldehyde at 4°C overnight and then selected tissue cut

into 5mm cubes. Samples of left upper lobe were taken from control animals, and of

macroscopically inflamed and/or consolidated lung (typically left lung) from animals

instilled with bacteria. Tissue cubes were left in 30% sucrose solution at 4°C overnight

for cryoprotection. The following day blocks of tissue were submerged in optimal

cutting temperature compound (OCT), frozen in Freon 22 and then plunged into liquid

nitrogen. ~3"m sections were cut using a Bright cryostat (Huntington) by Susan Harvey.

Tissue sections were stored at -20°C until used.

73

2.11 Rat model of haemorrhagic shock-induced acute lung injury

The rat haemorrhagic shock model was developed jointly with Gareth Clegg.

Experiments were performed jointly, but subsequent characterisation of the

haemorrhagic shock-induced lung injury and biochemical analysis of bronchoalveolar

lavage fluid presented in this thesis was my own work. The experimental protocol used

is outlined in figure 39.

2.11.1 Animal preparation

Specific pathogen-free male Sprague-Dawley rats (250-300g, Harlan) were

anaesthetised by intraperitoneal injection of midazolam (5mg/kg) and ketamine (75-

100mg/kg) and placed supine on a temperature-regulated heating pad. A transverse

incision of the neck was made and the trachea, right common carotid artery and left

jugular veins exposed by blunt dissection. The common carotid was clamped proximally

and cannulated using PE50 tubing (BD Medical). This allowed continuous measurement

of arterial blood pressure (modified Propaq Encore monitor, WelchAllyn) and

intermittent arterial blood sampling. The left internal jugular vein was cannulated in a

similar manner, without proximal clamping, allowing central venous pressure

monitoring (modified Propaq Encore monitor, WelchAllyn) and the administration of

drugs and fluids. Anaesthesia was maintained with an intravenous infusion of propofol

(0.5mg/kg/min) and muscle relaxation with pancuronium bromide (2mg/kg/h). Normal

saline was administered intravenously such that intravenous fluids, including

medications, totalled 1.5ml/h. A tracheostomy was performed, the trachea cannulated

(18G Monoject blunt needle, Harvard Apparatus), and rats ventilated using a volume-

controlled ventilator (Small Animal Ventilator Model 683, Harvard Apparatus): tidal

volume 6ml/kg; positive end expiratory pressure (PEEP) 5cmH20; fractioned inspired

oxygen (FiO2) 1.0; respiratory rate 55-60 breaths/min adjusted to maintain pH between

7.30 and 7.45.

74

2.11.2 Experimental protocol

After a 30min stabilisation period, blood was removed from the carotid artery in 500µl

aliquots lowering the mean arterial blood pressure (MAP) to 30-40mmHg. This level of

haemorrhagic shock was maintained for 30min followed by resuscitation over 15-30min

with warmed intravenous normal saline restoring MAP to #80mmHg, whilst maintaining

CVP*10cmH2O. Rats were subsequently ventilated for an additional 4h and then

exsanguinated. Blood was collected from the abdominal aorta and the lungs lavaged

with 2x10ml sterile endotoxin- and Ca/Mg-free PBS to obtain BALF for analysis.

Control animals underwent surgery without subsequent haemorrhage.

BAL fluid was analysed for protein content (section 2.2), total and differential cell

counts (section 2.10.6) and, using ELISA-based dot blot assays, levels of the cell

selective proteins RTI40, MMC6 and MMC4 (section 2.8).

2.12 Statistics

Unless otherwise stated, data are expressed as the mean (taken from at least 3

independent experiments) ± standard deviation. Statistical analysis was carried out using

Graphpad Instat, 3.0 for Macintosh. Comparison of two means was performed using

unpaired t-tests assuming a two-tail p value. Comparison of multiple means was

performed using one-way ANOVA with post-test analysis and Bonferroni’s multiple

comparisons test where appropriate. p * 0.05 was considered statistically significant.

Data was tested for normality using the Kolmogorov-Smirnov test. There were too few

values to determine normality in the following data sets: adherent cell viability under

normoxic and hyperoxic conditions (figures 14); semiquantitative determination of

RTI40, MMC6 and MMC4 expression under normoxic and hyperoxic conditions (figures

17 & 19); LDH release and apoptosis of cells in culture under normoxic and hyperoxic

conditions (figures 23 & 24); ROS generation in response to standardised oxidative

75

stress by cells cultured under normoxic and hyperoxic conditions (figure 21B); BALF

protein, total cell count, differential cell counts and RTI40 content following S. aureus

instillation (figures 26, 27, 28 & 29); the proportion of the alveolar epithelial surface

with ATI-, ATII- and transitional-staining membrane following S. aureus instillation

(figures 36 & 38); deformation-induced injury in repairing monolayer wounds (figure

48); BALF protein, total cell count, differential cell counts, RTI40 and MMC6 content

following haemorrhagic shock (figures 51, 52 & 53). Data from these experiments was

also analysed using unpaired t-tests or one-way ANOVA, on the assumption of

normality.

76

3. The effect of hyperoxia on alveolar epithelial cells in culture

3.1 Introduction & objectives

Oxygen therapy is a potentially life-saving intervention in severe respiratory failure.

However, the therapeutic use of high fractions of inspired oxygen to prevent

hypoxaemia results in exposure of the alveolar epithelium to significant levels of

hyperoxia and oxidative stress. Hyperoxia is known to be injurious to the normal lung,

with prolonged exposure leading to a pattern of injury similar to ARDS (Crapo 1986;

Fracica et al. 1991). Damage to the alveolar epithelium is a central feature of this

hyperoxia-induced injury (Crapo 1986; Kazzaz et al. 1996; De Paepe et al. 2005).

The established paradigm postulates that ATI cells are more susceptible to injury than

ATII cells, and that following injury, damaged alveolar epithelium is replaced by the

proliferation of ATII cells and the differentiation of a subpopulation of daughter cells

into ATI cells (Uhal 1997). Hyperoxia has been shown to inhibit ATII cell proliferation

in vivo (Crapo 1986; O'Reilly et al. 1998) and to inhibit the differentiation of other cell

types in vitro (Zhang et al. 1999), but the effect of hyperoxia on the transdifferentiation

of ATII cells into ATI cells is not known.

The objective of the work outlined in this chapter was to use a series of in vitro

experiments to investigate the hypothesis that hyperoxia inhibits normal epithelial repair

by inhibiting the transdifferentiation of ATII cells into ATI cells.

Primary rat ATII cells are known to gradually lose their mature morphologic and

biochemical characteristics and adopt an ATI-like phenotype with time in culture

(Dobbs et al. 1985; Dobbs 1990). This transition from an ATII-like phenotype to an

ATI-like phenotype has obvious parallels to the processes thought to underlie normal

alveolar epithelial repair in vivo, and offers an in vitro model for the study of ATII cells,

ATI-like cells and the factors influencing the transition between the two cell types in

culture.

The first aim of the studies outlined in this chapter was therefore to establish the

77

isolation and culture of primary rat ATII cells, and using a panel of cell-selective

antibodies, characterise the phenotype of alveolar epithelial cells in culture. Having

characterised this in vitro system, the subsequent aim was to investigate the effect of

hyperoxia on the transition of ATII-like cells into ATI-like cells in culture.

The use of multiple cell-selective biomarkers working in the same assay facilitates the

correct assignment of cell type using this approach. MMC6 is a novel monoclonal

antibody (raised by McElroy) that appears to be recognised by ATI cells

(immunofluoresence screening during development). Preliminary data suggest that the

MMC6 antigen is lung-specific and distinct from RTI40 (Franklin and McElroy,

unpublished). To investigate the possibility that MMC6 may be a useful biomarker, a

further objective of the work outlined in this chapter was to characterise the expression

of the MMC6 antigen in both the adult and developing lung and by primary alveolar

epithelial cells in culture.

3.2 Results

3.2.1 Characterisation of freshly isolated ATII cells

The viability of freshly isolated cell suspensions at the time of seeding, determined by

exclusion of crystal violet, was >90% in all isolations used in these experiments. ATII

cell purity, determined by modified Papanicolau staining of cytospun freshly isolated

cell suspensions, was 68.8 ± 3.6% (figure 2A), rising to 82.3 ± 3.8% after rinsing 6 h

post-seeding (n=12). The major contaminants of ATII cell isolations were macrophages,

lymphocytes and neutrophils. The relative proportions of these are shown in (figure 2B).

Freshly isolated ATII cells co-expressed the ATII cell markers RTII70 and MMC4 but

did not express the ATI markers RTI40 or aquaporin 5 (figure 3A) (n=6).

3.2.2 Change of morphology and phenotype of ATII cells in culture

ATII cells cultured under normoxic conditions for 3 days, flattened and spread to form a

78

confluent monolayer. These cells expressed the ATI cell-selective markers RTI40 and

aquaporin 5. After 3 days in culture, cells did not express the ATII cell-selective markers

RTII70 and MMC4 (figure 3B) (n=6).

3.2.3 Identification of a novel intermediate cell phenotype

Loss of expression of the ATII cell marker RTII70 occurred by 24 h in culture, before

significant expression of the ATI marker aquaporin 5 was detected. However, a

subpopulation of cells (5.9 ± 1.4%) co-expressed the ATII marker MMC4 and the ATI

marker RTI40 after 24h in culture (n=3). Three dimensional imaging of RTI40/MMC4 co-

expressing cells revealed a more flattened shape than typical MMC4-expressing ATII

cells (figure 4A), with RTI40 expression localised to the spreading edge of the cells

(figure 4B).

3.2.4 The immunoselectivity of the monoclonal antibody MMC6

Localisation of MMC6 antigen expression in the adult lung: In control lungs, the

majority of the apical alveolar epithelial surface was comprised of thin attenuated cells

staining positively for the ATI-selective marker RTI40. These cells co-expressed MMC6

(figures 5, 6 & 7A) (n=3). RTII70 & MMC4 co-expressing cells were typically located

in the corners of the alveolar airspaces and were flanked by RTI40/MMC6 expressing

cells. RTII70/MMC4 co-expressing cells did not express RTI40 or MMC6 (figures 6 &

7B) (n=3). Endothelial cells, Clara cells and bronchiolar epithelial cells did not stain

positively for MMC6.

Developmental expression of MMC6 antigen: Foetal rat sections at different stages of

development were stained with combinations of MMC6, ATI-selective anti-RTI40 and

ATII-selective MMC4 and anti-RTII70 (n=3 for each developmental stage and

79

combination of primary antibodies used).

E16: At embryonic day 16 (E16), the size of the embryo enabled imaging of an entire

saggital section. No RTI40 or MMC6 staining was detected in the lung. The choroid

plexus and parts of the developing central nervous system stained positively for RTI40 as

previously described by Williams and colleagues (Williams et al. 1996), but did not stain

for MMC6. No MMC4 staining was detected at this time point (figure 8).

E17: At E17, RTI40 staining was detected on the apical surface of cuboidal cells lining

tubules within the embryonic lung. No significant MMC6 or MMC4 staining was

detected (figure 9).

E20: By E20 the embryonic lung had discernible airspaces with thick cellular alveolar

septae. The airspaces were lined with squamous cells staining positively for both RTI40

and MMC6 (figure 10). Separating these RTI40/MMC6 co-expressing cells were small

clusters of cells staining positively for RTII70 and MMC4. These cells did not stain for

either RTI40 or MMC6 (figure 11).

Expression of MMC6 antigen by alveolar epithelial cells in culture: Freshly isolated

ATII cells co-expressed the ATII cell markers RTII70 and MMC4 but did not express the

ATI cell marker RTI40 as described previously. Freshly isolated ATII cells did not

express MMC6 (figure 12A) (n=3). Loss of expression of the ATII cell marker RTII70

occurred by 24h in culture. At this time point a subpopulation of cells co-expressed

RTI40/MMC4 but MMC4/MMC6 coexpression was not detected (n=3).

After 3 days in culture, cells had flattened and spread to form a confluent monolayer of

ATI-like cells expressing RTI40 and MMC6 (figure 12B) (n=3). These cells did not

express the ATII cell markers RTII70 or MMC4. Deconvolution and 3D reconstruction

of the images obtained from MMC6/RTI40 coexpressing cells suggest that MMC6 is an

apical membrane protein (figure 13).

80

3.2.5 The effect of hyperoxia on ATII cells in culture

To determine the effect of hyperoxia on alveolar epithelial cells in culture, freshly

isolated ATII cells were cultured under either normoxic (media pO2 16.3 ± 1.6 kPa) or

hyperoxic (media pO2 95.4 ± 4.3 kPa) conditions for 3 days as described in materials

and methods (sections 2.1.3 & 2.1.4).

Adherent cell viability: There was no statistically significantly difference in the viability

of adherent cells cultured under normoxic and hyperoxic conditions after 3 d in culture

(98.0 ± 1.0% normoxia vs 93.4 ± 0.6% hyperoxia, p>0.05, n=3) (figure 14).

Cell spreading: Hyperoxia markedly impaired cell spreading. Cells cultured for 3 days

under normoxic conditions flattened and spread to form a confluent monolayer. These

cells had a mean surface area of 1801 ± 463µm2. In contrast, cells cultured under

hyperoxic conditions did not spread, but remained as small non-confluent clusters of

cells with a mean surface area of 583 ± 245µm2 (p<0.0001, n=3) (figure 15).

Expression of ATI markers: Hyperoxia inhibited the expression of the ATI-selective cell

markers, RTI40, aquaporin 5 and MMC6. Immunofluorescence analysis of normoxic-

exposed cells demonstrated that RTI40 (n=5), aquaporin 5 (n=3) and MMC6 (n=3) were

expressed on flattened alveolar epithelial cells after 3 days in culture. In contrast,

expression of each of these markers was greatly reduced in hyperoxic-exposed cells

(figure 16). The decrease in both RTI40 and MMC6 expression was (semi-) quantified

using ELISA-based dot blot assays. The concentration of RTI40 present in cells cultured

under normoxic conditions for three days was 13.0 ± 1.4 du/mg protein compared to

0.25 ± 0.2 du/mg protein in cells cultured under hyperoxic conditions, p<0.0001 (n=3)

(figure 17A). The concentration of MMC6 was 11.4 ± 1.7 du/mg protein for cells

cultured under normoxic conditions compared to 1.5 ± 1.1 du/mg protein for cells

cultured under hyperoxic conditions, p<0.05 (n=3) (figure 17B).

81

RTI40 expression in ATI-like cells: To determine the effect of hyperoxia on RTI40

expression in ATI-like cells, primary ATII cells were cultured under normoxic

conditions for 5 days and then exposed to hyperoxia for a further 2 days. There was no

significant change in morphology or difference in the level of RTI40 expression

determined by an ELISA-based dot blot assay between cells cultured under normoxic

conditions for 7 days and those switched to hyperoxic culture conditions on day 5 (10.8

± 1.9 du/mg protein normoxia vs 9.8 ± 0.4 du/mg protein d5 switch, p=0.3, n=3).

Expression of ATII markers: Hyperoxic exposure appeared to maintain the expression of

the ATII cell-selective markers MMC4 (n=3) and pro-SP-C (n=3) but not RTII70 (n=3)

in cultured ATII cells, with MMC4 and pro-SP-C detectable in epithelial cells cultured

under hyperoxic conditions for 3 days by immunofluorescence (figure 18). RTII70 was

not detectable by immunofluorescence in either normoxic- or hyperoxic- exposed cells

after 3 days in culture. Using an ELISA-based dot blot assay, the concentration of

MMC4 protein was determined to be more than 3-fold higher in cells cultured under

hyperoxic conditions compared with cells cultured under normoxic conditions (MMC4

55 ± 2 du/mg protein normoxia vs 187 ± 56 du/mg protein hyperoxia, p<0.01, n=3)

(figure 19).

Expression of E-cadherin: Reddy and colleagues have speculated that the ATII cell

population isolated from hyperoxia-treated rats is heterogeneous, with E-cadherin-

expressing cells more sensitive to hyperoxia-induced injury than cells with low levels of

E-cadherin expression (Reddy et al. 2004). To investigate the hypothesis that E-cadherin

negative cells may be more resistant to hyperoxia-induced injury, E-cadherin expression

in cells cultured under normoxic and hyperoxic conditions was examined by

immunofluoresence. Both cells cultured under normoxic and hyperoxic conditions

appeared to express E-cadherin heterogeneously. This pattern of staining was

82

reproducible, with strong expression at certain cell-cell contacts and weak expression at

others. Cells cultured under hyperoxic conditions appeared to have lower levels of E-

cadherin expression, with fewer strongly E-cadherin positive cells compared to cells

cultured under normoxic conditions (n=3) (figure 20).

Generation of ROS species: Hyperoxia-induced cytotoxicity is thought to be mediated

by the generation of reactive oxygen species. To investigate the hypothesis that cells

resistant to hyperoxia-induced injury generate fewer ROS, steady state ROS production

and the generation of ROS in response to oxidative stress was investigated in cells

cultured under normoxic and hyperoxic conditions. Day-3 cells cultured under hyperoxic

conditions demonstrated significantly lower steady state levels of ROS production than

those cultured under normoxic conditions (16 ± 5 vs 8 ± 3 fu/mg protein/min, p<0.05,

n=3). Hyperoxic-exposed cells also generated fewer ROS in response to standardised

exposure to oxidative stress (72 ± 8 vs 26 ± 4 fu/mg protein/min, p<0.001, n=3) (figure

21).

Proliferation: A small percentage (3.5 ± 1.0 %) of normoxia-exposed cells had BrdU-

incorporating nuclei. No BrdU-positive nuclei were detected in hyperoxic-exposed cells

(figure 22) (n=3).

Hyperoxia-induced cell cytotoxicity: Cell death, as determined by quantification of LDH

release, was observed in cells cultured under both normoxic and hyperoxic conditions at

all time points. There was no significant difference in the level of cell death determined

after 6 h (64.5 ± 2.1% normoxia vs 64.3 ± 10.7% hyperoxia, p>0.05, n=3) and 24 h in

culture (76.0 ± 4.4% normoxia vs 73.3 ± 17.2% hyperoxia, p>0.05, n=3), but after 3

days, there was significantly greater LDH release in cells cultured under hyperoxic

conditions (23.3 ± 8.3% normoxia vs 72.0 ± 2.6% hyperoxia, p<0.05, n=3). Significantly

greater LDH release was also observed under hyperoxic conditions after 5 days in

83

culture (16.0 ± 6.1% normoxia vs 70.5 ± 3.5% hyperoxia, p<0.05, n=3) (figure 23).

Hyperoxia-induced apoptosis: There was no statistically significant increase in the level

of apoptosis determined by Fuelgen staining with time in culture under normoxic

conditions, but the rate of apoptosis did increase with time in culture for cells cultured

under hyperoxic conditions. The difference in rate of apoptosis between cells cultured

under normoxic and hyperoxic conditions was not statistically significant after 6 h (1.7 ±

0.6% normoxia vs 2.7 ± 0.6% hyperoxia, p>0.05, n=3) and 24 h in culture (3.0 ± 1.7%

normoxia vs 5.7 ± 2.1% hyperoxia, p>0.05, n=3), but after 3 days, there was

significantly greater apoptosis in cells cultured under hyperoxic conditions (1.7 ± 0.6%

normoxia vs 6.3 ± 2.1% hyperoxia, p<0.05, n=3). Significantly greater rates of apoptosis

were also observed after 5 d in culture (3.0 ± 1.0% normoxia vs 7.7 ± 1.5% hyperoxia,

p<0.05, n=3) (figure 24).

The dose-response effect of the partial pressure of oxygen: The partial pressure of

oxygen in the culture media in these experiments was 95.4 ± 4.3 kPa under hyperoxic

conditions, and 16.3 ± 1.6 kPa under normoxic conditions, suggesting that these

experiments were conducted over a range of clinically relevant partial pressures of

oxygen. The effect of cell culture under hypoxic conditions (culture media pO2 4.3 ± 0.5

kPa) was investigated qualitatively in preliminary studies and appeared to have no effect

on adherent cell viability or the rate of transition from an ATII- to an ATI-like

phenotype with time in culture. Exposure of cells to 60% oxygen (culture media pO2

62.3 ± 1.9 kPa) also had no effect on adherent cell viability, but did qualitatively appear

to have an inhibitory effect on cell spreading and the expression of the ATI cell-selective

proteins RTI40 and MMC6. This effect was less dramatic than the effects described

following exposure to 95% oxygen and was not quantified.

84

3.2.6 The reversibility of hyperoxia-induced effects on ATII cells in culture

To determine the reversibility of the effects of hyperoxia on alveolar epithelial cells in

culture, freshly isolated ATII cells were cultured under hyperoxic conditions for 3 days

and then switched to normoxic conditions for a further 2 days.

The effects of hyperoxia on alveolar epithelial cell morphology and phenotype was

reversible. Cells cultured under hyperoxic conditions for 3 days and then transferred to

normoxic conditions spread and became confluent by day 5 in culture. These ‘d3

switched’ cells were viable (adherent cell viability 96.3 ± 3.1%, n=3) (figure 14) and

expressed the ATI cell-selective markers RTI40 (n=5), aquaporin 5 (n=3) and MMC6

(n=3) (figures 16 & 17) but not the ATII cell-selective markers MMC4 (n=3) and

RTII70 (n=3) (figure 19). There was a significant increase in cell proliferation following

the switch from hyperoxia to normoxia (13.1 ± 1.9% BrdU positive nuclei in switched

cells vs 0% in hyperoxia exposed cells, p<0.0001, n=3) (figure 22).

3.3 Discussion

The yield, viability and purity of the primary ATII cells used in these experiments was

in keeping with those published by other investigators (Dobbs et al. 1986; Dobbs 1990;

Chen et al. 2004).

3.3.1 ATII cells transdifferentiate into ATI-like cells in culture

Freshly isolated ATII cells co-expressed the ATII cell-selective markers RTII70 and

MMC4 (aminopeptidase N) but not the ATI cell-selective markers RTI40, aquaporin 5 or

MMC6, reflecting an ATII-like phenotype in freshly isolated cells. After 3 days in

culture under normoxic conditions this pattern of cell-selective protein expression

changed, with cells expressing the ATI cell-selective markers RTI40, aquaporin 5 and

MMC6 but not the ATII cell-selective markers RTII70 and MMC4. This change in

expression of cell-selective proteins was accompanied by a change in morphology as

85

cells flattened and spread. The loss of characteristics associated with the ATII cell

phenotype and acquisition of characteristics of the ATI cell phenotype under normoxic

conditions supports the widely held belief that primary ATII cells transdifferentiate from

an ATII-like phenotype to an ATI-like phenotype with time in culture (Dobbs et al.

1985; Dobbs et al. 1988; Borok et al. 1998; Borok et al. 1998; Campbell et al. 1999).

3.3.2 Identification of a novel intermediate cell phenotype

The finding of a subpopulation of cells coexpressing the ATII cell-selective protein

MMC4 and the ATI cell-selective protein RTI40 after 1 day in culture and before cells

could be defined as ATI-like is consistent with this population of cells representing an

intermediate or transition cell phenotype in the ATII to ATI cell transition. Although

others have described putative intermediate cell phenotypes (Fehrenbach et al. 1999;

McElroy and Kasper 2004), the coexpression of MMC4 and RTI40 is a novel finding.

The functions of MMC4 and RTI40 are unknown, and it is not known whether the

coexpression of MMC4 and RTI40 has any functional significance, or simply reflects the

fact that the MMC4 antigen has a longer half-life than RTII70. 3D visualisation of

MMC4/RTI40 coexpressing cells demonstrates that these cells have begun to spread and

flatten with RTI40 expression appearing localised to the spreading cell edge. Coupled

with the findings that RTI40 appears to be associated with increased migration rates in

wounding experiments (Schacht et al. 2003) and is associated with organisation of the

actin cytoskeleton (Scholl et al. 1999), these data support the speculation that RTI40 has

a role in cell spreading and migration.

3.3.3 The MMC6 antigen appears to be ATI-selective

In control lungs, the MMC6 antigen appeared to be coexpressed on the apical surface of

RTI40-expressing ATI cells. The MMC6 antigen did not appear to be expressed by ATII

cells, lung parenchymal cells or inflammatory cells. In vitro data, showing that the

86

MMC6 antigen was expressed by alveolar epithelial cells with an ATI-like phenotype

but not by freshly isolated primary ATII cells, provides further support for the

hypothesis that the MMC6 antigen is ATI-selective.

The MMC6 antigen appeared to be developmentally regulated, being co-expressed with

RTI40 in epithelial cells by E20, but absent in RTI40-expressing cells at E17. The fact

that RTI40 expressing cells did not bind MMC6 MAb in E17 lungs or E16 choroid

plexus suggests that RTI40 and MMC6 are different antigens.

Together, these data suggest that the MMC6 MAb is ATI-selective and recognises an

antigen distinct from RTI40. This suggests that MMC6 may be a useful additional

biomarker for the assignment of alveolar epithelial cell type both in vivo and in vitro.

Work to characterise the antigen is ongoing (Franklin & McElroy).

3.3.4 Hyperoxia reversibly inhibits transdifferentiation in vitro

Hyperoxia inhibited both the expression of ATI cell-selective proteins and the change in

morphology characteristic of the transition of alveolar epithelial cells from an ATII- into

an ATI-like phenotype with time in culture. The inhibition of transdifferentiation by

hyperoxia is a novel finding in relation to alveolar epithelial cells, although hyperoxia

has been shown to inhibit the differentiation (stellation) of astrocytes in the retina

(Zhang et al. 1999).

Hyperoxia also appeared to inhibit loss of expression of the ATII cell-selective marker

MMC4, although it did not appear to inhibit loss of RTII70 expression. Whether

inhibition of the loss of MMC4 expression has any functional significance, or simply

reflects the fact that the MMC4 antigen has a longer half-life than RTII70 under

hyperoxic conditions is not known, but after 3 days in culture under hyperoxic

conditions, alveolar epithelial cells appeared relatively undifferentiated. It has been

suggested that the dedifferentiation of ATII cells, with the gradual lose of differentiated

morphological and biochemical characteristics, is independently regulated from the

subsequent transdifferentiation into ATI cells (Gonzalez et al. 2005). Whether

87

dedifferentiation is an in vitro phenomenon remains unknown, but these data support the

hypothesis that hyperoxia inhibits the acquisition of an ATI-like phenotype

(differentiation), and may also inhibit the loss of an ATII-like phenotype

(dedifferentiation) in culture.

The finding that hyperoxia did not significantly affect the morphology or expression of

RTI40 in ATI-like cells suggests that hyperoxia was not simply a signal resulting in loss

of alveolar epithelial cell differentiation. It also suggests that the observed inhibition of

ATII transdifferentiation is not solely attributable to a global hyperoxia-induced

inhibition of protein synthesis. The fact that the inhibition of transdifferentiation was

reversible, with cells switched from hyperoxia to normoxia able to spread, flatten and

adopt an ATI-like phenotype, also suggests that the effect of hyperoxic exposure was not

caused by irreversible cell cytotoxicity.

3.3.5 Hyperoxia is cytotoxic to alveolar epithelial cells

The release of greater levels of LDH in hyperoxia-exposed cells compared to those

cultured under normoxic conditions confirms that prolonged exposure to hyperoxia is

cytotoxic (at least to some cells) and results in significant alveolar epithelial cell

necrosis. The increase in the rate of apoptosis following prolonged exposure to

hyperoxia also suggests that hyperoxic exposure is pro-apoptotic, and that the

mechanism of hyperoxia-induced cell death is, at least in part, apoptotic. These data lend

support to the concept suggested by other in vitro studies that hyperoxia may activate a

cellular death pathway sharing features of both apoptosis and necrosis (Kazzaz et al.

1996; Wang et al. 2003; De Paepe et al. 2005).

3.3.6 A subpopulation of cells appears resistant to hyperoxia-induced injury

Cell death in vitro is typically accompanied by cell detachment from the culture surface,

and the finding that there was no difference in adherent cell membrane integrity

88

(viability) between cells cultured under hyperoxic and normoxic conditions supports

this. However, these data also suggest that hyperoxia is not universally cytotoxic, and

that some cells may have a greater propensity to withstand hyperoxic insult.

