Top Banner
Lymphocyte Responses in the Lung in Patients with Respiratory Disease Thesis presented for the degree of Doctor of Philosophy at the university of London Dr Simon Barry MBBS, BSc, MRCP, DTMH November 2002
189

Lymphocyte Responses in the Lung in Patients with ...

May 17, 2023

Download

Documents

Khang Minh
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Lymphocyte Responses in the Lung in Patients with ...

Lymphocyte Responses in the

Lung in Patients with Respiratory

Disease

Thesis presented for the degree of Doctor of Philosophy at the university of London

Dr Simon Barry

MBBS, BSc, MRCP, DTMH

November 2002

Page 2: Lymphocyte Responses in the Lung in Patients with ...

ProQuest Number: 10016089

All rights reserved

INFORMATION TO ALL USERS The quality of this reproduction is dependent upon the quality of the copy submitted.

In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if material had to be removed,

a note will indicate the deletion.

uest.

ProQuest 10016089

Published by ProQuest LLC(2016). Copyright of the Dissertation is held by the Author.

All rights reserved.This work is protected against unauthorized copying under Title 17, United States Code.

Microform Edition © ProQuest LLC.

ProQuest LLC 789 East Eisenhower Parkway

P.O. Box 1346 Ann Arbor, Ml 48106-1346

Page 3: Lymphocyte Responses in the Lung in Patients with ...

Declaration

This work undertaken in this thesis has been undertaken solely by the candidate, Dr

Simon Barry.

This thesis has not been submitted or accepted in any previous application for a

degree.

Sources of information have been acknowledged in the text.

Page 4: Lymphocyte Responses in the Lung in Patients with ...

Acknowledgements

This thesis would not have been possible without the considerable help and careful

guidance of the following people.

• Richard Tilling who patiently taught me flow cytometry and encouraged my

interest in immunology with stimulating discussions.

• Margaret Johnson who supported me for longer than she should have done and I

who can only thank for her faith in me. I hope that the persistent watering finally

resulted in a satisfactory bloom!

• To all the consultants in respiratory medicine and infectious disease who helped

my quest for patients with tuberculosis

• To the patients themselves, some of whom undenwent bronchoscopy despite

knowing that they had tuberculosis and especially the two patients who

volunteered for a repeat bronchoscopy following therapy.

• To llesh Jani with whom I shared many ideas, but mostly because he helped me

out of my curmudgeonly state with humour.

• To Sandra, Nick, Helen and Arabi for their help and humour also.

• To Len Poulter for his support and comments on my thesis.

• Most of all, my gratitude goes to my supervisor, George Janossy. He was always

entertaining, sometimes obscure, but I learnt that to listen and to question were

well rewarded. His breadth and depth of knowledge, together with a tremendous

ability to see the direction in which immunology was heading were invaluable in

stimulating my interest in the subject. He was always encouraging and his critical

analysis of my papers helped me to grow during this time. Without his direction

and support I am sure that I could not have completed this work.

Dedication

This thesis is dedicated to my father, who died in a tragic accident soon after I started

working and from whom I learnt the values of enthusiasm and application and with whom

I would have loved to have shared the joys of finishing this project.

Page 5: Lymphocyte Responses in the Lung in Patients with ...

AbstractThe initial promise generated by earlier studies of bronchoalveolar lavage (BAL) in the

differential diagnosis of lung disease has generally failed to be translated to routine

clinical practice. The reasons for this delay stem largely from the fact that the cytospin

techniques used to differentiate BAL leukocyte subpopulations are cumbersome, time-

consuming and imprecise. Nevertheless, flow cytometry (FCM) offers an alternative

technology that is rapid, precise and well suited to document complex changes in cellular

phenotype in fresh and cultured specimens.

The patients included in this thesis were all investigated for suspected respiratory

disease and FCM was undertaken in addition to routine diagnostic tests on the BAL

specimens. Three key findings were observed. First, a simple single four-colour panel

has been developed that enables the rapid enumeration of the major clinically relevant

leukocyte components in BAL, including the CD4/CD8 lymphocyte ratio. This technology

is shown to be superior to cytospin techniques in terms of precision and speed, and

should be adopted for routine clinical investigation.

Second, it has been demonstrated that the lung is a distinct immunological site when

compared to the blood. CD8 T lymphocytes have been investigated using the

discriminatory markers, CD27 and CD45RA, and it has been shown that there is a

preferential accumulation of mature memory CD8 cells in the lung.

Lastly, the differences between the lung and the blood have been further evaluated

by analysing antigen-specific responses in patients with tuberculosis. It has been shown

that powerful CD4 interferon-y and tumour necrosis factor-a synthetic responses to short

term incubation with purified protein derivative (PPD) in BAL, but not blood, can be used

for the rapid diagnosis of acute tuberculosis. This test is a candidate for routine clinical

application, particularly because patients with extra-pulmonary tuberculosis also

respond.

Most importantly, this thesis has demonstrated that the focused investigation of BAL

using a powerful tool such as FCM can deliver important immunological information with

direct clinical relevance. It therefore highlights the vital link between medicine and

laboratory services in order to define optimal diagnostic technologies on the basis of

modern research.

Page 6: Lymphocyte Responses in the Lung in Patients with ...

Publications arising from this thesis

Papers

1) Barry SM and Janossy G. Optimal gating strategies for determining

bronchoalveolar lavage CD4/CD8 lymphocyte ratios by flow cytometry. J Imm

Methods 2003 in press

2) Barry SM, Johnson MA and Janossy G. Increased proportions of activated

and proliferating memory CD8+ T lymphocytes in both lung and blodd are

associated with blood HIV viral load. JAIDS 2003 34(4): 351-7

3) Barry SM. The utility of bronchoalveolar lavage evaluation in patients with

respiratory disease. CPD Bulletin Immunology and Allergy. 2003, 3: 8-10

4) Barry SM, Lipman MCI, Bannister B, Johnson MA and Janossy G. Type-1

cytokine synthesising CD4 lymphocytes in the lung are a characteristic sign

of pulmonary and non-pulmonary tuberculosis. J Infect Dis 2002,187: 243-

50.

5) Barry SM, Condez A, Deery A, Johnson MA and Janossy G. Determination

of Bronchalveolar lavage Leukocyte Populations by Flow Cytometry in

Patients Investigated for Respiratory Disease. Clinical Cytometry 2002, 50:

291-297.

6) Barry SM, Lipman MCI, Deery AR, Johnson MA and Janossy G. Immune

reconstitution pneumonitis following Pneumocystis carinii pneumonia in HIV-

infected subjects. HIV Medicine 2002, 3: 207-211

7) Barry SM, Johnson MA and Janossy G. Cytopathology or immunopathology?

The puzzle of cytomegalovirus pneumonitis revisited. . Bone Marrow Transpl.

2002, 26: 591-598

8) Barry SM, Lipman MCI, Johnson MA and Prentice HG. Respiratory

infections in immunocompromised patients. CurrOpin Pulm Med 1999, 5:

168-173

Page 7: Lymphocyte Responses in the Lung in Patients with ...

Oral presentations

1) Barry SM and Janossy G. Flow cytometry in BAL and other tissue fluids.

European flow cytometry conference andworking group on clinical cell

analysis. Urbino, Italy 2002.

2) Barry SM and Janossy G. Applications of flow cytometry in bronchoalveolar

lavage from patients with respiratory disease. European flow cytometry

conference and working group on clinical cell analysis. Urbino, Italy 2001.

Poster presentations

1) Barry SM, Johnson MA and Janossy G. Analysis of lung and blood CD8 T

lymphocytes in patients with HIV infection. 13^ World AIDS conference,

Durban, South Africa 2001

Page 8: Lymphocyte Responses in the Lung in Patients with ...

Index of Tables

T able 1.1 Advantages and disadvantages of different methods for determining

antigen-specific lymphocyte responses...................................................... 20

Table 2.1 Fluorochromes available for use between the CytoronAbsolute and

FACSCalibur flow cytometers...................................................................... 46

Table 2.2 Monoclonal antibodies used in this thesis................................. ...................51

Table 2.3 Antigens and substances used to stimulate cytokine synthesis...................61

Table 3.1 Coefficients of variation for BAL lymphocyte, macrophage and

granulocyte percentages derived by flow cytometry and cytospin.............. 75

Table 4.1 Demographic details, diagnoses and CD4/CD8 ratios by different

FCM methods in the study population.................................................. 88

Table 5.1 Characteristics of BAL from study population.............................................. 101

Table 5.2 Main BAL diagnoses in HIV- and HIV+ patients...............................L........ 102

Table 5.3 Demographic and diagnostic features of patients with sarcoid....................104

Table 5.4 Demographic, diagnostic and BAL FCM data of patients with TB.............. 107

Table 5.5 BAL lymphocyte percentages from radiologically abnormal and normal

lung in patients with pulmonary TB.............................................................110

Table 6.1. Demographic, Immunological, Viral and Diagnostic Data of the HIV+

study Population.........................................................................................127

Table 7.1 Demographic and diagnostic data for patients undergoing CD8

phenotypic analysis in blood and BAL........................................................143

Table 8.1 Demographic and diagnostic results in patients with TB..............................159

Table 8.2 Demographic and diagnostic results of patients with non-tuberculous

respiratory disease..................................................................................... 161

Table 8.3 BAL lymphocyte percentages and CD4 type-1 cytokine responses at

diagnosis of TB and following completion of TB therapy ............167

Table 8.4 BAL IFN-y responses to PPD from radiologically affected and

unaffected lung in patients with TB.............................................................168

Page 9: Lymphocyte Responses in the Lung in Patients with ...

Index of Figures

Figure 1.1 FCM dotplots of lysed whole blood and BAL...............................................17

Figure 1.2 Cartoon of immune response to Mycobacterium tuberculosis (TB)............25

Figure 2.1 Photomicrographs of stained BAL cytospin preparations............................43

Figure 2.2 FCM dotplot of lysed whole blood............................................................... 45

Figure 2.3 FCM dotplot of CD45 panleukogating against side scatter (SSC) to

differentiate the major leukocyte populations in lysed whole blood............ 47

Figure 2.4 FCM dotplot from BAL demonstrating the separation of CD45+

leukocytes from CD45- non-leukocyte debris.............................................48

Figure 2.5 Epithelial cell contamination of BAL in FCM dotplots and cytospins...........49

Figure 2.6 FCM gating strategy for phenotyping of lymphocytes................................. 50

Figure 2.7 Mean fluorescence intensity (MFI) of CD4 expression on BAL

lymphocytes with incubation time................................................................ 55

Figure 2.8 Time course of BAL CD4 cytokine responses to PPD................................ 56

Figure 2.9 CD69 expression on CD4 and CD8 lymphocytes in fresh BAL.................. 57

Figure 2.10 Dose response curve of CD4 cytokine synthesis to PPD stimulation......... 59

Figure 2.11 BAL CD4 expression before and after permeabilisation............................. 60

Figure 3.1 CD45 panleukogating in BAL.......................................................................67

Figure 3.2 BAL eosinophil discrimination by FCM........................................................ 68

Figure 3.3 BAL lymphocyte determination by CD45 expression and light scatter

compared with lymphosum gating................................................................ 69

Figure 3.4 CD45 expression and light side scatter characteristics in BAL cells

expressing the dead cell marker, 7-AAD.....................................................72

Figure 3.5 Correlation plots comparing the enumeration of BAL lymphocytes,

granulocytes and macrophages by flow cytometry and cytospin................ 73

Figure 3.6 Bland-Altman plots comparing the enumeration of BAL lymphocytes,

granulocytes and macrophages by flow cytometry and cytospin............... 74

Figure 3.7 Correlation and Bland Altman plots comparing BAL lymphocyte

percentages by CD45 gating with lymphosum.............................................76

Figure 3.8 Immunofluoresence staining of BAL with an eosinophilia........................... 77

Figure 4.1 Optimum gating strategy for determining BAL CD4/CD8 ratios...................85

Figure 4.2 Simplified gating strategy for determining BAL CD4/CD8 ratios..................86

Figure 4.3 Standard gating strategy to determine the BAL CD4/CD8 ratios................ 86

Page 10: Lymphocyte Responses in the Lung in Patients with ...

Figure 4.4 Correlation between method 1 and method 2 for determining

the CD4/CD8 ratio determination................................................................. 89

Figure 4.5 Bland Altman comparisons between method 1 and 2 for the

determination of CD4/CD8 ratios................................................................ 89

Figure 4.6 Correlation between method 1 and method 3 for determining the BAL

CD4/CD8 ratios............................................................................................ 90

Figure 4.7 Bland Altman comparisons between method 1 and 3 for the

determination of CD4/CD8 ratios................................................................ 91

Figure 5.1 Percentage of BAL lymphocytes by FCM in patients with sacoidosis

according to the stage of their pulmonary disease.................................... 105

Figure 5.2 BAL CD4/CD8 ratios by FCM in patients with sacoidosis according to

the stage of their pulmonary disease.........................................................105

Figure 5.3 Percentage of BAL lymphocytes by FCM in patients with tuberculosis,

sacoidosis, and in healthy controls............................................................108

Figure 5.4 Percentage of BAL lymphocytes by FCM in patients with tuberculosis

with washings taken from cavities and radiologically normal lung............108

Figure 5.5 BAL lymphocyte percentages in all patients withTB, in symptomatic

TB patients without cavities and in healthy controls..................................110

Figure 5.6 BAL CD4/CD8 ratios in patients with TB, sarcoidosis and in controls...... 111

Figure 5.7 Blood CD4 count in HIV+ patients according to pathogens obtained in

BAL............................................................................................................. 112

Figure 5.8 BAL lymphocyte percentages in HIV+ patients without respiratory

Pathogens according their blood CD4 counts........................................... 113

Figure 5.9 CD4/CD8 ratios in blood and BAL in HIV+ patients without respiratory

Pathogens according their blood CD4 counts........................................... 114

Figure 5.10 Box and whisker plots comparing the percentage of CD4 lymphocytes

in BAL and blood according to different blood CD4 categories in HIV+

Patients without respiratory pathogens..................................................... 115

Figure 5.11 Box and whisker plots comparing the percentage of CD8 lymphocytes

in BAL and blood according to different blood CD4 categories in HIV+

Patients without respiratory pathogens..................................................... 115

Figure 5.12 Leukocyte discrimination and CD4/CD8 ratios by FCM in pleural fluid.

Ascetic fluid and cerebrospinal fluid...........................................................116

Page 11: Lymphocyte Responses in the Lung in Patients with ...

Figure 6.1 FCM gating strategy to determine the activation and proliferation staus

Of CD8+ memory lymphocytes..................................................................129

Figure 6.2 Determination of CD38 gating strategy by FCM........................................ 130

Figure 6.3 Comparison between the percentages of CD38+ CD8+ T lymphocytes

in BAL and blood in controls and HIV+ patients........................................ 133

Figure 6.4 Comparison between CD38+ CD8+ T lymphocytes from BAL of HIV+

Patients with and without respiratory pathogens.......................................134

Figure 6.5 Box and whisker plots comparing the percentage of Ki67+ CD8+ T

Lymphocytes in CD38+ and CD38- populations in BAL and blood...........135

Figure 7.1 FCM dotplots demonstrating CD8 naïve and memory subsets in BAL

In HIV infection and sarcoidosis................................................................ 145

Figure 7.2 Expression of of CD8 CD45 isoforms in BAL.............................................146

Figure 7.3 Box and whisker plots comparing memory CD8 lymphocytes in BAL

and blood....................................................................................................147

Figure 7.4 Pie charts of CD8 naïve and memory CD8 subpopulations in BAL and

Blood for the whole study population.........................................................149

Figure 7.5 Pie charts of CD8 naïve and memory CD8 subpopulations in BAL and

and blood from HIV+ patients and controls............................................... 151

Figure 8.1 IFN-y and TN F-a responses in BAL T lymphocytes following incubation

with PPD in a patient with TB.....................................................................163

Figure 8.2 CD4 IFN-y responses in BAL in patients with TB and non-TB

respiratory disease..................................................................................... 164

Figure 8.3 CD4 IFN-y responses in BAL in patients with pulmonary and non-

pulmonary TB..............................................................................................165

Figure 8.4 CD4 IFN-y responses in blood in patients with TB, BCG-vaccinated

healthy controls and non-BCG vaccinated patients without TB................ 168

Page 12: Lymphocyte Responses in the Lung in Patients with ...

Table of Contents

Acknowledgements....................................................................................................... 1

Dedication......................................................................................................................... 1

Publications arising from this thesis ........................................................................3

Papers............................. 3Oral presentations.................................................................................................................................. 4Poster presentations..............................................................................................................................4

Index of Tables................................................................................................................ 5

Index of Figures..............................................................................................................6

Table of Contents............................................................................................................9

1. Chapter 1............................................ 13

1.1 Background.....................................................................................................................................141.2 Flow cytometry and CD45 panleukogating.............................................................................. 171.3 Techniques for detecting antigen-specific I lymphocytes................................................... 191.4 Overview of recent developments in understanding lung immune responses................231.5 Summary of rationale and aims.................................................................................................. 281.6 References....................................................................................................................................... 29

2. Chapter 2.............................................................................................................40

2.1 Introduction.....................................................................................................................................412.2 Fibreoptic bronchoscopy and bronchoalveolar lavage.........................................................412.3 Preparation of BAL........................................................................................................................ 412.4 Cytospins......................................................................................................................................... 422.5 Immunofluoresence staining.......................................................................................................422.6 Flow cytometry: general introduction and gating strategies...............................................44

2.6.1 General characteristics of the flow cytometers used...................................................... 442.6.2 Mechanisms of analyte discrimination by FCM.............................................................. 442.6.3 CD45 directed panieukogating in blood and BAL........................................................... 462.6.4 Gating strategy to identify bronchiai epitheiiai and squamous celis in BAL by FCM....482.6.5 General gating strategy for lymphocyte phenotypic analysis: primary immunological gating.......................................................................................................................................... 49

2.7 Flow cytometry: Reagents, panels and protocols.................................................................502.7.1 Reagents and panels for three and four colour FCM.......................................................502.7.2 Protocols for staining of fresh whole blood and BAL......................................................522.7.3 Intracellular staining by FCM: fixation and permeabilisation of ceils............................ 52

2.8 Measurement of antigen-specific responses: cytokine synthesis assay..........................532.8.1 General introduction to the method.................................................................................. 532.8.2 Time course experiment for cytokine synthesis following incubation with PPD.......... 542.8.4 Use of CD69 in BAL........................................................................................................... 572.8.5 Dose response curve for purified protein derivative...................................................... 582.8.6 Optimisation of antibody surface staining sequence..................................................... 602.8.7 Method for the detection of intracellular cytokine synthesis in whole blood and BAL .602.8.8 Antigens used for the cytokine synthesis assay............................................................. 61

2.9 Statistics.......................................................................................................................................... 61

Page 13: Lymphocyte Responses in the Lung in Patients with ...

2.10 References..................................................................................................................................... 62

.............................................................................................................................Chapter 3........................................................................................................................................... 64

3.1 Introduction.....................................................................................................................................653.2 Material and methods.................................................................................................................... 66

3.2.1 Subjects..............................................................................................................................663.2.2 Bronchoalveolar lavage.................................................................................................... 663.2.3 Flow cytometry.................................................................................................................. 663.2.4 Cytospin.............................................................................................................................703.2.5 Freezing and thawing of BAL............................................................................................703.2.6 Immunofluorescence staining of BAL..............................................................................703.2.7 Statistical analysis.............................................................................................. 70

3.3 Results.............................................................................................................................................. 713.3.1 BAL diagnoses.................................................................................................................. 713.3.2 BAL leukocyte differential counts by FCM....................................................................... 713.3.3 7-AAD expression In BAL................................................................................................. 723.3.4 Correlation between leukocyte differentials by FCM and cytospin................................723.3.5 Coefficient of variation between FCM and cytospin........................................................ 753.3.6 Comparison between fresh and frozen BAL for leukocyte subset determination by FCM .....................................................................................................................................................753.3.7 Comparison between BAL lymphocyte percentages obtained by CD45 and light scatter with the sum of the lymphocyte subsets by FCM..................................................................... 763.3.8 Immunofluorescence staining of BAL..............................................................................77

3.4 Discussion....................................................................................................................................... 773.5 References....................................................................................................................................... 79

.............................................................................................................................Chapter 4 82

4.1 Introduction.....................................................................................................................................834.2 Methods............................................................................................................................................83

4.2.1 Patients...............................................................................................................................834.2.3 Bronchoalveolar lavage and pleural fluid.........................................................................844.2.4 Handling of samples......................................................................................................... 844.2.5 Flow cytometry.................................................................................................................. 844.2.6 Statistics.............................................................................................................................87

4.3 Results.............................................................................................................................................. 874.3.1 Diagnoses In the study population...................................................................................874.3.2 Comparison of CD4/CD8 ratios determined by the ‘gold standard’ (method 1) with the simplified technique (method 2)................................................................................................ 874.3.3 Differences between the BAL and pleural fluid CD4/CD8 ratios measured by method 1 and method 3 ..............................................................................................................................90

4.4 Discussion....................................................................................................................................... 914.5 References....................................................................................................................................... 93

Chapter 6........................................................................................................................................... 96

5.1 Introduction.....................................................................................................................................975.2 Methods............................................................................................................................................97

5.2.1 Patients...............................................................................................................................975.2.2 Bronchoalveolar lavage and bronchial biopsy................................................................ 985.2.3 Acquisition of pleural, peritoneal and cerebrospinal fluid samples...............................985.2.4 Routine analysis of Clinical Specimens........................................................................... 985.2.5 Preparation of Specimens.................................................................................................995.2.6 Flow Cytometry.................................................................................................................. 99

10

Page 14: Lymphocyte Responses in the Lung in Patients with ...