3.3.7 E-cadherin negative cells appear resistant to hyperoxia-induced injury

In a study examining the phenotype of ATII cells isolated from hyperoxia-treated rats,

Reddy and colleagues found that the isolated ATII cell population could be functionally

segregated based on their expression of E-cadherin (Reddy et al. 2004). The E-cadherin

positive subpopulation contained damaged cells and was non-proliferative with low

levels of telomerase activity, whilst in contrast, the E-cadherin negative subpopulation

was undamaged, proliferative and expressed high levels of telomerase activity. These

data suggest that the ATII cell population isolated from hyperoxia-treated rats is

heterogeneous, with E-cadherin expressing cells more sensitive to hyperoxia-induced

injury than cells with low levels of E-cadherin expression. On the basis of these

findings, Reddy and colleagues speculated that the E-cadherin negative subpopulation

might be the source of the transiently amplifying cells responsible for epithelial repair

following injury.

To investigate the hypothesis that E-cadherin negative alveolar epithelial cells may be

more resistant to hyperoxia-induced injury, E-cadherin expression in cells cultured under

normoxic and hyperoxic conditions was investigated in the studies outlined in this

chapter. The pattern of E-cadherin expression observed by immunofluoresence was

heterogeneous in both normoxia and hyperoxia-exposed cells, with strong expression at

certain cell-cell contact points but not at others. However there appeared to be less E-

cadherin expression, with fewer E-cadherin positive cells, in cells cultured under

hyperoxic conditions compared to those cultured under normoxic conditions. This

finding may simply reflect downregulation of E-cadherin by hyperoxia, but would also

support the concept that hyperoxia-induced injury of E-cadherin positive cells resulted in

cell death and detachment of these cells leaving an adherent cell population with a

89

greater proportion of E-cadherin negative cells.

3.3.8 Hyperoxia-resistant cells generate fewer ROS

In addition to differing levels of E-cadherin expression, cells cultured under hyperoxic

conditions appeared to have a lower steady state production of ROS than cells cultured

under normoxic conditions, and to generate fewer ROS in response to oxidative stress.

ROS are thought to play a key role in mediating hyperoxia-induced cell injury; both

through a direct cytotoxic effect and through activation of the redox-sensitive

transcription factor NF-'B. Reducing ROS production in the face of hyperoxic exposure

is therefore likely to have a cytoprotective effect. The finding of reduced ROS

production in cells cultured under hyperoxic conditions may reflect such an adaptive

response, with fewer mitochondria or upregulation of antioxidant defences.

Alternatively, lower ROS production may further support the concept of a

heterogeneous ATII cell population, and simply reflect the fact that the cells surviving 3

days of hyperoxic exposure are phenotypically distinct and constitutively generate fewer

ROS in response to oxidative stress.

In an attempt to understand the role of ROS and NF-'B in the mechanism of hyperoxia-

induced inhibition of ATII cell transdifferentiation, preliminary studies were conducted

in which hyperoxia-exposed cells were incubated with the antioxidant and free radical

scavenger N-acetylcysteine and NF-'B inhibited by the addition of sulfasalazine (SSA)

and the inhibitory peptide SN-50. Neither the addition of antioxidant or inhibition of

NF-'B appeared to attenuate the effect of hyperoxia on ATII cell transdifferentiation

determined by change in cell morphology and RTI40 expression.

3.3.9 Hyperoxia reversibly inhibits alveolar epithelial cell proliferation

Primary ATII cells are thought to proliferate to a limited degree in culture (Dobbs 1990),

and this concept is supported by the finding of low proliferative rates in cells cultured

90

under normoxic condition in these studies. Whilst exposure of animals to 100% oxygen

has previously been shown to inhibit ATII cell proliferation in vivo (Crapo 1986), the

effect of hyperoxia on the proliferation of primary ATII cells in culture does not appear

to have been investigated previously. The studies presented in this chapter demonstrate

for the first time that hyperoxia is a potent inhibitor of ATII cell proliferation in vitro.

Switching cells from hyperoxic to normoxic conditions was followed by a proliferative

response in these studies, with BrdU incorporating cells going on to express the ATI

cell-selective protein RTI40. This is a novel finding, and supports the concept that short

term exposure of ATII cells to sublethal hyperoxia inhibits proliferation, with removal of

this inhibitory effect upon switching cells to normoxic conditions being followed by

ATII cell proliferation and transdifferentiation (Adamson and Bowden 1974; Thet et al.

1986; Yee et al. 2006).

3.4 Summary

The data presented in this chapter demonstrate for the first time that clinically relevant

levels of hyperoxia reversibly inhibit the normal transition of ATII-like cells into ATI-

like cells with time in culture. This suggests that hyperoxia may significantly inhibit

alveolar epithelial repair following injury in vivo. The findings of these in vitro studies

also support the hypothesis that the ATII cell population harbours a subpopulation of

cells, with lower levels of E-cadherin expression and ROS generation, resistant to

hyperoxia-induced injury. This subpopulation of hyperoxia-resistant cells may reflect

the presence of a transiently amplifying cell population cell within the isolated ATII cell

population.

91

4. The effect of hyperoxia on alveolar epithelial repair in vivo

4.1 Introduction & objectives

The finding that hyperoxia inhibits the change in cell morphology and expression of

cell-selective proteins associated with the transition of alveolar epithelial cells from an

ATII-like phenotype to an ATI-like phenotype with time in culture suggests that

hyperoxia may significantly inhibit the transdifferentiation of ATII cells during alveolar

epithelial repair (chapter 3). However, a recent study demonstrating that freshly isolated

ATI cells, ATII cells and cultured ATII cells have distinct molecular phenotypes

(Gonzalez et al. 2005), highlights the potential limitations inherent in extrapolating

biological relevance from these in vitro studies.

To investigate the hypothesis that hyperoxia inhibits alveolar epithelial repair in vivo, the

studies outlined in this chapter examined the effect of hyperoxia on the resolution of

lung injury in a characterised rat model of Staphylococcus aureus (S. aureus)-induced

pneumonia.

The model used was based upon a model of S. aureus-induced lung injury established

and characterised by McElroy and colleagues, in which rats are inoculated by

intratracheal instillation of S. aureus and then allowed to recover in standard (normoxic)

conditions (Bachofen et al. 1987; McElroy et al. 1999; Clegg et al. 2005). S. aureus is a

common cause of both pneumonia and (direct) acute lung injury (sections 1.2.2 and

1.2.3), and in the McElroy model, animals appear to develop a reproducible

inflammatory injury with damage to the air-blood barrier and ATI cell necrosis a central

feature. Previous characterisation of this S. aureus-induced injury has shown significant

ATII cell hyperplasia at day 3 post-instillation which resolves by day 7 post-instillation

as a result of the transdifferentiation of ATII cells into ATI cells and (probably) ATII

cell apoptosis (Clegg et al. 2005).

To examine the effect of hyperoxia on the resolution of the ATII cell hyperplasia

observed at day 3 in this model, rats were inoculated with S. aureus and recovered for 3

days in standard (normoxic) conditions in an identical manner, but then randomised to

92

either normoxic (air) or hyperoxic (95% O2) custom-built animal chambers for a further

2 days as detailed in materials and methods (section 2.10) (figure 25). The hypothesis

was that hyperoxic exposure would inhibit the normal resolution of ATII hyperplasia.

Having established this model, the first objective of the studies described in this chapter

was to characterise the lung injury at day 5 following instillation, comparing the extent

of injury in animals exposed to hyperoxia with that in animals exposed to normoxia

alone. The second objective was to use a panel of ATI- (RTI40) and ATII-selective

(RTII70 and MMC4) antibodies, coupled with laser scanning confocal microscopy, to

determine the proportion of the alveolar surface associated with ATI, ATII and

intermediate (MMC4/RTI40-coexpressing) cells to quantify the effect of hyperoxic

exposure on the changes in these cell populations during the resolution of lung injury.

4.2 Results

A total of 24 animals were randomised to either PBS-instilled control (n=8, 8 analysed),

S. aureus installation followed by recovery in normoxia for 120h (n=8, 7 analysed) or S.

aureus installation with recovery in normoxia for 72h followed by 48h in hyperoxia

(n=8, 8 analysed). In the S. aureus-instilled groups, rats received a dose of (3.2 ± 1.1) x

108 cfu. One animal in the S. aureus normoxia group appeared distressed with marked

tachypnoea 24h following S. aureus installation and was withdrawn from the

experimental protocol for the purposes of animal welfare. This animal was excluded

from subsequent analysis.

4.2.1 S. aureus-induced lung injury and the effect of hyperoxia

To characterise the effect of hyperoxia on the resolution of S. aureus-induced injury

total BALF protein, BALF cell counts and BALF RTI40 were determined.

BALF protein: The total protein recovered from BALF of control (PBS-instilled) lungs

93

was 1.1 ± 0.2mg (n=5). In S. aureus-instilled lungs exposed to normoxia alone this rose

to 2.8 ± 1.0mg (n=4), whilst in lungs instilled with S. aureus recovered in normoxia for

3 days and then exposed to hyperoxia for 2 days the total amount of protein recovered

was 5.9 ± 2.4mg (n=5) (figure 26).

BALF cell counts: The total number of leucocytes recovered from BALF was 14 ± 7 x

104 in control lungs (n=5), 314 ± 169 x 10

4 in S. aureus-instilled lungs exposed to

normoxia alone (n=4) and 530 ± 303 x104 in S. aureus-instilled lungs exposed to

hyperoxia (n=5) (figure 27A). Differential cell counts revealed that the majority of

leucocytes recovered were alveolar macrophages, but the presence of significant

numbers of neutrophils in the S. aureus-instilled lungs (0.3 ± 0.6% control lungs, 8.0 ±

2.8% instilled lungs exposed to normoxia alone and 9.3 ± 7.9% instilled lungs exposed

to hyperoxia) confirmed the presence of an acute inflammatory injury in these lungs

(figure 27B).

BALF RTI40: To determine whether there was significant ATI cell injury, BALF RTI40

content was measured. The total amount of RTI40 in BALF recovered from control lungs

was 9.8 ± 3.0 du (n=5). In S. aureus-instilled lungs exposed to normoxia alone this fell

to 7.8 ± 3.6 du (n=4), whilst in lungs instilled with S. aureus and exposed to hyperoxia

the total amount of RTI40 recovered was 5.6 ± 4.5 du (n=5). The observed differences in

BALF RTI40 content were not statistically significant (p=0.4) (figure 28).

4.2.2 The phenotype of the alveolar epithelium in control lung

The method used for quantification of alveolar epithelial surface staining with individual

cell-selective markers was based upon that previously described by Clegg and

colleagues (Clegg et al. 2005), as detailed in materials and methods (section 2.9.6) and

illustrated in figure 30.

94

ATI (RTI40) staining membrane: In control lungs (n=3), 93.6 ± 1.6% of the apical

alveolar epithelial surface stained positively for the ATI cell-selective antibody RTI40

(figures 31 & 36A).

ATII (RTII70 and MMC4) staining membrane: 2.4 ± 0.4% of the alveolar epithelial

surface in control lungs stained positively for both of the ATII-selective proteins RTII70

& MMC4. RTII70 & MMC4 co-expressing cells were typically located in the corners of

the alveolar airspaces and were flanked by RTI40-expressing cells (figure 31 & 36B).

The lengths of alveolar epithelial surface co-expressing RTII70 & MMC4 in control lung

was normally distributed with a mean of 7.6 ± 3.6µm, representing the mean apical

surface length of ATII cells (figure 31 & 37). Small airways in control lung sections

were lined with cuboidal, non-ciliated epithelial cells, morphologically typical of Clara

cells. These cells stained positively for MMC4 but not for RTII70 or RTI40 as described

previously (Boylan et al. 2001) (figure 31).

Intermediate cell (RTI40 and MMC4) staining membrane: In control lungs, 0.6 ± 0.1% of

the alveolar epithelial surface stained positively for both RTI40 and MMC4 (figure 38).

The majority of this positively staining membrane was <1µm in length (mean 0.8 ±

1.0µm), and occurred at the junction of ATI and ATII cells.

4.2.3 The effect of hyperoxia on the proportion of ATI, ATII and intermediate cell-

staining membrane following S. aureus-induced lung injury

ATI (RTI40) staining membrane: In S. aureus-instilled lungs exposed to normoxia alone

(n=3), 84.4 ± 2.6% of the alveolar epithelial surface stained positively for the ATI-

selective protein RTI40. This fell to 73.4 ± 5.3% in S. aureus-instilled lungs exposed to

hyperoxia (n=3, p<0.05) (figures 32, 33, 34, 35 & 36).

95

ATII (RTII70 and MMC4) staining membrane: In S. aureus-instilled lungs exposed to

normoxia alone (n=3), 7.7 ± 1.1% of the alveolar epithelial surface stained positively for

both RTII70 and MMC4 (figures 32, 33 & 36). The mean length of RTII70/MMC4 co-

expressing membrane was 7.3 ± 6.8µm. However, the distribution of the lengths of

RTII70 and MMC4 staining membrane was markedly different from that observed in

control lungs, with a greater range and a negative skew (figure 37). In S. aureus-instilled

lungs exposed to hyperoxia (n=3), 15.5 ± 1.2% of the alveolar epithelial surface stained

positively for both RTII70 and MMC4 (figures 34, 35 & 36), with a mean length of

positively staining membrane of 9.6 ± 6.6µm. The distribution of lengths of positively

staining membrane was binomial, with peaks around (7, 14µm (figure 37).

Intermediate cell (RTI40 and MMC4) staining membrane: In S. aureus-instilled lungs

exposed to normoxia alone (n=3), 11.9 ± 4.2% of the alveolar epithelial surface co-

expressed RTI40 and MMC4 (figures 32, 33 & 38), with a mean length of positively

staining membrane of 6.5 ± 11.2µm. In S. aureus-instilled lungs exposed to hyperoxia

(n=3), 3.0 ± 0.8% of the alveolar epithelial surface stained positively for RTI40 and

MMC4 (figures 34, 35 & 38), with a mean length of co-expressing membrane of 3.3 ±

4.8µm.

4.2.4 The effect of hyperoxia on lung RTI40 concentration following S.aureus-

induced lung injury

Lung samples were obtained from the left upper lobe (controls) and macroscopically

inflamed regions (S. aureus-instilled lungs) and homogenised as detailed in materials

and methods (section 2.10.7). The concentration of RTI40 showed a two-fold decrease in

S. aureus-instilled lungs exposed to hyperoxia compared to those exposed to normoxia

alone (41.4 ± 19.8 vs 18.8 ± 10.4 du/mg protein, n=3, p<0.05). Both lungs exposed to

hyperoxia, and those exposed to normoxia alone, had significantly lower concentrations

of RTI40 than control lung (330.3 ± 88.0 du/mg protein, n=3, p<0.001) (figure 29). In

contrast, PBS-instilled (control) lungs exposed to hyperoxia had a significantly higher

96

concentration of RTI40 compared to control lungs exposed to normoxia alone (566.0 ±

32.5 vs 330.3 880 du/mg protein, n=3, p<0.05).

4.3 Discussion

4.3.1 The effect of hyperoxic exposure on the resolution of lung injury

S. aureus instillation resulted in an inflammatory injury to the lungs in animals

recovered in air (normoxia) alone and in those recovered in air for three days then

switched to hyperoxia (90-95% O2). This was evident by an increase in the total protein

content and number of leucoytes, both neutrophils and macrophages, in BALF recovered

from S. aureus-instilled lungs. Injury was most significant in the lungs of animals

exposed to hyperoxia. Previous characterisation of the model used in these studies has

demonstrated that, under normoxic conditions, the inflammatory response to S. aureus

(ie. the total number of BALF leucocytes) and alveolar wall damage (as assessed by the

total amount of protein in BALF) is maximal at day 1, resolving by day 3 and almost

completely resolved by day 7 following instillation (McElroy et al. 1999; Clegg et al.

2005). At day 3 following S. aureus instillation, the total protein recovered in BALF was

elevated 1.6-fold above control values in these studies compared to a 6.8-fold elevation

at day 1 post-instillation and the total number of BALF leucocytes was elevated 2.5-fold

at day 3 compared to a 57-fold elevation at day 1. This is consistent with the finding

that, although still elevated, the alveolar protein content and number of leucocytes

recovered from S. aureus-instilled lungs exposed to normoxia alone in these studies was

not significantly different from controls at day 5 following instillation. The fact that

hyperoxic exposure of S. aureus-instilled lungs resulted in a significant increase in these

markers of lung injury however, suggests that hyperoxia was either causing an

inflammatory lung injury itself and/or inhibiting the normal resolution of the pre-

existing lung injury.

Continuous exposure of rats to 100% oxygen has been shown in several studies to cause

a progressive inflammatory lung injury, with alveolar epithelial injury and ATI cell

97

necrosis a characteristic feature (Crapo et al. 1982; Crapo 1986; Fracica et al. 1991).

However, these studies refer to the effect of hyperoxia on the normal lung and not it’s

effect on injured lung, and the finding that the amount of RTI40 recovered from BALF, a

well-characterised marker of ATI cell necrosis (McElroy et al. 1995; McElroy et al.

1997), was not significantly elevated in hyperoxia-exposed animals suggests ongoing

ATI cell necrosis was not a central feature of the injury observed.

4.3.2 The phenotype of the alveolar epithelium in control lungs

In control lungs, the alveoli were lined with RTI40-positive (ATI) and RTII70/MMC4-

positive (ATII) cells as reported previously (Boylan et al. 2001; Clegg et al. 2005). The

distribution and extent of the alveolar epithelial surface covered by the two distinct

epithelial cell types determined using the immunotargeting methodology adopted in

these studies was in keeping with conventional (EM) morphological analysis, with Haies

and colleagues reporting an ATI:ATII surface area ratio of 39:1 in normal lungs, almost

identical to that observed in these studies (93.6% ATI: 2.4% ATII) (Haies et al. 1981;

Crapo et al. 1982).

The individual lengths of RTII70/MMC4 coexpressing membrane in control lungs were

normally distributed, with a mean (and mode) of (7µm, reflecting the mean apical

length of an ATII cell. This is also in keeping with the findings of the morphological

studies conducted by Haies and colleagues, which (excluding microvilli and assuming a

circular surface) suggest an apical ATII surface diameter of 8-9µm (Haies et al. 1981).

4.3.3 The phenotype of the alveolar epithelium in resolving S. aureus-induced lung

injury

Examination of the phenotype of S. aureus-instilled lungs exposed to normoxia alone

revealed a significant decrease in ATI cell-staining (RTI40-positive) membrane

consistent with a significant decrease in the proportion of the alveolar epithelial surface

98

covered by ATI cells in these lungs. The observed decrease in lung homogenate RTI40

concentration is consistent with this finding.

There was a 3-fold increase in ATII cell-staining (RTII70/MMC4-positive) alveolar

membrane in normoxia-exposed lungs in keeping with an increase in the population of

ATII cells following injury. However, there was also a dramatic increase in the

proportion of the epithelial surface coexpressing the ATI cell-selective protein RTI40 and

the ATII cell-selective protein MMC4 (11.9% vs 0.6% controls). Deconvolution of high

magnification images has previously confirmed that these proteins colocalise within the

limits of resolution of confocal microscopy ((0.5µm), confirming coexpression on the

same cell membrane, rather than apparent coexpression as an artefact of ATI cells

overlaying ATII cells (Clegg et al. 2005). The finding of RTI40/MMC4-coexpressing

cells both ex vivo in repairing lungs and in vitro as cultured ATII cells lose ATII cell

phenotypic characteristics and gain ATI-like characteristics (see chapter 3), supports the

concept that these cells are kinetically competent to be intermediate cells. The

observation that a significant number of the RTI40/MMC4-positive cells had a luminal

surface greater than that of ATII cells in control lungs, also suggests that these cells are

in the process of becoming attenuated ATI cells.

Together, these data are consistent with the accepted paradigm of alveolar epithelial

repair: damaged ATI cells being replaced by the proliferation of ATII cells and the

subsequent resolution of ATII cell hyperplasia by the transdifferentiation of a

subpopulation of these cells into ATI cells (Uhal 1997). Comparison of the data obtained

in these studies with that obtained by Clegg and colleagues from characterisation of

alveolar epithelium at day 3 following S. aureus instillation supports this interpretation,

suggesting that the alveolar epithelial repair occurring under normoxic conditions

between days 3 to 5 following instillation is associated with an increase in ATI-staining

membrane (from ~80% at day 3 in the studies of Clegg and colleagues to 84% in these

studies) and decrease in ATII-staining membrane (from ~13% at day 3 in the studies of

Clegg and colleagues to 8% in these studies) (Clegg et al. 2005).

99

4.3.4 The effect of hyperoxia on phenotype of the alveolar epithelium in resolving S.

aureus-induced lung injury

In S. aureus-instilled lungs exposed to hyperoxia, both the proportion of ATI cell-

staining membrane and lung RTI40 content were reduced significantly in comparison to

controls and to S. aureus-instilled animals exposed to normoxia alone. This is unlikely to

reflect downregulation of RTI40 expression by hyperoxia because PBS-instilled (control)

lungs exposed to hyperoxia and non-inflamed lung from hyperoxia exposed S. aureus-

instilled lungs showed a significant increase in the concentration of RTI40. Published

work by Cao and colleagues also suggests that RTI40 (T1$) in mice is upregulated by

hyperoxic exposure (Cao et al. 2003). It therefore seems likely that the observed

decrease in ATI cell-staining membrane reflects a significant decrease in the proportion

of the alveolar epithelial surface covered by ATI cells in hyperoxia-exposed S. aureus-

instilled lungs. Consistent with this, there was a 6-fold increase in the proportion of ATII

cell-staining alveolar membrane in these lungs.

In S. aureus-instilled lungs exposed to hyperoxia, not only was there a significant

increase in the proportion of ATII cell-staining membrane, but there was also a

difference in the distribution of RTII70/MMC4 coexpressing membrane lengths, with a

significant number of longer lengths of coexpressing membrane and a trinomial

distribution with peaks around 7, 14 and 22µm (figure 27). Assuming that the surface

area of ATII cells is comparable in both control and injured lungs, this finding is

consistent with the concept of significant ATII cell hyperplasia in the injured lung, with

2 and (less frequently) 3 ATII cells lying adjacent to one another.

Hyperoxia has been shown to inhibit ATII cell proliferation both in vivo (Crapo 1986;

O'Reilly et al. 1998; O'Reilly et al. 1998) and in vitro (see chapter 3), and this would

suggest that the ATII cell proliferation observed in hyperoxia-exposed lungs occurred

during the 3-day recovery of the S. aureus-instilled animals in normoxia and prior to

transfer into hyperoxic conditions. This, in combination with the fact that the proportion

of ATII cell-staining membrane was significantly greater in hyperoxia-exposed animals

compared to those exposed to normoxia alone suggests that hyperoxia significantly

100

inhibits the resolution of ATII hyperplasia and the restoration of normal cell numbers

which occurs during normal repair.

The ATII cell hyperplasia associated with normal alveolar epithelial repair is thought to

be resolved by a combination of apoptosis and differentiation of ATII cells into ATI

cells (Fehrenbach et al. 1999; Fine et al. 2000). Failure of this resolution has been

speculated to be a key event in the disruption of epithelial-fibroblast interactions and

progression to abnormal epithelial repair with fibrosis (see section 1.3.4) (Adamson et

al. 1988).

As hyperoxia is pro-apoptotic to ATII cells (see chapter 3) (Haddad and Land 2000;

Lee and Choi 2003; Buccellato et al. 2004), this suggests that the lack of resolution of

the ATII cell hyperplasia observed in hyperoxia-exposed S. aureus-instilled lungs is

largely due to the inhibition of the differentiation of ATII cells into ATI cells. This

concept is supported by the in vitro data presented in this thesis (see chapter 3), and also

by the finding of significantly less intermediate (RTI40/MCC4 coexpressing) membrane

in the lungs of animals exposed to hyperoxia compared to those exposed to normoxia

alone (11.9% vs 3.0%).

4.3.5 General discussion

The mechanism by which hyperoxia inhibits the differentiation of ATII cells into ATI

cells in the repairing lung was not investigated. However several studies have shown

that upregulation of insulin-like growth factor (IGF) components occurs during

hyperoxia-induced lung injury (Besnard et al. 2001; Chetty and Nielsen 2002;

Narasaraju et al. 2006), and a recent study has demonstrated that addition of antibodies

to the IGF-1 receptor to ATII cells in culture inhibits the transition of cells from an

ATII-like to an ATI-like phenotype with time in culture (Narasaraju et al. 2006). The

change in expression profile of the IGF system might therefore be speculated to have a

central role in the mechanism. It has also been suggested that transdifferentiation is

modulated by signalling through the Smad-dependent TGF-&1 pathway (Bhaskaran et al.

101

2007), raising the possibility that hyperoxia may inhibit transdifferentiation through

inhibition of TGF-&1 signalling.

Intratracheal instillation of S. aureus produced a reproducible, heterogeneous pattern of

injury most frequently affecting the left lung. This is in keeping with other published

models of pulmonary infection in the rat (Bakker-Woudenberg 2003). The left upper

lobe was taken consistently for biochemical and morphological analysis from control

animals, but in S. aureus-instilled animals macroscopically inflamed lung was readily

identifiable and was separated from non-inflamed lung prior to tissue processing as

described in materials and methods (section 2.10.7 & 8). A limitation of this approach,

inherent in the morphological analysis of any model producing a heterogeneous pattern

of injury, was that initial tissue sampling was based upon identification of

macroscopically inflamed areas and was not strictly random. This increases the

likelihood of sampling error. However, the aim of these studies was to compare discrete

areas of repairing lung with relatively homogeneous uninjured (control) lung. As such,

the data presented are not reflective of the whole lung, but rather a representative region

of injured repairing lung and its reponse to hyperoxia.

Hyperoxic exposure in these experiments was achieved by means of animal housing in

custom-built chambers as described in materials and methods (section 2.10.4). The

oxygen tension within the chambers was recorded continuously throughout hyperoxic

exposure and remained >90% at all times (mean 93 ± 2%). In spontaneously breathing

rats with normal (unconsolidated) lungs, this fraction of inspired oxygen would result in

an alveolar partial pressure of oxygen in the region of ( 90kPa. In the heterogeneously

injured lung however, in which there are consolidated and underventilated regions, it is

likely that there are alveoli in which the alveolar partial pressure of oxygen is

significantly less than this. As the partial pressure of oxygen within individual alveoli

cannot be determined, it is not possible to correlate the observed changes in alveolar

epithelial phenotype with a precise knowledge of the partial pressure of oxygen to which

the epithelium was exposed. However, despite this drawback, the model of an animal

with a heterogeneous lung injury breathing high concentrations of oxygen remains

102

clinically relevant, and it does not seem unreasonable to compare the effects of

‘hyperoxic’ and ‘normoxic’ exposure.

4.4 Summary

The studies outlined in this chapter demonstrate the novel use of immunotargetting to

investigate the effect of hyperoxia on the phenotype of the alveolar epithelium during

resolution of S. aureus-induced lung injury. The use of cell-selective markers, combined

with laser scanning confocal microscopy and image analysis of large contiguous areas of

lung tissue, enabled the extraction of quantitative data demonstrating the proportion of

the alveolar epithelial surface associated with ATI cells, ATII cells and cells with an

intermediate immunophenotype and the effect hyperoxic exposure has on the changes in

these cell populations during the resolution of lung injury.

Characterisation of the phenotype of the repairing epithelium during resolution of S.

aureus-induced injury in lungs exposed to normoxia alone revealed a reduction in the

proportion of ATI(RTI40)-staining epithelial surface with an increase in

ATII(RTII70/MMC4)-staining surface. Inflamed regions in these lungs were

characterised by a significant proportion of the epithelial surface having an intermediate

or transitional phenotype expressing both RTI40 and MMC4. This intermediate cell-

staining membrane was not present to any significant degree in control lungs, but was

observed in vitro as ATII-like cells became ATI-like cells with time in culture (chapter

3). Together, these data are consistent with the accepted paradigm of alveolar epithelial

repair: damaged ATI cells being replaced by the proliferation of ATII cells and the

subsequent resolution of ATII cell hyperplasia by the transdifferentiation of a

subpopulation of these cells into ATI cells (Uhal 1997).

In S. aureus-instilled animals exposed to hyperoxia, the repairing epithelium had a

different phenotype from both control and S. aureus-instilled lungs exposed to normoxia

alone. Inflamed regions in these lungs were still characterised by a reduction in the

proportion of ATI-staining surface and an increase in the proportion of ATII-staining

membrane, but the increase in ATII-staining membrane was significantly greater than

103

that observed in S. aureus-instilled lungs exposed to normoxia alone. As hyperoxia is

pro-apoptotic and known to inhibit ATII cell proliferation, and coupled with the finding

that hyperoxia-exposed lungs had significantly less intermediate cell-staining membrane,

these data suggest a reduction in resolution of ATII cell hyperplasia in hyperoxia-

exposed lungs due to inhibition of the transdifferentiation of ATII cells into ATI cells.

These data support the in vitro findings presented in this thesis (chapter 3), and are

consistent with the hypothesis that clinically relevant levels of hyperoxia inhibit normal

alveolar epithelial repair by inhibiting the differentiation of ATII cells into ATI cells.

104

5. Mechanical deformation-induced injury of alveolar epithelial cells

5.1 Introduction & objectives

The basement membrane underlying the alveolar epithelium undergoes marked

expansion as the lung is inflated toward total lung capacity (Tschumperlin and

Margulies 1999). This suggests that alveolar epithelial cells may be exposed to

significant mechanical deformation during ventilation at high lung volumes. Whilst

mechanical deformation of alveolar epithelial cells is known to have important

functional and developmental roles (Riley et al. 1990; Wirtz and Dobbs 2000), it is also

potentially injurious (Tschumperlin and Margulies 1998; Tschumperlin et al. 2000), and

deformation-induced epithelial injury is thought to play a central role in the disruption of

the air-blood barrier that occurs during the development of ventilator-induced lung

injury (VILI) (Tremblay and Slutsky 1998; Slutsky 1999). This epithelial injury is

thought to result from the alveolar overdistension in aerated alveoli during positive

pressure ventilation of the heterogeneously injured lung.

The clinical signficance of this is highlighted by the fact that following acute lung

injury, lung protective strategies of ventilation, designed to limit alveolar overdistension,

are associated with lower levels of epithelial and endothelial injury (Frank et al. 2002),

lower organ dysfunction scores and reduced mortality (Amato et al. 1998; Ranieri et al.

1999; ARDSNet 2000).

Whilst both clinical and animal studies have shown that mechanical ventilation can

worsen existing lung injury and produce an iatrogenic injury, the effect that mechanical

ventilation has on the mechanisms underlying alveolar epithelial repair is not known.

The Flexercell FX-4000T" is a commercially available apparatus that allows the

application of uniform radial and circumferential strain to cells cultured on flexible

silastic membranes in the BioFlex! cell culture system. Both the magnitude and

frequency of the strain applied to cells is computer regulated, allowing the exposure of

cells to uniform and quantifiable deformations in vitro and enabling a simple correlation

between deformation magnitude and cell response.

105

The first aim of the studies outlined in this chapter was to establish the culture of

primary alveolar epithelial cells in the BioFlex! cell culture system and to characterise

the use of the Flexercell FX-4000T by determining the vulnerability of primary alveolar

epithelial cell monolayers to deformation-induced injury. Having established the

relationship between cell injury and deformation magnitude, the objective was to use

this in vitro system to investigate the hypothesis that the migrating and flattening cells in

regions of ‘repairing’ epithelial cell monolayers are more susceptible to deformation-

induced injury than ‘normal’ epithelium.

5.2 Results

5.2.1 Culture of alveolar epithelial cells in a mechanically active environment

Primary alveolar epithelial cells were seeded at constant density on the fibronectin-

coated silastic membranes of BioFlex! cell culture plates. In all experiments, cell

attachment was successfully confined to the central portion of the membrane by seeding

within a removable silicon gasket placed in the centre of the culture well. After 6 h in

culture, cells appeared to be fully adherent to culture substratum. Gaskets were removed

24 h post-seeding. After 5 days in culture, cells formed a confluent monolayer

comprising two broadly distinct cell types: a central area of small densely grouped cells

(SA 490 ± 116µm2), surrounded by a peripheral region of larger cells (SA 1963 ±

527µm2), which had flattened and spread (figure 45).

Day-5 alveolar epithelial cell monolayers were subject to 1 h of cyclic equibiaxial

mechanical strain at a frequency of 15 cycles per minute using a Flexercell FX-4000T

strain unitTM

(figure 39). Deformation-induced injury was assessed 1 h after completion

of this strain protocol using a Live/DeadTM

cell viability assay.

Cells remained attached to the culture surface during all the deformation protocols used,

and by restricting the position of cell attachment to the central portion of the membrane

all cells were subjected to identical strains in both radial and circumferential directions

(figure 39).

106

5.2.2 Deformation-induced injury and magnitude of mechanical strain

To determine the extent of cell injury following exposure to a range of clinically

relevant mechanical deformations, day-5 alveolar epithelial cell monolayers were

subjected to 1 h of cyclic equibiaxial mechanical strain at physiological magnitudes of

12 and 22% #SA and at the pathological magnitude of 44% #SA. In the central region

of cell monolayers, there was no difference in cell viability between unstrained controls

and cells strained at 12% change in basement membrane surface area (#SA) (1.4 ± 0.7%

control vs 1.4 ± 0.6% 12% #SA, p>0.05, n=5). Cell death increased slightly at 22% #SA

but this was not statistically significant compared to unstrained controls (4.3 ± 0.8%,

p>0.05, n=5). At 44% #SA there was a significant increase in deformation-induced

injury (48.9 ± 12.5%, p<0.001, n=5) (figures 40 & 41).

5.2.3 Deformation-induced injury and cell phenotype

In the central region of day-5 epithelial cell monolayers, there was no significant

deformation-induced injury in cells strained at 12% #SA, however, even at this

physiological level there was significant cell death in cells located in the peripheral

region of the same monolayers (1.4 ± 0.6% central vs 49.5 ± 12.2% peripheral, p<0.001,

n=5) (figures 43 & 44).

Both centrally located cells and those towards the periphery of the monolayer lacked

lamellar bodies. Cells towards the periphery of the monolayer expressed the ATI-

selective protein RTI40 but not the ATII-selective proteins MMC4 or RTII70. Centrally

located cells did not express RTI40, but stained weakly for MMC4 (n=3) (figure 46).

5.2.4 Deformation-induced injury in repairing wound

Alveolar epithelial cell monolayers were ‘wounded’ by scoring across the central section

of day-3 monolayers with a 200µl pipette tip. Wounded monolayers were then cultured

107

under standard conditions for a further 48 h prior to mechanical deformation of

‘repairing’ wounds at 12% #SA.

Cells in the repairing region of epithelial cell monolayers appeared to express RTI40

(n=1) and spread to occupy a significantly greater surface area in comparison to cells in

the unwounded central portion of the same monolayers (1440 ± 319µm2 vs 487 ±

191µm2, p<0.05, n=3). Deformation caused significant injury of cells in repairing

wounds in comparison to the same region of repairing alveolar epithelium in unstrained

controls (38.0 ± 8% cell death vs 8.3 ± 1.6% unstrained controls, p < 0.005) (figures 47

& 48).

5.3 Discussion

5.3.1 Deformation-induced injury is a non-linear function of deformation

magnitude

Mechanical deformation-induced injury was found to be a non-linear function of

deformation magnitude in these studies, with no statistically significant cell death

occurring with cyclic (physiological) deformations up to and including 22% #SA, but

significant cell death occurring at (pathological) deformations of 44% #SA.

Reassuringly, these data are in broad agreement with the only other known report of

changes in day-5 alveolar epithelial cell viability with quantified deformation

(Tschumperlin and Margulies 1998). These findings also make intuitive sense when

interpreted in light of the changes in lung volumes that these deformations are thought to

represent (figure 42).

In the isolated rat lung, inflation from functional residual capacity (FRC) to 80% of total

lung capacity (TLC) has been shown to result in a change in the epithelial basement

membrane surface area of (25%, whilst inflation to 100% TLC results in a change of

(30-40% (Mercer and Crapo 1990; Tschumperlin and Margulies 1999). Mechanical

deformation producing a 22% #SA might therefore be reasonably interpreted as

physiological in magnitude, whilst a 44% #SA is greater than that experienced in

108

inflation of the normal lung to TLC and is likely to be pathological. A 44% #SA was

chosen for these experiments because it was thought to reflect the deformations likely to

occur during alveolar overdistension resulting from positive pressure ventilation of the

heterogeneously injured lung.

Whilst there are limitations in the use of alveolar epithelial cells in culture as a surrogate

for the alveolar epithelial barrier, it is reassuring that there appears to be a close

correlation between the finding of ATI cell injury in electron micrographs of rat lungs

inflated beyond TLC (Dreyfuss et al. 1988) and the findings in these studies of a

dramatic increase in deformation-induced injury when deformations exceed those

associated with inflation to TLC.

5.3.2 Spreading RTI40-expressing cells in the peripheral region of epithelial cell

monolayers are susceptible to deformation-induced injury

The finding that a physiological level of strain (12% #SA) produces significant cell

death in the peripheral region of an alveolar epithelial cell monolayer, but not in the

central region of the same monolayer, is a novel finding. Primary ATII cells typically

flatten and spread in culture, losing their lamellar bodies and expression of ATII-

selective proteins whilst simultaneously acquiring expression of ATI-selective proteins

(chapter 3) (Dobbs et al. 1985; Danto et al. 1992; Paine and Simon 1996). This

transition from an ATII- into an ATI-like phenotype typically occurs within 3 d of

seeding, suggesting that a monolayer of day-5 alveolar epithelial cells may uniformly

represent this ATI-like phenotype. However, in these studies, cells were seeded at

relatively high density within removable gaskets in order to constrain cell attachment to

the central portion of the culture membrane. Whilst this ensured that all cells within the

monolayer were exposed to uniform patterns of strain, an artefact of this method of

seeding was the development of different cell phenotypes within the monolayer, as upon

removal of the silicon gasket, cells in the peripheral region were free to flatten and

spread onto unoccupied culture surface, whilst cells in the central region were prevented

from doing so by neighbouring cells. After 5 d in culture, cells in the peripheral region

109

of the monolayer resembled ATI cells both morphologically and phenotypically, being

large, attenuated and RTI40-expressing. These findings are consistent with the

morphologic and phenotypic changes known to occur in ATII cells with time in culture.

However, cells in the central region of the monolayer appeared relatively

undifferentiated. They were less spread and demonstrated only weak MMC4 expression.

Cells in the central region of the monolayer did not express either RTII70 or RTI40. The

finding of different levels of cell injury in the two regions suggests that the different

alveolar epithelial cell phenotypes observed (relatively undifferentiated and ATI-like)

have different susceptibilities to deformation-induced injury, with the larger RTI40-

expressing (ATI-like) cells being more susceptible to injury.

Although there are limitations inherent in the extrapolation of any in vitro model, there

are clear parallels between the system of ATII cell culture used in these studies and the

mechanisms thought to operate in the alveolar epithelium during epithelial repair in vivo

– namely re-epithelialisation of denuded basement membrane by the transition of a

population of ATII cells into ATI cells, possibly via some kind of undifferentiated cell

phenotype (figure 1). Viewed in this light, it is clear that spreading RTI40-expressing

cells in the peripheral region of monolayers share many similarities with alveolar

epithelial cells in regions of epithelial repair. These data would then support the concept

that the spreading and flattening cells in regions of active epithelial repair are susceptible

to deformation-induced injury even at physiological levels of mechanical strain.

5.3.3 Epithelial cells in the region of repairing epithelial monolayers appear

susceptible to deformation-induced injury

The ‘repair’ of wounded d 3 epithelial cell monolayers appeared to occur by migration

and spreading of cells from the wound edge. By 48 h following wounding, cells in the

region of the repairing wound displayed similar characteristics to those of cells in the

peripheral region of the monolayers, having adopted an ATI-like (RTI40-expressing)

phenotype and spread to occupy a greater surface area. The finding that spreading RTI40-

expressing cells in regions of repairing wounds appear susceptible to deformation-

110

induced injury at physiological levels of deformation supports the concept that cells in

regions of active alveolar epithelial repair may also be highly susceptible to

deformation-induced injury in vivo. These data may explain the clinical observation that

patterns of mechanical ventilation which are not injurious in the normal lung appear to

cause significant injury in the already injured repairing lung (ARDSNet 2000; Frank et

al. 2002).

5.3.4 General discussion

Freshly isolated ATII cells did not attach well to uncoated silastic membranes, and the

BioFlex, culture plates were therefore pre-coated with extracellular matrix protein prior

to seeding. In preliminary experiments, pre-coating the culture plates with fibronectin,

collagen or laminin, the major components of the alveolar epithelial basement

membrane, did not appear to significantly alter either cell attachment or the pattern of

deformation-induced cell injury observed. Fibronectin was therefore chosen because it

has been shown to stimulate alveolar epithelial cell spreading in vitro and is found in

elevated concentrations in the basement membrane of the repairing lung (Sugahara et al.

1993).

Because cells in these studies were exposed to uniform patterns of strain, it is reasonable

to assume that the differences in the cellular response observed are a reflection of

differences in cellular properties. Cell morphology, phenotype, cytoskeletal structure,

cell-cell and cell-matrix attachments may all play a role in this heterogeneity of

response. Cells in the peripheral region of alveolar epithelial cell monolayers had a

significantly greater surface area than those in the central region, and Tschumperlin and

colleagues have previously reported a dependence of deformation-induced injury on

mean cell size, with injury of ATI-like cells being greatest in cells with largest surface

area (Tschumperlin et al. 2000). However, these investigators also found that day-1

alveolar epithelial (ATII-like) cells, with a significantly smaller size, were more

sensitive to deformation-induced injury than day-5 (ATI-like) cells suggesting that it is

not only cell surface area which determines the sensitivity of individual cells to injury.

111

Whilst previous studies suggest that ATII-like cells may be more sensitive to mechanical

deformation-induced injury than ATI-like cells in vitro, these studies have assumed the

phenotype of the cells based on time in culture alone rather than the expression of cell-

selective proteins. The correlation between the expression of RTI40 and increased

sensitivity to injury within the same cell culture is a novel finding.

The function of RTI40 is unknown, but it has been speculated to have a functional role in

cell spreading and migration (chapter 3) (Williams 2003). It increases migration rates in

wounding experiments (Schacht et al. 2003), and time-lapse studies of the transition of

ATII cells into ATI-like cells in culture, reveal that RTI40 is initially expressed in the

leading or spreading cell edge (McKechnie and Clegg unpublished data and figure 4).

The finding that RTI40 expressing cells are more susceptible to deformation-induced

injury may therefore lend support to the hypothesis that cells in the process of spreading

and migrating are more vulnerable to injury.

The outcome measure used to assess deformation-induced injury in these studies was

loss of cell membrane integrity, which leads to cell death. To determine whether cells

were able to recover from the membrane disruption allowing ethidium homodimer-1 to

enter cells, a number of wells were followed for 24 h following completion of

deformation with the addition of fresh calcein AM. Cells labelled with ethidium

homodimer-1 did not appear to recover their ability to concentrate calcein, suggesting

that the deformation-induced injury in these cells was irreversible.

Alveolar epithelial damage due to deformation is likely to occur as a continuum of cell

responses, from loss of normal cell function through to gross structural failure. Cell

death represents the extreme end of this continuum, and more subtle forms of injury,

such as disruption of tight junction function and alveolar epithelial transport, may well

contribute to deformation-induced damage of the air-blood barrier. These more subtle

forms of cell injury were not investigated in these studies.

In addition, the mechanism of cell injury was not formally examined. Although

mechanical strain has previously been shown to induce apoptosis in ATII cells (Edwards

et al. 1999), nuclear staining did not reveal significant numbers of apoptotic nuclei in

these experiments, and preliminary studies using a polyclonal anti-active caspase-3

112

antibody did not show significant caspase-3 activation. These findings suggest that the

deformation-induced cell injury in these studies was necrotic rather than apoptotic, most

likely resulting from plasma membrane stress failure (Vlahakis and Hubmayr 2000).

Oxidative stress secondary to stretch-induced antioxidant depletion and increased ROS

generation in response to mechanical deformation has been implicated as a potential

mechanism underlying deformation-induced epithelial cell injury (Jafari et al. 2004;

Chapman et al. 2005). These studies suggest that cells generating fewer ROS in response

to deformation may be more resistant to deformation-induced injury. Intriguingly, such

cells also appear resistant to hyperoxia-induced injury (section 3.3.8).

5.4 Summary

The findings of these in vitro studies support the hypothesis that at high lung volumes

alveolar epithelial cells undergo irreversible cell damage. The finding that different

alveolar epithelial phenotypes within the same culture show different sensitivities to

deformation-induced injury, with spreading RTI40-expressing cells more susceptible to

injury, suggests that epithelial cells in the process of spreading and adopting an ATI

phenotype in regions of epithelial repair may be more susceptible to deformation-

induced injury, and may be injured even at physiological levels of mechanical strain.

The increased susceptibility of cells in the region of ‘repairing’ epithelial monolayer

wounds to deformation-induced injury further supports this hypothesis. These data

suggest that alveolar epithelial deformation-induced injury may play a central role in the

development of ventilator-induced lung injury during mechanical ventilation of the

already injured lung.

113

6. Alveolar epithelial injury in a rat haemorrhagic shock-induced

model of acute lung injury

6.1 Introduction

Haemorrhagic shock is a common cause of acute lung injury. Unlike pneumonia

however, haemorrhage results in an indirect injury to the lung (Bernard et al. 1994;

Ware and Matthay 2000).

The distinction between direct and indirect lung injury has been recognised for some

time. Disorders resulting in ‘direct’ injury to the alveolar air-blood barrier (most

commonly gastric aspiration and pulmonary infection) appear to inflict damage to the

alveolar-capillary membrane through a local inflammatory response. Conversely

‘indirect’ injury to the lung (caused most often by sepsis, trauma or massive blood

transfusion) is thought to result from circulating mediators generated by extra-

pulmonary sources of inflammation. Although the distinctions in pathogenesis are

largely speculative, there are differences in the clinical behaviour of direct and indirect

injuries, and clinical observation supports the existence of two distinct pathogenic

pathways with different morphological aspects, respiratory mechanics and responses to

ventilatory strategies (Gattinoni et al. 1998; Pelosi et al. 2003).

There is experimental evidence that haemorrhagic shock and fluid resuscitation are

associated with lung endothelial injury (Todd et al. 1978; Michel et al. 1981), and that

severe haemorrhage alters alveolar epithelial barrier function (Pittet et al. 1996; Pittet et

al. 1996; Modelska et al. 1997), but few studies have examined whether haemorrhage is

injurious to the alveolar epithelium.

The use of cell-selective markers in investigating epithelial injury is not without

precedent (see section 1.4), and offers the advantage of indicating the degree to which

individual epithelial cell populations are affected during injury (McElroy and Kasper

2004). This approach has not been used to study haemorrhagic shock-induced lung

injury.

114

The initial objective of the work outlined in this chapter was to establish a (non-

recovery) rat model of haemorrhagic shock-induced lung injury, characterise the injury

and, using epithelial cell-selective proteins, assess the severity of epithelial injury.

Having achieved this, the objective was to develop the model into a recovery model that

could be used in combination with cell-selective biomarkers to compare the effects of

hyperoxia (± mechanical ventilation) on the phenotype of the repairing alveolar

epithelium following direct (pneumonia-induced, chapter 4) and indirect (haemorrhagic

shock-induced) injuries.

The haemorrhagic shock protocol chosen in this study (figure 49) was based upon that

established by Modelska et al. (Modelska et al. 1997). The ventilatory strategy used was

clinically applicable and has previously been shown not to cause lung injury in isolation

(Frank et al. 2002).

6.2 Results

A total of 6 animals, 3 undergoing haemorrhage and 3 controls (surgery without

haemorrhage), completed the experimental protocol (figure 49). Blood loss during

surgery was minimal. Only animals with normal acid-base status at the end of the

stabilisation period were included in the study. The blood pressure profile of a typical

animal undergoing surgery, haemorrhage and fluid resuscitation is shown in (figure 50).

6.2.1 Characterisation of haemorrhagic shock-induced acute lung injury

BALF Protein: BALF recovered from animals undergoing haemorrhage had

significantly (5-fold) higher levels of total protein (13.2 ± 2.1mg haemorrhage vs 2.7 ±

1.1 control, p<0.01) (figure 51), indicative of a loss of alveolar epithelial integrity.

115

BALF cell counts: There was no statistically significant difference in the total cell counts

in BALF between the two groups (86.2 ± 13.9x104

cells haemorrhage vs 97.3 ± 19.9

control, p=0.24) (figure 52A). However, differential cell counts revealed a significantly

higher proportion of neutrophils in the BALF recovered from animals undergoing

haemorrhage (12.7 ± 2.1% haemorrhage vs 0.1 ± 0.1% control, p<0.001), with a

decrease in the proportion of alveolar macrophages (82.3 ± 2.9% haemorrhage vs 98.3 ±

0.6% control, p<0.001) (figure 52B). There was no statistically significant difference

between the number of other cell types identified in the BALF in the two groups.

6.2.2 Alveolar epithelial cell injury following haemorrhagic shock

ATI-selective proteins: The amount of the ATI-selective integral membrane protein

RTI40 showed a 7-fold increase in BALF recovered from animals undergoing

haemorrhage (108 ± 54 du haemorrhage vs 15 ± 9 du control, p<0.05) (figure 53A).

There was also a 2-fold increase in the ATI-selective protein MMC6 (6.9 ± 0.8 du

haemorrhage vs 2.6 ± 0.8 control, p<0.05) (figure 53B). Neither RTI40 nor MMC6 were

detected in plasma samples from either control or haemorrhaged rats.

ATII-selective proteins: Although there was an increase in the level of the ATII-selective

protein MMC4 in BALF recovered from rats undergoing haemorrhage (125 ± 76 du

haemorrhage vs 63 ± 39 control, p=0.16), this did not reach statistical significance.

6.3 Discussion

6.3.1 Haemorrhagic shock results in acute lung injury

Loss of alveolar air-blood barrier integrity, with accumulation of protein-rich oedema

fluid and neutrophils within the airspaces, is a pathognomonic feature of the acute phase

116

of acute lung injury (Ware and Matthay 2000). The data presented in this chapter,

demonstrating a significant increase in the level of protein and number of neutrophils in

BALF following haemorrhage, confirm that a clinically relevant model of haemorrhagic

shock produces significant lung injury, and that it does so within six hours of the onset

of haemorrhage.

6.3.2 Haemorrhagic shock is associated with ATI cell injury

The use of alveolar epithelial cell-selective proteins as markers of acute lung injury

offers the advantage of determining which of the specific alveolar epithelial cell

populations is affected (McElroy and Kasper 2004). For example, elevation of the ATI-

selective integral membrane protein RTI40 in BALF has been shown to provide a

biochemical marker of ATI cell injury in a Pseudomonas aeruginosa-induced model of

lung injury. In this model, the degree of RTI40 elevation correlated with both the degree

of injury, assessed morphologically, and the severity of functional disruption of the

alveolar air-blood barrier (McElroy et al. 1995).

The 7-fold elevation of the ATI-selective protein RTI40 within 6 hours of the onset of

haemorrhage is a novel finding and suggests that haemorrhagic shock resulted in

significant alveolar type I cell injury in this model. Further evidence for this is provided

by the finding of a statistically significant increase in the level of MMC6.

The ATII-selective marker MMC4 was elevated 2-fold, which although suggestive of

alveolar type II cell injury, was not statistically significant. However,

immunophenotyping of injured lungs following S. aureus instillation (see chapter 4)

revealed that MMC4 is expressed by activated alveolar macrophages in addition to ATII

cells in the inflamed lung, suggesting that BALF MMC4 content is unlikely to prove a

useful marker of ATII cell injury.

The mechanism of haemorrhagic shock-induced alveolar epithelial injury was not

investigated in detail. The finding of significant alveolar neutrophil infiltration suggests

that the mechanism is, at least in part, inflammatory in nature. This would be consistent

117

with the prevailing view that neutrophils have an important early immunomodulatory

role in haemorrhage-induced acute lung injury, being responsible for the initial

activation of nuclear factor-'B (NF-'B) and subsequent increase in proinflammatory

cytokines such as interleukin-1& (IL-1&) and tumour necrosis factor-$ (TNF-$)

(Shenkar and Abraham 1993; Shenkar et al. 1996; Abraham et al. 2000).

Bacterial translocation, in which viable bacteria or bacterial endotoxins pass from the

gastrointestinal tract to extraintestinal sites as a consequence of gut ischaemia, has been

implicated in the pathogenesis of sepsis (Rush et al. 1988; Sori et al. 1988), and may

play a role in the pathogenesis of acute lung injury following haemorrhage. In a rat

model of haemorrhagic shock, Tani and colleagues demonstrated bacterial translocation

and upregulation of the proinflammatory cytokine tumour necrosis factor-$ (TNF-$)

within 1h of haemorrhage (Tani et al. 2000). However, these effects were prevented by

hyperoxia (inhaled 100% O2) early in the course of haemorrhagic shock, suggesting that

bacterial translocation is unlikely to have played a significant role in this model. This

hypothesis is supported by the fact that blood cultures taken from animals completing

the study were negative in all cases.

6.4 Summary

The objective of this study was to establish a reproducible model of indirect acute lung

injury and investigate the affect of this injury on the alveolar epithelium. It was hoped

the model could then be modified to establish a recovery model enabling the study of

alveolar epithelial repair following indirect lung injury. Unfortunately, the model proved

difficult to establish with a significant animal mortality rate, and only the biochemical

characterisation of the lung injury was possible.

Whilst the work presented in this chapter reflects only the early stages in the

development of a model of haemorrhagic shock-induced acute lung injury, it does

suggest that haemorrhage is injurious to the alveolar epithelium, leading to significant

118

ATI cell necrosis. As such, it establishes an important proof of principle and provides a

positive platform for further work.

119

7. Summary

Oxygen and mechanical ventilation are key therapeutic interventions in the supportive

treatment of respiratory failure following lung injury. Although both hyperoxia and the

mechanical deformations associated with mechanical ventilation are known to be

potentially injurious to the normal lung, their effects on alveolar epithelial repair

following injury are not known. The clinical impression however, is that poor outcome

is associated with exposure of injured (repairing) epithelium to these iatrogenic ‘hits’.

This thesis describes in vitro (chapter 3) and in vivo (chapter 4) studies investigating

the hypothesis that hyperoxia inhibits normal alveolar epithelial repair by inhibiting the

transdifferentiation of ATII cells into ATI cells, and in vitro studies addressing the

hypothesis that (spreading and flattening) alveolar epithelial cells in regions of epithelial

repair are susceptible to mechanical deformation-induced injury (chapter 5).

7.1 Hyperoxia inhibits the transdifferentiation of ATII cells into ATI cells

The in vitro data presented demonstrate for the first time that hyperoxia reversibly

inhibits the changes in morphology and cell-selective protein expression characteristic of

the transition of ATII-like cells into ATI-like cells with time in culture. These studies

also confirm that hyperoxia inhibits proliferation and is cytotoxic to alveolar epithelial

cells, with cell death occuring by both apoptosis and necrosis following prolonged

exposure. However, it appears that hyperoxia is not universally cytotoxic, and these

studies suggest that the (isolated) ATII cell population harbours a subpopulation of cells

that have proliferative potential and are less susceptible to hyperoxia-induced injury.

These cells appear to generate fewer ROS and have lower levels of E-cadherin

expression.

The effect of hyperoxia on alveolar epithelial repair in vivo was investigated using a

novel immunotargeting approach and quantitative comparison of the phenotype of

repairing alveolar epithelium following S. aureus-induced lung injury.

120

In S. aureus-instilled lungs exposed to normoxia alone, these studies revealed a

reduction in the proportion of ATI(RTI40)-staining epithelial surface and an increase in

ATII(RTII70/MMC4)-staining surface, consistent with ATI cell injury and ATII cell

proliferation. Inflamed regions in these lungs were also characterised by a significant

proportion of the alveolar epithelial surface having an intermediate immunophenotype

expressing both the ATI-selective RTI40 and the ATII-selective MMC4. This

RTI40/MMC4-coexpressing membrane was not present to any significant degree in

control lungs, but was observed in vitro as ATII-like cells became ATI-like cells with

time in culture. These data are consistent with the accepted paradigm of normal alveolar

epithelial repair: damaged ATI cells being replaced by the proliferation of ATII cells and

the subsequent differentiation of a subpopulation of daughter cells into ATI cells, with

this differentiation of ATII into ATI cells occurring via cells with an intermediate

(RTI40/MMC4-coexpressing) cell phenotype.