5.2.7 Statistics........................................................................................................................ 1005.3 Results............................................................................................................................................100

5.3.1 General characteristics of BAL.......................................................................................1005.3.2 Diagnoses in patients undergoing BAL.......................................................................... 1015.3.3 Sarcoidosis...................................................................................................................... 1035.3.4 Tuberculosis.................................................................. 1065.3.5 HIV....................................................................................................................................112

5.4 Analysis of leukocyte differentials in non-BAL flu ids......................................................... 1165.5 Discussion............................................... 1175.6 References..................................................................................................................................... 120

.................................... Chapter 6......................................................................................................................................... 124

6.1 introduction...................................................................................................................................1256.2 Methods......................................................................................................................................... 126

6.2.1 Patients.............................................................................................................................1266.2.2 Determination of HIV Viral Load......................................................................................1266.2.3 Standard Investigations for Respiratory Pathogens in BAL..........................................1266.2.4 Bronchoscopy and Sample Preparation........................................................................ 1286.2.5 Flow Cytometry and Gating strategies........................................................................... 1286.2.6 Statistical Analysis...........................................................................................................131

6.3 Results............................................................................................................................................1316.3.1 Diagnoses in the HIV* patients with respiratory disease and BAL lymphocyte percentages..............................................................................................................................1316.3.2 CD45 Isoform Expression of CDS* I lymphocytes in BAL and blood in HIV* Patients and control subjects................................................................................................................ 1316.3.3 CD38 expression in CD45RA CDS* lymphocytes from BAL and blood of HIV* patients and control subjects................................................................................................................ 1326.3.4 CD3S expression in CD45RA CDS* lymphocytes from BAL of HIV* patients with and without Respiratory Pathogens................................................................................................1335.3.5 Expression of KI67 in activated and unactivated CDS* lymphocytes in lung and blood ...................................................................................................................................................135

6.4 Discussion.....................................................................................................................................1366.5 References.....................................................................................................................................137

.............................................................................................................................Chapter 7.........................................................................................................................................141

7.1 Introduction...................................................................................................................................1427.2 Materials and Methods................................................................................................................142

7.2.1 Patients.............................................................................................................................142CD4............................................................................................................................................143HIV viral load’' ........................................................................................................................... 1437.2.2 Bronchoscopy................................................................................................................. 1447.2.3 Sample preparation......................................................................................................... 1447.2.4 Flow Cytometry and Gating strategies........................................................................... 1447.2.5 Statistical Analysis.......................................................................................................... 146

7.3 Results............................................................................................................................................1467.3.1 Comparison of the proportion of memory CDS+1 lymphocytes in the total CDS I cellpool in BAL and blood..............................................................................................................1467.3.3 Differences in CDS lymphocyte subpopulations between patients with HIV, sarcoidosis and healthy control subjects....................................................................................................149

7.4 Discussion.....................................................................................................................................1527.5 References.....................................................................................................................................154

Chapter 8.............................................................................................................. 167

11

Page 15: Lymphocyte Responses in the Lung in Patients with ...

8.1 Introduction...................................................................................................................................1588.2 Methods..........................................................................................................................................158

8.2.1 Patients............................................................................................................................. 1588.2.2 Bronchoalveolar lavage...................................................................................................1618.2.3 Sample preparation..........................................................................................................1628.2.4 PPD stimulation and FCM analysis................................................................................. 1628.2.5 Statistics........................................................................................................................... 1638.3.1 Comparison of IFN-y and TNF-a responses to PPD In BAL between IB-Infected and uninfected Individuals.............................................................................................................. 1648.3.3 Type-1 cytokine responses In PPD-stlmulated CD4 lymphocytes In BAL In patients with pulmonary and non-pulmonary TB..........................................................................................1658.3.4 Type-1 cytokine synthetic responses to PPD In BAL CD4 and CD8 lymphocytes In patients with TB........................................................................................................................ 1668.3.5 Persistence of type-1 cytokine synthetic responses to PPD In BAL following Initiationof treatment for TB...................................................................................................... 166Patient.......................................................................................................................................1678.3.6 Type-1 cytokine synthetic responses to PPD In BAL from radiologically normal and abnormal areas of lung In patients with TB............................................................................ 1678.3.7 Comparison of IFN-y and TNF-a responses In the blood of TB patients with BCG- vacclnated controls........................................................................................................... 167

8.4 Discussion..................................................................................................................................... 1698.5 References..................................................................................................................................... 172

............................................................................................................................ Chapter 9.........................................................................................................................................176

9.1 Discussion..................................................................................................................................... 1779.2 References..................................................................................................................................... 182

Glossary of Abréviations..........................................................................................184

Appendix 1....................................................................................................................185

Appendix 2....................................................................................................................186

12

Page 16: Lymphocyte Responses in the Lung in Patients with ...

1. Chapter 1

Introduction, Rationale and Aims

13

Page 17: Lymphocyte Responses in the Lung in Patients with ...

1.1 BackgroundThe burden of respiratory infections worldwide is enormous. The broad spectrum of

these diseases encompasses upper and lower respiratory tract infections through to

community and hospital acquired pneumonia and tuberculosis. Tuberculosis (TB) is

estimated to infect one third of the world’s population and causes eight million new

infections and nearly two million deaths each year [1]. Untreated, the mortality rate of

clinical disease has been estimated at 40-60% [2]. One of the terrible tragedies of this

disease is that effective chemotherapeutic regimes exist [3], although 95% of cases and

deaths occur in resource-poor countries [1] that often cannot afford the drugs and do not

have the health infrastructure to cope. Despite a commitment to reduce the death rate

from tuberculosis by 50% by the year 2010, the leaders of the world’s most powerful

countries have seemingly set themselves an impossible task. The rising TB pandemic in

Sub-Saharan Africa is fuelled by a number of factors including HIV co-infection [4, 5],

poor health infrastructures, famine, poverty and war. Nevertheless, sensible directly

observed therapy (DOTS) treatment programmes adapted to local situations have

proved highly effective [6-8] and have led some observers to be cautiously optimistic

about TB control [9].

Even more disastrous than TB in terms of mortality rates is pneumonia which is the

most frequent cause of death worldwide in children under five [10] and also carries a

high mortality rate in both resource-rich and poor settings in adults [11]. Viral respiratory

tract infections are generally less severe in the immunocompetent host, but they are a

very significant factor in exacerbations in patients with underlying asthma [12, 13] and

chronic obstructive pulmonary disease [14].

The burden of respiratory disease in patients who are immunocompromised either

due to HIV infection, organ transplantation, or immunosuppressive therapy is even

greater than in the immunocompetent patient. Worldwide HIV infection is by far the most

significant cause of immunosuppression, with an estimated 40 million infected individuals

in 2001 of whom 70% are from Sub-Saharan Africa [15]. Overall, the greatest burden of

respiratory disease in HIV infected individuals is that of tuberculosis with recent

estimates from some Sub-Saharan countries that 70% of patients with active

tuberculosis are also co-infected with HIV [16]. Tuberculosis is the leading cause of

death among people with HIV infection, accounting for a third of deaths world-wide [16].

In resource-poor settings, the burden of tuberculosis is a mixture of reactivation and re­

infection, with the latter thought to be increasingly more important in TB endemic areas

14

Page 18: Lymphocyte Responses in the Lung in Patients with ...

[17]. HIV co-infection dramatically increases the risk of reactivation in those who are

infected with TB but do not have clinical disease. It has been estimated that the rate of

reactivation of primary TB is only 5-10% for the lifetime of a non-HIV infected individual

[18]. In those co-infected with HIV the annual risk of developing active disease ranges

from 5% to 15% [19-21]. These features have been highly significant in fuelling the TB

pandemic in resource-poor countries, particularly in Sub-Saharan Africa. The high

prevalence of TB in HIV-infected patients is not confined to the developing world.

Increasing migration and immigration of persons from such countries has contributed to

rising rates of TB in the West [22-24]. A particular concern is that of multi-drug resistant

TB in HIV infection that has extremely high mortality rates [25]

In addition to increased susceptibility to mycobacterial infections, HIV+ patients are

also at increased risk of Pneumocystis carinii pneumonia (PCP) which historically

occurred in 60-80% in the resource-rich world prior to the advent of anti-retroviral and

anti-pneumocystis therapy [26]. However, infection with this opportunistic pathogen is

rare in adults from resource-poor settings, a finding that is largely explained by death

from other diseases before a sufficient drop in CD4 count is reached to increase the risk

of PCP [27]. In addition, HIV-infected adults and children are at increased risk of

developing bacterial pneumonia [28, 29] and bacteraemia complicating this [30, 31].

By contrast with the HIV-infected population, where cytomegalovirus (CMV) infection

is a rare respiratory pathogen [32, 33], patients who have undergone bone marrow

transplantation (BMT) are known to be particularly at risk of cytomegalovirus

pneumonitis (CMV-P). Infection with this pathogen had historical mortality rates of 30-

80% until the recent introduction of effective prophylactic therapy [34]. Fungal infections,

particularly with aspergiiius species are also frequent infectious hazards in the early post

transplant period characterized by neutropenia [34]. In lung transplant patients, CMV-P

is also a well-recognised infectious complication [35].

Therefore, one of the characteristic features of respiratory infections in these groups

reveals that different patterns emerge in the types of respiratory pathogens between

patients who are immunocompromised due to HIV infection from those that have had

BMT or solid organ transplantation. Fungal infections and cytomegalovirus frequently

cause respiratory infections in BMT patients, but only rarely in those with HIV, whilst

PCP and tuberculosis are more common in HIV infection than following BMT [34]

Underlying these clinical presentations are various defects in the host immune

response that, in combination with the direct pathogenic effects of the organism, lead to

15

Page 19: Lymphocyte Responses in the Lung in Patients with ...

different clinical outcomes. Unfortunately, relatively few studies have examined the

processes of the immune response in the lung in humans, but instead, investigations

frequently extrapolate from the findings seen with cells taken from peripheral blood

samples with the assumption that these are equally applicable to the responses in

tissues.

Interest in the lung as a distinct immunological site has been stimulated by the

investigation of diseases such as sarcoidosis in which lung involvement is a dominant

clinical presentation. Considerable effort has been invested over the last two decades in

determining the leukocyte differentials and CD4/CD8 lymphocyte ratios in the lung in

patients with lung disease. The impetus behind this drive was threefold. First, it was

discovered that the lymphocyte proportions obtained in BAL were similar to those

obtained from lung biopsy specimens in sarcoidosis patients, thus lending credence to

the use of BAL as an investigative sample [36, 37]. Second, sarcoidosis was shown to

be characterized by a BAL lymphocytosis and a raised CD4/CD8 ratio when compared

to healthy controls. [38, 39]. Lastly, it was documented that the changes noted in BAL

were largely absent in the blood [38]. Therefore, the investigation of cell populations in

the lung was thought to be of diagnostic relevance for diseases such as sarcoidosis and

provided a further impetus to study BAL lymphocyte differentials in other respiratory

diseases such as TB [40-42], cryptogenic organising pneumonia [43] and pulmonary

fibrosis [44-46].

One of the features that has handicapped the investigation of lung immunology has

been a conservatism in adopting new investigative tools and the consequent reluctance

for introducing new concepts into the evaluation of disease processes. For example, flow

cytometry, which has been the gold standard for enumerating CD4 counts and CD4/CD8

ratios in blood for twenty years [47], has yet to be adopted as a standard technique for

BAL lymphocyte analysis. Most of the studies investigating BAL CD4/CD8 lymphocyte

ratios have involved the use of immunofluoresence or peroxidase-anti-peroxidase

staining. These techniques are time consuming and suffer from inaccuracies due to the

low number of cells routinely counted. Furthermore, such a cumbersome technology

does not allow the convenient application of new ideas that aim to solve complex

problems of immunoregulation at the relevant tissue sites, in this case the lung.

16

Page 20: Lymphocyte Responses in the Lung in Patients with ...

1.2 Flow cytometry and CD45 panleukogating

Flow cytometry (FCM) is an alternative investigative tool to cytocentrifuge

preparations (cytospins) that has several advantages. First, it analyses data for

thousands of events and therefore reduces errors due to manual counting of small cell

numbers. Second, it is fast as FCM can be performed immediately after filtering,

centrifugation and staining of BAL samples. Third, and most importantly, the extensive

experience of analyzing leukocytes in blood with FCM has resulted in the development

of convenient and precise techniques for determining leukocyte differential counts. The

key strategy that has recently emerged has been morphospectral analysis using the

leukocyte marker, CD45 [48-50]. Importantly, these methods are readily exploitable for

the analysis of other tissue samples. In the past, it has been well documented that CD45

staining was optimal to differentiate lymphomas from anaplastic carcinomas in tissue

sections by immunohistology [51, 52]. In BAL, the use of CD45 enables leukocytes to be

differentiated from non-leukocyte components such as mucoid particles and epithelial

cells. Here, the adoption of CD45 pan-leukogating is particularly important because in

BAL the intrinsic cell parameters measured by FCM, size and granularity are not

sufficient to distinguish between the different leukocyte components and contaminating

debris (figure 1.1)Figure 1.1

B lo od B A L

SSC

Figure 1.1

FCM dotplots of fresh lysed whole blood and BAL. The intrinsic parameters of the

acquired events measured are forward scatter (FSC, size, y axis) and side scatter

(SSC, granularity, x axis). In the lysed blood, distinct leukocyte populations of

lymphocytes, monocytes and granulocytes are demonstrated. In BAL, no clear

populations are determined by these characteristics.

17

Page 21: Lymphocyte Responses in the Lung in Patients with ...

The concept of using CD45 during BAL analysis is not new. Several investigators

have adopted a gating approach that included CD45 for distinguishing lymphocytes from

the rest of the leukocyte pool in BAL [53-55]. However, these earlier methods have

included unnecessary complications that masked the advantages of using CD45 [54]. An

even more serious problem has been that in these previous studies the discrimination

between the relevant leukocyte components of the BAL fluid such as neutrophils and

eosinophils was neglected by FCM. This omission has been a significant factor in

ensuring that cytospins have generally remained the dominant method for BAL leukocyte

differential analysis.

Alveolar macrophages have also posed particular problems for flow cytometric

evaluation due to their autofluoresence [56] and heterogenous light scatter

characteristics. Some investigators have attempted to overcome the autofluoresence by

quenching with gentian violet [57]. However, It has remained unclear whether such

techniques have rendered these treated cells more amenable to phenotypic analysis by

FCM. A further problem has been the lack of a bona fide’ surface marker that would

identify macrophages in their various stages of differentiation. The only likely candidate

for the role of a pan-macrophage marker is the transmembrane glycoprotein CD68 [58].

Unfortunately, this marker is only suitable for histological or intracellular staining as the

molecule is not well expressed on the membrane of intact macrophages when studied in

suspension. This fact has led to an extra complication for the use of FCM to characterize

alveolar macrophages since an additional permeabilisation step is required for adequate

CD68 staining. As a result of these problems most, but not all analyses of alveolar

macrophages have still been performed by cytospin preparations.

In summary, despite the fact that FCM has been refined, simplified and accepted as

the gold standard method for the determination of leukocyte differentials in blood, this

technology has not yet been adequately applied to BAL. Therefore, the first aim of this

thesis was to develop a flow cytometric system that could distinguish all the relevant

leukocyte components in BAL. In particular, it was felt necessary that such a system

should be simpler, faster and more precise than the existing cytospin methods and thus

provide an impetus for adopting FCM as the routine diagnostic tool for BAL analysis.

This initial aim provided the platform for the further investigation of BAL by FCM and

the logical development of the other aims of this thesis. The second aim was to

investigate the differences between the lung and the blood T cell responses in terms of

both the major subsets of these lymphocytes and their particular phenotypic

18

Page 22: Lymphocyte Responses in the Lung in Patients with ...

characteristics found in each site. This second aim sought to determine to what extent

the lung was a distinct immunological compartment when compared to the blood, the

third aim of the thesis was to apply this comparative technique for the investigation of

antigen-specific responses in both the lung and blood compartments by making use of

recent advances in immunological techniques to detect such T lymphocyte responses.

1.3 Techniques for detecting antigen-specific I lymphocytes.Over the last six years there have been major advances in cellular immunology, the

most important of which has been the development of several techniques for the

accurate determination of antigen-specific lymphocytes [59-61] This has been a

revolutionary step as it has enabled the study of the functional performance of antigen-

specific CD4 and CDS T lymphocytes in vivo and also provided valuable insights into the

nature of immune responses to pathogens. Experiments using class I MHC tetramers

bound to Epstein-Barr virus (EBV) epitopes have demonstrated huge EBV-specific CDS

responses during acute infection that previous limiting dilution techniques had markedly

underestimated [62]. In the field of HIV, the detection of HIV-specific responses by

tetramers and cytokine production methods such as the ELISPOT have been

instrumental in understanding how the immune system responds to the virus [61, 63].

Each of these techniques has both advantages and disadvantages that are relevant to

their application as research tools (table 1.1).

Tetramers are major histocompatibility (MHC) class 1 molecules folded into a

tetrameric complex bound together with streptavidin to which relevant peptides can be

attached. This structure forms a stable unit that binds CD8+ T lymphocytes that

recognize the MHC-restricted peptide. This tetrameric complex has the advantage that it

binds specific CD8+ T lymphocytes with greater avidity than the natural monomeric

complex [64, 65]. The addition of a fluorochrome allows the CD8-tetramer complex to be

analysed by FCM [66].

To date the majority of tetrameric complexes have been made with class 1 MHC

molecules, although most recently class 2 MHC tetramers have also appeared as

research tools [67]. The advantages of using tetramers are that the peptide-specific CD8

lymphocytes and with class 2 tetramers, CD4 lymphocytes can be directly visualized by

FCM and the phenotype of these cells analysed using further discriminating monoclonal

antibodies. Nevertheless, some investigators have questioned the functional ability of the

tetramer-binding cells [68-70].

19

Page 23: Lymphocyte Responses in the Lung in Patients with ...

Table 1.1 Advantages and disadvantages of different methods for determining

antigen-specific lymphocyte responses

Method Advantages Disadvantages

Tetramer

1. Rapid detection (1 hour) of

Ag-specific response by FCM

2. Phenotypic analysis possible

1. Only measures response to

peptide present on tetramer

which may not be

immunodominant

2. Predominantly only CD8

responses since veiV few class-2

tetramers exist

3. Functionality of the cells not

determined

4. HI_A restriciton

5. Tetramer binding is temperature

dependent

6. Requires a flow cytometer

Elispot

1. Functional responses

measured.

2. low tech- responses can be

assessed with a microscope

3. Can use a variety of

stimulatory antigens so HLA

restriction not an issue

1. Cannot distinguish which

lymphocyte (CD4 or CD8) is

responding

2. Only measures the secretion of a

single cytokine thereby may

underestimate the Ag-specific

response

3. Phenotypic analysis not possible

Flow cytometric

1. Functional responses

measured

2. Can determine the

responding lymphocyte

subset

3. phenotypic analysis possible

4. Can distinguish a variety of

different cytokines

synthesized

5. Can use a variety of

stimulatory antigens so HLA

restriction is not an issue

Requires a flow cytometer.

Phenotypic analysis and multiple

cytokine detection is dependent

on the type of machine (number

of lasers) and the number of

fluorochromes used

20

Page 24: Lymphocyte Responses in the Lung in Patients with ...

The addition of a peptide stimulation step following tetramer staining has overcome this

problem by enabling the analysis of cytokine responses in the tetramer binding cells [71].

The major disadvantage of using tetramers to study CD8 lymphocytes is the HLA-

restriction of the response. Subjects must share the HLA haplotype of the tetramer and

there is no guarantee that the response generated is an immunodominant one.

Pathogens contain multiple epitopes that stimulate different responses between

individuals. Thus in order to approximate the natural response to many pathogens a

battery of different HLA-tetrameric complexes would need to constructed. Fortunately,

some well-studied viruses such as cytomegalovirus (CMV) appear to generate dominant

responses to conserved epitopes of the CMV matrix protein pp65 [72]. This restriction of

responses has facilitated the use of tetramers to study the immune response to this

pathogen in various clinical settings [73-75]. The responses to viruses such as HIV are

more complex and tetramer studies using separate HIV epitopes will only measure part

of the total immune response against the virus. These limitations, in addition to the fact

that tetramers are expensive and difficult to construct make them likely to have a limited

role beyond that of purely applied scientific research.

The two main additional well-standardized techniques for analysing antigen-specific

responses both detect cytokine responses following incubation with antigen. Both

techniques more closely mimic the natural immune response in the sense that antigen,

added to the culture medium is presented to cognate T lymphocytes by antigen-

presenting cells. The concept underlying these systems is that T cells, either CD4 or

CD8, that recognize antigen in the context of relevant MHC molecules, rapidly start

synthesizing cytokines. These cytokines can then be measured intracellularly by FCM

[60], visualized as spot forming colonies following secretion into a gel matrix containing

anti-cytokine antibodies [76], or detected by ELISA [77]. These methods both have the

advantage over the standard tetramer-binding assay that they directly measure the

functional responses of antigen-specific cells.

A potential problem with the cytokine production methods for quantifying the antigen-

specific lymphocyte populations are that a variety of different cytokines may be produced

by these cells on encounter with antigen. Conventionally, interferon-y (IFN-y) has been

measured by ELISPOT [61, 78] although this system can be used to detect other

cytokines such as interleukin-12 (IL-12) released from monocytes [79]. The flow

cytometric technique has also predominantly measured type-1 cytokine responses.

21

Page 25: Lymphocyte Responses in the Lung in Patients with ...

including tumour necrosis factor-a (TNF-a) [80]. However, focusing on such responses

ignores other cytokines that may be produced and therefore may underestimate the total

number of antigen-specific cells. This is a particularly important shortfall of the ELISPOT

method where only one cytokine response is measured. The FCM method has the

advantage of being able to discriminate a variety of different cytokine responses in

addition to providing a phenotypic analysis of responding cells. Whilst current flow

cytometers widely available on the market are set up for three of four colour analysis,

industry is rapidly responding with interest to this powerful tool. As a result machines are

in use that can perform 11 colour analysis, enabling the measurement of a large number

of different cytokine responses in addition to phenotyping the responding cells [81]. It is

likely that in the future cheaper multi-parameter flow cytometers will become widely

available.

The major advantage of the cytokine-production methods over the tetramer assay for

the detection of antigen-specific lymphocytes is that both CD4 and CD8 lymphocyte

responses can be measured. In these assays the size of the stimulating antigen

determines which T lymphocytes are preferentially stimulated. Complex antigens are

phagocytosed and then presented in the context of class-2 MHC molecules [82]. Studies

using peptides, rather than complex soluble antigens have demonstrated that larger

peptides of 15 amino acids or more stimulate CD4 lymphocyte responses whilst CD8

responses are optimally stimulated by short peptides of between 8-12 amino acids [83].

By constructing overlapping libraries of peptides of varying lengths to use as the

stimulating antigens the sum total of the CD4 and CD8 responses can be estimated [83,

84]. This technique has the great advantage that it overcomes the problem of HLA-

restriction of responses.