Markers of lung injury were elevated to a greater degree in S. aureus-instilled lungs

exposed to hyperoxia than in those exposed to normoxia alone, suggesting that

hyperoxia was either causing an inflammatory lung injury in the repairing lung itself

and/or inhibiting normal resolution of the pre-existing lung injury. Examination of the

alveolar epithelium in S. aureus-instilled lungs exposed to hyperoxia revealed significant

differences in the immunophenotype, with a greater proportion of ATII-staining alveolar

epithelial surface. As hyperoxia is known to be pro-apoptotic and to inhibit ATII cell

proliferation, both factors likely to reduce the ATII cell population, the observed

increase in ATII-staining alveolar epithelial surface following hyperoxic-exposure

suggests that hyperoxia inhibits normal epithelial repair by inhibiting the differentiation

of ATII cells into ATI cells, preventing effective resolution of ATII cell hyperplasia.

The finding that hyperoxia-exposed lungs had significantly less intermediate cell-

staining membrane is consistent with this interpretation.

The in vitro and in vivo studies described support the hypothesis that prolonged exposure

to clinically relevant levels of hyperoxia is injurious to the alveolar epithelium and

inhibits normal alveolar epithelial repair by inhibiting the differentiation of ATII cells

into ATI cells. This is a novel finding.

121

7.2 Spreading RTI40-expressing cells are susceptible to deformation-induced injury

The effect of mechanical deformation on alveolar epithelial cells was investigated by

examining changes in cell viability following exposure of cells to uniform, quantified

equibiaxial strain. The studies performed to characterise the in vitro system used for

these experiments demonstrated that deformation-induced injury was a non-linear

function of deformation magnitude, with significant injury occurring following exposure

of cells to strains greater than those associated with inflation of the lung to total lung

capacity (>40% #SA), but not over a range of more physiological levels of strain (<30%

#SA). These findings are in broad agreement with previously published data linking

deformation-induced injury with deformation magnitude and animal studies

demonstrating significant ATI cell injury following inflation of the lungs beyond total

lung capacity.

Subsequent studies demonstrated that different alveolar epithelial cell phenotypes within

the same culture system had different sensitivities to deformation-induced injury, with

spreading RTI40-expressing cells in regions of ‘repairing’ epithelial cell monolayers

susceptible to injury even at physiological levels of mechanical strain. This is a novel

observation.

Although there are limitations inherent in the extrapolation of any in vitro system, there

are parallels between the cell phenotypes observed in the cell culture system used for

these experiments and those present in the repairing epithelium, with a population of

ATII cells losing their mature ATII cell phenotype whilst spreading and flattening to

cover denuded basement membrane and acquiring an ATI (RTI40-expressing) cell

phenotype. Viewed in this light, these data are consistent with the hypothesis that

repairing epithelium is susceptible to deformation-induced injury even at physiological

levels of strain.

122

7.3 The multiple-hit hypothesis

Based upon clinical observation, it has been speculated that initial injury to the lung

‘primes’ the epithelium for an excessive or abnormal response following exposure to a

second insult (hit). This has been termed the ‘two-’ or ‘multiple-hit’ hypothesis (Moore

and Moore 1995; Vuichard et al. 2005). The data presented in this thesis support this

hypothesis by demonstrating that the clinically relevant ‘hits’ of mechanical deformation

and hyperoxia are potentially injurious to the alveolar epithelium and inhibit the

processes underlying normal alveolar epithelial repair.

123

8. Future work

The mechanism underlying the hyperoxia-induced inhibition of ATII cell

transdifferentiation observed in these studies remains unknown and warrants further

investigation. A recent study has suggested that transdifferentiation of ATII cells into

ATI cells is modulated by signalling through the Smad-dependent TGF-&1 pathway

(Bhaskaran et al. 2007). This raises the possibility that hyperoxia inhibits

transdifferentiation through inhibition of TGF-&1 signalling and suggests one avenue for

further work.

Several studies have demonstrated that upregulation of the insulin growth factor (IGF)

system occurs during hyperoxia-induced lung injury, and transcriptional regulation of

this system appears to play a role in the arrest of ATII cell proliferation induced by

hyperoxia. Interestingly, a recent study has demonstrated that addition of IGF-1 receptor

antibodies to ATII cells in culture also inhibits the ATII to ATI transition (Narasaraju et

al. 2006), and it seems reasonable to speculate that changes in the expression profile of

the IGF system may have a central role in the mechanism of hyperoxia-induced

inhibition of ATII cell transdifferentiation.

Hyperoxia-induced injury is thought to be mediated by a combination of ROS

cytotoxicity and activation of the redox-sensitive transcription factor NF-'B (D'Angio

and Finkelstein 2000; Haddad 2002; Haddad 2002). Studies in which hyperoxia-exposed

cells were incubated with the free radical scavenger N-acetylcysteine did not appear to

attenuate the effect of hyperoxia on ATII cell differentiation. Similarly, NF-'B

inhibition by sulfasalazine and the inhibitory peptide SN-50 appeared to have little effect

on the inhibition of ATII cell differentiation. However, these experiments were

preliminary and warrant further investigation.

Oxidative stress secondary to stretch-induced ROS generation has been implicated as a

potential mechanism underlying deformation-induced epithelial cell injury (Chapman et

al. 2005). The role of ROS generation in the deformation-induced injury observed in

these studies was not investigated but warrants further study. Intriguingly, it appears that

124

ROS generation by alveolar epithelial cells may play a central role in determining

sensitivity to both hyperoxia- and mechanical deformation-induced injury.

Integrin and extracellular matrix interactions have been shown to play an important role

in epithelial cell migration and spreading and in the repair of distal airway epithelium

(White et al. 1999). In vitro, laminin-rich culture surfaces favour retention of an ATII-

like cell phenotype whilst fibronectin-rich surfaces accelerate loss of the ATII phenotype

(Rannels and Rannels 1989). Although the precise role of integrin/matrix interactions in

the transdifferentiation of ATII cells is not known, these data suggest that

integrin/matrix interactions play an important role in the process of alveolar epithelial

cell repair. The effect of hyperoxia on these interactions is not known.

The data presented in this thesis suggest that functional segregation of the ATII cell

population into E-cadherin-positive and-negative populations may be possible prior to

hyperoxic-exposure. A similar approach has been used to segregate the population of

ATII cells isolated from hyperoxia-exposed rats (Reddy et al. 2004). This approach

would allow further investigation of the hyperoxia-resistant epithelial cell phenotype.

The ‘multiple-hit’ hypothesis postulates that initial injury to the lung primes the

epithelium for an excessive or abnormal response following exposure to a second insult

(hit) (Moore and Moore 1995; Vuichard et al. 2005). Both hyperoxia and mechanical

deformation are clinically relevant second ‘hits’ but the clinical reality is that the

epithelium is exposed to both with the use of supplemental oxygen and positive pressure

ventilation. Whether these hits act synergistically in the context of lung injury remains

unknown, and an obvious extension to the work presented in this thesis is investigation

of the effects of mechanical deformation in combination with hyperoxia (± hypoxia) in

the injured and repairing lung.

125

Figures

126

Figure 1 Schematic representation of alveolar epithelial repair. (A) The traditional model

of alveolar epithelial repair postulates that damaged ATI cells are replaced by the

proliferation of ATII cells and the differentiation of a subpopulation of daughter cells into

terminally differentiated ATI cells. In this model ATII cells are the progenitor cell for both

ATI and ATII cells. More recent studies have refined this to the current model shown in

(B). In this model ATII cells are the usual progenitor of both ATI and ATII cells. ATII cell

division gives rise to two relatively undifferentiated daughter cells that have the capacity

to assume either a mature ATI or ATII phenotype. This may occur through cells having

an intermediate phenotype. Transformation of ATII cells into ATI cells is potentially

reversible, but only ATII cells appear able to re-enter the cell cycle. It remains unclear

whether ATI cells are capable of replacing damaged ATII cells, but after severe

epithelial injury cells from outside the alveolus may contribute to alveolar epithelial

repair. (Adapted from Uhal 1997 & Clegg thesis)

A

B

A

B

Figure 2 Primary rat ATII cell isolation. (A) Modified Papanicolaou (Pap) stain of

cytospun final cell suspension from a typical ATII cell isolation. ATII cells (arrowheads)

were identified as cells containing ! 3 characteristic purple-staining inclusion bodies

(lamellar bodies). Lymphocytes (arrows) appear to be the main contaminant in this

isolation. Differential cell counts were performed at high magnification (100x oil

immersion objective) with ATII cells, macrophages, neutrophils and lymphocytes

(arrows) identified by their characteristic cell morphology. A minimum of 500 cells was

counted per isolation and differential cell counts expressed as a percentage of totals

(B). Data shown from 12 isolations.

127

10µm

A Freshly isolated cells

B Cells after 3 days in culture

Figure 3 The immunophenotype of alveolar epithelial cells in culture. Rat primary

alveolar epithelial cells in culture stained with anti-RTII70 (blue), MMC4 (red) and anti-

RTI40 (green). Day 3 cells are also stained with the nuclear stain TO-PRO-3. (A)

Freshly isolated ATII cells coexpressed the ATII-selective markers RTII70 and MMC4

(appearing pink-purple in the merged image) but did not express the ATI-selective

marker RTI40 (n=6). (B) After 3 days in cell culture under normoxic conditions,

epithelial cells had flattened and spread to form a confluent monolayer of attenuated

cells (~1!m thick) which were difficult to visualise by transmitted light microscopy.

These cells expressed the ATI-selective marker RTI40 but not the ATII-selective

markers RTII70 and MMC4 (n=6). These data are in keeping with the transition of

alveolar epithelial cells from an ATII-like phenotype to an ATI-like phenotype with time

in culture.

Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9).

128

20µm

20µm

MMC4

RTI40

RTII70

Merged

RTI40 Transmitted

MMC4 RTII70 & TO-PRO-3

Merged

Transmitted

A

B

Figure 4 (A) Rat primary ATII cells after 1 day in culture stained with the nuclear stain

TO-PRO-3 (blue), MMC4 (red) and anti-RTI40 (green). The majority of cells had begun

to flatten and spread with loss of MMC4 expression. Some cells had begun to express

RTI40 (arrows), with RTI40 expression appearing to be localised to the spreading celledge (broad arrow). A small population of cells (5.9 ± 1.4%) stained strongly for both

the ATII-selective MMC4 and the ATI-selective RTI40 (arrowhead) suggesting an

intermediate phenotype in the transition from ATII- to ATI-like cells (n=3).

(B) 3-dimensional imaging of the MMC4/RTI40 co-expressing cell (arrowhead in A),

demonstrating localisation of RTI40 to the spreading basal surface of the cell. Images

of RTI40/MMC4 coexpressing cells acquired in conjunction with Clegg (Clegg, Tyrell et

al. 2005).

Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9).

129

MMC4

RTI40

Transmitted

TO-PRO-3

Merged

xy

Merged

xy

z

TransmittedMerged

25µm 25µm

xy xy

xy xy

Figure 5 Immunofluorescent image of a frozen lung tissue section stained with TO-

PRO-3 (blue), MMC6 (red) and anti-RTI40 (green). ATI cells coexpressed the ATI cell-

selective marker RTI40 and the MMC6 antigen, with areas of coexpression appearing

yellow. ATII cells, identifiable from their location and morphology in the transmitted

image, stained with the nuclear stain TO-PRO-3 but did not stain with either anti-RTI40

or MMC6 (arrows). Sectioning artefact marked (*). Secondary-only and isotype-specific

negative controls were performed as described in materials and methods (section

2.9). (n=3).

130

Merged

MMC6

RTI40 TO-PRO-3

Transmitted

20µm

*

Figure 6 Immunofluorescent image of frozen lung section stained with MMC4 (red),

anti-RTI40 (green) and MMC6 (blue) demonstrating alveolar airspaces (Aa) lined with

ATI cells expressing the ATI-selective RTI40 (arrowhead) separated by ATII cells

expressing the ATII cell selective MMC4 (arrow). MMC6 was coexpressed with RTI40

(arrowhead) but not with MMC4 suggesting ATI cell-selectivity. The MMC6 antigen was

not detected in bronchiolar (Br) epithelium and was not expressed by MMC4-expressing

Clara cells (broad arrow). Secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9). (n=3).

131

Merged

RTI40

TransmittedMMC6

20µm

MMC4

Br

Aa

Figure 7 3D reconstruction of deconvoluted images taken by incremental z-plane

scanning of frozen lung tissue section demonstrating localisation of the MMC6

antigen. (A) Stained with MMC6 (red), anti-RTI40 (green) and the nuclear stain TO-

PRO-3 (blue). MMC6 was coexpressed with the ATI-selective apical membrane

protein RTI40, with areas of coexpression appearing yellow in the merged images.

ATII cells (arrow) did not express either MMC6 or RTI40. (B) Stained with MMC6

(red) and anti-RTII70 (blue). The MMC6 antigen was not coexpressed with the ATII-

selective protein RTII70. These data are consistent with expression of MMC6 by ATI

cells but not by ATII cells in the adult rat lung. Secondary-only and isotype-specific

negative controls were performed as described in materials and methods (section

2.9).

132

Merged

MMC6

RTI40 MMC6

RTI40 MMC6

Merged

A

B

Merged

RTII70

Figure 8 Immunofluorescent image of the choroid plexus in the developing rat at

embryonic day 16 (E16). Parasaggital frozen tissue section stained with MMC6 (red),

anti-RTI40 (green) and the nuclear stain TO-PRO-3. The choroid plexus (and part of the

thoracic spinal canal, not shown) stained positively for RTI40 (arrow) but not for the

MMC6 antigen. Positive controls, secondary-only and isotype-specific negative controls

were performed as described in materials and methods (section 2.9). (n=3).

133

Merged RTI40

TO-PRO-3

Transmitted

MMC6

20µm

Merged RTI40

TO-PRO-3

Transmitted

MMC6

20µm

Figure 9 Immunofluorescent image of foetal lung in the developing rat at embryonic

day 17 (E17). Frozen tissue section stained with MMC6 (red), anti-RTI40 (green) and

the nuclear stain TO-PRO-3. Tubules within the developing lung were lined by epithelial

cells expressing RTI40 (arrow). The MMC6 antigen was not detected in the foetal lung

at E17. Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9). (n=3).

134

Figure 10 Immunofluorescent image of foetal lung in the developing rat at embryonic

day 20 (E20). Frozen lung tissue section stained with MMC6 (red) and anti-RTII70

(blue). Developing alveolar airspaces (Aa) appeared lined by attenuated cells

expressing the MMC6 antigen (arrowhead) separated by ATII-like cells expressing the

ATII cell-selective protein RTII70 (broad arrow). Small areas of the alveolar epithelial

surface coexpressed the MMC6 antigen and RTII70 (arrow, appearing pink in merged

image). Bronchiolar (Br) epithelium did not express either RTII70 or the MMC6 antigen.

Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9). (n=3).

Figure 11 Immunofluorescent image of foetal lung in the developing rat at embryonic

day 20 (E20). Frozen lung tissue section stained with MMC6 (red), anti-RTI40 (green)

and the nuclear stain TO-PRO-3. Developing alveolar airspaces (Aa) appeared lined by

attenuated cells coexpressing the MMC6 antigen and RTI40 (yellow in merged image).

Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9). (n=3).

Merged

RTI40

TO-PRO-3 Transmitted

MMC6

20µm

Merged

RTII70

Transmitted

MMC6

50µm

Aa

Aa

Br

Aa

Figure 12 Immunofluorescent images of primary alveolar epithelial cells. (A)

Freshly isolated and cytospun cells stained with anti-RTI40 (green), MMC6 (red)

and anti-RTII70 (blue). Freshly isolated cells expressed the ATII cell-selective

marker RTII70 but did not express the ATI cell-selective marker RTI40. These

ATII-like cells did not express the MMC6 antigen (n=3). (B) Alveolar epithelial

cells after 3 days in culture stained with anti-RTI40 (green), MMC6 (red) and the

nuclear stain TO-PRO-3 (blue). After 3 days in culture epithelial cells had

flattened and spread to form a confluent monolayer of cells expressing the ATI-

selective marker RTI40. These ATI-like cells coexpressed the MMC6 antigen.

Positive controls, secondary-only and isotype-specific negative controls were

performed as described in materials and methods (section 2.9). (n=3).

135

20µmRTII70

Merged Transmitted

MMC6

Merged

RTI40

TO-PRO-3 Transmitted

MMC6

20µm

A Freshly isolated cells

B Cells after 3 days in culture

RTI40

Figure 13 3D reconstruction of deconvoluted images taken by incremental z-plane

scanning of alveolar epithelial cells after 3 days in culture. Cells stained with MMC6

(red), anti-RTI40 (green) and the nuclear stain TO-PRO-3 (blue). After 3 days in culture

alveolar epithelial cells were morphologically and phenotypically ATI-like, being

attenuated and expressing the ATI cell-selective protein RTI40. These images

demonstrate that the MMC6 antigen was coexpressed with RTI40 on the apical

membrane of these ATI-like cells, with areas of coexpression appearing yellow in the

merged images (left hand column).

136

Merged RTI40MMC6

A Normoxia B Hyperoxia

Figure 14 The effect of hyperoxia on adherent cell viability. Epifluorescent images

demonstrating the viability of day 3 adherent cells cultured under (A) normoxic and (B)

hyperoxic conditions. Viable cells stained with calcein AM appear green, dead cells

stained with ethidium homodimer-1 appear red. (C) Demonstrates the viability of

adherent cells cultured under hyperoxic conditions for 3 days then switched to normoxic

conditions for a further 2 days. There was no statistically significant difference in

adherent cell viability between conditions (98.0 ± 1.0% normoxia vs 93.4 ± 0.6%

hyperoxia vs 96.3 ± 3.1% d3 switched, p>0.05, n=3) (D).

137

C d3 Switched

D

50µm

A Normoxia

B Hyperoxia

Figure 15 The effect of hyperoxia on cell spreading. Phalloidin staining (green) of the

actin cytoskeleton of alveolar epithelial cells after 3 days in culture. Nuclei counter-

stained with TO-PRO-3 (blue). (A) Cells cultured under normoxic conditions flattened

and spread to form a confluent monolayer. (B) Cells cultured under hyperoxic

conditions were less spread, remaining as small non-confluent clusters. (C) Alveolar

epithelial cells cultured under normoxic conditions for 3 days had a mean surface areaof 1801 ± 463µm2, whilst cells cultured under hyperoxic conditions had a mean surface

area of 583 ± 245µm2 (p<0.0001, n=3).

138

C

Transmitted

Transmitted

20µm

Figure 16 The effect of hyperoxia on the expression of ATI cell-selective proteins.

Immunofluorescent images of alveolar epithelial cells stained for the ATI-selective

markers RTI40 (top row, n=5), aquaporin 5 (middle row, n=3) and MMC6 (bottom row,

n=5). Nuclei are counter-stained with TO-PRO-3 (blue). Day 3 cells cultured under

normoxic conditions are shown in the left hand column, cells cultured under hyperoxic

conditions in the middle column. Cells cultured under hyperoxic conditions for 3 days

then switched to normoxic conditions for a further 2 days (d3 switched) are shown in the

right hand column. Positive controls, secondary-only and isotype-specific negative

controls were performed as described in materials and methods (section 2.9).

Cells cultured under normoxic conditions expressed the ATI cell markers RTI40,

aquaporin 5 and MMC6 in keeping with the acquisition of an ATI-like phenotype with

time in culture. Hyperoxia inhibited the expression of each of these ATI cell markers.

Cells cultured under hyperoxic conditions for 3 days then switched to normoxic

conditions for a further 2 days did express RTI40, aquaporin 5 and MMC6, suggesting

that the inhibition of the expression of these proteins by hyperoxia is, at least partially,

reversible.

139

Normoxia Hyperoxia d3 switched

RTI40

Aqp 5

MMC6

20µm

A RTI40

Figure 17 Semi-quantitative determination of the effect of hyperoxia on the expression

of the ATI-selective proteins RTI40 and MMC6 using ELISA-based dot blot assays. (A)

The concentration of RTI40 present in cells cultured under normoxic conditions for three

days was significantly greater than in cells cultured under hyperoxic conditions (13.0 ±

1.4 vs 0.25 ± 0.2 du/mg protein, p<0.0001). Cells cultured under hyperoxic conditions

for 3 days then switched to normoxic conditions for a further 2 days (d3 switched) went

on to express significantly greater levels of RTI40 compared to those cultured under

hyperoxic conditions for 3 days (4.4 ± 2.1 du/mg protein , p<0.05). (n=5).

(B) The concentration of MMC6 was 11.4 ± 1.7 du/mg protein in cells cultured under

normoxic conditions compared to 1.5 ± 1.1 rdu/mg protein for cells cultured under

hyperoxic conditions, p<0.05. d3 Switched cells went on to express significantly greater

levels MMC6 (6.2 ± 2.0 du/mg protein) compared to those cultured under hyperoxic

conditions, p<0.05. (n=3).

140

B MMC6

Figure 19 Semi-quantitative determination of the effect of hyperoxia on the expression

of the ATII-selective protein MMC4 using an ELISA-based dot blot assay. The

concentration of MMC4 present in cells cultured under normoxic conditions for three

days was significantly less than in cells cultured under hyperoxic conditions (55 ± 3 vs

187 ± 56 du/mg protein, p<0.01). Cells cultured under hyperoxic conditions for 3 days

then switched to normoxic conditions for a further 2 days expressed significantly lower

levels of MMC4 (45 ± 4 du/mg protein) compared to those cultured under hyperoxic

conditions for 3 days, p<0.01. (n=3).

141

Normoxia

Hyperoxia

MMC4 Pro-SP-C RTII70

Figure 18 The effect of hyperoxia on ATII cell-selective protein expression.

Immunofluorescent images of alveolar epithelial cells stained for the ATII-selective

markers MMC4 (left column, n=3), pro-SP-C (middle column, n=3) and RTII70 (right

column, n=3). Nuclei are counter-stained with TO-PRO-3 (blue) in the images stained

for MMC4 and pro-SP-C but not those for RTII70. Day 3 cells cultured under normoxic

conditions are shown in the top row, cells cultured under hyperoxic conditions in the

bottom row. Positive controls, secondary-only and isotype-specific negative controls

were performed as described in materials and methods (section 2.9).

Cells cultured under normoxic conditions lost expression of the ATII cell markers

MMC4, pro-SP-C and RTII70 in keeping with the loss of an ATII-like phenotype with

time in culture. Cells cultured under hyperoxic conditions for 3 days appeared to retain

low level expression of MMC4 and pro-SP-C but not RTII70.

20µm

A Normoxia

Figure 20 E-cadherin expression in normoxia- (A) and hyperoxia-exposed (B)

primary alveolar epithelial cells after 3 days in culture. Cells stained with anti-E-

cadherin (green) and the nuclear stain TO-PRO-3. E-cadherin expression

appeared reproducibly heterogeneous in cultured cells, with strong expression at

certain cell-cell contact points (arrowheads) and weak expression at others

(broad arrows). There appeared to be less E-cadherin expression with fewer

strongly E-cadherin positive cells in cells cultured under hyperoxic conditions (B)

compared to those cultured under normoxic conditions (A). Positive controls,

secondary-only and isotype-specific negative controls were performed as

described in materials and methods (section 2.9). (n=3).

142

B Hyperoxia

Transmitted

Transmitted

20µm

143

A Steady state ROS production

B ROS generation in response to oxidative stress

Figure 21 The effect of hyperoxia on reactive oxygen species (ROS) generation. ROS

production estimated by intracellular oxidation of the probe CMH2DCFDA. (A) The

steady state production of ROS was significantly greater in cells cultured under

normoxic conditions compared to those cultured under hyperoxic conditions (16 ± 5 vs

8 ± 3 fu/mg protein/min, p < 0.05, n=3). CMH2DCFDA was omitted from otherwise

identically treated control wells, and fluorescence in these controls subtracted from

measurements obtained as described in materials and methods (section 2.6).

(B) ROS production following exposure to standardised oxidative stress. Controlled

exposure to H2O2 was produced by the addition of glucose oxidase to cell culture

wells, catalysing the conversion of glucose present in HBSS to D-glucono-1,5-lactone

and H2O2. Cells cultured under normoxic conditions demonstrated significantly greater

ROS production in response to oxidative stress compared to those cultured under

hyperoxic conditions (72 ± 8 vs 26 ± 4 fu/mg protein/min, p < 0.001, n=3). Control wells

contained HBSS and CMH2DCFDA without glucose oxidase, and fluorescence

detected in these controls was subtracted from measurements obtained as decsribed

in materials and methods (section 2.7).

A

B

Figure 22 The effect of hyperoxia on alveolar epithelial cell proliferation. (A)

Immunofluorescent images of day 3 alveolar epithelial cells cultured under normoxic

conditions, incubated with BrdU for 6 h then stained with anti-BrdU (green), anti-RTI40

(red) and the nuclear stain TO-PRO-3 (blue). Merged image demonstrates a proportion

of ATI-like cells (RTI40-positive) with BrdU-positive nuclei (TO-PRO-3-positive

organelles) indicative of cell proliferation. Positive controls, secondary-only and isotype-

specific negative controls were performed as described in materials and methods

(section 2.9).

(B) Quantification of cell proliferation rates by determination of the percentage of nucleiincorporating BrdU. A small proportion (3.5 ± 1%) of cells cultured under normoxic

conditions for 3 days incorporated BrdU. No BrdU-positive nuclei were observed in cells

cultured under hyperoxic conditions suggesting hyperoxia inhibited cell proliferation. In

cells cultured under hyperoxic conditions for 3 days then switched to normoxic

conditions (d3 switched) there was a statistically significant increase in the number ofBrdU-incorporting nuclei (13.1 ± 1.9%, p<0.0001), suggesting that the switch from

hyperoxia to normoxia was followed by a proliferative response. (n=3).

144

Merged

20µm

RTI40

TO-PRO-3

BrdU

Transmitted

Figure 23 Hyperoxia-induced cell cytotoxicity assessed by determination of LDH

release. The percentage of cell death was calculated by determining the LDH activity in

the supernatant of each well as a percentage of the total LDH activity (supernatant +

lysate) as described in materials and methods (section 2.4). There was no significant

difference in the level of cell death determined after 6 h (64.5 ± 2.1% normoxia vs 64.3

± 10.7% hyperoxia, p>0.05, n=3) and 24 h in culture (76.0 ± 4.4% normoxia vs 73.3 ±

17.2% hyperoxia, p>0.05, n=3), but after 3 days, there was significantly greater LDH

release in cells cultured under hyperoxic conditions compared to those cultured under

normoxic conditions (23.3 ± 8.3% normoxia vs 72.0 ± 2.6% hyperoxia, p<0.05, n=3).

Significantly greater LDH release was also observed under hyperoxic conditions after 5

days in culture (16.0 ± 6.1% normoxia vs 70.5 ± 3.5% hyperoxia, p<0.05, n=3).

145

146

A Normoxia B Hyperoxia

Figure 24 Hyperoxia-induced apoptosis. Phase contrast images demonstrating

apoptosis (arrowheads) in day 3 Fuelgen stained alveolar epithelial cells cultured under

(A) normoxic and (B) hyperoxic conditions. There was no statistically significant

increase in the level of apoptosis determined by Fuelgen staining with time in culture

under normoxic conditions, but the rate of apoptosis did increase with time in culture for

cells cultured under hyperoxic conditions. The difference in rate of apoptosis between

cells cultured under normoxic and hyperoxic conditions was not statistically significant

after 6 h (1.7 ± 0.6% normoxia vs 2.7 ± 0.6% hyperoxia, p>0.05, n=3) and 24 h in

culture (3.0 ± 1.7% normoxia vs 5.7 ± 2.1% hyperoxia, p>0.05, n=3), but after 3 days,

there was significantly greater apoptosis in cells cultured under hyperoxic conditions

(1.7 ± 0.6% normoxia vs 6.3 ± 2.1% hyperoxia, p<0.05, n=3). Significantly greater rates

of apoptosis were also observed after 5 d in culture (3.0 ± 1.0% normoxia vs 7.7 ±

1.5% hyperoxia, p<0.05, n=3) (C).

C

25µm

Figure 25 Experimental protocol for rat model of Staphylococcus aureus pneumonia.

Detailed protocol described in materials & methods (section 2.10).

147

Insertion of tracheal cannula and

intrapulmonary instillation of Staphylococcusaureus or PBS (controls)

Housing under standard conditions (air)

Induction ofanaesthesia

Bacterial instillation

72hr

48hr

Recovery

Randomisation

Intraperitoneal injection fentanyl, fluanisone &midazolam

Housing under normoxic (air) orhyperoxic (90-95% O2)

conditions

Euthanasia, bronchoalveolarlavage & specimen collection

Normoxia Hyperoxia

End experiment End experiment

A B

Figure 27 Bronchoalveolar lavage fluid (BALF) cell counts in PBS-instilled (control,

n=5) and S. aureus-instilled lungs 5 days post-instillation. (A) BALF total cell counts.

There was an increase in the total number of cells in BALF obtained from the lungs of

S. aureus-instilled animals recovered in normoxic conditions (SA+normoxia, n=4) butthis was not statistically significant compared to controls (14 ± 7 x104 control vs 314 ±

169 x104 SA+normoxia, p>0.05). The increase in total cell count from the lungs of

animals recovered in normoxic conditions for 3 days then switched to hyperoxic

conditions for 2 days (SA+hyperoxia, n=5) was significant in comparison to controls(530 ± 303 x104, p<0.05). (B) Differential cell count of BALF. The proportion of

neutrophils recovered was significantly increased in both SA+normoxia andSA+hyperoxia groups (0.3 ± 0.6% control vs 8.0 ± 2.8% SA+normoxia vs 9.3 ± 7.9%

SA+hyperoxia, p<0.05) consistent with an acute inflammatory injury. There was no

statistically significant difference in either total cell count or the proportion of

neutrophils recovered between SA+normoxia and SA+hyperoxia. Data analysed using

ANOVA.

148

Figure 26 Bronchoalveolar lavage fluid (BALF) protein content in PBS-instilled

(control, n=5) and S. aureus-instilled lungs 5 days post-instillation. The total protein

content of BALF increased in the lungs of S. aureus-instilled animals recovered in

normoxic conditions (SA+normoxia, n=4) but this was not statistically significantcompared to controls (1.1 ± 0.2mg control vs 2.8 ± 1.0mg, p>0.05). The increase in

protein content of BALF from the lungs of animals recovered in normoxic conditions for

3 days then switched to hyperoxic conditions for 2 days (SA+hyperoxia, n=5) wassignificant in comparison to controls (5.9 ± 2.4mg, p<0.01). The difference in BALF

protein content between the SA+normoxia and SA+hyperoxia groups was not

statistically significant. Data analysed using ANOVA.

Figure 29 Lung RTI40 concentration in control (PBS-instilled) and S. aureus-instilled

lungs 5 days post-instillation determined by ELISA-dased dot blot assay of lung

homogenate. Samples were obtained from the left upper lobe (controls) and

macroscopically inflamed regions (S. aureus-instilled lungs) and homogenised as

detailed in materials and methods (section 2.10.7). The concentration of RTI40 showed

a two-fold decrease in the lungs of animals recovered in normoxia for 3 days then

switched to hyperoxia (SA+hyperoxia) compared to those exposed to normoxia alone

(SA+normoxia) (41.4 ± 19.8 vs 18.8 ± 10.4 du/mg protein, p<0.05, n=5). Both lungs

exposed to hyperoxia, and those exposed to normoxia alone, had significantly lower

concentrations of RTI40 than control lung (330.3 ± 88.0 du/mg protein, p<0.001, n=5).

149

Figure 28 BALF RTI40 content in PBS-instilled (control, n=5) and S. aureus-instilled

lungs 5 days post-instillation determined by ELISA-dased dot blot assay. The BALF

content of RTI40 (a marker of ATI cell necrosis) was not statistically significant between

groups: 9.8 ± 3.0 du controls vs 7.8 ± 3.6 du in animals exposed to normoxia alone

(SA+normoxia, n=4) vs 5.6 ± 4.5 du in animals recovered in normoxia for 3 days then

switched to hyperoxia (SA+hyperoxia, n=5). (p=0.4).

Figure 30 Illustration of the quantification of alveolar epithelial surface staining in a

control lung section stained with anti-RTI40 (green) and MMC4 (red) (A). For detailed

explanation see materials and methods (section 2.9.6). Images for comparison (4x4tiles at 63x magnification ! 4096x4096 pixels) were acquired using confocal microscopy

with identical settings and were not digitally enhanced prior to analysis. Positive staining

with a fluorochrome (MMC4 (red) in this example) was determined using the Density

Slice… function in Openlab 3.5.2, with the lower threshold of positive staining

determined by polling the pixels of a positive area of staining using the HSI Colorspy

palette. This generated a binary map layer (D) showing regions of positive staining. A

white binary layer map of MMC4-positive staining is shown overlaid on the original

image in (B). Binary layers from different fluorochromes could be combined to show

regions of coexpression. After image calibration, the length of positively staining

membrane on the binary map was traced using a graphics tablet and the

Measurement… functions in Openlab 3.5.2. The length of positively stained membrane

was then expressed as a fraction of the entire epithelial surface (C). Illustration adapted

from Clegg thesis.

150

A

B

Figure 31 Expression of ATI and ATII cell-selective markers in control lungs. Frozenlung sections (~3µm thick) were stained with the ATI cell-selective marker anti-RTI40

(green) and the ATII cell-selective markers anti-RTII70 (blue) and MMC4 (red) (n=3). (A)

2x2 tiled image at 63x magnification demonstrating alveolar airspaces (Aa) lined by

RTI40 expressing ATI cells (arrowheads). ATII cells coexpressed RTII70 and MMC4

(arrows, appearing pink-purple in merged image) and were typically located in the

corners of the alveoli. Terminal bronchioles (Br) were lined with Clara cells expressing

MMC4 but not RTI40 or RTII70. (B) High magnification view demonstrating RTII70 and

MMC4 coexpression by ATII cells located between RTI40 expressing ATI cells. Themean length of RTII70/MMC4 coexpressing membrane in control lungs was 7.6 ± 3.6µm.

Secondary-only and isotype-specific negative controls were performed as described in

materials and methods (section 2.9).

151

MMC4 RTI40

25µm

Merged

Merged

Transmitted

TransmittedRTII70

10µm

Br Aa

Aa

Aa

Figure 32 High magnification view of 4x4 tiled image (shown in figure 33 at lower

magnification) showing lung tissue section from normoxia-exposed S. aureus-injured

lung 5 days post-instillation. Section stained with anti-RTI40 (green), anti-RTII70 (blue)

and MMC4 (red). The proportion of alveolar epithelial surface staining positively for the

ATI-selective protein RTI40 (broad arrow) was significantly reduced in comparison to

controls. There was an increase in the proportion of the epithelial surface coexpressing

the ATII cell-selective proteins RTII70 and MMC4 (arrows), and an increase in the

proportion of epithelial surface with an intermediate phenotype expressing a combination

of the ATI-selective RTI40 and the ATII-selective MMC4 (arrowheads). Note alveolar

macrophages expressing MMC4 in the alveolar airspaces (dashed arrow). Positive

controls, secondary-only and isotype-specific negative controls were performed as

described in materials and methods (section 2.9). (n=3).

152

100µm

A

B

Figure 33 Frozen lung tissue section of S. aureus-injured lung 5 days post-instillation

with the rat recovered in normoxic conditions. Section stained with anti-RTI40 (green),

anti-RTII70 (blue) and MMC4 (red). (A) 4x4 tiled image at 63x magnification used for

quantification of alveolar epithelial surface staining (shown at higher magnification in

figure 32). The proportion of alveolar epithelial surface staining positively for the ATI-

selective protein RTI40 (broad arrow) was significantly reduced in comparison to

controls. There was an increase in the proportion of the epithelial surface coexpressing

the ATII cell-selective proteins RTII70 and MMC4 (arrows), and an increase in the

proportion of epithelial surface with an intermediate phenotype expressing a

combination of the ATI-selective RTI40 and the ATII-selective MMC4 (arrowheads). (B)

Inset magnified, demonstrating an area of epithelial surface coexpressing RTI40 (green)

and MMC4 (red), with the combination appearing yellow in the merged image. Positive

controls, secondary-only and isotype-specific negative controls were performed as

described in materials and methods (section 2.9). (n=3).

153

100µm

RTII70

Transmitted

Transmitted

MMC4

RTI40

Merged

Merged

Figure 34 High magnification view of 4x4 tiled image (shown in figure 35 at lower

magnification) showing lung tissue section from hyperoxia-exposed S. aureus-injured

lung 5 days post-instillation. Section stained with anti-RTI40 (green), anti-RTII70 (blue)

and MMC4 (red). The proportion of alveolar epithelial surface staining positively for the

ATI-selective protein RTI40 (broad arrow) was significantly reduced in comparison to

both controls and to S. aureus-induced pneumonia recovered in normoxic conditions.

There was a marked increase in the proportion of the epithelial surface coexpressing

the ATII cell-selective proteins RTII70 and MMC4 (arrows) in keeping with significant

ATII cell hyperplasia. There was a non-significant increase in the proportion of epithelial

surface expressing a combination of the ATI-selective RTI40 and the ATII-selective

MMC4 (arrowheads) in comparison to controls. Note alveolar macrophages expressing

MMC4 (dashed arrow). Positive controls, secondary-only and isotype-specific negative

controls were performed as described in materials and methods (section 2.9). (n=3).

154

100µm

A

B

Figure 35 Frozen lung tissue section of S. aureus-injured lung 5 days post-instillation

with the rat recovered in normoxic conditions for 3 days then switched to hyperoxic

conditions for a further 2 days. Section stained with anti-RTI40 (green), anti-RTII70

(blue) and MMC4 (red). (A) 4x4 tiled image at 63x magnification used for quantification

of alveolar epithelial surface staining (shown in figure 34 at higher magnification). The

proportion of alveolar epithelial surface staining positively for the ATI-selective protein

RTI40 (broad arrow) was significantly reduced in comparison to both controls and to S.

aureus-induced pneumonia recovered in normoxic conditions. There was a marked

increase in the proportion of the epithelial surface coexpressing the ATII cell-selective

proteins RTII70 and MMC4 (arrows), suggestive of ATII cell hyperplasia. There was a

non-significant increase in the proportion of epithelial surface expressing a

combination of the ATI-selective RTI40 and the ATII-selective MMC4 (arrowheads) in

comparison to controls. (B) Inset magnified, demonstrating a significant increase in the

area of epithelial surface coexpressing RTII70 (blue) and MMC4 (red), with the

combination appearing purple-pink in the merged image. Positive controls, secondary-

only and isotype-specific negative controls were performed as described in materials

and methods (section 2.9). (n=3).

155

RTII70 Transmitted

MMC4 RTI40

Transmitted

100µm

Merged

Merged

A ATI cell-staining membrane

B ATII cell-staining membrane

Figure 36 ATI and ATII cell-staining membrane. The percentage of alveolar epithelial

surface expressing the ATI cell-selective marker RTI40 and the ATII cell-selective

markers RTII70 and MMC4 in control (PBS-instilled, n=3) and S. aureus-injured lungs 5

days post-instillation was determined as described in materials & methods (section

2.9.6). Of the animals instilled with S. aureus, one group was recovered under

normoxic conditions (SA+normoxia, n=3), the other was recovered under normoxic

conditions for 3 days then switched to hyperoxic conditions (95% O2) for a further 2

days (SA+hyperoxia, n=3). (A) RTI40 expressing (ATI) membrane. In control lungs, 93.6± 1.6% of the apical alveolar epithelial surface stained positively for RTI40. This

decreased significantly in S. aureus-instilled lungs, falling to 84.4 ± 2.6%

(SA+normoxia) and 73.4 ± 5.3% (SA+hyperoxia). The difference in the percentage of

RTI40-staining membrane between S. aureus-instilled lungs exposed to normoxia alone

and those exposed to normoxia followed by hyperoxia was statistically significant

(p<0.05, ANOVA). (B) RTII70 & MMC4 coexpressing (ATII) membrane. In control lungs,2.4 ± 0.4% of the apical alveolar epithelial surface stained positively for both RTII70 &

MMC4. This increased significantly in S. aureus-instilled lungs to 7.7 ± 1.1%

(SA+normoxia) and 15.5 ± 1.2% (SA+hyperoxia), suggesting ATII cell hyperplasia. The

difference in the percentage of RTII70/MMC4-staining membrane between S. aureus-

instilled lungs exposed to normoxia alone and those exposed to normoxia followed by

hyperoxia was statistically significant (p<0.001, ANOVA). These data are consistent

with the hypothesis that ATII cell hyperplasia has not resolved in S. aureus-injured

lungs exposed to hyperoxia.

156

Figure 37 Frequency histogram demonstrating the frequency of occurrence ofRTII70/MMC4 coexpressing (ATII cell-staining) membrane lengths per 100,000µm of

alveolar epithelial surface. In control lungs there was an almost normal distribution ofATII-staining membrane lengths, with a mode of ~7µm (mean 7.3 ± 6.8µm). This

reflects the normal apical surface length of an ATII cell. In S. aureus-instilled lungs

there was an increase in the proportion of ATII cell-staining membrane present and a

different pattern of distribution of membrane lengths. In animals recovered under

normoxic conditions for 3 days then switched to hyperoxic conditions for 2 days(SA+hyperoxia), the mean length of RTII70/MMC4 coexpressing membrane was 9.6 ±

6.6µm, but the distribution of positively staining membrane was trinomial, with peaks

occuring at ~7, 14 and 22µm. These data suport the concept of ATII cell hyperplasia,

with 2 and (less frequently) 3 ATII cells lying adjacent to one another in the injured lung.

In animals recovered under normoxic conditions (SA+normoxia), the mean length ofRTII70/MMC4 coexpressing membrane was 7.3 ± 6.8µm. There was a trinomial

distribution of positively staining membrane, with peaks occuring at ~8, 15 and 22µm

although this was less dramatic than in the SA+hyperoxia group. The striking feature of

the distribution of positively staining membrane in the SA+normoxia group was thefrequent occurrence of membrane lengths in the region of 1-7µm. This was not a

prominent feature of either the control or the SA+hyperoxia group, and is consistent

with the hypothesis of ATII cells transdifferentiating into ATI cells in the SA+normoxia

group.

157

Distribution of RTII70/MMC4 coexpressing membrane lengths

158

RTI40/MMC4 coexpressing membrane

Figure 38 RTI40 & MMC4 coexpressing (intermediate cell) membrane. The percentage

of alveolar epithelial surface coexpressing the ATI cell-selective marker RTI40 and the

ATII cell-selective marker MMC4 in control (PBS-instilled, n=3) and S. aureus-injured

lungs 5 days post-instillation is shown. Of the animals instilled with S. aureus, one

group was recovered under normoxic conditions (SA+normoxia, n=3), the other was

recovered under normoxic conditions for 3 days then switched to hyperoxic conditions(95% O2) for a further 2 days (SA+hyperoxia, n=3). In control lungs, 0.6 ± 0.1% of the

apical alveolar epithelial surface stained positively for both RTI40 & MMC4. Thisincreased significantly to 11.9 ± 4.2% in S. aureus-instilled lungs exposed to normoxia

alone, suggesting the presence of a significant number of cells with an intermediatephenotype as (hyperplastic) ATII cells differentiate into ATI cells. 3.0 ± 0.8% of the

alveolar epithelial surface coexpressed RTI40 & MMC4 in S. aureus-instilled lungs

exposed to normoxia followed by hyperoxia but this was not significantly different from

controls. The difference in the percentage of RTI40/MMC4-staining membrane between

S. aureus-instilled lungs exposed to normoxia alone and those exposed to normoxia

followed by hyperoxia was statistically significant (p<0.05, ANOVA).

159

Figure 39 (A) Diagram demonstrating the BioFlex® cell culture system. Up to four 6-

well BioFlex® culture plates are positioned on a BioFlex® baseplate such that the

silastic membrane base of each culture well sits directly over a loading post. A side-on

view of this arrangement is shown in (B). Culture plates form an air-tight seal with the

baseplate by seating within rubber gaskets (shown in red). The baseplate is connected

to a computer regulated vacuum pump which, via the vacuum inlets in the baseplate,

generates a controlled negative pressure on the underside of the culture plates. This

vacuum draws the silastic membrane down and across the face of the loading post,

exposing all cells cultured on the surface above the loading post to uniform equibiaxial

(radial and circumferential) strain.

Connection to

computer regulated

vacuum

Baseplate

Loading postVacuum inlet Gasket

BioFlex® 6-well

culture plate

Gasket

Loading post

A

B

Quantifiable vacuum

Quantifiable

equibiaxial strain

Top viewSide view

160

Figure 40 Epifluorescent and phase contrast images demonstrating the effect of 1 h

cyclic mechanical strain on the viability of primary alveolar epithelial cells after 5

days in culture. A & B unstrained controls. C & D strained at 12% change in

basement membrane surface area (!SA), E & F strained at 22% !SA and G & H at

44% !SA. Cell viability was determined 1 h following completion of mechanical

deformation. Live cells stained with calcein AM appear green, dead cells stained

with ethidium homodimer-1 appear red.(n=5).

D

A

E F

C

B

100µm

G H

Control

12% !SA

22% !SA

44% !SA

Figure 41 Deformation-induced injury in day 5 primary alveolar epithelial cells after 1 h

cyclic mechanical strain. Data are means ± SD. There was no significant difference in

cell viability between unstrained controls and cells strained at 12% !SA or 22% !SA (1.4

± 0.7% vs 1.4 ± 0.6% vs 4.3 ± 0.8% cell death, p>0.05), but significant cell death

occured following deformation at 44% (!SA 48.9 ± 12.5%, p<0.001, n=5).

161

% Total Lung Capacity

% C

ha

ng

e e

pith

elia

l b

ase

me

nt

me

mb

ran

e s

urf

ace

are

a

Figure 42 Relationship between lung volume and change in epithelial basement

membrane surface area in isolated rat lungs. Adapted from Tschumperlin and

Margulies 1999. Inflation of the isolated lung to ~60, 80 and 100% of total lung capacity

results in changes of epithelial basement membrane surface area of ~10, 20 and 30-

40% respectively. The 12, 22 and 44% !SA deformations used in the in vitro studies in

this thesis were selected to represent lung inflation to ~60, 80 and >100% respectively

on the basis of these data.

Figure 43 Epifluorescent and phase contrast images demonstrating the effect of 1 h

cyclic mechanical strain on the viability of day 5 primary alveolar epithelial cells at the

periphery of cell culture monolayers. A & B unstrained controls. C & D cells strained at

12% !SA. Cell viability was determined 1 h following completion of mechanical

deformation. Live cells stained with calcein AM appear green, dead cells stained with

ethidium homodimer-1 appear red. (n=5).

Figure 44 Deformation-induced injury in the central & peripheral regions of day 5

primary alveolar epithelial cell monolayers after 1 h 12% !SA cyclic mechanical strain.

Data are means ± SD. There was no significant difference in cell viability between

unstrained controls and strained cells located in the central region of cell monolayers.

However, in the peripheral region of epithelial cell monolayers 12% !SA resulted in

significant cell death compared to unstrained controls (3.3 ± 1.3% vs 49.5. ± 12.2% cell

death, p<0.001, n=5).

162

D

A

C

B

100µm

Central

Peripheral

B

A

Figure 45 Phalloidin staining of the actin cytoskeleton of day 5 primary alveolar

epithelial cell monolayers demonstrating the difference in cell morphology between

densely packed cells in the central region of the monolayer (A) compared to thoseflattened and spread in the peripheral region (B) (SA 490 ± 116µm2 vs 1963 ± 527µm2,

p<0.0001, n=3).

50!m

BA

Figure 46 Day 5 primary alveolar epithelial cell monolayer stained with MMC4 (red),

anti-RTI40 (green), RTII70 (blue) and TO-PRO-3 (blue). Cells in the central region (A) of

the monolayer did not express the ATI-selective marker RTI40 but stained weakly with

the ATII-selective marker MMC4 (arrows). Cells in the peripheral region (B) expressed

RTI40 (arrow heads) but not MMC4. Positive controls, secondary-only and isotype-

specific negative controls were performed as described in materials and methods

(section 2.9). (n=3).

163

100µm

Figure 47 Epifluorescent images demonstrating the effect of mechanical deformation on

cell viability of repairing wounds (wound edges marked by arrow heads) in alveolar

epithelial cell monolayers. Primary alveolar epithelial cells were isolated and cultured for

3 days. The resulting cell monlayers were then wounded and cultured for a further 2

days to simulate alveolar epithelial repair. Repairing monolayers were randomised toeither (A) unstrained control or (B) 1 h cyclic equibiaxial mechanical strain at 12% !SA.

Cell viability was determined 1 h following completion of mechanical deformation. Live

cells stained with calcein AM appear green, dead cells stained with ethidium

homodimer-1 appear red. (n=3).

A B

Figure 48 Deformation-induced injury in repairing alveolar epithelial cell wounds

following exposure to 1 h cyclic mechanical strain at 12% !SA. Significant cell death

occurred in the region of strained repairing alveolar epithelium in comparison to thesame region in unstrained controls (38.0 ± 8% cell death vs 8.3 ± 1.6% unstrained

controls, p < 0.005, n=3).

164

100µm

Figure 49 Experimental protocol for rat model of haemorrhagic shock-induced acute

lung injury. Detailed protocol described in materials & methods (section 2.11).

165

Surgical insertion of:- Carotid cannula for measurement of meanarterial blood pressure, arterial blood gassampling and blood removal- Jugular cannula for drug administration andfluid resuscitation- Tracheostomy for mechanical ventilation

Withdrawal of blood over ~15 mins to reducemean arterial pressure to 30-40mmHg (~30-40% blood volume)

Infusion of warmed intravenous fluid over ~15

mins to raise mean arterial blood pressure to !80 mmHg

Induction ofanaesthesia

Stabilisation (30 min)

Surgery

Haemorrhagic shock

Reperfusion (4 hrs)

End experiment

Haemorrhage

Fluid resuscitation

Intraperitoneal injection midazolam & ketamine

Maintenance of anaesthesia, normothermia and

mean arterial blood pressure ! 80 mmHg

Euthanasia, bronchoalveolar lavage & specimencollection

Spontaneouslybreathing room air

Intermittent positivepressure ventilation

FiO2 1.0; RR 60-80; VT

6 ml/Kg; PEEP 5cmH2O

Mean arterial pressure 30-40 mmHg 30 min

Figure 51 Bronchoalveolar lavage fluid (BALF) protein content in rats completing the

the haemorrhagic shock protocol. There was a significant increase in the level of BALF

protein in animals exposed to haemorrhagic shock (13.2 ± 2.1 mg, n=3) in comparison

to controls (2.7 ± 1.1 mg, n=3, p<0.01) consistent with significant alveolar protein leak.

166

Figure 50 Blood pressure profile of an animal undergoing surgery, haemorrhage and

fluid resuscitation during rat model of haemorrhagic shock-induced acute lung injury.

Data acquired from continuous intraarterial pressure monitoring as described in

materials and methods (section 2.11.2).

Figure 52 Bronchoalveolar lavage fluid (BALF) cell counts in control animals and

animals exposed to haemorrhagic shock (haemorrhage). (A) Total BALF cell counts.There was no statistically significant difference in total cell counts (86.2 ± 13.9 x 104

haemorrhage vs 97.3 ± 19.9 control, n=3, p=0.24). (B) BALF differential cell counts.

There was a significant increase in the proportion of neutrophils in the BALF recoveredfrom animals exposed to haemorrhagic shock compared to controls (12.7 ± 2.1%

haemorrhage vs 0.1 ± 0.1% control, n=3, p<0.001), with a commensurate decrease in

the proportion of alveolar macrophages (82.3 ± 2.9 haemorrhage vs 98.3 ± 0.6 control,

n=3, p<0.001).

167

A

B

Figure 53 Bronchoalveolar lavage fluid (BALF) content of the ATI-selective markers

RTI40 and MMC6 determined by ELISA-based dot blot assay. (A) BALF RTI40 content.

There was a 7-fold increase in the amount of RTI40 in the BALF recovered from animalsexposed to haemorrhagic shock (haemorrhage) compared to controls (108 ± 54 du

haemorrhage vs 15 ± 9 control, n=3, p<0.05). (B) BALF MMC6 content. There was a 2-

fold increase in the amount of MMC6 in the BALF recovered from animals exposed tohaemorrhagic shock compared to controls (6.9 ± 0.8 du haemorrhage vs 2.6 ± 0.8

control, n=3, p<0.05). These data are consistent with significant ATI cell necrosis in

animals exposed to haemorrhagic shock.

168

A : RTI40

B : MMC6

169

Appendix I

(A) Alveolar epithelial type I cell markers in adult lung

Protein Expression in cells other than ATI cells

ATI cell-specific

RTI40 or T1$ None

HTI56 None

ATI cell-selective

Caveolin-1/-2 Bronchial, endothelial, fibroblasts, smooth muscle cells

Na+/K

+-ATPase

Aquaporin 5 Bronchial

Cytochrome P450 2B1 Bronchial

Carboxypeptidase M Alveolar macrohages

ATI cell-selective/-associated

ICAM-1 Macrophages, bronchial, endothelial, ATII?

Connexin 43 ATII

P-glycoprotein Smooth muscle cells, bronchial, ATII?

RAGE Endothelial, ATII?

PAI-1 ATII?

P2X4 ATII?

CDKN2B ATII?

ATB0+

Ciliated cells, ATII?

Cytochrome P450 1A1 Bronchial, ATII

Eotaxin Airway, macrophages, lymphocytes, endothelial

%GT Clara cells, ATII

IGFR-2

Bronchial = bronchial epithelial cells; Airway = airway epithelial cells; Endothelial = endothelial

cells. Adapted from McElroy and Kasper 2004.

170

(B) Alveolar epithelial type II cell markers in adult lung

Protein Expression in cells other than ATII cells

Aminopeptidase N or MMC4 or p146 Clara cells, alveolar macrophages

p172 Clara cells

RTII70

Glycoproteins GP330 & GP600

Ca1 Clara cells, non-ciliated bronchiolar epithelium

Alkaline phosphatase Clara cells

$-Glucosidase

SP-A Clara cells, alveolar macrophages

SP-B Clara cells, alveolar macrophages

Pro-SPC/SP-C

SP-D Clara cells, alveolar macrophages

Pneumocin Clara cells

CCSP/CC10 Clara cells, non-ciliated bronchiolar epithelium

Macular Promifera agglutinin Clara cells

Adapted from Boylan thesis and Dobbs 1990.

171

Appendix II Manufacturers and Suppliers

Amersham Biosciences http://www.amershambiosciences.com

BD Biosciences http://www.bdbeurope.com

BD Medical http://www.bdeurope.com

Bio-Rad Laboratories http://www.bio-rad.com

Billups-Rothenberg http://www.brincubator.com

Bitplane AG http://www.bitplane.com

BOC Gases http://www.boc-gases.com

Dako http://www.dako.com

Dunn Labortechnik GmbH http://www.dunnlab.de

Dynatech Laboratories http://dynatechlaboratories.com

Fisher Scientific http://www.fisher.co.uk

Fluka and Riedel http://www.sigmaaldrich.com

Genusxpress http://www.touchaberdeen.com

Grace Bio Labs http://www.gracebio.com

Greiner Bio-One http://www.greinerbioone.com

Harlan (UK) http://www.arcned.nl

Harvard Apparatus http://www.harvardapparatus.co.uk

Improvision http://www.improvision.com

Invitrogen http://www.invitrogen.com

Portex http://www.smiths-medical.com

Roche http://www.roche-applied-science.com

Millipore http://www.millipore.com

Molecular Devices http://www.moleculardevices.com

Molecular Probes http://www.invitrogen.com

Schleicher & Schuell http://www.schleicher-schuell.com

Sigma-Aldrich http://www.sigmaaldrich.com

Stratech Scientific http://www.stratech.co.uk

172

Thermo Scientific http://www.thermo.com

WelchAllyn http://www.welchallyn.com

Zeiss http://www.zeiss.com/micro

173

Appendix III Buffers and Solutions

ATII cell isolation stock solutions: Made up with DDW and filter sterilised

Name FW Concentration Grams/Mgs Volume

NaCl 58.4 9% (10x stock) 45g 500ml

KCl 74.6 0.15M 5.6g 500ml

CaCl2.2H20 147.0 0.11M 1.62g 100ml

MgSO4.7H20 246.5 0.15M 2.45g 100ml

Sodium phosphate - 0.1M 0.28g mono-, 1.13g dibasic to pH 7.4 200ml

Hepes 260.3 0.2M 4.77g to pH 7.4 100ml

TRIS base 121.1 50mM 6.05g to pH 9.4 1000ml

ATII cell isolation working solutions: Filter sterilised, stored at 4°C and used <48 h

(a) Isolation buffer:

(b) Ca/Mg-free isolation buffer:

Name Quantity of stock

solution

NaCl 40 ml

KCl 16 ml

CaCl2 4 ml

MgSO4 4 ml

PO4 12 ml

Hepes 24 ml

Glucose 464 mg

DDW 360 ml

Name Quantity of stock

solution

NaCl 40 ml

KCl 16 ml

PO4 12 ml

Hepes 24 ml

Glucose 464 mg

EGTA 34.4 mg

DDW 360 ml

174

Immunoglobulin G (IgG) solution & IgG coating of bacteriological plates

5mg reagent grade rat IgG (Sigma) dissolved in 10mls of 50mM tris base. Solution filter

sterilised. 5mls of IgG solution placed in bacteriological plates, agitated for 30 min at

room temperature then plates stored at 4°C prior to use within 24 hours. IgG solution

removed prior to use (solution filter sterilised and reused up to 4x), and plates rinsed 5x

with Ca/Mg free PBS.