Nevertheless, despite the proliferation of recent studies examining antigen-specific

responses, these have been, with few exceptions [85] confined to looking at whole

blood, or peripheral blood mononuclear cells (PBMC). In particular, there have been few

attempts to examine antigen-specific responses in the lung, despite the high burden of

pulmonary pathology. The reason behind this undoubtedly relates to the difficulty in

obtaining lung specimens for examination when compared to the ease of evaluating

peripheral blood. The investigation of lung responses in humans requires a BAL

specimen in order to obtain sufficient leukocytes for immunological analysis. Fortunately,

BAL is often routinely performed in cases with suspected respiratory infections where a

diagnosis is not rapidly obtained from sputum samples or where an unusual organism.

22

Page 26: Lymphocyte Responses in the Lung in Patients with ...

such as mycobacterium tuberculosis is suspected. The threshold for performing BAL is

lower in immunocompromised patients because the range of potential pathogens is

greater and the treatment options are more complex. Therefore, BAL performed on

these patients should provide an adequate specimen for both routine laboratory testing

in microbiology, virology and cytology in addition to an aliquot for cellular analysis.

1.4 Overview of recent developments in understanding lung

immune responsesAn integrative understanding of lung immune responses has been elusive, in part due

to the paucity of knowledge of the role played by antigen-presenting dendritic cells (DC)

in orchestrating the immune response. Animal studies have demonstrated that DC reside

throughout the respiratory tract in epithelial tissue [86] and more recently the function of

DC s has been more clearly defined.

It is now well documented that DC s determine the type of immunological response

by secreting cytokines that influence the subsequent development of CD4 and CDS

effector phenotypes. Interleukin 12 (IL-12) is the key cytokine determining differentiation

towards Th1 responses [87-89] and interleukin-10 (IL-10) drives Th2 responses. The

signals that encourage these critical cytokines to be produced by DCs are unclear,

however, there is evidence that lung DCs in the rat preferentially stimulate Th2

responses and require additional signals such as TN F-a to switch to IL-12 production

[90]. Furthermore, there is a growing body of evidence demonstrating that IL-12

production can be suppressed by a variety of microenvironmental tissue factors such as

prostaglandin E2 (PGE2) [91], nitrous oxide [92] and histamines [93], as well as by drugs

such as P2 agonists [94]. Although these studies were performed on blood monocytes

and macrophages rather than alveolar cells, the findings are suggestive that a number of

different mechanisms exist in vivo to control Thi responses.

It has been argued that type-1 immune responses in such a delicate tissue as the

lung must be carefully controlled as the foreign antigen load is high and there is a

potential for damaging the fragile alveolar compartment vital for gas exchange [95].

Indeed, sarcoidosis, a disease characterized by strong type-1 responses and granuloma

formation in the lung is associated with lung fibrosis, a restrictive lung defect and

eventual respiratory failure in severe cases.

23

Page 27: Lymphocyte Responses in the Lung in Patients with ...

Most of the studies of DC and macrophage function have been performed on cells

isolated from lung epithelial tissue sections in animal models. However, most antigens

that escape the mucociliary escalator in the large airways will be likely to first encounter

alveolar macrophages that comprise approximately 90-95% of the alveolar cells [39].

These cells phagocytose the antigens, but their role as antigen presenters is uncertain.

Several studies have suggested that these cells are poor antigen presenters and argue,

like Holt, that this could be an adaptive response to minimise lung injury [96, 97].

However, other investigators have demonstrated that alveolar macrophages are good

antigen presenting cells [98]. Recently, this issue has been resolved by the

demonstration of low percentages of cells with phenotypic and functional characteristics

of DCs that were distinct from the main macrophage population from BAL in humans [99,

100]. It is likely that following lung infection, DC recruitment into BAL from epithelial

tissue is enhanced. Evidence from animal models suggests that this population of DCs

that is initially rare can be increased dramatically by intratracheal BCG inoculation [101].

Intriguingly, infection with BCG or with live mycobacterium tuberculosis (MTB) also

resulted in maturation and activation of the DCs [101-103].

Taken together, these findings suggest that antigen presentation occurs

predominantly in the alveolar space. It is likely that signals, such as TNF-a and GM-CSF

released by macrophages that have phagocytosed antigen encourage both the migration

into and the maturation of DCs in the alveoli. The antigen-loaded DCs must then migrate

back into lung epithelial tissues and thence to the regional lymph nodes. The key events

taking place in the immune response to mycobacteria are summarised in figure 1.2

Within the draining lymph nodes the key aspects of lymphocyte recruitment,

proliferation and maturation into effector cells is determined. The role of chemokines in

the recruitment of lymphocytes both to the lymph nodes and then to the lung is becoming

better understood [104, 105] and clearly plays a crucial role in the inflammatory

response. Of considerable interest has been the recent discovery that the chemokine

receptor CCR7, which is expressed on naïve and a subset of memory T cells is also

upregulated during DC maturation [106]. The localization of naïve T cells and DCs in the

lymph node is then mediated by the expression of two T cell zone expressed

chemokines, secondary lymphoid tissue chemokine (SLC) and EBL1 ligand chemokine

(ELC) which bind to CCR7 [107, 108]. Thus brought together in the T cell zones of the

24

Page 28: Lymphocyte Responses in the Lung in Patients with ...

Figure 1.2

GranulomaTB

AM AMAlveolar space

ifn -y ( e • TNF-a \T cell

TNF-a

DC DC

DC DC

IL-12

m atu ration proliferation

T cell

T cellBALTLung interstitium

Blood

J cel T cell J cell

Figure 1.2

Cartoon of immune response to Mycobacterium tuberculosis (TB). Macrophages

release TNF-a on encounter with TB, resulting in migration into and maturation of

dendritic cells (DC) in the alveolar space. Antigen-loaded DC orchestrate antigen-

specific naïve and memory T cells maturation to effector cells in bronchial

associated lymphoid tissue (BALT) under the influence of IL-12. Secretion of IFN-y

and TNF-a by T cells are crucial for the formation of granulomas to control TB.

25

Page 29: Lymphocyte Responses in the Lung in Patients with ...

lung lymph nodes, naïve CD4 and CD8 T lymphocytes that recognize antigen presented

in the context of relevant class-1 and class-2 MHC on mature DCs will undergo

proliferation. These proliferating, antigen-specific lymphocytes then develop their effector

phenotype under the influence of the DC derived cytokines, IL-12 or IL-10. Consequent

upon encounter with antigen, CD4 and CD8 lymphocytes undergo changes in their

surface markers as well as in their expression of cytokines. The CD45 isoform changes

from RA+ RO- to RA- R0+ [109, 110] and in CD8 lymphocytes there is a progressive

loss of CD27 [111, 112]. Both CD4 and CD8 lymphocytes emigrating from the lymph

nodes lose CCR7 expression [113].

One of the final important pieces in the immunological jigsaw puzzle has been the

discovery of chemokine receptors on different lymphocyte subsets that mediate the

recruitment of these cells to the sites of infection or inflammation. CD8 Lymphocytes with

predominant Thi characteristics have been demonstrated to express CXCR3 receptors

and to accumulate in the lungs of HIV infected subjects [114]. By contrast Th2 type

lymphocytes may preferentially express receptors for different chemokines such as

CCR3 and CCR4[104].

These studies, taken as a whole have contributed greatly to our understanding of the

immune response to pathogens in the lung. A picture has emerged of the role of antigen

presenting cells, interactions in regional lymph nodes and the recruitment and

differentiation of lymphocytes to the lung. However, there have been only a few attempts

to characterize the antigen-specific lymphocyte populations in the lung.

The immune responses to tuberculosis in the lung are undoubtedly the best studied

of all lung infections in humans. Investigators have examined the different leukocyte

populations found in radiologically normal and abnormal lung [41, 115], thus giving

insights into the pathogenesis of the disease. More interestingly, two studies have

estimated the antigen-specific component of the lung responses by measuring

lymphocyte proliferation to TB antigens. Both studies separated T lymphocytes from

BAL and incubated them with either irradiated PBMC or isolated autologous monocytes

in the presence of various MTB antigens. Increased proliferative responses to

tuberculosis antigens in BAL, but not PBMC measured by [^H]-methyl thymidine

incorporation were demonstrated when T cells from TB patients were incubated with TB

antigens [116, 117].

26

Page 30: Lymphocyte Responses in the Lung in Patients with ...

These complex experimental designs presumably reflected fears that the

macrophages from BAL would suppress the antigen-specific responses. Indeed, in one

of the studies the authors demonstrated that addition of alveolar macrophages

suppressed the BAL I cell proliferative responses to phytohaemaglutinin (PHA) from TB

patients [117]. The later study also used an ELISPOT system to measure IFN-y, IL-4

and IL-10 responses to PPD a in a subgroup of six patients. This is the first study to

utilize one of the new antigen-specific techniques to examine lung immune responses.

The authors demonstrated increased IFN-y spot-forming colonies in the BAL from TB

patients, but not from BAL from healthy subjects [117]. In three patients with TB, BAL

was taken from radiologically unaffected areas of the lung and in these samples the

number of spot-forming colonies were similar to those in the healthy controls. In this

paper the dominance of anti-TB responses in the lung, but not in the blood has been

clearly demonstrated by both the proliferative assays and the ELISPOT tests.

However, the ELISPOT system may not be the optimum technique for the delineation

of BAL antigen-specific lung responses. The proportions of lymphocytes and the

CD4/CD8 ratios may be highly variable in BAL from patients with active TB [40, 115].

Since complex antigens such as PPD will predominantly stimulate CD4 lymphocytes in

the short incubation period of the ELISPOT assay, then the number of spots detected by

this method will be depend on the proportion of CD4 lymphocytes in the BAL sample.

For example, a low number of spot-forming colonies could be obtained from BAL from a

tuberculous cavity in which the predominant leukocyte subset are neutrophils and only a

small proportion CD4 lymphocytes. In fact, the proportion of antigen-specific CD4

lymphocytes from such a sample could be very high but this would be more accurately

determined by a flow cytometric system.

A flow cytometric experimental system has been used to examine lymphocyte

responses in the lung from mice infected with MTB [118]. Following intravenous

inoculation with M. tuberculosis, lungs and spleens were removed at different time points

following infection and mononuclear cells separated by density centrifugation. The tissue

cells were then incubated with brefeldin A to prevent the secretion of cytokines and the

cells were stimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin.

Following incubation, the cells were permeabilised and intracellular cytokine staining in

combination with surface staining for CD4 and CD8 was performed. The proportion of

IFN-y producing T lymphocytes from the lung and spleen were then measured by FCM.

27

Page 31: Lymphocyte Responses in the Lung in Patients with ...

In resistant C57BL/6 mice infected with virulent M. tuberculosis, there was an early

and persistent production of IFN-y by CD4 and CD8 lymphocytes in the lung that

controlled the mycobacteria. By contrast, in susceptible mice, these IFN-y producing

responses in the lung were both delayed and attenuated, and there was failure to control

the mycobacterial load. This study elegantly demonstrates the importance of IFN-y

producing lymphocytes in the control of TB in a murine model and supports the previous

studies using IFN-y knockout mice [119]. However, in this study lymphocyte activation

was achieved in cells taken from the Tb-infected mice by using phorbol mystral acetate

(PMA) and ionomycin instead of specific antigens in order to boost cytokine synthesis.

The use of such powerful immune activators may by-pass certain physiological steps

that occur in antigen-specific systems and therefore these observations may generate a

misleading picture of the true antigen-specific cytokine response.

The conclusion of this review is therefore that the technology exists for the detection

of antigen specific responses in a tissue fluid such as BAL. A consideration of the merits

and disadvantages of each method has led to the conclusion that the optimum technique

is to detect intracellular cytokine synthesis following incubation with antigen by flow

cytometry. This is because the ELISPOT assay suffers from the lack of information about

the type of cells that respond by cytokine synthesis and the detection of only one

cytokine in the secreted product. This is likely to make this method far less sensitive than

FCM when using a tissue fluid such as BAL as the proportion of lymphocytes may be

highly variable during episodes of respiratory disease. The limitations imposed by HLA-

restriction and the current inability to measure CD4 lymphocyte responses that would

require class-2 tetramers excludes the direct antigen binding assays from use in this

investigation.

1.5 Summary of rationale and aimsStudies from the 1980’s, that focused on the lung as a target of immunological

investigation have confirmed that the lymphocyte responses in BAL differ considerably

from those in the blood. This holds true both for the simple lymphocyte percentages and

CD4/CD8 ratios demonstrated in sarcoidosis and also for the recently published fledgling

antigen-specific studies in patients with tuberculosis. These early comparative studies in

patients with sarcoidosis have demonstrated close correlations between the lymphocyte

percentages in BAL and those seen in biopsy specimens from the lung interstitium.

28

Page 32: Lymphocyte Responses in the Lung in Patients with ...

Consequently, samples of BAL can be regarded as a ‘window’ for lung immunity. From

these findings a strong case can be made that lymphocyte responses in BAL are likely to

be especially informative both in terms of understanding immune responses to

pathogens and also as diagnostic tools.

These investigations into lung immune responses are particularly topical due to the

recent development of exciting new techniques in the field of immunology. The most

important of these has been the ability to precisely and rapidly detect antigen-specific

responses and flow cytometry has emerged as optimum tool for this purpose.

The main impetus for this thesis was the exploration of the immune responses to

pathogens in BAL by flow cytometry. Encompassing this broad aim were three

objectives. First, to establish a simple and reliable panel for the detection of the most

clinically relevant parameters in BAL by flow cytometry. This was an important primary

objective since a narrow focus on lymphocytes alone would have ignored other

leukocyte responses that are relevant both for immunopathological and diagnostic

reasons.

The second objective was to examine more closely T lymphocyte differentiation by

investigating the various phenotypic alternatives of these populations during bacterial

and viral infections in the lung and the blood. Although it has been previously

demonstrated that the lung serves as a repository for T cells of ‘memory’ type, it has not,

so far, been investigated whether these cells can undergo local stimulation and show

special alterations in phenotypic and activation markers. Such investigations are timely

in the light of recent advances that have generated a more complete picture of

lymphocyte differentiation patterns.

The third objective was to introduce the tests of antigen-driven stimulation of cytokine

synthesis into clinical diagnosis using BAL samples. In this area the most important task

was to assess whether such as system would be relevant for the diagnosis of

tuberculosis.

Altogether, these objectives represent the first comprehensive array of technical

innovations that aim to place clinical flow cytometry of the lung using BAL samples into

the realms of practical thoracic medicine.

1.6 References

29

Page 33: Lymphocyte Responses in the Lung in Patients with ...

1. Dye C, Scheele S, Dolin P, et al. Consensus statement. Global burden of

tuberculosis: estimated incidence, prevalence, and mortality by country. WHO

Global Surveillance and Monitoring Project. JAMA 1999;282:677-86.

2. Styblo K. Epidemiology of tuberculosis. Vol. 24. The Hague, 1991 Royal

Netherlands Tuberculosis Association Selected Papers

3. Mitchison DA. The action of antituberculosis drugs in short-course chemotherapy.

Tubercle 1985;66:219-25.

4. Harries AD, Hargreaves NJ, Kemp J, et al. Deaths from tuberculosis in sub-

Saharan African countries with a high prevalence of HIV-1. Lancet

2001;357:1519-23.

5. Drobniewski FA, Pozniak AL and Uttley AH. Tuberculosis and AIDS. J Med

Microbiol 1995;43:85-91.

6. Wilkinson D. High-compliance tuberculosis treatment programme in a rural

community. Lancet 1994;343:647-8.

7. Wilkinson D, Anderson E, Davies GR, et al. Efficacy of twice weekly treatment for

tuberculosis given under direct observation in Africa. Trans R Soc Trop Med Hyg

1997;91:87-9.

8. Floyd K, Wilkinson D and Gilks C. Comparison of cost effectiveness of directly

observed treatment (DOT) and conventionally delivered treatment for

tuberculosis: experience from rural South Africa. BMJ 1997;315:1407-11.

9. Nunn P. The global control of tuberculosis: what are the prospects? Scand J

Infect Dis 2001 ;33:329-32.

10. Douglas RM. Acute respiratory infections in children in the developing world.

Semin Respir Infect 1991;6:217-24.

11. Sow O, Frechet M, Diallo AA, et al. Community acquired pneumonia in adults: a

study comparing clinical features and outcome in Africa (Republic of Guinea) and

Europe (France). Thorax 1996;51:385-8.

12. Peebles RS, Jr., Hartert TV. Respiratory viruses and asthma. Curr Opin Pulm

Med 2000;6:10-4.

13. Papadopoulos NG, Johnston SL. The role of viruses in the induction and

progression of asthma. Curr Allergy Asthma Rep 2001;1:144-52.

14. Glezen WP, Greenberg SB, Atmar RL, et al. Impact of respiratory virus infections

on persons with chronic underlying conditions. JAMA 2000;283:499-505.

30

Page 34: Lymphocyte Responses in the Lung in Patients with ...

15. UNAIDS. AIDS epidemic update: December 2001.

www.unaids.org/epidemic_update/report_dec2001/index.html accessed 050602

16. WHO. Tuberculosis: Strategy and operations.

www.who.int/gtb/policyrd/TBHIV.htm accessed 050602.

17. Fine PE, Small PM. Exogenous reinfection in tuberculosis. N Engl J Med

1999;341:1226-7.

18. Sutherland I. Recent studies in the epidemiology of tuberculosis, based on the

risk of being infected with tubercle bacilli. Adv Tuberc Res 1976;19:1-63.

19. Selwyn PA, Sckell BM, Alcabes P, et al. High risk of active tuberculosis in HIV-

infected drug users with cutaneous anergy. JAMA 1992;268:504-9.

20. Allen S, Batungwanayo J, Kerlikowske K, et al. Two-year incidence of

tuberculosis in cohorts of HIV-infected and uninfected urban Rwandan women.

Am Rev Respir Dis 1992;146:1439-44.

21. Antonucci G, Girardi E, Raviglione MC and Ippolito G. Risk factors for

tuberculosis in HIV-infected persons. A prospective cohort study. The Gruppo

Italiano di Studio Tubercolosi e AIDS (GISTA). JAMA 1995;274:143-8.

22. Rose AM, Watson JM, Graham C, et al. Tuberculosis at the end of the 20th

century in England and Wales: results of a national survey in 1998. Thorax

2001;56:173-9.

23. Del Amo J, Petruckevitch A, Phillips AN, et al. Risk factors for tuberculosis in

patients with AIDS in London: a case-control study. IntJ Tuberc Lung Dis

1999;3:12-7.

24. Bowen EF, Rice PS, Cooke NT, et al. HIV seroprevalence by anonymous testing

in patients with Mycobacterium tuberculosis and in tuberculosis contacts. Lancet

2000;356:1488-9.

25. Drobniewski F. Is death inevitable with multiresistant TB plus HIV infection?

Lancet 1997;349:71-2.

26. Guidelines for prophylaxis against Pneumocystis carinii pneumonia for persons

infected with human immunodeficiency virus. MMWR Morb Mortal Wkly Rep

1989;38 SuppI 5:1-9

27. Barry SM, Johnson MA. Pneumocystis carinii pneumonia: a review of current

issues in diagnosis and management. HIV Mecf 2001;2:123-32.

28. Gilks CF. Acute bacterial infections and HIV disease. Br Med Bull 1998;54:383-

93.

31

Page 35: Lymphocyte Responses in the Lung in Patients with ...

29. Moroni M, Franzetti F. Bacterial pneumonia in adult patients with HIV infection. J

C/?emof/7er 1995;7:292-306.

30. Gilks CF, Brindle RJ, Otieno LS, et al. Life-threatening bacteraemia in HIV-1

seropositive adults admitted to hospital in Nairobi, Kenya. Lancet 1990;336:545-

9.

31. Frankel RE, Virata M, Hardalo C, et al. Invasive pneumococcal disease: clinical

features, serotypes, and antimicrobial resistance patterns in cases involving

patients with and without human immunodeficiency virus infection. Clin Infect Dis

1996;23:577-84.

32. Millar AB, Patou G, Miller RF, et al. Cytomegalovirus in the lungs of patients with

AIDS. Respiratory pathogen or passenger? Am Rev Respir Dis 1990;141:1474-7

33. d'Arminio Monforte A, Mainini F, Testa L, et ai. Predictors of cytomegalovirus

disease, natural history and autopsy findings in a cohort of patients with AIDS.

Aids 1997;11:517-24

34. Barry SM, Lipman MC, Johnson MA and Prentice HG. Respiratory infections in

immunocompromised patients. Curr Opin Pulm Med 1999;5:168-73.

35. Zeevi A, Morel P, Spichty K, et ai. Clinical significance of CMV-specific I helper

responses in lung transplant recipients. Hum Immunol 1998;59:768-75

36. Semenzato G, Chilosi M, Ossi E, et ai. Bronchoalveolar lavage and lung

histology. Comparative analysis of inflammatory and immunocompetent cells in

patients with sarcoidosis and hypersensitivity pneumonitis. Am Rev Respir Dis

1985;132:400-4.

37. Campbell DA, Poulter LW and du Bois RM. Immunocompetent cells in

bronchoalveolar lavage reflect the cell populations in transbronchial biopsies in

pulmonary sarcoidosis. Am Rev Respir Dis 1985;132:1300-6.

38. Drent M, Wagenaar SS, Mulder PH, et ai. Bronchoalveolar lavage fluid profiles in

sarcoidosis, tuberculosis, and non-Hodgkin's and Hodgkin's disease. An

evaluation of differences. Chest 1994;105:514-9.

39. Bronchoalveolar lavage constituents in healthy individuals, idiopathic pulmonary

fibrosis, and selected comparison groups. The BAL Cooperative Group Steering

Committee. Am Rev Respir Dis 1990;141:8169-202.

40. Hoheisel GB, Tabak L, Teschler H, et ai. Bronchoalveolar lavage cytology and

immunocytology in pulmonary tuberculosis. Am J Respir Crit Care Med

1994;149:460-3.

32

Page 36: Lymphocyte Responses in the Lung in Patients with ...

41. Ainslie GM, Solomon JA and Bateman ED. Lymphocyte and lymphocyte subset

numbers in blood and in bronchoalveolar lavage and pleural fluid in various forms

of human pulmonary tuberculosis at presentation and during recovery. Thorax

1992;47:513-8.

42. Baughman RP, Dohn MN, Loudon RG and Frame FT. Bronchoscopy with

bronchoalveolar lavage in tuberculosis and fungal infections. Chest 1991;99:92-

7.

43. Poletti V, Cazzato 8, Minicuci N, at a/. The diagnostic value of bronchoalveolar

lavage and transbronchial lung biopsy in cryptogenic organizing pneumonia. Eur

Respir J 1996;9:2513-6.