ATII culture medium media

Dulbecco’s modified eagle’s medium (DMEM) (Sigma-Aldrich) 450ml

Foetal bovine serum (Gibco) 40ml filter sterilised

Penicillin G (10,000 units/ml) and streptomycin sulphate (10,000 g/ml) (Gibco) 5ml

1% L-glutamine (Gibco) 5ml filter sterilised

4% Paraformaldehyde

4g paraformaldehyde dissolved in 100mls Ca/Mg-free PBS in water bath at 70°C (30

min). Filter sterilised, stored at 4°C & used < 1 month or aliquoted and stored at -20°C.

Tris(hydroxymethyl)aminomethane-buffered saline (TBS)

(a) 20x TBS stock solution: 90g NaCl and 24.2g Tris base dissolved in 450ml DDW.

pH adjusted to 8.2 with HCl and final volume brought up to 500ml with DDW.

Stored at room temperature.

(b) TBS: 25ml 20x TBS to final volume 500ml with DDW

(c) TBS-T: 0.25ml Tween-20 added to 500ml TBS

Homogenisation buffer

1x Complete" protease inhibitor cocktail tablet (Roche) freshly dissolved in 50ml TBS

175

Blocking buffer (BB)

(a) Blocking buffer (BB): 44ml fish gelatin mixed in 100ml Ca/Mg-free PBS. 20ml

goat serum added to 80ml of resulting fish gelatine solution. Aliquoted & stored -20°C.

(b) Blocking buffer with triton (BBT): 10µl 10% tritonX (in Ca/Mg-free PBS) added

to 1ml BB. Used fresh.

30% Sucrose

15g sucrose dissolved in 50mls Ca/Mg-free PBS. Filter sterilised and used fresh.

Bouin’s Fixative (Modified)

25 ml glacial acetic acid added to 425 ml methanol followed by 50 ml 40%

formaldehyde. Stored at room temperature.

LDH stock solutions

(a) Tris buffer: 2.1g Tris base dissolved in 50ml DDW, adjusted to pH 8.5 with (6.3ml

5M HCl and made up to 100ml with DDW.

(b) LDH substrate: 0.01mol l-lactic acid made up to 50ml with DDW, adjusted to pH

5.5 with 1M NaOH and made up to 100ml with DDW.

(c) Chromagen solution: 12.5mg NAD added to 5mg iodotetrazolium chloride (INT)

dissolved in 2ml DDW. Immediately prior to use, 2.5mg of phenazine methosulphate

(PMS) weighed out and dissolved in 0.5ml DDW. 0.5ml of this PMS solution added to

the INT/NAD solution to make completed chromagen solution.

(d) Blank oxalate solution: 0.2g K2C2O4.H2O and 0.2g EDTA Na2H2.2H2O dissolved

in DDW and made up to 100ml.

176

Bibliography

(1999). "International consensus conferences in intensive care medicine: Ventilator-associated

Lung Injury in ARDS. This official conference report was cosponsored by the American

Thoracic Society, The European Society of Intensive Care Medicine, and The Societe de

Reanimation de Langue Francaise, and was approved by the ATS Board of Directors, July

1999." Am J Respir Crit Care Med 160(6): 2118-24.

Abel, S. J., S. J. Finney, S. J. Brett, B. F. Keogh, C. J. Morgan and T. W. Evans (1998).

"Reduced mortality in association with the acute respiratory distress syndrome (ARDS)." Thorax

53(4): 292-4.

Abraham, E., A. Carmody, R. Shenkar and J. Arcaroli (2000). "Neutrophils as early

immunologic effectors in hemorrhage- or endotoxemia-induced acute lung injury." Am J Physiol

Lung Cell Mol Physiol 279(6): L1137-45.

Abraham, V., M. L. Chou, K. M. DeBolt and M. Koval (1999). "Phenotypic control of gap

junctional communication by cultured alveolar epithelial cells." Am J Physiol 276(5 Pt 1): L825-

34.

Adamson, I. Y. and D. H. Bowden (1974). "The type 2 cell as progenitor of alveolar epithelial

regeneration. A cytodynamic study in mice after exposure to oxygen." Lab Invest 30(1): 35-42.

Adamson, I. Y. and D. H. Bowden (1975). "Derivation of type 1 epithelium from type 2 cells in

the developing rat lung." Lab Invest 32(6): 736-45.

Adamson, I. Y., C. Hedgecock and D. H. Bowden (1990). "Epithelial cell-fibroblast interactions

in lung injury and repair." Am J Pathol 137(2): 385-92.

Adamson, I. Y., L. Young and D. H. Bowden (1988). "Relationship of alveolar epithelial injury

and repair to the induction of pulmonary fibrosis." Am J Pathol 130(2): 377-83.

Adhikari, N., K. Burns and M. Meade (2004). "Pharmacologic therapies for adults with acute

lung injury and acute respiratory distress syndrome." Cochrane Database Syst Rev(4):

CD004477.

Akira, S. and K. Takeda (2004). "Toll-like receptor signalling." Nat Rev Immunol 4(7): 499-

511.

Alberts, B. (2002). "Molecular biology of the cell, 4th Edition."

Alcorn, D., T. M. Adamson, T. F. Lambert, J. E. Maloney, B. C. Ritchie and P. M. Robinson

(1977). "Morphological effects of chronic tracheal ligation and drainage in the fetal lamb lung."

J Anat 123(Pt 3): 649-60.

Amato, M. B., C. S. Barbas, D. M. Medeiros, R. B. Magaldi, G. P. Schettino, G. Lorenzi-Filho,

R. A. Kairalla, D. Deheinzelin, C. Munoz, R. Oliveira, T. Y. Takagaki and C. R. Carvalho

177

(1998). "Effect of a protective-ventilation strategy on mortality in the acute respiratory distress

syndrome." N Engl J Med 338(6): 347-54.

ARDSNet (2000). "Ventilation with lower tidal volumes as compared with traditional tidal

volumes for acute lung injury and the acute respiratory distress syndrome. The Acute

Respiratory Distress Syndrome Network." N Engl J Med 342(18): 1301-8.

Argiras, E. P., C. R. Blakeley, M. S. Dunnill, S. Otremski and M. K. Sykes (1987). "High PEEP

decreases hyaline membrane formation in surfactant deficient lungs." Br J Anaesth 59(10):

1278-85.

Armstrong, L., A. R. Medford, K. M. Uppington, J. Robertson, I. R. Witherden, T. D. Tetley and

A. B. Millar (2004). "Expression of functional toll-like receptor-2 and -4 on alveolar epithelial

cells." Am J Respir Cell Mol Biol 31(2): 241-5.

Ashbaugh, D. G., D. B. Bigelow, T. L. Petty and B. E. Levine (1967). "Acute respiratory distress

in adults." Lancet 2(7511): 319-23.

ATS (2000). "American Thoracic Society. Idiopathic pulmonary fibrosis: diagnosis and

treatment. International consensus statement. American Thoracic Society (ATS), and the

European Respiratory Society (ERS)." Am J Respir Crit Care Med 161(2 Pt 1): 646-64.

Bacallao, R., A. Garfinkel, S. Monke, G. Zampighi and L. J. Mandel (1994). "ATP depletion: a

novel method to study junctional properties in epithelial tissues. I. Rearrangement of the actin

cytoskeleton." J Cell Sci 107 ( Pt 12): 3301-13.

Bachofen, H., S. Schurch, M. Urbinelli and E. R. Weibel (1987). "Relations among alveolar

surface tension, surface area, volume, and recoil pressure." J Appl Physiol 62(5): 1878-87.

Bachofen, M. and E. R. Weibel (1982). "Structural alterations of lung parenchyma in the adult

respiratory distress syndrome." Clin Chest Med 3(1): 35-56.

Bai, C., N. Fukuda, Y. Song, T. Ma, M. A. Matthay and A. S. Verkman (1999). "Lung fluid

transport in aquaporin-1 and aquaporin-4 knockout mice." J Clin Invest 103(4): 555-61.

Bakker-Woudenberg, I. A. (2003). "Experimental models of pulmonary infection." J Microbiol

Methods 54(3): 295-313.

Bals, R. and P. S. Hiemstra (2004). "Innate immunity in the lung: how epithelial cells fight

against respiratory pathogens." Eur Respir J 23(2): 327-33.

Barazzone, C., Y. R. Donati, A. F. Rochat, C. Vesin, C. D. Kan, J. C. Pache and P. F. Piguet

(1999). "Keratinocyte growth factor protects alveolar epithelium and endothelium from oxygen-

induced injury in mice." Am J Pathol 154(5): 1479-87.

Barazzone, C., S. Horowitz, Y. R. Donati, I. Rodriguez and P. F. Piguet (1998). "Oxygen

toxicity in mouse lung: pathways to cell death." Am J Respir Cell Mol Biol 19(4): 573-81.

178

Bardales, R. H., S. S. Xie, R. F. Schaefer and S. M. Hsu (1996). "Apoptosis is a major pathway

responsible for the resolution of type II pneumocytes in acute lung injury." Am J Pathol 149(3):

845-52.

Bartlett, J. G., P. O'Keefe, F. P. Tally, T. J. Louie and S. L. Gorbach (1986). "Bacteriology of

hospital-acquired pneumonia." Arch Intern Med 146(5): 868-71.

Beers, M. F. and A. B. Fisher (1992). "Surfactant protein C: a review of its unique properties and

metabolism." Am J Physiol 263(2 Pt 1): L151-60.

Beers, M. F. and C. Lomax (1995). "Synthesis and processing of hydrophobic surfactant protein

C by isolated rat type II cells." Am J Physiol 269(6 Pt 1): L744-53.

Beers, M. F., A. Wali, M. F. Eckenhoff, S. I. Feinstein, J. H. Fisher and A. B. Fisher (1992). "An

antibody with specificity for surfactant protein C precursors: identification of pro-SP-C in rat

lung." Am J Respir Cell Mol Biol 7(4): 368-78.

Behrendt, C. E. (2000). "Acute respiratory failure in the United States: incidence and 31-day

survival." Chest 118(4): 1100-5.

Bellamy, C. O., A. R. Clarke, A. H. Wyllie and D. J. Harrison (1997). "p53 Deficiency in liver

reduces local control of survival and proliferation, but does not affect apoptosis after DNA

damage." Faseb J 11(7): 591-9.

Bernard, G. R., A. Artigas, K. L. Brigham, J. Carlet, K. Falke, L. Hudson, M. Lamy, J. R.

Legall, A. Morris and R. Spragg (1994). "The American-European Consensus Conference on

ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination." Am J

Respir Crit Care Med 149(3 Pt 1): 818-24.

Berthiaume, Y., O. Lesur and A. Dagenais (1999). "Treatment of adult respiratory distress

syndrome: plea for rescue therapy of the alveolar epithelium." Thorax 54(2): 150-60.

Besnard, V., S. Corroyer, G. Trugnan, K. Chadelat, E. Nabeyrat, V. Cazals and A. Clement

(2001). "Distinct patterns of insulin-like growth factor binding protein (IGFBP)-2 and IGFBP-3

expression in oxidant exposed lung epithelial cells." Biochim Biophys Acta 1538(1): 47-58.

Bhaskaran, M., N. Kolliputi, Y. Wang, D. Gou, N. R. Chintagari and L. Liu (2007). "Trans-

differentiation of alveolar epithelial type II cells to type I cells involves autocrine signaling by

transforming growth factor beta 1 through the Smad pathway." J Biol Chem 282(6): 3968-76.

Bitterman, P. B. (1992). "Pathogenesis of fibrosis in acute lung injury." Am J Med 92(6A): 39S-

43S.

Borok, Z., S. I. Danto, R. L. Lubman, Y. Cao, M. C. Williams and E. D. Crandall (1998).

"Modulation of t1alpha expression with alveolar epithelial cell phenotype in vitro." Am J

Physiol 275(1 Pt 1): L155-64.

179

Borok, Z., R. L. Lubman, S. I. Danto, X. L. Zhang, S. M. Zabski, L. S. King, D. M. Lee, P. Agre

and E. D. Crandall (1998). "Keratinocyte growth factor modulates alveolar epithelial cell

phenotype in vitro: expression of aquaporin 5." Am J Respir Cell Mol Biol 18(4): 554-61.

Bouvry, D., C. Planes, L. Malbert-Colas, V. Escabasse and C. Clerici (2006). "Hypoxia-induced

cytoskeleton disruption in alveolar epithelial cells." Am J Respir Cell Mol Biol 35(5): 519-27.

Boylan, G. M., J. G. Pryde, L. G. Dobbs and M. C. McElroy (2001). "Identification of a novel

antigen on the apical surface of rat alveolar epithelial type II and Clara cells." Am J Physiol

Lung Cell Mol Physiol 280(6): L1318-26.

Bradford, M. M. (1976). "A rapid and sensitive method for the quantitation of microgram

quantities of protein utilizing the principle of protein-dye binding." Anal Biochem 72: 248-54.

BTS (1992). "The aetiology, management and outcome of severe community-acquired

pneumonia on the intensive care unit. The British Thoracic Society Research Committee and

The Public Health Laboratory Service." Respir Med 86(1): 7-13.

BTS (2001). "BTS Guidelines for the Management of Community Acquired Pneumonia in

Adults." Thorax 56 Suppl 4: IV1-64.

BTS and T. B. T. S. R. C. a. T. P. H. L. Service (1992). "The aetiology, management and

outcome of severe community-acquired pneumonia on the intensive care unit." Respir Med

86(1): 7-13.

Buccellato, L. J., M. Tso, O. I. Akinci, N. S. Chandel and G. R. Budinger (2004). "Reactive

oxygen species are required for hyperoxia-induced Bax activation and cell death in alveolar

epithelial cells." J Biol Chem 279(8): 6753-60.

Buckley, S., L. Barsky, B. Driscoll, K. Weinberg, K. D. Anderson and D. Warburton (1998).

"Apoptosis and DNA damage in type 2 alveolar epithelial cells cultured from hyperoxic rats."

Am J Physiol 274(5 Pt 1): L714-20.

Calandra, T., J. Bernhagen, C. N. Metz, L. A. Spiegel, M. Bacher, T. Donnelly, A. Cerami and

R. Bucala (1995). "MIF as a glucocorticoid-induced modulator of cytokine production." Nature

377(6544): 68-71.

Campbell, L., A. J. Hollins, A. Al-Eid, G. R. Newman, C. von Ruhland and M. Gumbleton

(1999). "Caveolin-1 expression and caveolae biogenesis during cell transdifferentiation in lung

alveolar epithelial primary cultures." Biochem Biophys Res Commun 262(3): 744-51.

Cao, Y. X., M. I. Ramirez and M. C. Williams (2003). "Enhanced binding of Sp1/Sp3

transcription factors mediates the hyperoxia-induced increased expression of the lung type I cell

gene T1alpha." J Cell Biochem 89(5): 887-901.

Carden, D. L., J. S. Alexander and R. B. George (1998). "The pathophysiology of the acute

respiratory distress syndrome." Pathophysiology 5: 1-13.

180

Cavanaugh, K. J., Jr., J. Oswari and S. S. Margulies (2001). "Role of stretch on tight junction

structure in alveolar epithelial cells." Am J Respir Cell Mol Biol 25(5): 584-91.

Cazals, V., B. Mouhieddine, B. Maitre, Y. Le Bouc, K. Chadelat, J. S. Brody and A. Clement

(1994). "Insulin-like growth factors, their binding proteins, and transforming growth factor-beta

1 in oxidant-arrested lung alveolar epithelial cells." J Biol Chem 269(19): 14111-7.

Chapman, K. E., S. E. Sinclair, D. Zhuang, A. Hassid, L. P. Desai and C. M. Waters (2005).

"Cyclic mechanical strain increases reactive oxygen species production in pulmonary epithelial

cells." Am J Physiol Lung Cell Mol Physiol 289(5): L834-41.

Chastre, J. and J. Y. Fagon (2002). "Ventilator-associated pneumonia." Am J Respir Crit Care

Med 165(7): 867-903.

Chen, J., Z. Chen, T. Narasaraju, N. Jin and L. Liu (2004). "Isolation of highly pure alveolar

epithelial type I and type II cells from rat lungs." Lab Invest 84(6): 727-35.

Chetty, A. and H. C. Nielsen (2002). "Regulation of cell proliferation by insulin-like growth

factor 1 in hyperoxia-exposed neonatal rat lung." Mol Genet Metab 75(3): 265-75.

Christensen, P. J., S. Kim, R. H. Simon, G. B. Toews and R. Paine, 3rd (1993). "Differentiation-

related expression of ICAM-1 by rat alveolar epithelial cells." Am J Respir Cell Mol Biol 8(1):

9-15.

Cines, D. B., E. S. Pollak, C. A. Buck, J. Loscalzo, G. A. Zimmerman, R. P. McEver, J. S.

Pober, T. M. Wick, B. A. Konkle, B. S. Schwartz, E. S. Barnathan, K. R. McCrae, B. A. Hug, A.

M. Schmidt and D. M. Stern (1998). "Endothelial cells in physiology and in the pathophysiology

of vascular disorders." Blood 91(10): 3527-61.

Clark, R. A., J. M. Lanigan, P. DellaPelle, E. Manseau, H. F. Dvorak and R. B. Colvin (1982).

"Fibronectin and fibrin provide a provisional matrix for epidermal cell migration during wound

reepithelialization." J Invest Dermatol 79(5): 264-9.

Clegg, G. R., C. Tyrrell, S. R. McKechnie, M. F. Beers, D. Harrison and M. C. McElroy (2005).

"Coexpression of RTI40 with alveolar epithelial type II cell proteins in lungs following injury:

identification of alveolar intermediate cell types." Am J Physiol Lung Cell Mol Physiol 289(3):

L382-90.

Clement, A., M. Edeas, K. Chadelat and J. S. Brody (1992). "Inhibition of lung epithelial cell

proliferation by hyperoxia. Posttranscriptional regulation of proliferation-related genes." J Clin

Invest 90(5): 1812-8.

Clerch, L. B. and D. Massaro (1993). "Tolerance of rats to hyperoxia. Lung antioxidant enzyme

gene expression." J Clin Invest 91(2): 499-508.

Cooper, P., S. Potter, B. Mueck, S. Yousefi and G. Jarai (2001). "Identification of genes induced

by inflammatory cytokines in airway epithelium." Am J Physiol Lung Cell Mol Physiol 280(5):

L841-52.

181

Corroyer, S., B. Maitre, V. Cazals and A. Clement (1996). "Altered regulation of G1 cyclins in

oxidant-induced growth arrest of lung alveolar epithelial cells. Accumulation of inactive cyclin

E-DCK2 complexes." J Biol Chem 271(41): 25117-25.

Cortes, G., D. Alvarez, C. Saus and S. Alberti (2002). "Role of lung epithelial cells in defense

against Klebsiella pneumoniae pneumonia." Infect Immun 70(3): 1075-80.

Crandall, E. D. and M. A. Matthay (2001). "Alveolar epithelial transport. Basic science to

clinical medicine." Am J Respir Crit Care Med 163(4): 1021-9.

Crapo, J. D. (1986). "Morphologic changes in pulmonary oxygen toxicity." Annu Rev Physiol

48: 721-31.

Crapo, J. D., B. E. Barry, H. A. Foscue and J. Shelburne (1980). "Structural and biochemical

changes in rat lungs occurring during exposures to lethal and adaptive doses of oxygen." Am

Rev Respir Dis 122(1): 123-43.

Crapo, J. D., B. E. Barry, P. Gehr, M. Bachofen and E. R. Weibel (1982). "Cell number and cell

characteristics of the normal human lung." Am Rev Respir Dis 126(2): 332-7.

Crapo, J. D., B. A. Freeman, B. E. Barry, J. F. Turrens and S. L. Young (1983). "Mechanisms of

hyperoxic injury to the pulmonary microcirculation." Physiologist 26(3): 170-6.

Crapo, J. D., S. L. Young, E. K. Fram, K. E. Pinkerton, B. E. Barry and R. O. Crapo (1983).

"Morphometric characteristics of cells in the alveolar region of mammalian lungs." Am Rev

Respir Dis 128(2 Pt 2): S42-6.

Crystal, R. G., J. D. Fulmer, W. C. Roberts, M. L. Moss, B. R. Line and H. Y. Reynolds (1976).

"Idiopathic pulmonary fibrosis. Clinical, histologic, radiographic, physiologic, scintigraphic,

cytologic, and biochemical aspects." Ann Intern Med 85(6): 769-88.

D'Angio, C. T. and J. N. Finkelstein (2000). "Oxygen regulation of gene expression: a study in

opposites." Mol Genet Metab 71(1-2): 371-80.

Daly, H. E., C. M. Baecher-Allan, A. T. Paxhia, R. M. Ryan, R. K. Barth and J. N. Finkelstein

(1998). "Cell-specific gene expression reveals changes in epithelial cell populations after

bleomycin treatment." Lab Invest 78(4): 393-400.

Danto, S. I., S. M. Zabski and E. D. Crandall (1992). "Reactivity of alveolar epithelial cells in

primary culture with type I cell monoclonal antibodies." Am J Respir Cell Mol Biol 6(3): 296-

306.

Davis, W. B., S. I. Rennard, P. B. Bitterman and R. G. Crystal (1983). "Pulmonary oxygen

toxicity. Early reversible changes in human alveolar structures induced by hyperoxia." N Engl J

Med 309(15): 878-83.

De Paepe, M. E., B. D. Johnson, K. Papadakis and F. I. Luks (1999). "Lung growth response

after tracheal occlusion in fetal rabbits is gestational age-dependent." Am J Respir Cell Mol Biol

21(1): 65-76.

182

De Paepe, M. E., Q. Mao, Y. Chao, J. L. Powell, L. P. Rubin and S. Sharma (2005). "Hyperoxia-

induced apoptosis and Fas/FasL expression in lung epithelial cells." Am J Physiol Lung Cell

Mol Physiol 289(4): L647-59.

De Paepe, M. E., Q. Mao and F. I. Luks (2004). "Expression of apoptosis-related genes after

fetal tracheal occlusion in rabbits." J Pediatr Surg 39(11): 1616-25.

Delclaux, C. and E. Azoulay (2003). "Inflammatory response to infectious pulmonary injury."

Eur Respir J Suppl 42: 10s-14s.

Denker, B. M. and S. K. Nigam (1998). "Molecular structure and assembly of the tight junction."

Am J Physiol 274(1 Pt 2): F1-9.

Deterding, R. R., A. M. Havill, T. Yano, S. C. Middleton, C. R. Jacoby, J. M. Shannon, W. S.

Simonet and R. J. Mason (1997). "Prevention of bleomycin-induced lung injury in rats by

keratinocyte growth factor." Proc Assoc Am Physicians 109(3): 254-68.

Dobbs, L. G. (1990). "Isolation and culture of alveolar type II cells." Am J Physiol 258(4 Pt 1):

L134-47.

Dobbs, L. G., E. F. Geppert, M. C. Williams, R. D. Greenleaf and R. J. Mason (1980).

"Metabolic properties and ultrastructure of alveolar type II cells isolated with elastase." Biochim

Biophys Acta 618(3): 510-23.

Dobbs, L. G., R. Gonzalez, M. A. Matthay, E. P. Carter, L. Allen and A. S. Verkman (1998).

"Highly water-permeable type I alveolar epithelial cells confer high water permeability between

the airspace and vasculature in rat lung." Proc Natl Acad Sci U S A 95(6): 2991-6.

Dobbs, L. G., R. Gonzalez and M. C. Williams (1986). "An improved method for isolating type

II cells in high yield and purity." Am Rev Respir Dis 134(1): 141-5.

Dobbs, L. G., M. S. Pian, M. Maglio, S. Dumars and L. Allen (1997). "Maintenance of the

differentiated type II cell phenotype by culture with an apical air surface." Am J Physiol 273(2

Pt 1): L347-54.

Dobbs, L. G., M. C. Williams and A. E. Brandt (1985). "Changes in biochemical characteristics

and pattern of lectin binding of alveolar type II cells with time in culture." Biochim Biophys

Acta 846(1): 155-66.

Dobbs, L. G., M. C. Williams and R. Gonzalez (1988). "Monoclonal antibodies specific to apical

surfaces of rat alveolar type I cells bind to surfaces of cultured, but not freshly isolated, type II

cells." Biochim Biophys Acta 970(2): 146-56.

Donnelly, S. C., C. Haslett, P. T. Reid, I. S. Grant, W. A. Wallace, C. N. Metz, L. J. Bruce and

R. Bucala (1997). "Regulatory role for macrophage migration inhibitory factor in acute

respiratory distress syndrome." Nat Med 3(3): 320-3.

Donnelly, S. C., R. M. Strieter, P. T. Reid, S. L. Kunkel, M. D. Burdick, I. Armstrong, A.

Mackenzie and C. Haslett (1996). "The association between mortality rates and decreased

183

concentrations of interleukin-10 and interleukin-1 receptor antagonist in the lung fluids of

patients with the adult respiratory distress syndrome." Ann Intern Med 125(3): 191-6.

dos Santos, C. C., B. Han, C. F. Andrade, X. Bai, S. Uhlig, R. Hubmayr, M. Tsang, M. Lodyga,

S. Keshavjee, A. S. Slutsky and M. Liu (2004). "DNA microarray analysis of gene expression in

alveolar epithelial cells in response to TNFalpha, LPS, and cyclic stretch." Physiol Genomics

19(3): 331-42.

Dos Santos, C. C. and A. S. Slutsky (2000). "Invited review: mechanisms of ventilator-induced

lung injury: a perspective." J Appl Physiol 89(4): 1645-55.

Douzinas, E. E., P. D. Tsidemiadou, M. T. Pitaridis, I. Andrianakis, A. Bobota-Chloraki, K.

Katsouyanni, D. Sfyras, K. Malagari and C. Roussos (1997). "The regional production of

cytokines and lactate in sepsis-related multiple organ failure." Am J Respir Crit Care Med

155(1): 53-9.

Doyle, I. R., A. D. Bersten and T. E. Nicholas (1997). "Surfactant proteins-A and -B are elevated

in plasma of patients with acute respiratory failure." Am J Respir Crit Care Med 156(4 Pt 1):

1217-29.

Doyle, I. R., T. E. Nicholas and A. D. Bersten (1995). "Serum surfactant protein-A levels in

patients with acute cardiogenic pulmonary edema and adult respiratory distress syndrome." Am J

Respir Crit Care Med 152(1): 307-17.

Dreyfuss, D., G. Basset, P. Soler and G. Saumon (1985). "Intermittent positive-pressure

hyperventilation with high inflation pressures produces pulmonary microvascular injury in rats."

Am Rev Respir Dis 132(4): 880-4.

Dreyfuss, D. and G. Saumon (1998). "Ventilator-induced lung injury: lessons from experimental

studies." Am J Respir Crit Care Med 157(1): 294-323.

Dreyfuss, D., P. Soler, G. Basset and G. Saumon (1988). "High inflation pressure pulmonary

edema. Respective effects of high airway pressure, high tidal volume, and positive end-

expiratory pressure." Am Rev Respir Dis 137(5): 1159-64.

Duane, P. G., J. B. Rubins, H. R. Weisel and E. N. Janoff (1993). "Identification of hydrogen

peroxide as a Streptococcus pneumoniae toxin for rat alveolar epithelial cells." Infect Immun

61(10): 4392-7.