44. Hunninghake GW, Kawanami O, Ferrans VJ, et al. Characterization of the

inflammatory and immune effector cells in the lung parenchyma of patients with

interstitial lung disease. Am Rev Respir Dis 1981;123:407-12.

45. Haslam PL, Poulter LW, Rossi GA, et al. The clinical role of BAL in idiopathic

pulmonary fibrosis. Eur Respir J 1990;3:940-2, 961-9.

46. Costabel U, Guzman J. Bronchoalveolar lavage in interstitial lung disease. Curr

Opin Pulm Med 2001 ;7:255-61.

47. Mandy F NJ, Autran B and Janossy G. T-Cell Subset Counting and Fight Against

AIDS: Reflections Over a 20-Year Struggle. Clinical Cytometry 2002;50:39-45

48. Loken MR, Brosnan JM, Bach BA and Ault KA. Establishing optimal lymphocyte

gates for immunophenotyping by flow cytometry. Cytometry 1990;11:453-9.

49. Glencross DK SL, Jani IV, Barnett D and Janossy G. CD45-Assisted

Panleukogating for Accurate, Cost-Effective Dual-Platform CD4+ T-Cell

Enumeration. Cytometry (Clinical Cytometry) 2002;50:69-78

50. Janossy G Jl, Kahan M, Barnett D, Mandy F, and Shapiro H. Precise CD4 T-Cell

Counting Using Red Diode Laser Excitation: For Richer, for Poorer. Cytometry

(Clinical Cytometry) 2002;50:78-85.

51. Pizzolo G, Sloane J, Beverley P, et ai. Differential diagnosis of malignant

lymphoma and nonlymphoid tumors using monoclonal anti-leucocyte antibody.

Cancer 1980;46:2640-7.

52. Gatter KC, Abdulaziz Z, Beverley P, et al. Use of monoclonal antibodies for the

histopathological diagnosis of human malignancy. J Clin Pathol 1982;35:1253-67.

33

Page 37: Lymphocyte Responses in the Lung in Patients with ...

53. Padovan CS, Behr J, Allmeling AM, et al. Immunophenotyping of lymphocyte

subsets in bronchoalveolar lavage fluid. Comparison of flow cytometric and

immunocytochemical techniques. J Immunol Methods 1992;147:27-32.

54. Brandt B, Thomas M, von Eiff M and Assmann G. Immunophenotyping of

lymphocytes obtained by bronchoalveolar lavage: description of an all-purpose

tricolor flow cytometric application. J Immunol Methods 1996;194:95-102.

55. Dauber JH, Wagner M, Brunsvold S, et al. Flow cytometric analysis of

lymphocyte phenotypes in bronchoalveolar lavage fluid: comparison of a two-

color technique with a standard immunoperoxidase assay. Am J Respir Cell Mol

B/o/1992;7:531-41.

56. Aubin JE. Autofluorescence of viable cultured mammalian cells. J Histochem

Cytochem 1979;27:36-43.

57. Umino I , Skold CM, Pirruccello SJ, et al. Two-colour flow-cytometric analysis of

pulmonary alveolar macrophages from smokers. Eur Respir J 1999;13:894-9.

58. Pulford KA, Rigney EM, Micklem KJ, et al. KP1 : a new monoclonal antibody that

detects a monocyte/macrophage associated antigen in routinely processed tissue

sections. J Clin Pathol 1989;42:414-21.

59. Altman JD, Moss PAH, Goulder PJR, et al. Phenotypic analysis of antigen-

specific T lymphocytes Science 1996;274:94-6

60. Maine VC, Picker LJ. Identification of functional subsets by flow cytometry:

intracellular detection of cytokine expression. Cytometry 1998;34:207-15

61. Rowland-Jones SL, Dong T, Fowke KR, et al. Cytotoxic T cell responses to

multiple conserved HIV epitopes in HIV-resistant prostitutes in Nairobi. J Clin

Invest 1998;102:1758-65.

62. Callan MF, Tan L, Annels N, et al. Direct visualization of antigen-specific CD8+ T

cells during the primary immune response to Epstein-Barr virus In vivo. J Exp

/Wed 1998;187:1395-402

63. Ogg GS, Jin X, Bonhoeffer 8, et al. Quantitation of HIV-1-specific cytotoxic T

lymphocytes and plasma load of viral RNA. Science 1998;279:2103-6

64. Matsui K, Boniface JJ, Steffner P, et al. Kinetics of T-cell receptor binding to

peptide/l-Ek complexes: correlation of the dissociation rate with T-cell

responsiveness. Proc Natl Acad Sci U S A 1994;91:12862-6.

65. Valitutti 8, Muller 8, Celia M, et al. 8erial triggering of many T-cell receptors by a

few peptide-MHC complexes. Nature 1995;375:148-51.

34

Page 38: Lymphocyte Responses in the Lung in Patients with ...

66. McMichael AJ, O'Callaghan CA. A new look at T cells. J Exp Med

1998;187:1367-71.

67. Novak EJ, Liu AW, Gebe JA, at a i Tetramer-guided epitope mapping: rapid

identification and characterization of immunodominant CD4+ T cell epitopes from

complex antigens. J Immunol 2001;166:6665-70.

68. Gallimore A, Glithero A, Godkin A, at a i Induction and exhaustion of lymphocytic

choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble

tetrameric major histocompatibility complex class l-peptide complexes. J Exp

/Wed 1998;187:1383-93.

69. Zajac AJ, Blattman JN, Murali-Krishna K, at a i Viral immune evasion due to

persistence of activated T cells without effector function. J Exp Mad

1998;188:2205-13.

70. Lee PP, Yee C, Savage PA, at a i Characterization of circulating T cells specific

for tumor-associated antigens in melanoma patients. Nat Mad 1999;5:677-85.

71. Appay V, Nixon DF, Donahoe SM, at a i HIV-specific CD8(+) T cells produce

antiviral cytokines but are impaired in cytolytic function. J Exp Mad 2000;192:63-

75

72. Bitmansour AD, Waldrop SL, Pitcher CJ, at a i Clonotypic structure of the human

CD4+ memory T cell response to cytomegalovirus. J Immunol 2001;167:1151-63.

73. Cwynarski K, Ainsworth J, Cobbold M, at a i Direct visualization of

cytomegalovirus-specific T-cell reconstitution after allogeneic stem cell

transplantation. Blood 2001 ;97:1232-40.

74. Komanduri KV, Donahoe SM, Moretto WJ, at a i Direct measurement of CD4+

and CD8+ T-cell responses to CMV in HIV-1-infected subjects. Virology

2001;279:459-70.

75. Engstrand M, Tournay C, Peyrat MA, at a i Characterization of CMVpp65-specific

CD8+ T lymphocytes using MHC tetramers in kidney transplant patients and

healthy participants. Transplantation 2000;69:2243-50

76. Lalvani A, Brookes R, Hambleton S, at a i Rapid effector function in CD8+

memory T cells. J Exp Mad 1997;186:859-65.

77. Petrovsky N, Harrison LC. Cytokine-based human whole blood assay for the

detection of antigen-reactive T cells. J Immunol Methods 1995;186:37-46.

35

Page 39: Lymphocyte Responses in the Lung in Patients with ...

78. Lalvani A, Pathan AA, McShane H, et al. Rapid detection of Mycobacterium

tuberculosis infection by enumeration of antigen-specific T cells. Am J Respir Crit

Care Med 2001;163:824-8.

79. Ozenci V, Kouwenhoven M, Press R, et ai. IL-12 elispot assays to detect and

enumerate IL-12 secreting cells. Cytokine 2000;12:1218-24.

80. Badovinac VP, Harty JT. Intracellular staining for TNF and IFN-gamma detects

different frequencies of antigen-specific CD8(+) T cells. J immunoi Methods

2000;238:107-17.

81. De Rosa SC, Roederer M. Eleven-color flow cytometry. A powerful tool for

elucidation of the complex immune system, d in Lab Med 2001;21:697-712, vii.

82. Nakagawa TY, Rudensky AY. The role of lysosomal proteinases in MHC class II-

mediated antigen processing and presentation, immunoi Rev 1999;172:121-9.

83. Maecker H I, Dunn MS, Suni MA, et ai. Use of overlapping peptide mixtures as

antigens for cytokine flow cytometry. J immunoi Methods 2001;255:27-40.

84. Kern F, Faulhaber N, Frommel C, et ai. Analysis of CD8 T cell reactivity to

cytomegalovirus using protein-spanning pools of overlapping pentadecapeptides.

Eur J immunoi 2000;30:1676-82.

85. Tan LC, Mowat AG, Fazou C, et ai. Specificity of T cells in synovial fluid: high

frequencies of CD8(+) T cells that are specific for certain viral epitopes. Arthritis

Res 2000;2:154-64.

86. Schon-Hegrad MA, Oliver J, McMenamin PG and Holt PG. Studies on the

density, distribution, and surface phenotype of intraepithélial class II major

histocompatibility complex antigen (la)-bearing dendritic cells (DC) in the

conducting airways. J Exp Med 1991;173:1345-56.

87. Sinigaglia F, D'Ambrosio D, Panina-Bordignon P and Rogge L. Regulation of the

IL-12/IL-12R axis: a critical step in T-helper cell differentiation and effector

function, immunoi Rev 1999;170:65-72.

88. Murphy KM, Guyang W, Farrar JD, et ai. Signaling and transcription in T helper

development. Annu Rev immunoi 2000;18:451-94.

89. O'Garra A. Cytokines induce the development of functionally heterogeneous T

helper cell subsets, immunity 1998;8:275-83.

90. Stumbles PA, Thomas JA, Pimm CL, et ai. Resting respiratory tract dendritic cells

preferentially stimulate T helper cell type 2 (Th2) responses and require

36

Page 40: Lymphocyte Responses in the Lung in Patients with ...

obligatory cytokine signals for induction of Th1 immunity. J Exp Med

1998;188:2019-31.

91. van der Pouw Kraan TC, Boeije LC, Smeenk RJ, et al. Prostaglandin-E2 is a

potent inhibitor of human interleukin 12 production. J Exp Med 1995;181:775-9.

92. Huang FP, Niedbala W, Wei XQ, et al. Nitric oxide regulates Thi cell

development through the inhibition of IL-12 synthesis by macrophages. EurJ

Immunol 1998;28:4062-70.

93. van der Pouw Kraan TC, Snijders A, Boeije LC, et al. Histamine inhibits the

production of interleukin-12 through interaction with H2 receptors. J Clin Invest

1998;102:1866-73.

94. Panina-Bordignon P, Mazzeo D, Lucia PD, et al. Beta2-agonists prevent Thi

development by selective inhibition of interleukin 12. J Clin Invest

1997;100:1513-9.

95. Holt PG. Antigen presentation in the lung. Am J Respir Crit Care Med

2000;162:S151-6.

96. Chelen CJ, Fang Y, Freeman GJ, et al. Human alveolar macrophages present

antigen ineffectively due to defective expression of B7 costimulatory cell surface

molecules. J Clin Invest 1995;95:1415-21.

97. Blumenthal RL, Campbell DE, Hwang P, et al. Human alveolar macrophages

induce functional inactivation in antigen-specific CD4 T cells. J Allergy Clin

Immunol 2001;107:258-64.

98. Vecchiarelli A, Dottorini M, Pietrella D, et al. Role of human alveolar

macrophages as antigen-presenting cells in Cryptococcus neoformans infection.

Am J Respir Cell Mol Biol 1994; 11:130-7.

99. van Haarst JM, Hoogsteden HC, de Wit HJ, et al. Dendritic cells and their

precursors isolated from human bronchoalveolar lavage: immunocytologic and

functional properties. Am J Respir Cell Mol Biol 1994;11:344-50.

100. Havenith CE, van Haarst JM, Breedijk AJ, et al. Enrichment and characterization

of dendritic cells from human bronchoalveolar lavages. Clin Exp Immunol

1994;96:339-43.

101. Havenith CE, Breedijk AJ and Hoefsmit EC. Effect of Bacillus Calmette-Guerin

inoculation on numbers of dendritic cells in bronchoalveolar lavages of rats.

Immunobiology 1992;184:336-47.

37

Page 41: Lymphocyte Responses in the Lung in Patients with ...

102. Henderson RA, Watkins SC and Flynn JL. Activation of human dendritic cells

following infection with Mycobacterium tuberculosis. J Immunol 1997;159:635-43.

103. Demangel C, Bean AG, Martin E, et al. Protection against aerosol

Mycobacterium tuberculosis infection using Mycobacterium bovis Bacillus

Calmette Guerin-infected dendritic cells. Eur J Immunol 1999;29:1972-9.

104. D'Ambrosio D, Mariani M, Panina-Bordignon P and Sinigaglia F. Chemokines

and their receptors guiding T lymphocyte recruitment in lung inflammation. Am J

Respir Crit Care Med 2001 ; 164:1266-75.

105. Sinigaglia F, D'Ambrosio D. Regulation of helper T cell differentiation and

recruitment in airway inflammation. Am J Respir Crit Care Med 2000;162:S157-

60.

106. Sallusto F, Mackay CR and Lanzavecchia A. The role of chemokine receptors in

primary, effector, and memory immune responses. Annu Rev Immunol

2000;18:593-620.

107. Cyster JG. Leukocyte migration: scent of the I zone. Curr Biol 2000;10:R30-3.

108. Cyster JG. Chemokines and the homing of dendritic cells to the I cell areas of

lymphoid organs. J Exp Med 1999;189:447-50.

109. Akbar AN, Terry L, Timms A, et al. Loss of CD45R and gain of UCHL1 reactivity

is a feature of primed T cells. J Immunol 1988;140:2171-8.

110. Trowbridge IS, Thomas ML. CD45: an emerging role as a protein tyrosine

phosphatase required for lymphocyte activation and development. Annu Rev

Immunol 1994;12:85-116.

111. Hendriks J, Gravestein LA, Tesselaar K, et al. CD27 is required for generation

and long-term maintenance of T cell immunity. Nat Immunol 2000,^ :433-40.

112. Hamann D, Baars PA, Rep MH, et al. Phenotypic and functional separation of

memory and effector human CD8+ T cells. J Exp Med 1997;186:1407-18.

113. Sallusto F, Lenig D, Forster R, et al. Two subsets of memory T lymphocytes with

distinct homing potentials and effector functions. Nature 1999;401:708-12.

114. Agostini C, Facco M, Siviero M, et al. CXC chemokines IP-10 and mig expression

and direct migration of pulmonary CD8+/CXCR3+ T cells in the lungs of patients

with HIV infection and T-cell alveolitis. Am J Respir Crit Care Med

2000;162:1466-73.

38

Page 42: Lymphocyte Responses in the Lung in Patients with ...

115. Condos R, Rom WN, Liu YM and Schluger NW. Local immune responses

correlate with presentation and outcome in tuberculosis. Am J Respir Crit Care

Med 1998;167:729-35.

116. Faith A, Schellenberg DM, Rees AD and Mitchell DM. Antigenic specificity and

subset analysis of T cells isolated from the bronchoalveolar lavage and pleural

effusion of patients with lung disease. Clin Exp Immunol 1992;87:272-8.

117. Schwander SK, Torres M, Sada E, et ai. Enhanced responses to Mycobacterium

tuberculosis antigens by human alveolar lymphocytes during active pulmonary

tuberculosis. J Infect Dis 1998;178:1434-45.

118. Chackerian AA, Perera TV and Behar SM. Gamma interferon-producing CD4+ T

lymphocytes in the lung correlate with resistance to infection with Mycobacterium

tuberculosis. Infect Immun 2001;69:2666-74.

119. Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in

resistance to Mycobacterium tuberculosis infection. J Exp Med 1993;178:2249-

54.

Page 43: Lymphocyte Responses in the Lung in Patients with ...

2. Chapter 2

Methods

40

Page 44: Lymphocyte Responses in the Lung in Patients with ...

2.1 IntroductionThe methods described in this chapter cover the techniques used in the thesis as a

whole. Bronchoscopy and cytospin preparation are comrnonly used and the details of

these methods are well established and therefore given only a brief description here. By

contrast, considerable attention has been devoted in this thesis to describing flow

cytometry in general and CD45-directed gating in particular. The method for determining

antigen-specific analysis in BAL has also been described in detail including modifications

of this technique to enable it to be applied to tissue fluids such as BAL.

2.2 FIbreoptic bronchoscopy and bronchoalveolar lavageBronchoscopy was performed in a fully equipped endoscopy suite at the Royal Free

Hospital. British Thoracic Society (BTS) guidelines [1] regarding the safe practice of

bronchoscopy were adhered to. Bronchoscopies were generally performed through the

oral, rather than nasal approach which was more comfortable for the patient.

Bronchoalveolar lavage (BAL) was performed with a maximum of 200ml of warmed,

sterile 0.9% normal saline introduced in aliquots of 20 or 30ml. The bronchoscope was

wedged into a subsegmental bronchus directed to an area of radiological abnormality. In

patients or control subjects with radiologically normal lung parenchyma, standard BAL

was performed from the right middle lobe.

Bottles for acquiring BAL were siliconized glass containers that had been autoclaved

to ensure sterility prior to the procedure. Immediately following acquisition of the sample,

the bottles were placed on ice and sent to the laboratory.

2.3 Preparation of BALThe BAL specimen was kept on ice and all samples were analysed within two hours of

their acquisition. BAL was performed for the investigation of respiratory disease and the

appropriate samples were therefore sent to the relevant diagnostic laboratories. BAL

specimens were divided, placed into universal containers and sent to microbiology,

cytology and virology laboratories in most cases. The remaining BAL was used for

immunological analysis. BAL was centrifuged at 430g for 8 minutes and decanted. The

pellet was resuspended in phosphate buffered saline (PBS) and filtered through a

100pm filter (CellTricks, Partec GmBH, Munster, Germany) and centrifuged again. The

pellet was resuspended up to 1ml in either culture medium (RPMI 1640 with 10% PCS)

41

Page 45: Lymphocyte Responses in the Lung in Patients with ...

or PBS depending on whether simple leukocyte differentials were to be determined or

whether further culture and functional assays were planned.

2.4 CytospinsCytospin preparations were made in order to obtain BAL leukocyte differentials which

could then be compared with those obtained by a flow cytometric method. Following the

washing and filtering of BAL described in 2.3 above, an absolute cell count of the

number of leukocytes/ml was obtained using the CytoronAbsolute flow cytometer (see

below). A 50|xl aliquot of the BAL suspension containing 3-5 X 10® cells/ml was then

used to prepare a cytospin slide by standard methods. 50pl of BAL was introduced onto

glass microscope slides placed within a cytospin machine (Shandon cytospin 11,

Shandon scientific Ltd, Runcorn, UK) and spun for two minutes at 800 rpm. The slides

were then air-dried for one hour and fixed for ten minutes in a 50:50 mixture of

chloroform and acetone before air drying again.

The slides were then stained using a modified May-Grunwald Giemsa stain

(DiffQuik), left to dry and then mounted in DPX (BDH Chemicals Ltd, Poole, Dorset).

Formal cell differentiation into lymphocytes, alveolar macrophages, neutrophils,

eosinophils and any other cell types was then performed by light microscopy (figure 2.1)

by an expert cytologist. 500 leukocytes were counted per sample and the leukocyte

differentials recorded.

2.5 Immunofluoresence stainingIn several samples, immunofluoresence staining was performed on cytospin

preparations in order to validate the use of antibodies to discriminate eosinophils and

neutrophils by FCM. Cytospins were made as described above but were not fixed and

dried. Immunofluoresence staining was performed in a moist staining chamber to ensure

that the cytospins did not dry out. 50pl of the following antibodies: CD15 FITC (Dako,

Ely, UK) and CD23 FITC (Caltag Medsystems, Towcester, UK) both at a dilution of 1:10

with PBS were added to the cytospin preparations. Staining was performed in the dark

for 45 minutes and then the cytospins were washed twice with PBS before fixing in 4%

paraformaldehyde for five minutes. After fixation, the slides were washed again with PBS

and then examined using a fluorescence microscope with barrier filters appropriate for

FITC conjugated antibody staining.

42

Page 46: Lymphocyte Responses in the Lung in Patients with ...

Figure 2.1

#

Lymphocyte

W

Macrophage m

Neutrophil% t f t \

#* \

Eosinophil

m

Figure 2.1

Digital photographs of stained cytospin preparations from BAL at 400x

magnification. The morphological and staining characteristics of the major BAL

cell populations-alveolar macrophages, lymphocyte and neutrophils (a) and

eosinophils (b) are shown.

43

Page 47: Lymphocyte Responses in the Lung in Patients with ...

2.6 Flow cytometry: general introduction and gating strategies

2.6.1 General characteristics of the fiow cytometers used

FCM was performed by both three colour (CytoronAbsolute, Ortho diagnostics, High

Wycombe, UK) and four colour machines (FACSCalibur, Beckton Dickinson, San Diego,

California, USA). The CytoronAbsolute acquired a known volume of sample and

therefore an absolute count of the number of cells acquired was determined by

volumetry. From this and the total volume of the wash-outs, the absolute number of cells

in the original sample could be calculated. This flow cytometer was employed when

absolute cell counts were required for antigen-specific analysis (see below). The

FACSCalibur machine was utilized to define the percentages of the cells expressing a

given phenotype within a population with the increased discrimination of 4-colour

immunofluoresence. Instead of using microspheres on the FACSCalibur, the absolute

counts on the Cytoron in parallel samples were obtained for major populations of CD4

and CDS T lymphocytes when absolute numbers were required.

2.6.2 Mechanisms of analyte discrimination by FCM

Flow cytometry discriminates cells or other analytes by the virtue of both their size

and granularity and also by detecting antibodies bound to their surfaces or intracellular

components. Cells in suspension pass through a laser beam and this beam is scattered

dependant on the physical qualities of the cells. Size is measured by the forward

deflection of the laser beam when it hits the particles and is referred to as the forward

scatter (FSC). Some of the laser beam is deflected at right angles and the amount of this

reflected light corresponds to the intracellular features or granularity of the cell. This light,

detected by a separate photomultiplier is referred to as side scatter (SSC). Taken

together, size and granularity refer to the intrinsic qualities of a cell (or other analyte).

These intrinsic features can be used to differentiate the major leukocyte populations in

lysed whole blood (figure 2.2)

The second mechanism by which FCM can discriminate cells is through the

detection of light released by fluorochrome labelled monoclonal antibodies. Different

fluorochromes exist that absorb light from the laser beam and emit fluorescent light

within a particular wavelength band that is detected by different photomultiplier

detectors. Thus cells may be distinguished by their fluorescence characteristics

44

Page 48: Lymphocyte Responses in the Lung in Patients with ...

dependant on which monoclonal antibodies are bound to their surface. This

fluorochrome labelled antibody discrimination of a cell relates to its extrinsic features.

The investigation of both the intrinsic and extrinsic characteristics of an analyte may be

F ig u re 2.2

oCO

termed morphospectral analysis.SSC

Figure 2.2

FCM dotplot of lysed whole blood demonstrating that the intrinsic cell parameters

size (forward scatter, FSC) and granularity (side scatter, SSC) can be used to

distinguish lymphocytes (a), monocytes (b) and granulocytes (c)

The number and type of different fluorochromes that can be used is dependant on

their various absorption and emission spectra as well as the number of lasers present in

the machine. A single laser machine such as the CytoronAbsolute can perform analysis

with three fluorochromes, whereas with the FACSCalibur, the two lasers allow the

routine use of four fluorochromes (table 2.1). The use of four fluorochromes is not only

more informative than three colour analysis but is also more economical because fewer

tubes are required to investigate complex patterns of differentiation antigen expression.

The four colour analysis is also important in BAL where lymphocyte populations may be

scanty.

45

Page 49: Lymphocyte Responses in the Lung in Patients with ...

Table 2.1 Fluorochromes available for use between the CytoronAbsolute and

FACSCalibur flow cytometers

Detector Cytoron FACSCalibur

1 FITC' FITC

2 PE^ PE

3 PEcyS, PerCP^ PerCP, PEcyS.S, PEcy7

4 - APC^

Footnotes

1. FITC=Fluorescein isothiocyanate

2. PE=Phycoerythrin

3. PerCP=Perididine chlorophyll protein

4. APC=Allophycocyanin

2.6.3 CD45 directed panleukogating in blood and BAL

A number of different strategies have been developed for the discrimination of leukocyte

subsets by FCM. Lymphocytes, monocytes, neutrophils and eosinophils all carry unique

antigens by which they may be distinguished flow cytometrically using monoclonal

antibodies. However, the use of large numbers of different fluorochrome labelled

antibodies often introduces unnecessary complications and cost to the analysis. More

recently, the features of CD45 staining, well established since 1979 [2] when the first

CD45 reagent was made are coming back to prominence for two reasons: their simplicity

and their reliability on stored samples. These simple protocols investigate both the

intrinsic and extrinsic parameters of analytes. CD45-directed gating allows leukocytes to

be distinguished from non-leukocyte parenchymal cells and debris, a factor that is

particularly important for the analysis of tissue fluids such as BAL where many of the

acquired events are not leukocytes.

One of the basic features of CD45 staining is that on leukocyte populations in blood,

CD45 is expressed at high density on lymphocytes with medium density on monocytes

and more weakly on granulocytes. This feature, combined with the differences in tbe

46

Page 50: Lymphocyte Responses in the Lung in Patients with ...

intrinsic parameters of these populations allows CD45 and side scatter to precisely

differentiate these major populations in lysed whole blood (figure 2.3).

It is not known however, whether these features of discriminating intensity of

staining are maintained in BAL samples where in this thesis the various aspects of CD45

labelling will be investigated (see below). It appears that when using BAL lymphocytes

form an easily gateable population but the alveolar macrophages cannot be easily

distinguished from the granulocytes since macrophages have very heterogenous side

scatter characteristics (figure 2.4). Clearly the preliminary observations indicate that

additional granulocyte markers such as CD15 may be needed to achieve macrophage-

granulocyte discrimination.

Figure 2.3

LO

OU

SSC

Figure 2.3

FCM dotplot of CD45 panleukogating against side scatter (SSC) to differentiate the

major leukocyte populations in lysed whole blood. Lymphocytes (a) have low side

scatter and express CD45 brightly whilst granulocytes (c) have high side scatter

but are CD45 dim. Monocytes (b) are intermediate for both characteristics.

47

Page 51: Lymphocyte Responses in the Lung in Patients with ...

F i g u r e 2.4

in é

QO &

ssc1023

Figure 2.4

FCM dotpiot from BAL demonstrating the separation of CD45+ leukocytes from

CD45- non-leukocyte debris. CD45 bright lymphocytes form a gateable population

due to their low side scatter, but the other BAL populations cannot be clearly

distinguished.

2.6.4 Gating strategy to identify bronchial epithelial and squamous cells in

BAL by FCM

Effective BAL adequately samples the alveolar cellular component of the washed lobe.

However, BAL may be of variable quality and often specimens may include high

proportions of bronchial epithelial and squamous cells, indicating sampling from the

airways rather than the alveoli. These cells are readily identifiable by light microscopy,

thereby enabling the cytologist to comment on the adequacy of the BAL sample.

Therefore, it was felt that that a flow cytometric system should be devised that could also

identify these cells from the non-leukocyte component.

An epithelial marker conjugated to FITC (Ber-EP4, Dako) was used in tandem with

CD45. This epithelial antigen consists of two glycoproteins of 34 and 39 Kda and is

expressed on a broad range of epithelial tissues, but not on mésothélial cells [3]. In poor

quality BAL the epithelial+ cells could be gated and were demonstrated to be CD45-

(figure 2.5).

48

Page 52: Lymphocyte Responses in the Lung in Patients with ...

Figure 2.5

10* 1 0 ’ 10^ 1 0 ’ 10*

Epithelial

Bronchial epithelial cell

. '

Squamous cell

Epithelia l '

d• 1

• . ^

■ #

Figure 2.5

FCM dotpiots and images of cytospins of BAL indicating upper airways cellular

contamination (a,c) and a good alveolar specimen (b,d). The gated populations in

the dotpiots represent the epithelial+, CD45- bronchial epithelial and squamous

cell component. The staining aligned around the 45° is a feature of non-specific

labeling, including dead cells and debris.

2.6.5 General gating strategy for lymphocyte phenotypic analysis: primary

immunological gating

For all lymphocyte phenotypic analysis by FCM, primary immunological gating using the

relevant discriminatory monoclonal antibody (CD4, CDS or CDS) against side scatter

was performed. These gated events were subsequently sent to a lymphoid scatter gate.

Events that lay outside this gate were excluded from the analysis. Care was taken to

include larger lymphoid blast cells (figure 2.6). The events with lower forward scatter

49

Page 53: Lymphocyte Responses in the Lung in Patients with ...

lying outside this gate were assumed to be apoptotic lymphocytes. This strategy of

primary immunological gating followed by back gating to assess scatter characteristics

has been demonstrated to be the optimum method for flow cytometric discrimination of

lymphocyte and leukocyte subpopulations [2, 4], Further strategies for lymphocyte gating

are discussed in the relevant results chapters.

Figure 2.6

ssc

R:

Gated by R1

FSC

Figure 2.6

Flow cytometric gating strategy for lymphocyte subsets for phenotypic analysis.

Primary immunological gating of CD4+ events with low side scatter (R1, plot a) are

then sent to a forward scatter, side scatter plot (b) to ensure that they lie within a

characteristic lymphoid gate (R2). Events with high FSC within R2 are blast cells.

2.7 Flow cytometry: Reagents, panels and protocols

2.7.1 Reagents and panels for three and four colour FCM

The monoclonal antibodies, their manufacturers and the fluorochromes to which they

were conjugated to are shown in table 2.2. All antibodies were used in optimised pre­

titrated saturating concentrations.

50

Page 54: Lymphocyte Responses in the Lung in Patients with ...

Table 2.2 Monoclonal antibodies used In this thesis

CD Fluorochrome Clone Isotype Manufacturer Staining type

CD45 FITC 2D1 igGi Southern Biotechnology

CD45 ARC H130 igGi BD PharMingen

CD15 FITC C3D-1 igM Dako

CD15 PE V 1M C 6 igM Caltag

CD15 FITC MCS-1 igG Cytognos

CD23 FITC TU 1 igG3 Caltag

CD4 FITC RFT-4 igGi Royal Free Hospital

CD3 PEcyS UCHT1 igGi Dako

CD3 PEcyS.5 S4.1 lgG2a Caltag

MembraneCD3 Pecy7 S4.1 lgG2a Caltag

CD8 PE RFT-8 lgG1 Royal Free Hospital

CD8 PEcy5.5 3B5 lgG2a Caltag

CD8 Pecy7 3B5 lgG2a Caltag

CD27 FITC CLB-27/1 lgG2a Caltag

CD45RA ARC Sn130 lgG1 Southern Biotechnology

CD38 PE HIT2 lgG1 Caltag

CD56 FITC NCAM16.2 lgG1 BD

CD22 PE S-HCL-1 lgG2b BD

Epithelial FITC Ber-EP4 lgGl Dako

KI67 FITC MIB-1 lgGl Immunotech

Perforin PE ÔG9 lgG2b BD PharMingenIntracellular

IFN-y PE B27 lgGl Caltag

TN F-a ARC M P9-20A4 igGl Caltag

51

Page 55: Lymphocyte Responses in the Lung in Patients with ...

The monoclonal antibodies were used in panels of three reagents for the Cytoron, or four

reagents for the FACSCalibur. On the Cytoron, the following panels were used:

1. CD45 FITC / CD15 PE / 7-AAD

2. CD4 FITC / CD8 PE/ CD3 PEcyS

For the FACSCalibur the following panels were used:

3. CD23 FITC / CD15 PE / CD45 APC

4. Epith FITC / CD45 APC

5. CD15 FITC / CD4 PE / CD8 PEcy7 / CD45 APC

6 . CD4 FITC / CD8 PE / CD3 PEcy7 / CD45 APC

7. CD56 FITC / CD22 PE / CD3 PerCP / CD45 APC

8 . CD27 FITC / Perforin PE / CD8 PerCP / CD45RA APC

9. CD27 FITC / CD8 PE / CD3 PerCP / CD45RA APC

10. KI67 FITC / CD38 PE / CD8 PerCP / CD45RA APC

11. CD4 FITC / IFN-y PE / CD3 PerCP / TNF-a APC

2.7.2 Protocols for staining of fresh whole blood and BAL

Parallel blood and BAL samples were run using panels 1 and 2 above to determine the

leukocyte subsets and CD4/CD8 ratios in these fluids. 50pl of whole blood or 50pl of

prepared BAL (see 2.3) was added to the antibody panels in separate flow cytometry

tubes. The samples were stained at room temperature in the dark for 15 minutes.

Following this QSOpI of lysis buffer (ammonium chloride 8.26%, potassium bicarbonate

1% and EDTA tetra sodium salt 0.036%, pH 7.5) was added to the blood and 950pl of

PBS added to the BAL. The samples were left for a further 15 minutes at room

temperature to allow lysis of the red cells and then run immediately on the Cytoron using

an absolute counting protocol.

Absolute cell counts of the CD4 and CD8 lymphocyte subsets as well as the

CD4/CD8 ratio, and the percentages of lymphocytes granulocytes and

monocytes/macrophages were calculated for both blood and BAL (see chapter 3 for

further discussion)

2.7.3 Intracellular staining by FCM: fixation and permeabilisation of cells

In addition to characterising the phenotype of cells by their surface antibody staining,

intracellular components could also be identified following permeabilisation of the cells.

This technique was necessary for demonstrating the presence of cytokines following

52

Page 56: Lymphocyte Responses in the Lung in Patients with ...

incubation with antigen or for the presence of cytotoxic markers such as perforin or

nuclear markers of cell proliferation such as Ki67. Several different techniques exist for

the permeabilisation of cells [5].

In this study fixation and permeabilisation was performed using fix and perm (Caltag).

200|il of reagent A (fix) was added to SOOpI of whole blood or BAL in a universal

container and left for 15 minutes in the dark at room temperature. The samples were

then washed in PBS at 430g for 8 minutes. Following the wash step, the containers were

decanted allowing red cells to separate away from the cell pellet. 200pl of reagent B

(perm) was then added, the samples vortexed and then incubated in the dark for a

further 15 minutes. Following a second wash step, the pellet was resuspended up to

200pl with PBS and then added to the relevant antibody panel requiring intracellular

staining (panels 5 and 6 ). Staining was performed at 4°C for 30 minutes and followed by

a final wash step. By this stage almost all red cells were removed during the decanting

phases. Samples were run on the FACSCalibur machine.

2.8 Measurement of antigen-specific responses: cytokine

synthesis assay2.8.1 General introduction to the method

Several investigators have described methods for the detection of antigen specific

lymphocyte responses by measuring the production of cytokines in response to antigen

added in assays of whole blood or PBMC [6-9]. These assays used short (6-48) hour

incubations and the cytokines synthesised were measured by ELISA [6 ], visually by

detecting spot forming colonies in a gel matrix [8 , 10], or by flow cytometry [7].

The underlying assumption in all of these systems is that cognate T lymphocytes

present in the blood samples respond to antigen by producing cytokines. Therefore the

measurement of the proportion of lymphocytes synthesising cytokines is a measure of

the antigen-specific response. The caveat to this assumption is that a variety of different

cytokines may be produced following the encounter of cognate lymphocytes with antigen

and usually only one, or sometimes two cytokines are actually measured. This method

may therefore underestimate the true population of antigen-specific responses.

Nevertheless, these techniques have distinct advantages over the use of class 1 HLA

tetramers to detect antigen-specific responses since they do not require HLA matching

53

Page 57: Lymphocyte Responses in the Lung in Patients with ...

of the subject with the tetramer. Moreover, the detection of cytokine synthesis gives an

indication of the functionality of the responding lymphocytes.

FCM is a powerful technique that enables the rapid and accurate elucidation of both

the subtypes of responding lymphocyte (CD4 or CD8 ) as well as being able to

discriminate a number of different cytokines synthesised by the addition of further

fluorochrome labelled anti-cytokine antibodies.

The flow cytometric method, first described by Suni, Picker and Maino [7] used a

whole blood assay with a short incubation period of six hours in total before the samples

were lysed, fixed and permeabilised and stained prior to acquisition on the flow

cytometer. Integral to this method was the addition of the co-stimulatory antibody CD28

to augment the cytokine responses and the secretion blocking agent Brefeldin A to keep

the synthesised cytokines within lymphocytes [1 1 , 1 2 ], and therefore optimise their

detection by FCM following cell permeabilisation.

The aim was to modify and simplify the existing described method so that it would be

appropriate for use in bronchoalveolar lavage. One important consideration was that the

timings of the assay should be appropriate for the analysis of samples of BAL that were

collected following the routine clinical bronchoscopy lists.

The following issues were addressed when adapting this method for the

detection of antigen specific responses in BAL;

1. What was the optimum incubation time with antigen in order to maximise cytokine

synthesis?

2. Should a marker of cell activation such as CD69 be used in BAL?

3. What was the dose response curve for PPD?

4. What was the best sequence of staining of surface antigens in order to achieve

optimum flow cytometric discrimination of the T lymphocyte subpopulations?

2.8.2 Time course experiment for cytokine synthesis following incubation

with PPD

In this experiment a single patient with tuberculosis was investigated and the BAL CD4

IFN-y and TNF-a synthetic responses were measured at different time points following

incubation with PPD, ESAT- 6 or no antigen. A standard initial 2 hour incubation was

performed to allow antigen presentation and then 5pg of brefeldin A was added.

Samples were then incubated for a further 4, 8 , 16, 24 or 36 hours and the cytokines

synthesized were measured by FCM for each of the different time points.

54

Page 58: Lymphocyte Responses in the Lung in Patients with ...

The time course experiment demonstrated several important features of this assay

that were relevant to a precise determination of the antigen-specific response by FCM.

The first feature was that the CD4 molecules were progressively down regulated with

increasing incubation time so that at later time points it was difficult to distinguish CD4+

from CD4- events (figure 2.7). By contrast, CD3 molecules were well preserved on the

lymphocyte surface (MPI at four hours 280.9 and at 36 hours 174.0). The second feature

was that the scatter characteristics of the lymphocytes changed with time.

'5 50 n

I % 4 0

I I 30.S I (0 > '

201 8

10 -

Figure 2.7

10 20 30 40

Incubation time (hours)

50

4 hours

iSiiif;16 hours

SSC

36 hours

Figure 2.7

Decline in mean fluorescence intensity (MFi) of CD4 expression on BAL

lymphocytes demonstrated graphically and by FCM dotpiots with incubation time

following brefeldin administration. At 36 hours CD4+ lymphocytes were difficult to

distinguish from CD4- events.

55

Page 59: Lymphocyte Responses in the Lung in Patients with ...

At up to 8 hours incubation well preserved lymphoid scatter was noted, but by 24 hours

many of these cells had increased side scatter. A viability dye was not used in these

experiments, but many of these lymphocytes may have been undergoing apoptosis.

The percentage of CD4+ T lymphocytes synthesizing either IFN-y or TNF-a was

determined in CD3+ CD4+ lymphocytes (figure 2.8). The IFN-y and TNF-a responses

were maximal at 8 hours, but did not start to decline until after 24 hours incubation. It is

difficult to interpret why there was a drop in the 16 hour response.

Figure 2.8

20-1COto0)

<uc2 10 -

oQÜ

360 8 16 244

IFN-gTMF-a

Incubation time (hours)

Figure 2.8

IFN-g and TNF-a CD4 responses to PPD In BAL at different Incubations times

following the addition of brefeldin to the culture medium. The control samples had

no antigen added and show low responses.

The conclusion of this time course experiment is that the optimum incubation time for

maximizing the CD4 cytokine responses, lies between 8 and 24 hours following the

addition of brefeldin. However, CD4 down-regulation at longer incubation periods is a

significant problem for the accurate gating of CD4 lymphocyte responses. In addition, it

is not clear to what extent apoptosis may become a problem with longer incubation. The

final important factor was that the assay should be appropriate for routine analysis.

Therefore, BAL specimens were incubated with brefeldin for two hours and then for an

additional 14 hours overnight to allow the practical evaluation of the responses the

following day.

56

Page 60: Lymphocyte Responses in the Lung in Patients with ...

2.8.4 Use o f CD69 in BAL

The proportions of T lymphocytes responding to antigens has traditionally been

measured by determining the proportion of lymphocytes (CD4 or CDS) that have both

synthesised the cytokine of interest in addition to expressing the activation marker CD69

[7, 13, 14]. Despite the convention of using CD69 as an activation marker, it is not clear

that this provides any additional useful information than the cytokine synthetic response

alone for determining the antigen-specific response. In fact, some cognate cells may

take up to 3 days to maximally express this marker after encounter with antigen [15], so

exclusively counting the cells both expressing CD69 and cytokine may underestimate

the true antigen-specific response. A more serious problem with the use of CD69 in

Figure 2.9

CD 69

Control

71%

CDS A

CD 69

Sarcoid ds99%

CDS 110 10 10 10 10'

Figure 2.9

Histograms of expression of the activation marker, CD69 on CD4 lymphocytes

(a,c) and CDS lymphocytes (b,d) from fresh, unactivated BAL from a healthy

control subject and a patient with pulmonary sarcoidosis. Very high percentages

of CD69 expression are demonstrated on both CD4 and CDS lymphocytes.

57

Page 61: Lymphocyte Responses in the Lung in Patients with ...

tissue specimens such as BAL is that a high proportion of unactivated T lymphocytes

express this marker. Almost three-quarters of CD4 and CD8 BAL lymphocytes in a

healthy individual and even more in a patient with sarcoidosis expressed CD69 when the

BAL preparations were examined fresh without the addition of antigen (figure 2.9). These

findings render CD69 unsuitable for use as an activation marker in BAL.

2.8.5 Dose response curve for purified protein derivative

Different doses of PPD were added to BAL as the stimulatory antigen in order to

establish the optimum dose. The parameters measured by the standardised flow

cytometric cytokine synthesis assay were both the percentage of lymphocytes producing

cytokines and the mean fluorescence intensity (MFI) of the synthesised cytokines. Using

the standardised gating strategy described above, the percentage of CD4+ T

lymphocytes producing either IFN-y or TN F-a following incubation with PPD was

measured in comparison with the control samples to which no antigen was added. The

following doses of PPD were used 1pg, 2 pg, 5pg, 10pg and 20 pg. The standard 16

hour incubation was used for all samples and the percentages of CD4+ T lymphocytes

staining for intracellular IFN-y and TN F-a were measured for each dose of antigen by

FCM. The dose titration was performed on two patients with TB and the response

reached a plateau at a dose of lOpg of PPD in one patient and 5pg in the other (figure

2.10). For each dose of stimulatory antigen, the percentage CD4+ T lymphocytes

synthesising TN F-a was greater than those synthesising IFN-y (figure 2.10).

The MFI for the relevant cytokine was determined by flow cytometry using winMDI

software (M Trotter, free software). Single parameter histograms of cytokine synthesis by

CD4+ T lymphocytes were used to allow accurate gating of the positive events and the

mean values of these events calculated using the software. The MFI increased with

increasing doses of PPD (figure 2.9). What was most noticeable was the much greater

MFI for TN F-a expression than for IFN-y, particularly in one subject. However the

increased fluorescence noted with the APC-conjugated TN F-a antibody cannot be

directly correlated with increased production of that cytokine when compared to IFN-y

since APC fluorescence is greater than PE.

Based on these observations a dose of lOpg of PPD was chosen although 5pg

could have been adequate.

58

Page 62: Lymphocyte Responses in the Lung in Patients with ...

Figure 2.10

75 -,

25-

0 1 2 10 205

40-1

30-

20-

IFN-g

0 2 5 10 20

Dose of PPD Dose of PPD

10000

5 | 10004

s

1001 2 5 10 20

Doœof PPD

lOOOOi

| | «nw

s

100

TT\F-a— - à

IFNg

2 5 10 20Doœof PPD

Figure 2.10

The percentage of CD4 lymphocytes synthesizing IFN-7 and TNF-a in response to

different doses of PPD (dose in ^g) in BAL from two patients with TB (top two

graphs). The bottom two graphs demonstrate the mean fluorescence intensity

(MFi) of the synthesized cytokines.

59

Page 63: Lymphocyte Responses in the Lung in Patients with ...

2.8.6 Optimisation of antibody surface staining sequence

Following incubation, the surface antigens used to delineate the lymphocyte subsets

may become downregulated, thus affecting the ability of monoclonal antibodies to

reliably distinguish such populations. This process of downregulation may be

exaggerated by permeabilisation of the cells. Therefore, the MFI of CD4 and CD8

expression was compared when antibodies against these antigens were added before

fixation and permeabilisation (pre-staining), or when added at the same time as the

cytokine antibodies.

There was a clear advantage for CD4 discrimination with pre-staining as compared to

staining with the cytokines (figure 2.11). No difference was noted for CD8 staining

between these two sequences.