Edwards, Y. S. (2001). "Stretch stimulation: its effects on alveolar type II cell function in the

lung." Comp Biochem Physiol A Mol Integr Physiol 129(1): 245-60.

Edwards, Y. S., L. M. Sutherland and A. W. Murray (2000). "NO protects alveolar type II cells

from stretch-induced apoptosis. A novel role for macrophages in the lung." Am J Physiol Lung

Cell Mol Physiol 279(6): L1236-42.

Edwards, Y. S., L. M. Sutherland, J. H. Power, T. E. Nicholas and A. W. Murray (1999). "Cyclic

stretch induces both apoptosis and secretion in rat alveolar type II cells." FEBS Lett 448(1): 127-

30.

184

Effros, R. M., C. Darin, E. R. Jacobs, R. A. Rogers, G. Krenz and E. E. Schneeberger (1997).

"Water transport and the distribution of aquaporin-1 in pulmonary air spaces." J Appl Physiol

83(3): 1002-16.

Egan, E. A. (1982). "Lung inflation, lung solute permeability, and alveolar edema." J Appl

Physiol 53(1): 121-5.

Evans, M. J., L. J. Cabral, R. J. Stephens and G. Freeman (1975). "Transformation of alveolar

type 2 cells to type 1 cells following exposure to NO2." Exp Mol Pathol 22(1): 142-50.

Evans, M. J., R. J. Stephens, L. J. Cabral and G. Freeman (1972). "Cell renewal in the lungs of

rats exposed to low levels of NO2." Arch Environ Health 24(3): 180-8.

Evans, M. J., L. S. Van Winkle, M. V. Fanucchi and C. G. Plopper (1999). "The attenuated

fibroblast sheath of the respiratory tract epithelial-mesenchymal trophic unit." Am J Respir Cell

Mol Biol 21(6): 655-7.

Fang, K. C. (2000). "Mesenchymal regulation of alveolar repair in pulmonary fibrosis." Am J

Respir Cell Mol Biol 23(2): 142-5.

Fehrenbach, H. (2001). "Alveolar epithelial type II cell: defender of the alveolus revisited."

Respir Res 2(1): 33-46.

Fehrenbach, H., M. Kasper, R. Koslowski, T. Pan, D. Schuh, M. Muller and R. J. Mason (2000).

"Alveolar epithelial type II cell apoptosis in vivo during resolution of keratinocyte growth

factor-induced hyperplasia in the rat." Histochem Cell Biol 114(1): 49-61.

Fehrenbach, H., M. Kasper, T. Tschernig, T. Pan, D. Schuh, J. M. Shannon, M. Muller and R. J.

Mason (1999). "Keratinocyte growth factor-induced hyperplasia of rat alveolar type II cells in

vivo is resolved by differentiation into type I cells and by apoptosis." Eur Respir J 14(3): 534-44.

Fine, A., Y. Janssen-Heininger, R. P. Soultanakis, S. G. Swisher and B. D. Uhal (2000).

"Apoptosis in lung pathophysiology." Am J Physiol Lung Cell Mol Physiol 279(3): L423-7.

Fishman, A. P. (1982). "Endothelium: a distributed organ of diverse capabilities." Ann N Y

Acad Sci 401: 1-8.

Flecknoe, S., R. Harding, G. Maritz and S. B. Hooper (2000). "Increased lung expansion alters

the proportions of type I and type II alveolar epithelial cells in fetal sheep." Am J Physiol Lung

Cell Mol Physiol 278(6): L1180-5.

Flecknoe, S. J., M. J. Wallace, R. Harding and S. B. Hooper (2002). "Determination of alveolar

epithelial cell phenotypes in fetal sheep: evidence for the involvement of basal lung expansion."

J Physiol 542(Pt 1): 245-53.

Folkesson, H. G., M. A. Matthay, C. A. Hebert and V. C. Broaddus (1995). "Acid aspiration-

induced lung injury in rabbits is mediated by interleukin-8-dependent mechanisms." J Clin

Invest 96(1): 107-16.

185

Fracica, P. J., M. J. Knapp, C. A. Piantadosi, K. Takeda, W. J. Fulkerson, R. E. Coleman, W. G.

Wolfe and J. D. Crapo (1991). "Responses of baboons to prolonged hyperoxia: physiology and

qualitative pathology." J Appl Physiol 71(6): 2352-62.

Frank, J. A., J. A. Gutierrez, K. D. Jones, L. Allen, L. Dobbs and M. A. Matthay (2002). "Low

tidal volume reduces epithelial and endothelial injury in acid-injured rat lungs." Am J Respir Crit

Care Med 165(2): 242-9.

Frank, J. A., C. M. Wray, D. F. McAuley, R. Schwendener and M. A. Matthay (2006). "Alveolar

macrophages contribute to alveolar barrier dysfunction in ventilator-induced lung injury." Am J

Physiol Lung Cell Mol Physiol 291(6): L1191-8.

Franklin, L., A. D. Cronshaw, D. J. Harrison and M. C. McElroy (2004). "The type II and clara

cell specific monoclonal antibody (MMC4) recognises aminopeptidase N." Eur Respir J 24:

Suppl. 48: 719s.

Fujikawa, L. S., C. S. Foster, T. J. Harrist, J. M. Lanigan and R. B. Colvin (1981). "Fibronectin

in healing rabbit corneal wounds." Lab Invest 45(2): 120-9.

Funkhouser, J. D. and R. D. Peterson (1989). "Immunotargeting: a contemporary approach to the

study of lung development." Am J Physiol 257(6 Pt 1): L311-7.

Funkhouser, J. D., S. D. Tangada, M. Jones, S. J. O and R. D. Peterson (1991). "p146 type II

alveolar epithelial cell antigen is identical to aminopeptidase N." Am J Physiol 260(4 Pt 1):

L274-9.

Gattinoni, L., P. Pelosi, P. M. Suter, A. Pedoto, P. Vercesi and A. Lissoni (1998). "Acute

respiratory distress syndrome caused by pulmonary and extrapulmonary disease. Different

syndromes?" Am J Respir Crit Care Med 158(1): 3-11.

Geiser, T. (2003). "Idiopathic pulmonary fibrosis--a disorder of alveolar wound repair?" Swiss

Med Wkly 133(29-30): 405-11.

Giangreco, A., S. D. Reynolds and B. R. Stripp (2002). "Terminal bronchioles harbor a unique

airway stem cell population that localizes to the bronchoalveolar duct junction." Am J Pathol

161(1): 173-82.

Gil, J. and A. Martinez-Hernandez (1984). "The connective tissue of the rat lung: electron

immunohistochemical studies." J Histochem Cytochem 32(2): 230-8.

Girod, C. E., D. H. Shin, M. B. Hershenson, J. Solway, R. Dahl and Y. E. Miller (1996). "p172:

An alveolar type II and Clara cell specific protein with late developmental expression and

upregulation by hyperoxic lung injury." Am J Respir Cell Mol Biol 14(6): 538-47.

Gon, Y., M. R. Wood, W. B. Kiosses, E. Jo, M. G. Sanna, J. Chun and H. Rosen (2005). "S1P3

receptor-induced reorganization of epithelial tight junctions compromises lung barrier integrity

and is potentiated by TNF." Proc Natl Acad Sci U S A 102(26): 9270-5.

186

Gonzalez, R., Y. H. Yang, C. Griffin, L. Allen, Z. Tigue and L. Dobbs (2005). "Freshly isolated

rat alveolar type I cells, type II cells, and cultured type II cells have distinct molecular

phenotypes." Am J Physiol Lung Cell Mol Physiol 288(1): L179-89.

Gonzalez-Mariscal, L., B. Chavez de Ramirez and M. Cereijido (1985). "Tight junction

formation in cultured epithelial cells (MDCK)." J Membr Biol 86(2): 113-25.

Goodenough, D. A. (1999). "Plugging the leaks." Proc Natl Acad Sci U S A 96(2): 319-21.

Greene, K. E., J. R. Wright, K. P. Steinberg, J. T. Ruzinski, E. Caldwell, W. B. Wong, W. Hull,

J. A. Whitsett, T. Akino, Y. Kuroki, H. Nagae, L. D. Hudson and T. R. Martin (1999). "Serial

changes in surfactant-associated proteins in lung and serum before and after onset of ARDS."

Am J Respir Crit Care Med 160(6): 1843-50.

Gregory, T. J., W. J. Longmore, M. A. Moxley, J. A. Whitsett, C. R. Reed, A. A. Fowler, 3rd, L.

D. Hudson, R. J. Maunder, C. Crim and T. M. Hyers (1991). "Surfactant chemical composition

and biophysical activity in acute respiratory distress syndrome." J Clin Invest 88(6): 1976-81.

Guest, J. F. and A. Morris (1997). "Community-acquired pneumonia: the annual cost to the

National Health Service in the UK." Eur Respir J 10(7): 1530-4.

Gunther, A., C. Siebert, R. Schmidt, S. Ziegler, F. Grimminger, M. Yabut, B. Temmesfeld, D.

Walmrath, H. Morr and W. Seeger (1996). "Surfactant alterations in severe pneumonia, acute

respiratory distress syndrome, and cardiogenic lung edema." Am J Respir Crit Care Med 153(1):

176-84.

Guo, J., E. S. Yi, A. M. Havill, I. Sarosi, L. Whitcomb, S. Yin, S. C. Middleton, P. Piguet and T.

R. Ulich (1998). "Intravenous keratinocyte growth factor protects against experimental

pulmonary injury." Am J Physiol 275(4 Pt 1): L800-5.

Gutierrez, J. A., R. Ertsey, L. M. Scavo, E. Collins and L. G. Dobbs (1999). "Mechanical

distention modulates alveolar epithelial cell phenotypic expression by transcriptional

regulation." Am J Respir Cell Mol Biol 21(2): 223-9.

Gutierrez, J. A., R. F. Gonzalez and L. G. Dobbs (1998). "Mechanical distension modulates

pulmonary alveolar epithelial phenotypic expression in vitro." Am J Physiol 274(2 Pt 1): L196-

202.

Gutierrez, J. A., V. V. Suzara and L. G. Dobbs (2003). "Continuous mechanical contraction

modulates expression of alveolar epithelial cell phenotype." Am J Respir Cell Mol Biol 29(1):

81-7.

Haddad, J. J. (2002). "Oxygen homeostasis, thiol equilibrium and redox regulation of signalling

transcription factors in the alveolar epithelium." Cell Signal 14(10): 799-810.

Haddad, J. J. (2002). "Science review: Redox and oxygen-sensitive transcription factors in the

regulation of oxidant-mediated lung injury: role for nuclear factor-kappaB." Crit Care 6(6): 481-

90.

187

Haddad, J. J. and S. C. Land (2000). "The differential expression of apoptosis factors in the

alveolar epithelium is redox sensitive and requires NF-kappaB (RelA)-selective targeting."

Biochem Biophys Res Commun 271(1): 257-67.

Haies, D. M., J. Gil and E. R. Weibel (1981). "Morphometric study of rat lung cells. I.

Numerical and dimensional characteristics of parenchymal cell population." Am Rev Respir Dis

123(5): 533-41.

Hammerschmidt, S., H. Kuhn, T. Grasenack, C. Gessner and H. Wirtz (2004). "Apoptosis and

necrosis induced by cyclic mechanical stretching in alveolar type II cells." Am J Respir Cell Mol

Biol 30(3): 396-402.

Harding, R., S. B. Hooper and V. K. Han (1993). "Abolition of fetal breathing movements by

spinal cord transection leads to reductions in fetal lung liquid volume, lung growth, and IGF-II

gene expression." Pediatr Res 34(2): 148-53.

Hayatdavoudi, G., J. J. O'Neil, B. E. Barry, B. A. Freeman and J. D. Crapo (1981). "Pulmonary

injury in rats following continuous exposure to 60% O2 for 7 days." J Appl Physiol 51(5): 1220-

31.

Held, H. D., S. Boettcher, L. Hamann and S. Uhlig (2001). "Ventilation-induced chemokine and

cytokine release is associated with activation of nuclear factor-kappaB and is blocked by

steroids." Am J Respir Crit Care Med 163(3 Pt 1): 711-6.

Hermans, C. and A. Bernard (1999). "Lung epithelium-specific proteins: characteristics and

potential applications as markers." Am J Respir Crit Care Med 159(2): 646-78.

Herridge, M. S., A. M. Cheung, C. M. Tansey, A. Matte-Martyn, N. Diaz-Granados, F. Al-Saidi,

A. B. Cooper, C. B. Guest, C. D. Mazer, S. Mehta, T. E. Stewart, A. Barr, D. Cook and A. S.

Slutsky (2003). "One-year outcomes in survivors of the acute respiratory distress syndrome." N

Engl J Med 348(8): 683-93.

Hillyer, P., E. Mordelet, G. Flynn and D. Male (2003). "Chemokines, chemokine receptors and

adhesion molecules on different human endothelia: discriminating the tissue-specific functions

that affect leucocyte migration." Clin Exp Immunol 134(3): 431-41.

Hippenstiel, S., B. Opitz, B. Schmeck and N. Suttorp (2006). "Lung epithelium as a sentinel and

effector system in pneumonia--molecular mechanisms of pathogen recognition and signal

transduction." Respir Res 7: 97.

Hirani, N. A. and J. T. Macfarlane (1997). "Impact of management guidelines on the outcome of

severe community acquired pneumonia." Thorax 52(1): 17-21.

Hooper, S. B., V. K. Han and R. Harding (1993). "Changes in lung expansion alter pulmonary

DNA synthesis and IGF-II gene expression in fetal sheep." Am J Physiol 265(4 Pt 1): L403-9.

Hsia, C. C. (2004). "Signals and mechanisms of compensatory lung growth." J Appl Physiol

97(5): 1992-8.

188

Ikegami, M. and A. H. Jobe (1998). "Surfactant protein metabolism in vivo." Biochim Biophys

Acta 1408(2-3): 218-25.

Imai, Y., J. Parodo, O. Kajikawa, M. de Perrot, S. Fischer, V. Edwards, E. Cutz, M. Liu, S.

Keshavjee, T. R. Martin, J. C. Marshall, V. M. Ranieri and A. S. Slutsky (2003). "Injurious

mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an

experimental model of acute respiratory distress syndrome." Jama 289(16): 2104-12.

Isakson, B. E., G. J. Seedorf, R. L. Lubman, W. H. Evans and S. Boitano (2003). "Cell-cell

communication in heterocellular cultures of alveolar epithelial cells." Am J Respir Cell Mol Biol

29(5): 552-61.

Ishizawa, K., H. Kubo, M. Yamada, S. Kobayashi, M. Numasaki, S. Ueda, T. Suzuki and H.

Sasaki (2004). "Bone marrow-derived cells contribute to lung regeneration after elastase-induced

pulmonary emphysema." FEBS Lett 556(1-3): 249-52.

Jafari, B., B. Ouyang, L. F. Li, C. A. Hales and D. A. Quinn (2004). "Intracellular glutathione in

stretch-induced cytokine release from alveolar type-2 like cells." Respirology 9(1): 43-53.

Janeway, C. A., Jr. and R. Medzhitov (2002). "Innate immune recognition." Annu Rev Immunol

20: 197-216.

Jiao, G., E. Li and R. Yu (2002). "Decreased expression of AQP1 and AQP5 in acute injured

lungs in rats." Chin Med J (Engl) 115(7): 963-7.

Joe, P., L. D. Wallen, C. J. Chapin, C. H. Lee, L. Allen, V. K. Han, L. G. Dobbs, S. Hawgood

and J. A. Kitterman (1997). "Effects of mechanical factors on growth and maturation of the lung

in fetal sheep." Am J Physiol 272(1 Pt 1): L95-105.

Johanson, W. G., Jr., J. H. Higuchi, D. E. Woods, P. Gomez and J. J. Coalson (1985).

"Dissemination of Pseudomonas aeruginosa during lung infection in hamsters. Role of oxygen-

induced lung injury." Am Rev Respir Dis 132(2): 358-61.

Johnson, M. D., J. H. Widdicombe, L. Allen, P. Barbry and L. G. Dobbs (2002). "Alveolar

epithelial type I cells contain transport proteins and transport sodium, supporting an active role

for type I cells in regulation of lung liquid homeostasis." Proc Natl Acad Sci U S A 99(4): 1966-

71.

Joshi, P. C., G. V. Poole, V. Sachdev, X. Zhou and Q. Jones (2000). "Trauma patients with

positive cultures have higher levels of circulating macrophage migration inhibitory factor

(MIF)." Res Commun Mol Pathol Pharmacol 107(1-2): 13-20.

Kapanci, Y., E. R. Weibel, H. P. Kaplan and F. R. Robinson (1969). "Pathogenesis and

reversibility of the pulmonary lesions of oxygen toxicity in monkeys. II. Ultrastructural and

morphometric studies." Lab Invest 20(1): 101-18.

Kawano, T., S. Mori, M. Cybulsky, R. Burger, A. Ballin, E. Cutz and A. C. Bryan (1987).

"Effect of granulocyte depletion in a ventilated surfactant-depleted lung." J Appl Physiol 62(1):

27-33.

189

Kazzaz, J. A., J. Xu, T. A. Palaia, L. Mantell, A. M. Fein and S. Horowitz (1996). "Cellular

oxygen toxicity. Oxidant injury without apoptosis." J Biol Chem 271(25): 15182-6.

Kikkawa, Y. and K. Yoneda (1974). "The type II epithelial cell of the lung. I. Method of

isolation." Lab Invest 30(1): 76-84.

King, L. S. and P. Agre (1996). "Pathophysiology of the aquaporin water channels." Annu Rev

Physiol 58: 619-48.

Kitterman, J. A. (1996). "The effects of mechanical forces on fetal lung growth." Clin Perinatol

23(4): 727-40.

Kleeberger, W., A. Versmold, T. Rothamel, S. Glockner, M. Bredt, A. Haverich, U. Lehmann

and H. Kreipe (2003). "Increased chimerism of bronchial and alveolar epithelium in human lung

allografts undergoing chronic injury." Am J Pathol 162(5): 1487-94.

Kobayashi, J. and S. Kitamura (1995). "KL-6: a serum marker for interstitial pneumonia." Chest

108(2): 311-5.

Kohno, N., M. Akiyama, S. Kyoizumi, M. Hakoda, K. Kobuke and M. Yamakido (1988).

"Detection of soluble tumor-associated antigens in sera and effusions using novel monoclonal

antibodies, KL-3 and KL-6, against lung adenocarcinoma." Jpn J Clin Oncol 18(3): 203-16.

Korzeniewski, C. and D. M. Callewaert (1983). "An enzyme-release assay for natural

cytotoxicity." J Immunol Methods 64(3): 313-20.

Kotton, D. N., A. J. Fabian and R. C. Mulligan (2005). "Failure of bone marrow to reconstitute

lung epithelium." Am J Respir Cell Mol Biol 33(4): 328-34.

Kreda, S. M., M. C. Gynn, D. A. Fenstermacher, R. C. Boucher and S. E. Gabriel (2001).

"Expression and localization of epithelial aquaporins in the adult human lung." Am J Respir Cell

Mol Biol 24(3): 224-34.

Kumar, N. M. and N. B. Gilula (1996). "The gap junction communication channel." Cell 84(3):

381-8.

Kumar, V., A. Abbas and N. Fausto (2004). "Robbins & Cotran's Pathologic Basis of Disease."

Kurahashi, K., O. Kajikawa, T. Sawa, M. Ohara, M. A. Gropper, D. W. Frank, T. R. Martin and

J. P. Wiener-Kronish (1999). "Pathogenesis of septic shock in Pseudomonas aeruginosa

pneumonia." J Clin Invest 104(6): 743-50.

Laufe, M. D., R. H. Simon, A. Flint and J. B. Keller (1986). "Adult respiratory distress

syndrome in neutropenic patients." Am J Med 80(6): 1022-6.

Lee, P. J. and A. M. Choi (2003). "Pathways of cell signaling in hyperoxia." Free Radic Biol

Med 35(4): 341-50.

190

Lee, W. L. and G. P. Downey (2001). "Neutrophil activation and acute lung injury." Curr Opin

Crit Care 7(1): 1-7.

Lehmann, L. E., U. Novender, S. Schroeder, T. Pietsch, T. von Spiegel, C. Putensen, A. Hoeft

and F. Stuber (2001). "Plasma levels of macrophage migration inhibitory factor are elevated in

patients with severe sepsis." Intensive Care Med 27(8): 1412-5.

Lentsch, A. B., B. J. Czermak, N. M. Bless, N. Van Rooijen and P. A. Ward (1999). "Essential

role of alveolar macrophages in intrapulmonary activation of NF-kappaB." Am J Respir Cell

Mol Biol 20(4): 692-8.

Lewandowski, K., J. Metz, C. Deutschmann, H. Preiss, R. Kuhlen, A. Artigas and K. J. Falke

(1995). "Incidence, severity, and mortality of acute respiratory failure in Berlin, Germany." Am

J Respir Crit Care Med 151(4): 1121-5.

Lewis, J. F. and A. H. Jobe (1993). "Surfactant and the adult respiratory distress syndrome." Am

Rev Respir Dis 147(1): 218-33.

Li, L. F., B. Ouyang, G. Choukroun, R. Matyal, M. Mascarenhas, B. Jafari, J. V. Bonventre, T.

Force and D. A. Quinn (2003). "Stretch-induced IL-8 depends on c-Jun NH2-terminal and

nuclear factor-kappaB-inducing kinases." Am J Physiol Lung Cell Mol Physiol 285(2): L464-75.

Liggins, G. C., G. A. Vilos, G. A. Campos, J. A. Kitterman and C. H. Lee (1981). "The effect of

spinal cord transection on lung development in fetal sheep." J Dev Physiol 3(4): 267-74.

Lines, A., L. Nardo, I. D. Phillips, F. Possmayer and S. B. Hooper (1999). "Alterations in lung

expansion affect surfactant protein A, B, and C mRNA levels in fetal sheep." Am J Physiol

276(2 Pt 1): L239-45.

Low, F. N. (1952). "Electronmicroscopy of the rat lung." Anat Rec 113: 437-4443.

Luhr, O. R., K. Antonsen, M. Karlsson, S. Aardal, A. Thorsteinsson, C. G. Frostell and J. Bonde

(1999). "Incidence and mortality after acute respiratory failure and acute respiratory distress

syndrome in Sweden, Denmark, and Iceland. The ARF Study Group." Am J Respir Crit Care

Med 159(6): 1849-61.

Lwebuga-Mukasa, J. S. (1991). "Matrix-driven pneumocyte differentiation." Am Rev Respir Dis

144(2): 452-7.

Ma, T., N. Fukuda, Y. Song, M. A. Matthay and A. S. Verkman (2000). "Lung fluid transport in

aquaporin-5 knockout mice." J Clin Invest 105(1): 93-100.

Martin, T. R. (2002). "Neutrophils and lung injury: getting it right." J Clin Invest 110(11): 1603-

5.

Martinon, F. and J. Tschopp (2005). "NLRs join TLRs as innate sensors of pathogens." Trends

Immunol 26(8): 447-54.

191

Mason, R. J. and M. C. Williams (1991). "The Lung: Scientific Foundations edited by R. G.

Crystal, J. B. West et al." 235-246.

Massaro, G. D. and D. Massaro (1983). "Morphologic evidence that large inflations of the lung

stimulate secretion of surfactant." Am Rev Respir Dis 127(2): 235-6.

Matthay, M. A., H. G. Folkesson and C. Clerici (2002). "Lung epithelial fluid transport and the

resolution of pulmonary edema." Physiol Rev 82(3): 569-600.

Matthay, M. A., L. Robriquet and X. Fang (2005). "Alveolar epithelium: role in lung fluid

balance and acute lung injury." Proc Am Thorac Soc 2(3): 206-13.

Matthay, M. A. and J. P. Wiener-Kronish (1990). "Intact epithelial barrier function is critical for

the resolution of alveolar edema in humans." Am Rev Respir Dis 142(6 Pt 1): 1250-7.

Maunder, R. J., W. P. Shuman, J. W. McHugh, S. I. Marglin and J. Butler (1986). "Preservation

of normal lung regions in the adult respiratory distress syndrome. Analysis by computed

tomography." Jama 255(18): 2463-5.

McCullers, J. A. and E. I. Tuomanen (2001). "Molecular pathogenesis of pneumococcal

pneumonia." Front Biosci 6: D877-89.

McElroy, M. C., H. R. Harty, G. E. Hosford, G. M. Boylan, J. F. Pittet and T. J. Foster (1999).

"Alpha-toxin damages the air-blood barrier of the lung in a rat model of Staphylococcus aureus-

induced pneumonia." Infect Immun 67(10): 5541-4.

McElroy, M. C. and M. Kasper (2004). "The use of alveolar epithelial type I cell-selective

markers to investigate lung injury and repair." Eur Respir J 24(4): 664-73.

McElroy, M. C., J. F. Pittet, L. Allen, J. P. Wiener-Kronish and L. G. Dobbs (1997).

"Biochemical detection of type I cell damage after nitrogen dioxide-induced lung injury in rats."

Am J Physiol 273(6 Pt 1): L1228-34.

McElroy, M. C., J. F. Pittet, S. Hashimoto, L. Allen, J. P. Wiener-Kronish and L. G. Dobbs

(1995). "A type I cell-specific protein is a biochemical marker of epithelial injury in a rat model

of pneumonia." Am J Physiol 268(2 Pt 1): L181-6.

McEver, R. P., K. L. Moore and R. D. Cummings (1995). "Leukocyte trafficking mediated by

selectin-carbohydrate interactions." J Biol Chem 270(19): 11025-8.

McHugh, L. G., J. A. Milberg, M. E. Whitcomb, R. B. Schoene, R. J. Maunder and L. D.

Hudson (1994). "Recovery of function in survivors of the acute respiratory distress syndrome."

Am J Respir Crit Care Med 150(1): 90-4.

Mead, J., T. Takishima and D. Leith (1970). "Stress distribution in lungs: a model of pulmonary

elasticity." J Appl Physiol 28(5): 596-608.

Mercer, R. R. and J. D. Crapo (1990). "Spatial distribution of collagen and elastin fibers in the

lungs." J Appl Physiol 69(2): 756-65.

192

Mercer, R. R., J. M. Laco and J. D. Crapo (1987). "Three-dimensional reconstruction of alveoli

in the rat lung for pressure-volume relationships." J Appl Physiol 62(4): 1480-7.

Michel, R. P., M. Laforte and J. C. Hogg (1981). "Physiology and morphology of pulmonary

microvascular injury with shock and reinfusion." J Appl Physiol 50(6): 1227-35.

Michiels, C., E. Minet, D. Mottet and M. Raes (2002). "Regulation of gene expression by

oxygen: NF-kappaB and HIF-1, two extremes." Free Radic Biol Med 33(9): 1231-42.

Modelska, K., M. A. Matthay, M. C. McElroy and J. F. Pittet (1997). "Upregulation of alveolar

liquid clearance after fluid resuscitation for hemorrhagic shock in rats." Am J Physiol 273(2 Pt

1): L305-14.

Montgomery, A. B., M. A. Stager, C. J. Carrico and L. D. Hudson (1985). "Causes of mortality

in patients with the adult respiratory distress syndrome." Am Rev Respir Dis 132(3): 485-9.

Moore, F. A. and E. E. Moore (1995). "Evolving concepts in the pathogenesis of postinjury

multiple organ failure." Surg Clin North Am 75(2): 257-77.

Nahum, A., J. Hoyt, L. Schmitz, J. Moody, R. Shapiro and J. J. Marini (1997). "Effect of

mechanical ventilation strategy on dissemination of intratracheally instilled Escherichia coli in

dogs." Crit Care Med 25(10): 1733-43.

Narasaraju, T. A., H. Chen, T. Weng, M. Bhaskaran, N. Jin, J. Chen, Z. Chen, M. R. Chinoy and

L. Liu (2006). "Expression profile of IGF system during lung injury and recovery in rats exposed

to hyperoxia: a possible role of IGF-1 in alveolar epithelial cell proliferation and differentiation."

J Cell Biochem 97(5): 984-98.

Neff, S. B., R. Z'Graggen B, T. A. Neff, M. Jamnicki-Abegg, D. Suter, R. C. Schimmer, C.

Booy, H. Joch, T. Pasch, P. A. Ward and B. Beck-Schimmer (2006). "Inflammatory response of

tracheobronchial epithelial cells to endotoxin." Am J Physiol Lung Cell Mol Physiol 290(1):

L86-96.