Figure 2.11

SSC

Figure 2.11

FCM dotpiots of BAL following 16 hour incubation demonstrating that optimum

CD4 discrimination is achieved when surface staining is performed prior to

fixation and permeabilisation (a) rather than after this process (b)

2.8.7 Method for the detection of intracellular cytokine synthesis in whole

blood and BAL

Aliquots of the BAL suspension containing 1X10^ CD4+ lymphocytes in 1ml of culture

medium were placed into sterile 5ml polypropylene tubes (Thermo Life Sciences, UK). In

addition, 1 ml of peripheral blood from the same patient collected into lithium heparin

60

Page 64: Lymphocyte Responses in the Lung in Patients with ...

tubes was also placed into polypropylene tubes. To one of the BAL and blood samples,

10|ig of PPD (Statens Serum Institute, Copenhagen, Denmark) was added. The other

tubes were unstimulated control samples. The samples were incubated for two hours at

37°C and 5% CO2 , after which time 5pg of Brefeldin A (Epicentre Technologies,

Cambridge, UK) was added and the samples incubated for a further 14 hours.

Following incubation, the samples were vortexed vigorously to detach cells from the

walls of the tube. First, lymphocyte surface markers were stained using CD4-FITC

(Royal Free Hospital) and CD3-PerCP (Becton Dickinson) for 15 minutes in the dark at

room temperature and the samples were washed. Fixation and permeabilisation of the

cells was performed as described in 2.8.3 above using Fix-and-Perm (Caltag). Following

this, IFN-y-PE (Caltag) and TNF-a-APC (Becton Dickinson) were added and the samples

stained at 4°C for 30 minutes, followed by a final wash step. The acquisition and

analysis of the stained preparations is described in chapter 7.

2.8.8 Antigens used for the cytokine synthesis assay

The cytokine synthetic responses to a variety of antigens other than PPD were assessed

in both BAL and blood (table 2.3).

Table 2.3 Antigens and substances used to stimulate cytokine synthesis

Antigen Indication Manufacturer

Staphylococcal enterotoxin B Positive control Ag Sigma Aldrich

(SEE)

Purified protein derivative (PPD) TB-specific responses Statens serum institute

Tetanus toxoid Control antigen Pasteur Merieux

2.9 StatisticsMost of the data generated such as leukocyte differentials in BAL, and cytokine synthetic

responses were not normally distributed. Therefore median values and inter-quartile

ranges or full ranges were recorded in the text. Comparisons between data sets were

made using Mann-Whitney analysis.

61

Page 65: Lymphocyte Responses in the Lung in Patients with ...

Results generated using different techniques, for example the BAL leukocyte

differentials by cytospin and FCM were compared using both the Spearmans correlation

coefficient, and by Bland-Altman analysis.

2.10 References

1. Honeybourne D, Babb J, Bowie P, et al. British Thoracic Society guidelines on

diagnostic flexible bronchoscopy. Thorax 2001 ;56:i1-i21

2. Loken MR, Brosnan JM, Bach BA and Ault KA. Establishing optimal lymphocyte

gates for immunophenotyping by flow cytometry. Cytometry 1990;11:453-9.

3. Latza U, Niedobitek G, Schwarting R, et el. Ber-EP4: new monoclonal antibody

which distinguishes epithelia from mésothélial. J Clin Pathol 1990;43:213-9.

4. Mandy F Nicholson J, Autran B and Janossy G. T-Cell Subset Counting and

Fight Against AIDS: Reflections Over a 20-Year Struggle. Clinical Cytometry

2002;60:39-45

5. Kappelmayer J, Gratama JW, Karaszi E, et al. Flow cytometric detection of

intracellular myeloperoxidase, CD3 and CD79a. Interaction between monoclonal

antibody clones, fluorochromes and sample preparation protocols. J Immunol

Methods 2000;242:53-65.

6 . Petrovsky N, Harrison LC. Cytokine-based human whole blood assay for the

detection of antigen-reactive T cells. J Immunol Methods 1995;186:37-46.

7. Suni MA, Picker LJ and Maino VC. Detection of antigen-specific T cell cytokine

expression in whole blood by flow cytometry. J Immunol Methods 1998;212:89-

98.

8 . Rowland-Jones SL, Dong I , Fowke KR, et al. Cytotoxic I cell responses to

multiple conserved HIV epitopes in HIV-resistant prostitutes in Nairobi. J Clin

Invest 1998;102:1758-65.

9. Lalvani A, Pathan AA, Durkan H, et al. Enhanced contact tracing and spatial

tracking of Mycobacterium tuberculosis infection by enumeration of antigen-

specific I cells. Lancet 2001;357:2017-21.

10. Lalvani A, Pathan AA, McShane H, et al. Rapid detection of Mycobacterium

tuberculosis infection by enumeration of antigen-specific I cells. Am J RespirCrit

Care Med 2001;163:824-8.

62

Page 66: Lymphocyte Responses in the Lung in Patients with ...

11. Lippincott-Schwartz J, Yuan LC, Bonifacino JS and Klausner RD. Rapid

redistribution of Golgi proteins into the ER in cells treated with brefeldin A:

evidence for membrane cycling from Golgi to ER. Cell 1989;66:801-13.

12. Klausner RD, Donaldson JG and Lippincott-Schwartz J. Brefeldin A: insights into

the control of membrane traffic and organelle structure. J Cell Biol

1992;116:1071-80.

13. Kern F, Faulhaber N, Frommel C, et al. Analysis of CD8 T cell reactivity to

cytomegalovirus using protein-spanning pools of overlapping pentadecapeptides.

EurJ/mmuno/2000;30:1676-82.

14. Waldrop SL, Pitcher CJ, Peterson DM, et al. Determination of antigen-specific

memory/effector CD4+ T cell frequencies by flow cytometry: evidence for a novel,

antigen-specific homeostatic mechanism in HIV-associated immunodeficiency. J

Clin Invest 1997;99:1739-50.

15. Gibbons DC, Evans TG. CD69 expression after antigenic stimulation. Cytometry

1996;23:260-1.

63

Page 67: Lymphocyte Responses in the Lung in Patients with ...

Chapter 3

Comparison of Flow Cytometry with

Cytospin for the Determination of

Bronchoalveolar Lavage Leukocyte

Populations in Patients Investigated with

Respiratory Disease

64

Page 68: Lymphocyte Responses in the Lung in Patients with ...

3.1 IntroductionBronchoalveolar lavage is a recognized procedure for the diagnosis of respiratory

disease of both infective and inflammatory etiologies. Not only does the directed

washing of an affected area of lung provide specimens that may enable a microbiological

or cytological diagnosis to be made, but effective BAL also samples the cellular

component of the lower bronchial tree. Increases in BAL cell lymphocyte populations

have been found to be helpful in the diagnosis of inflammatory conditions such as

sarcoidosis [1, 2], interstitial lung diseases [3] and in cryptogenic organizing pneumonias

[4], while increasing neutrophil concentrations in BAL may be evidence of a bacterial

infection [5, 6 ], in types of pulmonary fibrosis [7], or as a response to lung transplantation

[8 . 9].

The existing standard method for the evaluation of BAL leukocyte populations is

predominantly through the investigation of cytospin preparations. Simple differential

counts of lymphocytes, macrophages and granulocytes can be achieved by counting

stained cells by microscopy. Further identification of subpopulations of lymphocytes or

macrophages can also be performed by the use of immunoperoxidase [1 0 ] or

immunofluorescence staining.

Similarly, flow cytometric (FCM) techniques have been developed to distinguish the

BAL leukocyte component from the non-cellular and non-leukocyte events. Several

investigators have used the pan-leukocyte marker, CD45 to determine the lymphocyte

events acquired by FCM [11, 12]. Lymphocytes form homogenous populations of small

cells that express high levels of CD45 and are easily gateable by FCM. Whilst

lymphocytes can be accurately determined by CD45 expression and light scatter

characteristics, this is not true of neutrophils, eosinophils and macrophages.

Macrophages in particular are notoriously difficult to distinguish by FCM, although

fortunately the enumeration of the total macrophage pool in BAL is rarely of clinical

significance. By contrast, BAL neutrophilia and eosinophilia are diagnostically important.

A granulocyte marker, CD15 was therefore used to distinguish these cells from the

macrophages by FCM. The macrophage population was then derived as those CD45+

events remaining after subtraction of the gated lymphocytes and granulocytes.

The main aim of this study was to develop a simple flow cytometric method for

determining the proportions of the clinically relevant leukocyte populations in BAL. The

results obtained by FCM were then compared with those from cytospin preparations

under optimum conditions using a highly experienced cytologist counting 500 -cells, A

65

Page 69: Lymphocyte Responses in the Lung in Patients with ...

significant improvement on the standard visual counts in which 2 0 0 cells are routinely

counted. A further aim of this study was to assess whether both techniques could reliably

determine BAL leukocyte differentials on samples frozen in liquid nitrogen.

3.2 Material and methods3.2.1 Subjects

Patients undergoing bronchoscopy for the diagnosis of suspected non-malignant

respiratory disease were included. 100 BAL were performed on 92 patients. 53 BAL

were performed on HIV-infected patients. The remaining 47 episodes included four BAL

undertaken on patients following bone marrow transplantation, three on patients

immunocompromised with haematological malignancies and two on subjects in intensive

care.

3.2.2 Bronchoalveolar lavage

Bronchoscopies were performed under sedation using a flexible bronchoscope

wedged into a subsegmental bronchus. BAL was site-directed in cases with

radiologically defined areas of abnormality, but otherwise the right middle lobe was

washed. Sterile normal saline was introduced through the bronchoscope to a maximum

volume of 200ml and the fluid aspirated into a siliconized glass container on ice. BAL

specimens were analysed within two hours of their acquisition. Aliquots of BAL were sent

to the relevant laboratories and the remaining fluid (normally >25ml) was centrifuged at

430g for 8 minutes, decanted and the pellet resuspended. The sample was then filtered

through a 100pm filter (Cell Tries, Partec GmBH, Münster, Germany), centrifuged again

and the pellet resuspended in 1 ml of Phosphate buffered saline (PBS).

3.2.3 Flow cytometry

50pl of the BAL suspension was added to a flow cytometry tube containing the following

monoclonal antibodies; CD45-FITC (Beckton Dickinson, Oxford, UK), CD15-PE (Caltag

Medsystems, Towcester, UK) and 7-AAD (7 amino-actinomycin D, Pharmingen, San

Diego. California, USA). These antibodies were pretitrated and added in saturating

concentrations. The samples were stained at room temperature in the dark for 15

minutes and PBS added to a volume of 1ml. In samples that were visibly bloodstained,

lysis buffer (0.17M NH4CI) was added instead of PBS and left for 15 minutes to ensure

66

Page 70: Lymphocyte Responses in the Lung in Patients with ...

red cell lysis. Samples were run on a CytoronAbsolute flow cytometer (Ortho Diagnostic

Systems, Raritan, New Jersey, USA) using an absolute counting protocol.

List mode data were analysed using lmmunoCount-2 software. Primary immunological

gating of CD45+ events against side scatter was performed and a tight gate placed

around the low side scatter lymphocytes (Figure 3.1, plot a). Second, a dot plot of CD45

against GDI 5 was produced and a gate placed around the CD45+, CD15+ granulocytes

(Figure 3.1, plot b). The percentages of the lymphocyte and granulocyte populations

were calculated as the number of gated events divided by the total number of CD45+

events.

Figure 3.1

ssc GDIS

Figure 3.1

FCM dotpiots of BAL demonstrating CD45 panleukogating (R1, plot a) to

differentiate leukocytes from debris. Lymphocytes (R2, plot a) express CD45

brightly and have low side scatter. Granulocytes can be distinguished from

macrophages within the panleukogate by their expression of G DI5 (R3, plot b).

The macrophage pool was derived from the total number of GD45+ cells after

subtraction of lymphocytes and granulocytes.

In selected patients in whom eosinophils were identified by cytospin a further aliquot of

BAL was stained with the following antibodies; GD23-FITG (Galtag), GD15-PE (Galtag)

and GD45-APG (Pharmingen). The samples were run on a FAGScalibur flow cytometer

(Becton Dickinson) after a wash step following antibody staining. G D I5+ granulocytes

within the GD45 pan-leukogate were gated and sent to a GD45 GD23 dotpiot (Figure

67

Page 71: Lymphocyte Responses in the Lung in Patients with ...

3.2). Eosinophils were characterized by the dual expression of CD15 and the IgE

receptor antigen, CD23, whereas neutrophils were CD23 negative.

F ig u re 3.2

CD 15

Gated by R2

10* "W 10-CD 23

%

A■ Eosinophil

‘ è /

Figure 3.2

FCM dotpiots and a photomicrograph of BAL from a patient with an eosinophilia.

CD45+ CD15+ granulocytes (R2) are demonstrated to be predominantly CD23+

eosinophils (R3) with few CD23- neutrophils (R4, plot b). The photograph (c)

confirms eosinophilia in the cytospin preparation.

The accuracy of the BAL lymphocyte gating strategy of CD45 expression and scatter

characteristics was compared with lymphocyte enumeration by counting the sum of the

various lymphocyte subsets in BAL by FCM (lymphosum).

68

Page 72: Lymphocyte Responses in the Lung in Patients with ...

In 15 samples, BAL was stained with T cell (CD3), B cell (CD19) and NK cell (CD56)

markers in addition to CD45. A cocktail of the following antibodies were used; CD56-

FITC (Beckton Dickinson), CD19-PE (Beckton Dickinson), CD3-perCP (Beckton

Dickinson) and CD45-APC (Beckton Dickinson). The samples were stained as described

above and washed before running on a FAGScalibur. First, Lymphocyte percentages

were derived by CD45 expression and side scatter characteristics as described above.

These values were then compared with the sum of the percentages of T, B and NK cells.

Individual lymphocyte subsets were calculated by gating the number of CD3, CD56 or

GDI9 bright events with lymphoid side scatter characteristics (figure 3.3).

Figure 3.3

R2

QOssc

G a t e d by R 2

C D 56

S S C ssc

Figure 3.3

FCM dotpiots of BAL lymphocyte determination by CD45 expression and light

scatter (a) compared with gating strategies to determine the numbers of NK (b), T

(c) and B (d) lymphocytes. The numbers of each of the lymphocyte subsets were

expressed as a percentage of the total CD45 panleukogate (R1). These

percentages were added together to give a sum value of the lymphocyte

percentage and this was compared with the lymphocyte percentage derived from

plot a. NK cells (b) were CD56+ but CD3-.

69

Page 73: Lymphocyte Responses in the Lung in Patients with ...

NK cells were only counted as such if they were CD56+ but CD3-. The numbers of each

lymphocyte subset were then expressed as a percentage of the total number of CD45+

events and these were added together to give the total lymphocyte percentage.

3.2.4 Cytospin

BAL was adjusted to a concentration of 2-5 x 10® CD45+ cells/ml. lOOpI of BAL was

centrifuged for 2 minutes at SOOrpm in a cytospin machine (Shandon, Runcorn, UK) and

the resultant slide air dried, fixed in chloroform and acetone for 10 minutes and stained

with Haematoxylin and eosin. 500 cells were counted by light microscopy and the BAL

cells differentiated by morphological and staining characteristics. The cytologist was

blinded to the BAL differentials achieved by FCM.

3.2.5 Freezing and thawing of BAL

15 BAL’s were frozen in liquid nitrogen after the samples were run on the flow cytometer

as described above. 500pl of the BAL suspension was added to 500pl of freeze mixture

(10% DMSO in 20% fetal calf serum in RPMI 1640) in a cryovial. The sample was

vortexed rapidly and then placed in the vapour phase of liquid nitrogen to ensure

freezing at the rate of 1°G/minute. After 12 hours the samples were stored in liquid

nitrogen. For reanalysis after freezing, the samples were rapidly defrosted by pippetting

with warm RPMI and 10% FCS into a universal container. The thawed sample was then

centrifuged, decanted and the pellet resuspended in RPMI and FCS before a further

centrifugation step to ensure complete removal of the freeze mixture.

3.2.6 Immunofluorescence staining of BAL

Cytospins were prepared from a sample with a BAL neutrophilia and a further sample

with an eosinophilia. After fixation, the cytospins were stained with either 5pl CD15-FITC

(Dako) or CD23-FITC (Caltag) in 45pl of PBS. The stained preparations were examined

using a fluorescence microscope.

3.2.7 Statistical analysis

Comparisons between FCM and cytospin cell differentials were made using Pearsons

correlation. Bland-Altman plots were used to analyse the degree of variation between the

two techniques. Coefficients of variation (CV) were assessed by the parallel analysis of

10 cytopsins and 10 FCM tubes from the same BAL specimen. This process was

70

Page 74: Lymphocyte Responses in the Lung in Patients with ...

undertaken with 5 different BAL samples. The microscopist was unaware that the

cytospins were from the same BAL specimen. The CV’s for cytospin and FCM were

estimated using the analysis of variance (ANOVA), after controlling for the difference

between the mean percentage of each leukocyte subset for the different samples.

Otherwise, data was expressed as mean values with 95% confidence limit adjustments

included.

3.3 Results3.3.1 BAL diagnoses

46 BAL specimens yielded a diagnosis of which tuberculosis was the most common,

occurring in 21 cases. A bacterial organism was only cultured in 8 specimens, partly

reflecting prior antibiotic usage. For the HIV-infected population, the most common BAL

diagnosis was tuberculosis, occurring in 9 cases. Pneumocystis carinii was found in 4

patients and rare diagnoses included strongyloidiasis, cryptococcosis and

cytomegalovirus infection. One patient was co-infected with pneumocystis carinii,

mycobacterium tuberculosis and cytomegalovirus and went on to have 3 further BAL.

The median blood CD4 count in the HIV-infected subjects was 78 cells/pl.

3.3.2 BAL leukocyte differential counts by FCM

The median recovery of saline introduced during BAL was 90ml (50%). Using CD45 to

differentiate the leukocyte from the non-leukocyte populations, a mean of 61.6% (95%

Cl; 56.6% to 66.6%) of the events acquired by FCM were leukocytes when using fresh

BAL. The majority of the CD45 negative events were non-cellular debris, although a

variable proportion consisted of bronchial epithelial cells and squamous cells. The mean

absolute number of leukocytes counted by FCM was 9305 cells (95% Cl: 7740 to

10870).

There was a marked variability in the leukocyte proportions between different

patients with lymphocyte percentages varying from 0.3% to 86% of CD45+ events and

the granulocyte percentages varying from 0.2% to 94%.

71

Page 75: Lymphocyte Responses in the Lung in Patients with ...

3.3.3 7-AAD expression in BAL

The mean number of CD45+ events that co-expressed the dead cell marker 7-AAD

was 37.2% (95% Cl: 32.7% to 41.7%). When these 7-AAD+ events were further

analysed, the majority were found to by non-lymphoid. 7-AAD+ lymphocytes maintained

their light scatter characteristics and formed a similar gateable population to 7-AAD-

lymphocytes (figure 3.4).

Figure 3.4

ssc

G ated by R3

SSC

R3

7-AAD

Figure 3.4

FCM dotpiots of fresh BAL demonstrating CD45 expression and light side scatter

before (a) and after (b) gating by the dead cell marker, 7-AAD (R3, histogram c).

CD45+ events that co-express the dead cell marker 7-AAD (b) maintain light side

scatter characteristics and lymphoctes (R2) can be as easily differentiated from

non-lymphocytes as in the non-7-AAD gated leukocytes (a).

3.3.4 Correlation between leukocyte differentials by FCM and cytospin

The correlation between each of the leukocyte proportions in BAL enumerated by FCM

and cytospin were close with R values of 0.92 for lymphocytes, 0.95 for granulocytes

and 0.86 for macrophages (figure 3.5). Bland-Altman analysis (figure 3.6) demonstrated

72

Page 76: Lymphocyte Responses in the Lung in Patients with ...

Figure 3.5100

ç■q.

I0È

1oa.

80 -

♦♦

60

40 0.922

20

20 1000 40 60 80

% Lymphocytes by FCM

100c■q.

13C2 CD

80

60

R* = 0.945

40

♦ ♦20

8 0 1000 20 40 6 0

% Granulocytes by FCM

1 0 0c■q.

IÈ$

♦80

♦ ♦ ♦

60= 0.861

4 0•§.ii

♦ ♦

20

1000 20 40 60 80

% Derived macrophages by FCM

Figure 3.5

Correlation plots comparing the enumeration of BAL lymphocytes, granulocytes

and macrophages by flow cytometry and cytospin.

73

Page 77: Lymphocyte Responses in the Lung in Patients with ...

Figure 3.6

20 •

10 ■ 4.8%

««.CV* *** * * % ^-3.0%(95%CI-3.7to-2.2). # « .» • ••••••• .»^»......." y ................................................. ..

* # ♦ ♦ , ♦ ♦ ♦ ♦ , ♦ ♦«» * ** .* . .

-10.8%

10 20 30 40 50 60 70 80 90 100

Average lymphocyte %30

2 0 -

g■q.

I

iu_ç

î

-20

-30I

30

20

10

0

-10

-20

-30

17.5% ♦

♦ %. . . y . . . . . . y . . . .

♦ ♦«»

' . . '♦ * +24% (095% 0.9 to+3.9) * ♦ ; * *

-12.7%♦ ♦

10 20 30 40 50 60 70 8 0 90 100

Average granulocyte %

+24.7%

+3.4%(CI95%1.3tp5.5)....................

♦ ♦ ♦ - ... • .r

-17.9%

10 20 30 40 50 60 70 80 90 100Average macrophage %

Figure 3.6

Bland-Altman plots comparing the enumeration of BAL lymphocytes,

granulocytes and macrophages by flow cytometry and cytospin.

74

Page 78: Lymphocyte Responses in the Lung in Patients with ...

a small but statistically significant tendency by cytospin to underestimate the percentage

of lymphocytes by 3% (95% Cl: -3.7% to -2.2%) when compared to FCM. There was a

significant tendency for cytospin to overestimate the percentage of macrophages by

3.4% (95% Cl: 1.3% to 5.5%). For granulocytes, the overestimation on cytospin was

2.4% (95% Cl: 0.9% to 3.9%).

3.3.5 Coefficient of variation between FCM and cytospin

The coefficient of variation for lymphocyte determination was 2.67% by FCM and 13.3%

by cytospin, whilst for macrophages the figures were 2.60% and 10.9% and for

granulocytes, 2.78% and 23.0% respectively (table 3.1).

Table 3.1 Coefficients of variation for BAL lymphocyte, macrophage and

granulocyte percentages derived by flow cytometry and cytospin

BAL Leukocyte subset FCM Cytospin

Lymphocytes 2.67% 13.3%

Macrophages 2.60% 10.9%

Granulocytes 2.78% 23.0%

3.3.6 Comparison between fresh and frozen BAL for leukocyte subset

determination by FCM

Frozen BAL specimens were analysed using the FCM method described above and the

leukocyte differentials were compared with those obtained previously from the same

fresh BAL sample. A correlation of 0.978 was achieved for lymphocytes (n=11), with

Bland Altman analysis demonstrating limits of agreement between 11.9% and -7.4% for

lymphocyte percentages derived from fresh and frozen samples. Similar close

correlations were reached for granulocyte and macrophage proportions by FCM. In

cytospins made from frozen BAL samples lymphocytes retained their morphology, but it

was difficult to count macrophages and neutrophils due to the deterioration of their

cellular architecture.

75

Page 79: Lymphocyte Responses in the Lung in Patients with ...

3.3.7 Comparison between BAL lymphocyte percentages obtained by CD45

and light scatter with the sum of the lymphocyte subsets by FCM

Excellent correlations between lymphocyte enumeration by CD45-light scatter and

lymphosum were obtained (r=0.9986). Bland Altman analysis demonstrated very close

limits of agreement between these two methods (figure 3.7) thus confirming the

adequacy of the CD45-light scatter method of calculating BAL lymphocyte percentages

Figure 3.7

>. >- J3 0

|1tl

I I

1 0 0

so

t o

4 0

2 0

0

0 2 0 4 0 6 0 6 0 100% Lymphocytes by lymphosum

Q.

1.7%

0.12% (95% Cl: -0.28-0.52)-1.46%

vn

0 20 60 60 100

Average lymphocyte %

Figure 3.7

Correlation plot (a) and Bland Altman plot (b) for the total percentage of

lymphocytes in BAL derived by CD45 expression and low side scatter compared

with the lymphocyte percentage derived by adding the percentages of T, B and NK

cells (lymphosum).

76

Page 80: Lymphocyte Responses in the Lung in Patients with ...

3.3.8 Immunofluorescence staining of BAL

Positively stained cells with characteristic granulocyte morphology were demonstrated

using both CD23 and CD15 reagents in samples with an eosinophilia (figure 3.8) As

expected, CD23 staining was negative on CD15-positive granulocytes in samples with a

neutrophilia. These findings confirm that CD23 allows eosinophils to be distinguished

from neutrophils and validates the FCM gating strategy (Figure 3.2).

Figure 3.8

Figure 3.8

Immunofluoresence staining of BAL from a patient with a BAL eosinophilia. Green

fluorescent cells with a characteristic eosinophil appearance are noted following

staining with CD23 FITC (a) and also following staining with CD15 FITC (b).

3.4 DiscussionCytospin remains the most frequently used method of analysing BAL leukocytes,

despite the fact that FCM readily discriminates lymphocytes, macrophages and

granulocytes in addition to providing details about lymphocyte subsets. Previous studies

using FCM have provided a biased view of BAL analysis by concentrating on the

characteristics of the easily distinguishable lymphocytes and their subsets [11-13]. Such

an approach has failed to provide information on the relative proportions of the clinically

77

Page 81: Lymphocyte Responses in the Lung in Patients with ...

important granulocyte component. Here a more complete picture has been provided by

recording the proportions of lymphocytes, granulocytes and macrophages in BAL in

patients with respiratory disease.

The precision of cytospin versus FCM has not previously been compared under

optimal conditions. In this study cytospins were evaluated by a cytologist of 15 years

experience who counted 500 leukocytes in each preparation. FCM was undertaken

using CD45 directed gating to distinguish leukocytes from debris and epithelial cells. The

use of CD45 panleukogating and light scatter characteristics has been established as

the optimum method for lymphocyte enumeration in blood [14]. The addition of a

granulocyte marker is required in BAL, but not blood, because light scatter and CD45

expression alone cannot reliably distinguish granulocytes from macrophages.

Some investigators have argued that the best strategy for BAL lymphocyte

analysis is to eliminate damaged cells from the analysis by using a DMA dye, LDS-751

after first gating the CD45+ low side scatter lymphocytes [12]. Whilst the exclusion of

damaged cells may be necessary for functional and phenotypic analysis of lymphocytes,

our study demonstrates that CD45 expression and light scatter characteristics are

remarkably robust for BAL leukocytes and are not significantly affected by freezing.

Moreover, the exclusion of damaged cells also skews the BAL leukocyte differentials.

We used a nuclear dye, 7-AAD to recognize early apoptotic and dying cells [15]. In fresh

BAL, the majority of 7AAD+ leukocytes were non-lymphoid, but the 7-AAD+ lymphoid

and non-lymphoid components could still differentiated from each other by light scatter

characteristics (figure 3.4). Thus, the exclusion of 7-AAD+ events will overestimate the

proportion of lymphocytes and underestimate macrophages and granulocytes that are

more prone to cell death.

Comparisons between FCM and cytospin were made using three statistical

techniques. First, the coefficient of variation for each method in determining leukocyte

subsets was assessed by the analysis of 10 parallel preparations of cytospins and FCM

tubes from the same BAL sample repeated with 5 different samples. The coefficients of

variation by FCM were considerably lower than by cytospin for each leukocyte subset

(Table 3.1), demonstrating the superior precision of FCM. Such a finding is unsurprising

as a mean of 9305 CD45+ events were counted by FCM in this study compared with 500

cells by cytospin. Second, correlation plots between the two methods for the

enumeration of lymphocytes, granulocytes and macrophages were performed to

demonstrate the excellent overall agreement of the mean values. Third, the two methods

78

Page 82: Lymphocyte Responses in the Lung in Patients with ...

were scrutinized using the Bland-Altman test (figure 3.6) to detect any consistent

variation between the two techniques. Bland-Altman analysis demonstrated a slight

selective accumulation of the larger macrophages and the reciprocal depletion of the

smaller lymphocytes by cytospin when compared to FCM. Such a phenomena has

previously been described when stimulated large blast cells preferentially accumulated

on cytospins at the expense of small lymphocytes [16].

The FCM method described here distinguishes granulocytes from macrophages by

virtue of CD15 expression. CD15+ granulocytes consist of neutrophils, eosinophils and

basophils. Basophils are rare populations in BAL, but the presence of eosinophils can be

diagnostically helpful. Eosinophils were found to co-express both CD15 and the IgE

receptor antigen, CD23 (Figure 3.2), whereas neutrophils were CD15+ but did not

express CD23.

In summary, a simple flow cytometric technique has been described for

distinguishing the BAL leukocyte populations that avoids the potential complications

recorded by previous investigators [12]. This system, using primary immunological gating

of CD45+ events in conjunction with the granulocyte marker CD15 and the eosinophil

marker CD23 represents is an effective antibody panel for the delineation of the clinically

relevant leukocyte subsets in BAL with apoptotic markers such as 7AAD demonstrated

to be unhelpful. Statistical analysis has confirmed close correlations between the

leukocyte populations demonstrated by FCM and by cytospin preparations while also

documenting that FCM is superior in terms of precision, reliability and robustness. These

features, combined with its speed and the ability to perform simple additional lymphocyte

phenotyping panels argue strongly in favor of FCM being adopted as a standard method

for BAL analysis.

3.5 References1. Campbell DA, Poulter LW and du Bois RM. Immunocompetent cells in

bronchoalveolar lavage reflect the cell populations in transbronchial biopsies in

pulmonary sarcoidosis. Am Rev Respir Dis 1985;132:1300-6.

2. Agostini C, Trentin L, Zambello R, et al. CDS alveolitis in sarcoidosis: incidence,

phenotypic characteristics, and clinical features. Am J Med 1993;95:466-72.

3. Hunninghake GW, Kawanami O, Ferrans VJ, et al. Characterization of the

inflammatory and immune effector cells in the lung parenchyma of patients with

interstitial lung disease. Am Rev Respir Dis 1981;123:407-12.

79

Page 83: Lymphocyte Responses in the Lung in Patients with ...

4. Poletti V, Castrilli G, Romagna M, et al. Bronchoalveolar lavage, histological and

immunohistochemical features in cryptogenic organizing pneumonia. Monaldi

Arch Chest Dis 1996;51:289-95.

5. Jensen BN, Lisse IM, Gerstoft J, et al. Cellular profiles in bronchoalveolar lavage

fluid of HIV-infected patients with pulmonary symptoms: relation to diagnosis and

prognosis. Aids 1991;5:527-33

6. Sternberg Rl, Baughman RP, Dohn MN and First MR. Utility of bronchoalveolar

lavage in assessing pneumonia in immunosuppressed renal transplant recipients.

Am J Med 1993;95:358-64

7. Wells AU, Mansell DM, Haslam PL, et ai. Bronchoalveolar lavage cellularity: lone

cryptogenic fibrosing alveolitis compared with the fibrosing alveolitis of systemic

sclerosis. Am J Respir Cht Care Med 1998;157:1474-82.

8. Tiroke AH, Bewig B and Haverich A. Bronchoalveolar lavage in lung

transplantation. State of the art. Clin Transplant 1999;13:131-57.

9. Ward 0, Whitford H, Snell G, et ai. Bronchoalveolar lavage macrophage and

lymphocyte phenotypes in lung transplant recipients. J Heart Lung Transplant

2001;20:1064-74.

10. Costabel U, Bross KJ and Matthys H. Diagnosis by bronchoalveolar lavage of

cause of pulmonary infiltrates in haematological malignancies. Br Med J (Clin

Res Ed) 1985;290:1041.

11. Padovan OS, Behr J, Allmeling AM, et ai. Immunophenotyping of lymphocyte

subsets in bronchoalveolar lavage fluid. Comparison of flow cytometric and

immunocytochemical techniques. J Immunol Methods 1992;147:27-32.

12. Brandt B, Thomas M, von Eiff M and Assmann G. Immunophenotyping of

lymphocytes obtained by bronchoalveolar lavage: description of an all-purpose

tricolor flow cytometric application. J Immunol Methods 1996;194:95-102.

13. Dauber JH, Wagner M, Brunsvold S, et al. Flow cytometric analysis of

lymphocyte phenotypes in bronchoalveolar lavage fluid: comparison of a two-

color technique with a standard immunoperoxidase assay. Am J Respir Cell Mol

8/0/1992;7:531-41.

14. Loken MR, Brosnan JM, Bach BA and Ault KA. Establishing optimal lymphocyte

gates for immunophenotyping by flow cytometry. Cytometry 1990;11:453-9.

80

Page 84: Lymphocyte Responses in the Lung in Patients with ...

15. Schmid I, Krall WJ, Uittenbogaart CH, et al. Dead cell discrimination with 7-

amino-actinomycin D in combination with dual color immunofluorescence in

single laser flow cytometry. Cytometry 1992;13:204-8.

16. Janossy G. Polyclonal activation of murine and human T and B lymphocytes.

University College London, 1974 PhD thesis.

81

Page 85: Lymphocyte Responses in the Lung in Patients with ...

Chapter 4

Optimal Gating Strategies for

Determining Bronchoalveolar Lavage

CD4/CD8 Lymphocyte Ratios by Flow

Cytometry

82

Page 86: Lymphocyte Responses in the Lung in Patients with ...

4.1 IntroductionThe aim of this chapter was to compare different gating strategies for the determination

of CD4/CD8 ratios in BAL with a gold standard’ flow cytometric method. The impetus for

this investigation was to generate a single, 4-colour monoclonal antibody panel that

could determine the lymphocyte hetereogeneity based on the CD4/CD8 ratios in addition

to the relevant BAL leukocyte components discussed in the previous chapter.

Raised BAL CD4/CD8 lymphocyte ratios have been known to be associated with

diseases such as sarcoidosis and berryliosis and respiratory physicians have found

these parameters helpful in diagnostic decision making [1-4]. Nevertheless, these early

studies have used cumbersome immunofluoresence or immunoperoxidase methods for

lymphocyte subset analysis that are both time consuming and labour intensive. Flow

cytometry has been demonstrated to be an alternative method for BAL lymphocyte

subsetting [5-7] . However, it remains unclear what are the optimum combinations of

antibodies and gating strategies in order to perform BAL lymphocyte subset analysis.

Since the publication of these early studies on the use of FCM in BAL, there have been

few subsequent published reports dedicated to exploring this issue. By contrast there

has been a considerable degree of research interest in the generation of simple new

protocols for the determination of CD4 and CD8 lymphocyte subsets in blood [8, 9] . In

view of these developments, a simplified, CD45 panleukogating system where CD45+

low side scatter lymphocytes were gated and the CD4 and CD8 subsets directly

investigated without the use of the T cell marker, CD3. The CD4 and CD8 subsets

derived by this method were compared with several different techniques. As a gold

standard’ comparator, a more complex panel including an anti-CD3 antibody and a

precise gating strategy designed for optimum CD4 counting in blood was used [10]. In

addition, the CD4/CD8 ratios were also analysed using a CD3 gating method, but

without CD45 discrimination on a different flow cytometer.

4.2 Methods4.2.1 Patients

Immunocompetent patients undergoing BAL for suspected respiratory disease

were included. In addition, pleural fluid was also analysed from two patients. The

majority of patients (table 4.1) had either sarcoidosis or tuberculosis. HIV+

patients were not included since the BAL CD4/CD8 ratios were reduced and

83

Page 87: Lymphocyte Responses in the Lung in Patients with ...

therefore any differences between the two techniques would have been less easy

to detect.

4.2.3 Bronchoalveolar lavage and pleural fluid

BAL and pleural aspirations were performed for clinical indications. Standard

techniques for BAL and pleural aspirations were undertaken. Aliquots of BAL and

pleural fluid were sent to the relevant laboratories and the remainder was used to

perform differential cell counts.

4.2.4 Handling of samples

BAL and pleural fluid samples were washed and filtered as described in chapter

3.

4.2.5 Flow cytometry

The following monoclonal antibodies were used in optimised, pretitrated saturating

concentrations. For the first panel (method 1): CD4 FITC (Royal Free Hospital), CD8 PE

(Cymbus), CDS PECyT (Caltag) and CD45 APC (Pharmingen). For the second panel

(method 2): CD15 FITC (Cytognos, Salamanca, Spain), CD4 PE (Cymbus), CD8 PECy7

(Caltag) and CD45 APC (Pharmingen) were used. For the third panel (method 3): CD4

FITC (Royal Free Hospital), CD8-PE (Royal Free Hospital) and CD3-PEcy5 (Dako). SOpI

of BAL or pleural fluid was added to the different antibody panels and the samples

stained at room temperature in the dark for 15 minutes. For the first two panels a wash

step was then perfomed and the pellets resuspended up to a volume of 200pl with PBS-

A before running on a FACSCalibur flow cytometer (Becton Dickinson). The wash step

was omitted for the last panel and the samples were made up to 1ml with PBS-A before

running directly on the CytoronAbsolute flow cytometer (Ortho diagnostics).

20,000 CD45+, low side scatter lymphocytes were acquired for each sample run on

the FACSCalibur. The listmode data generated were analysed in the following fashion.

For the first panel, the gating strategy adopted was the same as that described by

Bergeron et al [10] . Briefly, CD45+ low side scatter lymphocytes were first gated and

then sent to a second dotplot to discriminate CD3+ T cells with low side scatter from

non-T cell lymphocytes. The CD3+ CD45+ events were then backgated to a CD45 side

84

Page 88: Lymphocyte Responses in the Lung in Patients with ...

scatter plot to ensure that apoptotic lymphocytes were excluded. Lastly, the lymphocytes

were further scrutinised by their expression of either CD3 and CD4 or CD3 and CD8

(figure 4.1).

Figure 4.1

ssc

R2& Gated by R1

COt3Ü s

i I

Gated by R2

10"

CD45

SSC

&

QO &

—►

Gated by R3

# ' »

CD3

00 ÛO ^

Gated by R3

- '

CD3

Figure 4.1

FCM dotplots demonstrating the optimum gating strategy for determining

CD4/CD8 ratios (method 1). CD46+ low side scatter lymphocytes (R l) were

analysed in terms of the CD3+ T cell component (R2). R2 events were confirmed to

be lymphoid cells by backgating to a CD45 side scatter plot (R3). Finally, R3

events were scrutinized by their expression of CD3 and CD4, or CD3 and CD8. The

number of events in the upper right hand quadrant of each of these latter dotplots

was used to calculate the CD4/CD8 ratios.

85

Page 89: Lymphocyte Responses in the Lung in Patients with ...

For the simplified second panel (method 2), CD45+ low side scatter lymphocytes were

analysed directly in terms of CD4 and CD8 expression (figure 4.2). The final 3-colour

panel run on the cytoron was analysed as follows (method 3).

Figure 4.2

2

2

2COQO ^

Gated by R1#f

SSC

Figure 4.2

Dotplots demonstrating the simplified gating strategy for determining the BAL

CD4/CD8 ratios (method 2). CD46+ low side scatter lymphocytes were sent directly

to a second dotplot to differentiate the CD4 and CD8 components.

Figure 4.3

G ated by R2

C D 4 ------------------------------- 1

G ated by R1

ssc

Figure 4.3

Dotplots demonstrating the gating strategy to determine the BAL CD4/CD8 ratios

on samples run on the CytoronAbsolute (method 3). Cells with lymphoid forward

and side scatter (R1) were analysed in terms of their CD3 expression (R2) and

these CD3+ T lymphocytes were finally differentiated into CD4 and CD8 subsets.

86

Page 90: Lymphocyte Responses in the Lung in Patients with ...

Lymphocytes in BAL were gated on their intrinsic properties (figure 4.3) and sent to a

further dotplot in which CD3+ T cells were gated. This CD3+ lymphoid population was

then directly differentiated into its CD4+ and CD8+ components.

4.2.6 Statistics

The different methods for determining the BAL CD4/CD8 ratios were compared

by Spearmans correlation coefficient and by Bland Altman analysis.

4.3 Results4.3.1 Diagnoses in the study population

31 subjects were included in the analysis (table 4.1). Of these 15 had mycobacterium

tuberculosis diagnosed by culture confirmation and in one mycobacterium avium

intracellulari was grown. In 6 patients, sarcoidosis was diagnosed by a combination of

clinical suspicion, typical histological appearances on endobronchial or transbronchial

biopsies and failure to culture mycobacterium tuberculosis. Two bone marrow transplant

patients undenwent BAL for respiratory symptoms and in one cytomegalovirus was

detected by polymerase chain reaction (PCR). Cytomegalovirus was also detected in

BAL from a patient with chronic renal failure (patient 20). In 7 patients, no diagnosis was

determined from the BAL. Of the two pleural fluid specimens analysed, one was from a

patient with tuberculosis and in the other a pathological cause was not identified.

4.3.2 Comparison of CD4/CD8 ratios determined by the ‘gold standard’

(method 1 ) with the simplified technique (method 2 )

The BAL and pleural fluid CD4/CD8 ratios in the study population were compared using

method 1 and method 2 as described above. Comparisons were made both by

determining the correlation between the two methods (figure 4.4) and by Bland Altman

analysis (figure 4.5). An excellent close correlation was achieved (r=0.992). More

importantly. Bland Altman analysis demonstrated a very minimal difference between the

two techniques for BAL and pleural fluid CD4/CD8 ratio determination. When compared

to the method 1, the simplified gating strategy overestimated the CD4/CD8 ratio by only

0.08. Close levels of agreement were demonstrated between the two techniques

87

Page 91: Lymphocyte Responses in the Lung in Patients with ...

Table 4.1 Demographic details, diagnoses and BAL and pleural fluid CD4/CD8

ratios by different methods in the study population

Patient Age/sex DiagnosisCD4/CD8 ratio

Method 1

CD4/CD8 ratio

Method 2

CD4/CD8 ratio

Method 3

1 M45 Sarcoidosis 5.11 5.67 5.19

2 M51 Sarcoidosis 2.59 3.12 3.0

3 M40 Sarcoidosis 10.4 9.88 9.39

4 M52 Sarcoidosis 10.5 11.3 -

5 M30 Sarcoidosis 16.8 16.5 15.8

6 M33 Sarcoidosis 5.72 4.83 5.42

7 F 60 Tubercuiosis 1.56 1.65 1.62

8 F 37 Tubercuiosis 1.15 1.15 1.05

9 F 29 Tuberculosis 3.50 3.94 3.35

10 F 31 Tuberculosis 5.06 5.03 5.19

11 F 46 MAI 4.69 4.99 4.69

12 F 36 Tubercuiosis 3.47 3.32 3.44

13 F 41 Tuberculosis 5.33 5.05 4.12

14 M32 Tuberculosis 8.20 8.92 7.91

15 M24 Tuberculosis 2.62 2.83 2.57

16 F 42 Tuberculosis 2.63 2.37 2.61

17 M32 Tuberculosis 6.43 6.95 7.35

18 M 37 Tuberculosis 0.19 0.19 0.35

19 M27 Tubercuiosis 6.40 6.39 -

20 F 35 Tuberculosis 4.12 4.60 4.24

21 F 38 Tuberculosis 5.01 5.91 4.74

22 F 63 Tuberculosis 1.26 1.20 1.35

23 M27 Cytomegalovirus 0.64 0.63 0.66

24 M 31 Cytomegalovirus 0.52 0.53 0.44

25 M 52 NAD= 1.23 1.26 1.18

26 M29 NAD 0.47 0.45 0.44

27 M43 NAD 3.74 3.49 -

28 M 28 NAD 1.52 1.65 -

29 M38 NAD 1.96 2.53 -

30 F 63 Pleurai fluid NAD 6.37 5.97 8.06

31 M 32 Pleural fluid Tuberculosis 5.42 4.82 5.74

Footnotes MAI= Mycobacterium avium intracellulari

NAD=nothing abnormal detected

Page 92: Lymphocyte Responses in the Lung in Patients with ...

Figure 4 4

181

16-CNIT3 14-

12 -

R?=0.902O 10-

co

0 2 8 10 12 16 184 6 14Œ)4/Œ38 ratio methcd 1

Figure 4.4

Correlation plot for CD4/CD8 ratio determination between the complex

technique (method 1 ) and a more simplified approach (method 2 )

Figure 4.5

5

4

3

| o

i::-3

-4

-5

■0.77

......* ....... ♦.*....... .....................-0.08 (95% Cl:-0.26 to 0.1) • ♦ ^

-0.93

10

Log average CD4/CD8 ratio

Figure 4.5

Bland Altman comparisons between method 1 and 2 for the determination of

CD4/CD8 ratios.