Nelson, S., S. M. Belknap, R. W. Carlson, D. Dale, B. DeBoisblanc, S. Farkas, N.

Fotheringham, H. Ho, T. Marrie, H. Movahhed, R. Root and J. Wilson (1998). "A randomized

controlled trial of filgrastim as an adjunct to antibiotics for treatment of hospitalized patients

with community-acquired pneumonia. CAP Study Group." J Infect Dis 178(4): 1075-80.

Newman, V., R. F. Gonzalez, M. A. Matthay and L. G. Dobbs (2000). "A novel alveolar type I

cell-specific biochemical marker of human acute lung injury." Am J Respir Crit Care Med 161(3

Pt 1): 990-5.

Nicholas, T. E., J. H. Power and H. A. Barr (1982). "Surfactant homeostasis in the rat lung

during swimming exercise." J Appl Physiol 53(6): 1521-8.

Nicholas, T. E., J. H. Power and H. A. Barr (1982). "The pulmonary consequences of a deep

breath." Respir Physiol 49(3): 315-24.

193

Nielsen, S., L. S. King, B. M. Christensen and P. Agre (1997). "Aquaporins in complex tissues.

II. Subcellular distribution in respiratory and glandular tissues of rat." Am J Physiol 273(5 Pt 1):

C1549-61.

NIH (1977). "Conference report: Mechanisms of acute respiratory failure." Am Rev Respir Dis

115(6): 1071-8.

Normand, I. C., R. E. Olver, E. O. Reynolds and L. B. Strang (1971). "Permeability of lung

capillaries and alveoli to non-electrolytes in the foetal lamb." J Physiol 219(2): 303-30.

O'Donovan, D. J. and C. J. Fernandes (2004). "Free radicals and diseases in premature infants."

Antioxid Redox Signal 6(1): 169-76.

O'Reilly, M. A., R. J. Staversky, B. R. Stripp and J. N. Finkelstein (1998). "Exposure to

hyperoxia induces p53 expression in mouse lung epithelium." Am J Respir Cell Mol Biol 18(1):

43-50.

O'Reilly, M. A., R. J. Staversky, R. H. Watkins and W. M. Maniscalco (1998). "Accumulation

of p21(Cip1/WAF1) during hyperoxic lung injury in mice." Am J Respir Cell Mol Biol 19(5):

777-85.

Ohwada, A., Y. Tsutsumi-Ishii, Y. Yoshioka, K. Iwabuchi, I. Nagaoka and Y. Fukuchi (2003).

"Acid exposure potentiates intercellular adhesion molecule-1 and e-cadherin expression on A549

alveolar lining epithelial cells." Exp Lung Res 29(6): 389-400.

Olsen, C. O., B. E. Isakson, G. J. Seedorf, R. L. Lubman and S. Boitano (2005). "Extracellular

matrix-driven alveolar epithelial cell differentiation in vitro." Exp Lung Res 31(5): 461-82.

Otto, W. R. (2002). "Lung epithelial stem cells." J Pathol 197(4): 527-35.

Paine, R., 3rd and R. H. Simon (1996). "Expanding the frontiers of lung biology through the

creative use of alveolar epithelial cells in culture." Am J Physiol 270(4 Pt 1): L484-6.

Panos, R. J., P. M. Bak, W. S. Simonet, J. S. Rubin and L. J. Smith (1995). "Intratracheal

instillation of keratinocyte growth factor decreases hyperoxia-induced mortality in rats." J Clin

Invest 96(4): 2026-33.

Park, W. Y., R. B. Goodman, K. P. Steinberg, J. T. Ruzinski, F. Radella, 2nd, D. R. Park, J.

Pugin, S. J. Skerrett, L. D. Hudson and T. R. Martin (2001). "Cytokine balance in the lungs of

patients with acute respiratory distress syndrome." Am J Respir Crit Care Med 164(10 Pt 1):

1896-903.

Parker, J. C., L. A. Hernandez and K. J. Peevy (1993). "Mechanisms of ventilator-induced lung

injury." Crit Care Med 21(1): 131-43.

Parker, J. C. and M. I. Townsley (2004). "Evaluation of lung injury in rats and mice." Am J

Physiol Lung Cell Mol Physiol 286(2): L231-46.

194

Pelosi, P., D. D'Onofrio, D. Chiumello, S. Paolo, G. Chiara, V. L. Capelozzi, C. S. Barbas, M.

Chiaranda and L. Gattinoni (2003). "Pulmonary and extrapulmonary acute respiratory distress

syndrome are different." Eur Respir J Suppl 42: 48s-56s.

Petty, T. L. and D. G. Ashbaugh (1971). "The adult respiratory distress syndrome. Clinical

features, factors influencing prognosis and principles of management." Chest 60(3): 233-9.

Pison, U., U. Obertacke, W. Seeger and S. Hawgood (1992). "Surfactant protein A (SP-A) is

decreased in acute parenchymal lung injury associated with polytrauma." Eur J Clin Invest

22(11): 712-8.

Pittet, J. F., T. J. Brenner, K. Modelska and M. A. Matthay (1996). "Alveolar liquid clearance is

increased by endogenous catecholamines in hemorrhagic shock in rats." J Appl Physiol 81(2):

830-7.

Pittet, J. F., S. Hashimoto, M. Pian, M. C. McElroy, G. Nitenberg and J. P. Wiener-Kronish

(1996). "Exotoxin A stimulates fluid reabsorption from distal airspaces of lung in anesthetized

rats." Am J Physiol 270(2 Pt 1): L232-41.

Pittet, J. F., R. C. Mackersie, T. R. Martin and M. A. Matthay (1997). "Biological markers of

acute lung injury: prognostic and pathogenetic significance." Am J Respir Crit Care Med 155(4):

1187-205.

Pugin, J., I. Dunn, P. Jolliet, D. Tassaux, J. L. Magnenat, L. P. Nicod and J. C. Chevrolet (1998).

"Activation of human macrophages by mechanical ventilation in vitro." Am J Physiol 275(6 Pt

1): L1040-50.

Pugin, J., G. Verghese, M. C. Widmer and M. A. Matthay (1999). "The alveolar space is the site

of intense inflammatory and profibrotic reactions in the early phase of acute respiratory distress

syndrome." Crit Care Med 27(2): 304-12.

Ramirez, M. I., G. Millien, A. Hinds, Y. Cao, D. C. Seldin and M. C. Williams (2003).

"T1alpha, a lung type I cell differentiation gene, is required for normal lung cell proliferation

and alveolus formation at birth." Dev Biol 256(1): 61-72.

Ranieri, V. M., P. M. Suter, C. Tortorella, R. De Tullio, J. M. Dayer, A. Brienza, F. Bruno and

A. S. Slutsky (1999). "Effect of mechanical ventilation on inflammatory mediators in patients

with acute respiratory distress syndrome: a randomized controlled trial." Jama 282(1): 54-61.

Rannels, D. E. and S. R. Rannels (1989). "Influence of the extracellular matrix on type 2 cell

differentiation." Chest 96(1): 165-73.

Reddy, R., S. Buckley, M. Doerken, L. Barsky, K. Weinberg, K. D. Anderson, D. Warburton

and B. Driscoll (2004). "Isolation of a putative progenitor subpopulation of alveolar epithelial

type 2 cells." Am J Physiol Lung Cell Mol Physiol 286(4): L658-67.

Reynolds, S. D., A. Giangreco, K. U. Hong, K. E. McGrath, L. A. Ortiz and B. R. Stripp (2004).

"Airway injury in lung disease pathophysiology: selective depletion of airway stem and

195

progenitor cell pools potentiates lung inflammation and alveolar dysfunction." Am J Physiol

Lung Cell Mol Physiol 287(6): L1256-65.

Riley, D. J., D. E. Rannels, R. B. Low, L. Jensen and T. P. Jacobs (1990). "NHLBI Workshop

Summary. Effect of physical forces on lung structure, function, and metabolism." Am Rev

Respir Dis 142(4): 910-4.

Rouby, J. J., L. Puybasset, A. Nieszkowska and Q. Lu (2003). "Acute respiratory distress

syndrome: lessons from computed tomography of the whole lung." Crit Care Med 31(4 Suppl):

S285-95.

Rubenfeld, G. D., E. Caldwell, E. Peabody, J. Weaver, D. P. Martin, M. Neff, E. J. Stern and L.

D. Hudson (2005). "Incidence and outcomes of acute lung injury." N Engl J Med 353(16): 1685-

93.

Rubins, J. B., P. G. Duane, D. Clawson, D. Charboneau, J. Young and D. E. Niewoehner (1993).

"Toxicity of pneumolysin to pulmonary alveolar epithelial cells." Infect Immun 61(4): 1352-8.

Rush, B. F., Jr., A. J. Sori, T. F. Murphy, S. Smith, J. J. Flanagan, Jr. and G. W. Machiedo

(1988). "Endotoxemia and bacteremia during hemorrhagic shock. The link between trauma and

sepsis?" Ann Surg 207(5): 549-54.

Sadikot, R. T., W. Han, M. B. Everhart, O. Zoia, R. S. Peebles, E. D. Jansen, F. E. Yull, J. W.

Christman and T. S. Blackwell (2003). "Selective I kappa B kinase expression in airway

epithelium generates neutrophilic lung inflammation." J Immunol 170(2): 1091-8.

Saitou, M., Y. Ando-Akatsuka, M. Itoh, M. Furuse, J. Inazawa, K. Fujimoto and S. Tsukita

(1997). "Mammalian occludin in epithelial cells: its expression and subcellular distribution." Eur

J Cell Biol 73(3): 222-31.

Sannes, P. L. (1984). "Differences in basement membrane-associated microdomains of type I

and type II pneumocytes in the rat and rabbit lung." J Histochem Cytochem 32(8): 827-33.

Schacht, V., M. I. Ramirez, Y. K. Hong, S. Hirakawa, D. Feng, N. Harvey, M. Williams, A. M.

Dvorak, H. F. Dvorak, G. Oliver and M. Detmar (2003). "T1alpha/podoplanin deficiency

disrupts normal lymphatic vasculature formation and causes lymphedema." Embo J 22(14):

3546-56.

Schittny, J. C., V. Djonov, A. Fine and P. H. Burri (1998). "Programmed cell death contributes

to postnatal lung development." Am J Respir Cell Mol Biol 18(6): 786-93.

Schneeberger, E. E. (1991). "The Lung: Scientific Foundations edited by R. G. Crystal, J. B.

West et al." 229-234.

Schneeberger, E. E. (1991). "The Lung: Scientific Foundations edited by R. G. Crystal, J. B.

West et al." 205-214.

Schneeberger, E. E. and R. D. Lynch (1992). "Structure, function, and regulation of cellular tight

junctions." Am J Physiol 262(6 Pt 1): L647-61.

196

Schneeberger, E. E. and K. M. McCarthy (1986). "Cytochemical localization of Na+-K+-

ATPase in rat type II pneumocytes." J Appl Physiol 60(5): 1584-9.

Scholl, F. G., C. Gamallo, S. Vilar inverted question marko and M. Quintanilla (1999).

"Identification of PA2.26 antigen as a novel cell-surface mucin-type glycoprotein that induces

plasma membrane extensions and increased motility in keratinocytes." J Cell Sci 112 ( Pt 24):

4601-13.

Seeger, W., R. G. Birkemeyer, L. Ermert, N. Suttorp, S. Bhakdi and H. R. Duncker (1990).

"Staphylococcal alpha-toxin-induced vascular leakage in isolated perfused rabbit lungs." Lab

Invest 63(3): 341-9.

Selman, M. and A. Pardo (2002). "Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-

talk disorder." Respir Res 3: 3.

Selman, M. and A. Pardo (2006). "Role of epithelial cells in idiopathic pulmonary fibrosis: from

innocent targets to serial killers." Proc Am Thorac Soc 3(4): 364-72.

Selman, M., V. J. Thannickal, A. Pardo, D. A. Zisman, F. J. Martinez and J. P. Lynch, 3rd

(2004). "Idiopathic pulmonary fibrosis: pathogenesis and therapeutic approaches." Drugs 64(4):

405-30.

Shenkar, R. and E. Abraham (1993). "Effects of hemorrhage on cytokine gene transcription."

Lymphokine Cytokine Res 12(4): 237-47.

Shenkar, R., M. D. Schwartz, L. S. Terada, J. E. Repine, J. McCord and E. Abraham (1996).

"Hemorrhage activates NF-kappa B in murine lung mononuclear cells in vivo." Am J Physiol

270(5 Pt 1): L729-35.

Simionescu, M. (1997). "The Lung: Scientific Foundations Edited by R.G Crystal, J.B West et

al." (Second Edition): 615-628.

Sirianni, F. E., F. S. Chu and D. C. Walker (2003). "Human alveolar wall fibroblasts directly

link epithelial type 2 cells to capillary endothelium." Am J Respir Crit Care Med 168(12): 1532-

7.

Slutsky, A. S. (1999). "Lung injury caused by mechanical ventilation." Chest 116(1 Suppl): 9S-

15S.

Slutsky, A. S. and L. N. Tremblay (1998). "Multiple system organ failure. Is mechanical

ventilation a contributing factor?" Am J Respir Crit Care Med 157(6 Pt 1): 1721-5.

Song, Y., N. Fukuda, C. Bai, T. Ma, M. A. Matthay and A. S. Verkman (2000). "Role of

aquaporins in alveolar fluid clearance in neonatal and adult lung, and in oedema formation

following acute lung injury: studies in transgenic aquaporin null mice." J Physiol 525 Pt 3: 771-

9.

Sori, A. J., B. F. Rush, Jr., T. W. Lysz, S. Smith and G. W. Machiedo (1988). "The gut as source

of sepsis after hemorrhagic shock." Am J Surg 155(2): 187-92.

197

Spellerberg, B., C. Rosenow, W. Sha and E. I. Tuomanen (1996). "Pneumococcal cell wall

activates NF-kappa B in human monocytes: aspects distinct from endotoxin." Microb Pathog

20(5): 309-17.

Springer, T. A. (1995). "Traffic signals on endothelium for lymphocyte recirculation and

leukocyte emigration." Annu Rev Physiol 57: 827-72.

Stone, K. C., R. R. Mercer, P. Gehr, B. Stockstill and J. D. Crapo (1992). "Allometric

relationships of cell numbers and size in the mammalian lung." Am J Respir Cell Mol Biol 6(2):

235-43.

Sue, R. D., J. A. Belperio, M. D. Burdick, L. A. Murray, Y. Y. Xue, M. C. Dy, J. J. Kwon, M. P.

Keane and R. M. Strieter (2004). "CXCR2 is critical to hyperoxia-induced lung injury." J

Immunol 172(6): 3860-8.

Sugahara, K., T. Kiyota, R. A. Clark and R. J. Mason (1993). "The effect of fibronectin on

cytoskeleton structure and transepithelial resistance of alveolar type II cells in primary culture."

Virchows Arch B Cell Pathol Incl Mol Pathol 64(2): 115-22.

Suratt, B. T., C. D. Cool, A. E. Serls, L. Chen, M. Varella-Garcia, E. J. Shpall, K. K. Brown and

G. S. Worthen (2003). "Human pulmonary chimerism after hematopoietic stem cell

transplantation." Am J Respir Crit Care Med 168(3): 318-22.

Suzuki, Y. J., H. J. Forman and A. Sevanian (1997). "Oxidants as stimulators of signal

transduction." Free Radic Biol Med 22(1-2): 269-85.

Takahashi, T., M. Munakata, I. Suzuki and Y. Kawakami (1998). "Serum and bronchoalveolar

fluid KL-6 levels in patients with pulmonary alveolar proteinosis." Am J Respir Crit Care Med

158(4): 1294-8.

Tani, T., M. Fujino, K. Hanasawa, T. Shimizu, Y. Endo and M. Kodama (2000). "Bacterial

translocation and tumor necrosis factor-alpha gene expression in experimental hemorrhagic

shock." Crit Care Med 28(11): 3705-9.

Taylor, A. E. and K. A. Gaar, Jr. (1970). "Estimation of equivalent pore radii of pulmonary

capillary and alveolar membranes." Am J Physiol 218(4): 1133-40.

Thannickal, V. J. and B. L. Fanburg (2000). "Reactive oxygen species in cell signaling." Am J

Physiol Lung Cell Mol Physiol 279(6): L1005-28.

Thet, L. A., S. C. Parra and J. D. Shelburne (1986). "Sequential changes in lung morphology

during the repair of acute oxygen-induced lung injury in adult rats." Exp Lung Res 11(3): 209-

28.

Todd, T. R., E. Baile and J. C. Hogg (1978). "Pulmonary capillary and permeability during

hemorrhagic shock." J Appl Physiol 45(2): 298-306.

198

Tomashefski, J. F. J. (2003). Pulmonary Pathology of the Acute Respiratory Distress Syndrome

- Diffuse Alveolar Damage. Acute Respiratory Distress Syndrome. M. A. Matthay. New York,

Marcel Dekker. 179: 75-114.

Torikata, C., B. Villiger, C. Kuhn, 3rd and J. A. McDonald (1985). "Ultrastructural distribution

of fibronectin in normal and fibrotic human lung." Lab Invest 52(4): 399-408.

Tosi, M. F., J. M. Stark, C. W. Smith, A. Hamedani, D. C. Gruenert and M. D. Infeld (1992).

"Induction of ICAM-1 expression on human airway epithelial cells by inflammatory cytokines:

effects on neutrophil-epithelial cell adhesion." Am J Respir Cell Mol Biol 7(2): 214-21.

Towne, J. E., K. S. Harrod, C. M. Krane and A. G. Menon (2000). "Decreased expression of

aquaporin (AQP)1 and AQP5 in mouse lung after acute viral infection." Am J Respir Cell Mol

Biol 22(1): 34-44.

Tremblay, L., F. Valenza, S. P. Ribeiro, J. Li and A. S. Slutsky (1997). "Injurious ventilatory

strategies increase cytokines and c-fos m-RNA expression in an isolated rat lung model." J Clin

Invest 99(5): 944-52.

Tremblay, L. N., D. Miatto, Q. Hamid, A. Govindarajan and A. S. Slutsky (2002). "Injurious

ventilation induces widespread pulmonary epithelial expression of tumor necrosis factor-alpha

and interleukin-6 messenger RNA." Crit Care Med 30(8): 1693-700.

Tremblay, L. N. and A. S. Slutsky (1998). "Ventilator-induced injury: from barotrauma to

biotrauma." Proc Assoc Am Physicians 110(6): 482-8.

Tschumperlin, D. J. and S. S. Margulies (1998). "Equibiaxial deformation-induced injury of

alveolar epithelial cells in vitro." Am J Physiol 275(6 Pt 1): L1173-83.

Tschumperlin, D. J. and S. S. Margulies (1999). "Alveolar epithelial surface area-volume

relationship in isolated rat lungs." J Appl Physiol 86(6): 2026-33.

Tschumperlin, D. J., J. Oswari and A. S. Margulies (2000). "Deformation-induced injury of

alveolar epithelial cells. Effect of frequency, duration, and amplitude." Am J Respir Crit Care

Med 162(2 Pt 1): 357-62.

Turrens, J. F. (2003). "Mitochondrial formation of reactive oxygen species." J Physiol 552(Pt 2):

335-44.

Tutor, J. D., C. M. Mason, E. Dobard, R. C. Beckerman, W. R. Summer and S. Nelson (1994).

"Loss of compartmentalization of alveolar tumor necrosis factor after lung injury." Am J Respir

Crit Care Med 149(5): 1107-11.

Uhal, B. D. (1997). "Cell cycle kinetics in the alveolar epithelium." Am J Physiol 272(6 Pt 1):

L1031-45.

Uhlig, S. (2002). "Ventilation-induced lung injury and mechanotransduction: stretching it too

far?" Am J Physiol Lung Cell Mol Physiol 282(5): L892-6.

199

Ulich, T. R., E. S. Yi, K. Longmuir, S. Yin, R. Biltz, C. F. Morris, R. M. Housley and G. F.

Pierce (1994). "Keratinocyte growth factor is a growth factor for type II pneumocytes in vivo." J

Clin Invest 93(3): 1298-306.

Vaccaro, C. A. and J. S. Brody (1981). "Structural features of alveolar wall basement membrane

in the adult rat lung." J Cell Biol 91(2 Pt 1): 427-37.

Veldhuizen, R. A., A. S. Slutsky, M. Joseph and L. McCaig (2001). "Effects of mechanical

ventilation of isolated mouse lungs on surfactant and inflammatory cytokines." Eur Respir J

17(3): 488-94.

Verkman, A. S., M. A. Matthay and Y. Song (2000). "Aquaporin water channels and lung

physiology." Am J Physiol Lung Cell Mol Physiol 278(5): L867-79.

Vlahakis, N. E. and R. D. Hubmayr (2000). "Invited review: plasma membrane stress failure in

alveolar epithelial cells." J Appl Physiol 89(6): 2490-6;discussion 2497.

Vlahakis, N. E., M. A. Schroeder, A. H. Limper and R. D. Hubmayr (1999). "Stretch induces

cytokine release by alveolar epithelial cells in vitro." Am J Physiol 277(1 Pt 1): L167-73.

von Bethmann, A. N., F. Brasch, R. Nusing, K. Vogt, H. D. Volk, K. M. Muller, A. Wendel and

S. Uhlig (1998). "Hyperventilation induces release of cytokines from perfused mouse lung." Am

J Respir Crit Care Med 157(1): 263-72.

Vuichard, D., M. T. Ganter, R. C. Schimmer, D. Suter, C. Booy, L. Reyes, T. Pasch and B.

Beck-Schimmer (2005). "Hypoxia aggravates lipopolysaccharide-induced lung injury." Clin Exp

Immunol 141(2): 248-60.

Wang, F., B. Daugherty, L. L. Keise, Z. Wei, J. P. Foley, R. C. Savani and M. Koval (2003).

"Heterogeneity of claudin expression by alveolar epithelial cells." Am J Respir Cell Mol Biol

29(1): 62-70.

Wang, R., O. Ibarra-Sunga, L. Verlinski, R. Pick and B. D. Uhal (2000). "Abrogation of

bleomycin-induced epithelial apoptosis and lung fibrosis by captopril or by a caspase inhibitor."

Am J Physiol Lung Cell Mol Physiol 279(1): L143-51.

Wang, X., S. W. Ryter, C. Dai, Z. L. Tang, S. C. Watkins, X. M. Yin, R. Song and A. M. Choi

(2003). "Necrotic cell death in response to oxidant stress involves the activation of the

apoptogenic caspase-8/bid pathway." J Biol Chem 278(31): 29184-91.

Ward, N. S., A. B. Waxman, R. J. Homer, L. L. Mantell, O. Einarsson, Y. Du and J. A. Elias

(2000). "Interleukin-6-induced protection in hyperoxic acute lung injury." Am J Respir Cell Mol

Biol 22(5): 535-42.

Ware, L. B. and M. A. Matthay (2000). "The acute respiratory distress syndrome." N Engl J Med

342(18): 1334-49.

200

Ware, L. B. and M. A. Matthay (2001). "Alveolar fluid clearance is impaired in the majority of

patients with acute lung injury and the acute respiratory distress syndrome." Am J Respir Crit

Care Med 163(6): 1376-83.

Waters, C. M., P. H. Sporn, M. Liu and J. J. Fredberg (2002). "Cellular biomechanics in the

lung." Am J Physiol Lung Cell Mol Physiol 283(3): L503-9.

Weatherall, D., E. J. Benz, D. A. Warrell, T. M. Cox and F. J. D (2003). "Oxford Textbook of

Medicine."

Webb, H. H. and D. F. Tierney (1974). "Experimental pulmonary edema due to intermittent

positive pressure ventilation with high inflation pressures. Protection by positive end-expiratory

pressure." Am Rev Respir Dis 110(5): 556-65.

Webster, N. R., A. T. Cohen and J. F. Nunn (1988). "Adult respiratory distress syndrome--how

many cases in the UK?" Anaesthesia 43(11): 923-6.

Weibel, E. R. (1984). The pathway for oxygen. Cambridge, Massachusetts and London, Harvard

University Press.

Weibel, E. R. and D. M. Gomez (1962). "Architecture of the human lung. Use of quantitative

methods establishes fundamental relations between size and number of lung structures." Science

137: 577-85.

Weinert, C. R., C. R. Gross, J. R. Kangas, C. L. Bury and W. A. Marinelli (1997). "Health-

related quality of life after acute lung injury." Am J Respir Crit Care Med 156(4 Pt 1): 1120-8.

West, J. B. (2004). "Repiratory Physiology - the essentials."

White, S. R., D. R. Dorscheid, K. F. Rabe, K. R. Wojcik and K. J. Hamann (1999). "Role of very

late adhesion integrins in mediating repair of human airway epithelial cell monolayers after

mechanical injury." Am J Respir Cell Mol Biol 20(4): 787-96.

Wiener-Kronish, J. P., K. H. Albertine and M. A. Matthay (1991). "Differential responses of the

endothelial and epithelial barriers of the lung in sheep to Escherichia coli endotoxin." J Clin

Invest 88(3): 864-75.

Williams, M. C. (2003). "Alveolar type I cells: molecular phenotype and development." Annu

Rev Physiol 65: 669-95.

Williams, M. C., Y. Cao, A. Hinds, A. K. Rishi and A. Wetterwald (1996). "T1 alpha protein is

developmentally regulated and expressed by alveolar type I cells, choroid plexus, and ciliary

epithelia of adult rats." Am J Respir Cell Mol Biol 14(6): 577-85.

Williams, M. C. and L. G. Dobbs (1990). "Expression of cell-specific markers for alveolar

epithelium in fetal rat lung." Am J Respir Cell Mol Biol 2(6): 533-42.

Winkelstein, J. A. and A. Tomasz (1978). "Activation of the alternative complement pathway by

pneumococcal cell wall teichoic acid." J Immunol 120(1): 174-8.

201

Wirtz, H. R. and L. G. Dobbs (1990). "Calcium mobilization and exocytosis after one

mechanical stretch of lung epithelial cells." Science 250(4985): 1266-9.

Wirtz, H. R. and L. G. Dobbs (2000). "The effects of mechanical forces on lung functions."

Respir Physiol 119(1): 1-17.

Woodhead, M. A., J. T. Macfarlane, J. S. McCracken, D. H. Rose and R. G. Finch (1987).

"Prospective study of the aetiology and outcome of pneumonia in the community." Lancet

1(8534): 671-4.

Woodhead, M. A., J. T. Macfarlane, F. G. Rodgers, A. Laverick, R. Pilkington and A. D. Macrae

(1985). "Aetiology and outcome of severe community-acquired pneumonia." J Infect 10(3): 204-

10.

Wyncoll, D. L. and T. W. Evans (1999). "Acute respiratory distress syndrome." Lancet

354(9177): 497-501.

Yamada, M., H. Kubo, S. Kobayashi, K. Ishizawa, M. Numasaki, S. Ueda, T. Suzuki and H.

Sasaki (2004). "Bone marrow-derived progenitor cells are important for lung repair after

lipopolysaccharide-induced lung injury." J Immunol 172(2): 1266-72.

Yee, M., P. F. Vitiello, J. M. Roper, R. J. Staversky, T. W. Wright, S. A. McGrath-Morrow, W.

M. Maniscalco, J. N. Finkelstein and M. A. O'Reilly (2006). "Type II epithelial cells are critical

target for hyperoxia-mediated impairment of postnatal lung development." Am J Physiol Lung

Cell Mol Physiol 291(5): L1101-11.

Zhang, H., G. P. Downey, P. M. Suter, A. S. Slutsky and V. M. Ranieri (2002). "Conventional

mechanical ventilation is associated with bronchoalveolar lavage-induced activation of

polymorphonuclear leukocytes: a possible mechanism to explain the systemic consequences of

ventilator-induced lung injury in patients with ARDS." Anesthesiology 97(6): 1426-33.

Zhang, Y., R. M. Porat, T. Alon, E. Keshet and J. Stone (1999). "Tissue oxygen levels control

astrocyte movement and differentiation in developing retina." Brain Res Dev Brain Res 118(1-

2): 135-45.

Zimmerman, G. A., K. H. Albertine, H. J. Carveth, E. A. Gill, C. K. Grissom, J. R. Hoidal, T.

Imaizumi, C. G. Maloney, T. M. McIntyre, J. R. Michael, J. F. Orme, S. M. Prescott and M. S.

Topham (1999). "Endothelial activation in ARDS." Chest 116(1 Suppl): 18S-24S.