89

Page 93: Lymphocyte Responses in the Lung in Patients with ...

with the differences ranging from + 0.77 to -0.93! The widest limits of agreement for the

CD4/CD8 ratios determined by these two methods were demonstrated in those with the

highest values (mostly in patients with sarcoidosis). At lower values, the limits of

agreement were tighter and there was very little variation when the CD4/CD8 ratios were

<2.5.

4.3.3 Differences between the BAL and pleural fluid CD4/CD8 ratios

measured by method 1 and method 3

This analysis was performed to assess whether a standard method for CD4/CD8

ratio determination in BAL and pleural fluid using CDS to identify T lymphocytes

performed as well as the ‘gold standard’, method 1. 24 BAL specimens and the

two pleural fluid samples were analysed by both methods. The correlation

between these two methods was close (R=0.9B8, figure 4.6).

Figure 4.6

18

16CO

14

12 = 0.98810

8006

4

2

00 2 6 8 10 164 12 14 18

CD4/CD8 ratios method 1

Figure 4.6

Correlation plot between the CD4/CD8 ratios derived on the FACSCalibur by a

complex gating system (method 1) and those derived by a different method on the

Cytoron (method 3).

90

Page 94: Lymphocyte Responses in the Lung in Patients with ...

Bland Altman analysis demonstrated an insignificant underestimation of the CD4/CD8

ratio by the cytoron of 0.02. The limits of agreement were slightly wider (-1.1 to 1.1) than

between method 1 and method 2 that were both performed on the FACSCalibur.

However, at lower CD4/CD8 ratios the limits of agreement where much closer, and the

wider limits of agreement were a feature of higher CD4/CD8 values.

Figure 4.7

5

4

3

2

1

00.02 (95% 01: -1.1 to 1.1)

1

2

3

-4

•50.1 1 10

Log a v e ra g e C D 4 /C D 8 ratio

Figure 4.7

Bland Altman plot comparing the CD4/CD8 ratios in BAL and pleural fluid between

method 1 and method 3

4.4 DiscussionThis study has been the first to assess the precision of flow cytometry for determining

tissue fluid CD4/CD8 ratios by comparing different flow cytometric methods. As the ‘gold

standard’, a recently published method using a combination of CD45-directed gating in

conjunction with CDS gating was used [10] . This gating strategy was designed as a

universal template to produce accurate absolute CD4 and CD8 T cell counts, but was

equally applicable for deriving the CD4/CD8 ratios. However, it was clear that optimum

precision was not an overriding concern for CD4/CD8 ratio measurement in tissue fluids.

Rather, the method should be able to distinguish between those with normal and high

91

Page 95: Lymphocyte Responses in the Lung in Patients with ...

ratios, with the upper limit of normal generally agreed to lie between 2.5 to 3 [11] .

Therefore, the primary aim was to develop a simplified gating strategy with adequate

precision to determine the CD4/CD8 ratios in BAL for clinical diagnostic purposes.

Interestingly, close agreement between these methods was demonstrated by Bland

Altman analysis with the simplified method overestimating the CD4/CD8 ratios by only

0.08. More importantly, the limits of agreement between these two techniques varied

between only 0.77 and -0.93, with the main variability occurring at the higher CD4/CD8

ratios. The conclusion, therefore, is that the simplified method offers no loss of precision

for CD4/CD8 ratio analysis. Such a finding is reassuring, since it might be expected that

the omission of a CD3 antibody could introduce an error into the CD4/CD8 ratios

analysis. The reason for this is that NK cells, which weakly express CD8 but do not

express CD3 may be erroneously included as CD8 cells in the simplified method. It is of

interest, therefore that this analysis included a bone marrow transplant patient (patient

24, table 4.1), in whom BAL was performed only three months after the transplant. Since

it is known that NK cells reconstitute early after transplantation followed by a slower

recovery of T cells [12] , the BAL cellular constituents were examined more closely in

this patient in order to determine the NK cell component. NK cells, defined as CD56+

and CD3-, constituted 20.1% of the total CD45+ low side scatter BAL lymphocyte pool.

Nevertheless, despite this high proportion of NK cells, the CD4/CD8 ratios determined by

both the optimum and the simplified method were virtually identical. The explanation for

this finding is that the gate to differentiate the CD8 component in the simplified method

was placed to include only the CD8 "^ * lymphocytes, thus excluding NK cells.

Both methods 1 and 2 described above were performed on the FACSCalibur flow

cytometer. In this study an additional panel was run using a different flow cytometer, the

CytoronAbsolute. This last method has been extensively employed for deriving CD4 and

CD8 counts in both blood [13] and BAL [14] using a CD3 gating strategy without the

use of panleukogating. Again, close correlations between the ‘gold standard’ method

and the Cytoron method were observed for the generation of CD4/CD8 ratios. Bland

Altman analysis demonstrated that the latter method underestimated the lymphocyte

ratios by only 0.02 and the limits of agreement ranged between -1.1 to 1.1. As with the

first comparison, the greatest variation occurred with the highest CD4/CD8 ratios.

Taken together, these observations demonstrate the remarkable precision of flow

cytometry. Here three different gating methods were employed and two different flow

cytometers used. Despite this, there was clinically insignificant variability between the

92

Page 96: Lymphocyte Responses in the Lung in Patients with ...

different techniques. It has been demonstrated in chapter 3 that CD45 directed gating, in

conjunction with only one other antibody, CD15 to determine the granulocyte component

in BAL was able to discriminate the major leukocyte subpopulations in BAL. Therefore, a

single 4-colour antibody panel can be constructed enabling the differentiation of not only

the lymphocyte, granulocyte and macrophage populations but also the CD4 and CD8

lymphocyte subset ratios. The following monoclonal antibodies were used: CD45, CD15,

CD4 and CD8. This single panel could therefore provide the maximum clinically relevant

information in a rapid and simple manner. Nevertheless, such a panel does not provide

information on the relative proportions of neutrophils and eosinophils within the total

granulocyte pool. The importance of eosinophils discrimination in BAL was discussed in

the previous chapter where it was also demonstrated that monoclonal antibodies against

the IgE receptor, CD23 could be used to discriminate eosinophils from neutrophils within

the CD15+ granulocyte pool. Preliminary observations have suggested that CD23 may

be included in the single panel by staining with CD15 and either CD4 or CD8 conjugated

to the same fluorochrome. This is possible since the difference in the scatter

characteristics between lymphocytes and granulocytes allow these two populations to be

easily distinguished within the CD45 panleukogate. Whether such a panel is

demonstrated to be equally reliable for lymphocyte and leukocyte subsetting remains to

be determined.

In summary, it has been demonstrated here that CD45-directed morphospectral gating

of lymphocytes is sufficient before CD4 and CD8 discrimination to assess the clinically

relevant CD4/CD8 ratio in both BAL and pleural fluid. The ratios generated by this

method varied by a clinically insignificant amount when compared to an optimal gating

strategy. Therefore, these results have made feasible a simple, single panel protocol for

the determination of both the major leukocyte components and the CD4/CD8 lymphocyte

subsets in BAL.

4.5 References1. Epstein PE, Dauber JH, Rossman MD,Daniele RP. Bronchoalveolar lavage in a

patient with chronic berylliosis: evidence for hypersensitivity pneumonitis. Ann

Intern Med 1982;97:213-6.

2. Costabel U, Bross KJ, Guzman J, et al. Predictive value of bronchoalveolar T cell

subsets for the course of pulmonary sarcoidosis. Ann N Y Acad Sol

1986;465:418-26.

93

Page 97: Lymphocyte Responses in the Lung in Patients with ...

3. Drent M, Wagenaar SS, Mulder PH, et al. Bronchoalveolar lavage fluid profiles in

sarcoidosis, tuberculosis, and non-Hodgkin's and Hodgkin's disease. An

evaluation of differences. Chest 1994;105:514-9.

4. Ward K, O'Connor 0, Odium 0 ,Fitzgerald MX. Prognostic value of

bronchoalveolar lavage in sarcoidosis: the critical influence of disease

presentation. Thorax 1989;44:6-12.

5. Dauber JH, Wagner M, Brunsvold S, et al. Flow cytometric analysis of

lymphocyte phenotypes in bronchoalveolar lavage fluid: comparison of a two-

color technique with a standard immunoperoxidase assay. Am J Respir Cell Mol

B/o/1992;7:531-41.

6. Brandt B, Thomas M, von Eiff M, Assmann G. Immunophenotyping of

lymphocytes obtained by bronchoalveolar lavage: description of an all-purpose

tricolor flow cytometric application. J Immunol Methods 1996;194:95-102.

7. Padovan OS, Behr J, Allmeling AM, et al. Immunophenotyping of lymphocyte

subsets in bronchoalveolar lavage fluid. Comparison of flow cytometric and

immunocytochemical techniques. J Immunol Methods 1992;147:27-32.

8. Glencross DK SL, Jani IV, Barnett D and Janossy G. CD45-Assisted

Panleukogating for Accurate, Cost-Effective Dual-Platform CD4+ T-Cell

Enumeration. Cytometry (Clinical Cytometry) 2002;50:69-78

9. Janossy G, Jani IV, Bradley NJ, et al. Affordable CD4(+)-T-cell counting by flow

cytometry: CD45 gating for volumetric analysis. Clin Diagn Lab Immunol

2002;9:1085-94.

10. Bergeron M, Faucher S, Ding T, Phaneuf S,Mandy F. Evaluation of a universal

template for single-platform absolute T-lymphocyte subset enumeration.

Cytometry 2002;50:62-8.

11. Bronchoalveolar lavage constituents in healthy individuals, idiopathic pulmonary

fibrosis, and selected comparison groups. The BAL Cooperative Group Steering

Committee. Am Rev Respir Dis 1990;141:8169-202.

12. Barry SM, Johnson MA,Janossy G. Cytopathology or immunopathology? The

puzzle of cytomegalovirus pneumonitis revisited. Bone Marrow Transplant

2000;26:591-7.

13. Mercolino TJ, Connelly MC, Meyer EJ, et al. Immunologic differentiation of

absolute lymphocyte count with an integrated flow cytometric system: a new

concept for absolute T cell subset determinations. Cytometry 1995;22:48-59.

94

Page 98: Lymphocyte Responses in the Lung in Patients with ...

14. Whitehead BF, Stoehr C, Pinkie C, et al. Analysis of bronchoalveolar lavage from

human lung transplant recipients by flow cytometry. Respir Med 1995;89:27-34.

95

Page 99: Lymphocyte Responses in the Lung in Patients with ...

Chapter 5

Bronchoalveolar Lavage and Other

Tissue Fluid Leukocyte Differentials

Assessed by Flow Cytometry in Patients

with Distinct Clinical Syndromes

96

Page 100: Lymphocyte Responses in the Lung in Patients with ...

5.1 IntroductionIn the previous chapters, flow cytometry was used to simplify and optimize the

discrimination of both the BAL leukocyte differentials and CD4/CD8 ratios. In this

chapter, these parameters were determined in BAL from a large number of patients

investigated for respiratory disease. The results were then correlated with the clinical

findings in order to assess their diagnostic relevance.

Such a study is not new as numerous previous investigators have examined BAL

leukocyte differentials and shown certain characteristic features in a variety of clinical

diseases such as sarcoidosis [1-6], tuberculosis [7-9] and interstitial lung diseases [10,

11]. Nevertheless, these previous studies have exclusively used cytospin techniques to

determine the differentials. Although there have been some investigations using FCM on

BAL from patients with various interstitial lung diseases, these have focused mainly on

analysing either the CD4/CD8 I lymphocyte subset ratios [12, 13], or the lymphocyte

proportions using CD45 panleukogating and light scatter characteristics [14, 15]. Whilst

the BAL lymphocyte percentages and CD4/CD8 ratio are undoubtedly the most useful

cellular characteristics for the diagnosis of sarcoidosis, the omission of details of the

neutrophil component is serious as a BAL neutrophilia in this disease may be an adverse

prognostic factor [16]. Similarly, raised neutrophils in interstitial lung diseases and

tuberculosis may also be relevent to the disease process.

Therefore, a comprehensive FCM system has not previously been applied to the

routine investigation of BAL in patients with respiratory disease and the leukocyte

differentials thus derived assessed for their diagnostic significance. The same flow

cytometric analysis was also performed on a small number of clinical specimens other

than BAL in order to demonstrate that such a system has more widespread clinical

applicability. These specimens included pleural, peritoneal, ascitic and cerebrospinal

fluid.

5.2 Methods5.2.1 Patients

Samples were analysed from patients undergoing routine bronchoscopy for the

investigation of respiratory disease of presumed infectious or inflammatory aetiology. 5

healthy control subjects also underwent bronchoscopy (median age 38; range 25-56, 2

smokers, 3 non-smokers). A small number of specimens of pleural, peritoneal or

97

Page 101: Lymphocyte Responses in the Lung in Patients with ...

cerebrospinal fluid were also investigated after samples had been sent for routine

diagnostic analysis. In total 167 subjects including the controls had BAL differentials

measured by FCM. In addition, five had pleural fluid, one peritoneal fluid and one

cerebrospinal fluid measured by the same procedure.

5.2.2 Bronchoalveolar lavage and bronchial biopsy

BAL was performed as previously detailed and according to British Thoracic Society

guidelines. In cases with focal abnormalities detected on thoracic radiographs or

computed tomograms, the BAL was site-directed to these areas. In those in whom either

diffuse radiological abnormalities were noted, or the chest radiography was normal but

respiratory pathology was still suspected, standard right middle lobe BAL’s were

performed. In several cases with pulmonary tuberculosis, BAL was undertaken from both

a radiologically abnormal and normal area. In these latter cases, 150ml of normal saline

was instilled into the radiologically affected area and 50ml into the right middle lobe.

10ml of blood was collected into a lithium heparinised tube at the time of bronchoscopy.

In patients suspected of having sarcoidosis, endobronchial, and in most cases

transbronchial biopsies were performed in addition to BAL.

5.2.3 Acquisition of pleural, peritoneal and cerebrospinal fluid samples

Samples obtained from pleural, peritoneal and cerebrospinal sites were obtained

using standard sterile procedures by medical staff investigating patients with suspected

clinical disease. In these cases, aliquots were sent to the relevant diagnostic laboratories

and the remainder analysed by flow cytometry.

5.2.4 Routine analysis of Clinical Specimens

All samples other than those from the normal control subjects who underwent BAL

were sent for routine analysis. Since the patients who underwent BAL and other

diagnostic procedures were being investigated for presumed infectious or inflammatory

conditions, an aliquot from all samples was sent to microbiology. Standard culture was

performed with an additional Ziehl-Neelson smear for acid-alcohol fast bacilli,

polymerase chain reaction (PGR) and culture on Lowenstein-Jensen medium if

tuberculosis (IB) was suspected. Fungal culture was also performed in those at high risk

such as bone marrow transplant patients. In almost all specimens a further sample was

sent to cytology. Stained cytology specimens were examined for the presence of acid-

98

Page 102: Lymphocyte Responses in the Lung in Patients with ...

alcohol fast bacilli, pneumocystis carinii, fungal hyphae and viral inclusion bodies. In

addition a comment was made on the relative proportions of the leukocyte populations

although a formal leukocyte differential was not performed. Lastly, in BAL and other

samples where a viral aetiology was considered, an aliquot was sent to virology where

relevant immunofluorescence or enzyme linked immunoabsorbant (ELISA) assays were

performed. In BAL samples, adenovirus, influenza and parainfluenza viruses as well as

respiratory syncitial virus were routinely tested for. In selected samples from patients

who had undergone bone marrow transplantation, or those severely

immunocompromised due to HIV, a cytomegalovirus direct antigen fluorescent foci test

(DEAFF) and cytomegalovirus PCR were performed. All HIV+ BAL specimens were sent

to microbiology, cytology and virology.

Endobronchial, and transbronchial biopsies from patients with sarcoidosis were

examined for the characteristic histological features of the disease. Some cases

presenting with stage 1 pulmonary disease with hilar adenopathy underwent mediastinal

lymph node biopsies following failed endobronchial and transbronchial biopsy

procedures. Patients with suspected sarcoidosis also had their serum angiotensin

converting enzyme (SACE) levels measured routinely in biochemistry.

The pleural, peritoneal and cerebrospinal samples were analysed in a similar manner.

Formal leukocyte differential counts were determined in the microbiology laboratory for

cerebrospinal and peritoneal fluid samples and expressed as the number of cells per

mm . In addition, the protein and glucose concentrations were also determined in these

samples. Further tests such as lactate dehydrogenase levels and pH were performed on

pleural fluid samples.

5.2.5 Preparation of Specimens

BAL specimens were collected on ice and analysed within two hours. Aliquots were sent

for routine analysis and the remainder was prepared for flow cytometric analysis as

detailed in chapter 3. The non-BAL specimens were also prepared in the same way.

Briefly, this involved centrifugation, filtering and a further centrifugation step before the

cell pellets were resuspended up to a volume of 1 ml in phosphate buffered saline

(PBS).

5.2.6 Flow Cytometry

Flow cytometry was performed following staining with CD45 FITC and CD15 PE on both

the prepared BAL samples as well as peripheral blood as detailed in chapter 3. In

99

Page 103: Lymphocyte Responses in the Lung in Patients with ...

addition, a second panel containing CD4-FITC (Royal Free Hospital), CD8-PE (Royal

Free Hospital) and CD3-PEcy5 (Dako, Ely, UK) was also used to assess the CD4/CD8

ratios as described in chapter 4. The samples were run on a Cytoron flow cytometer

using an absolute counting protocol. Gating strategies using a CD45 panleukogate and a

CD15+ granulocyte gate were performed and the lymphocyte, macrophage/monocyte

and granulocyte proportions calculated as detailed previously in chapter 3. The

CD4/CD8 ratios were determined as detailed in chapter 4.

5.2.7 Statistics

Data on leukocyte differentials and CD4/CD8 ratios was not normally distributed and

therefore median values and interquartile ranges were quoted in the text. Comparison

between data sets was performed using Mann-Whitney analysis.

5.3 Results5.3.1 General characteristics of BAL

The median volume of saline instilled during BAL was 180ml (IQR: 180-200ml) and

the median return was 50% (IQR: 40%-55.6%), Table 5.1. Following removal of aliquots

for routine analysis, the remaining BAL (normally more than 25ml) was left for analysis.

When the BAL sample had been washed, filtered and the pellet resuspended up to a

volume of 1 ml, only 50pl was used for the leukocyte differentials and 50pl for the

CD4/CD8 ratios. The remaining 0.9ml of BAL was reserved for further phenotypic and

functional analysis in the HIV-infected group and those with suspected tuberculosis.

Therefore, the number of events acquired following staining with CD45 and GDI 5

represented only 5% of the available sample. The median total number of CD45+

leukocytes acquired from the 50pl sample even after resuspension up to 1ml was 8312

(IQR: 4867-17440).

These findings confirm that routine clinical BAL specimens provide more than

adequate cells for the simple flow cytometric analysis described here. The majority of

samples were of good quality as determined by the percentage of CD45+ leukocytes in

the total number of events acquired by FCM. A median of 82.0% (IQR: 57.4-92.3%) of all

events fell within the panleukogate. The non-leukocyte cells were a mixture of epithelial

cells and non-cellular debris.

1 0 0

Page 104: Lymphocyte Responses in the Lung in Patients with ...

Since an absolute counting FCM was used (CytoronAbsolute), the absolute number of

events in the total (1 ml) BAL sample could be calculated. The median number of CD45+

BAL leukocytes was 7.5 X 10® (IQR: 3.6-14.4 X 10®).

Table 5.1 Characteristics of BAL from study population

Characteristic Median value Inter-quartile range

Saline instilled at BAL (ml) 180 180-200

% return of saline 50% 40-55.6%

Number of leukocytes acquired by FCM 8312 4867-17440

% Leukocytes in total number of BAL events 82.0 % 57.4-92.3%

Total number of leukocytes in BAL sample 7.5X10® 3.6-14.4 X 10®

5.3.2 Diagnoses in patients undergoing BAL

162 patients underwent BAL of whom 70 (43.2%) were HIV+, reflecting the particular

cohort of respiratory patients seen at the Royal Free Hospital. A pathological result was

obtained from the BAL, or endobronchial or transbronchial biopsies in 99 (61.1%), table

5.2. In several patients with sarcoidosis, the BAL provided additional diagnostic

information in terms of the lymphocyte percentages and CD4/CD8 ratio, even though the

endobronchial or transbronchial specimens were non-diagnostic.

The most common BAL diagnosis in both HIV+ and HIV- patients was tuberculosis,

accounting for 51 (31.5%) of all BAL. TB was diagnosed in 12 (17.1%) of BAL from HIV+

patients and 39 (42.4%) of those that were HIV-. Sarcoidosis was diagnosed in 15

patients. Bacterial organisms other than mycobacteria were only cultured in seven

patients (4.3%). The low frequency of bacterial culture positivity may in part have

reflected prior antibiotic usage. Nevertheless, a number of patients in whom a bacterial

infection was suspected, but not proven had a marked increase in BAL neutrophil count.

Two HIV- individuals with Pneumocystis carinii pneumonia (PCP) were

immunocompromised on therapy for lymphoma and of the two HIV- patients with

1 0 1

Page 105: Lymphocyte Responses in the Lung in Patients with ...

Table 5.2 Main BAL diagnoses In HIV- and HIV+ patients

HIV status BAL Diagnosis^ Number

No pathogen 25

Mycobacterium tuberculosis 39

sarcoid 15

Bacterial infection 1

HIV- (n=95) Atypical mycobacteria 2

Pneumocystis carinii 2

Cytomegalovirus 2

other 6

Total 92

No pathogen 41

Mycobacterium tuberculosis 12

Pneumocystis carinii 6

HIV+ (n=70) Bacterial infection 6

Cytomegalovirus 3

other 4

Total 72

Footnotes

1. Diagnosis determined by pathological investigation of BAL (see methods).

2. One HIV+ subject was co-infected with cytomegalovirus and TB.

cytomegalovirus infection, one was a bone marrow recipient and the other had chronic

renal failure. For further analysis, the BAL differentials were only considered in those in

whom a firm clinical diagnosis was determined in order to assess their diagnostic

relevance. The major clinical disease groups studied were those with sarcoidosis,

tuberculosis and HIV.

102

Page 106: Lymphocyte Responses in the Lung in Patients with ...

5.3.3 Sarcoidosis

15 patients were diagnosed with pulmonary saroidosis. The demographic, diagnostic

and clinical features together with the BAL leukocyte differentials and CD4/CD8 ratios

are displayed in table 5.3. Patients were divided dependent on the radiological staging of

their disease into those with stage 1, stage 2 or stage 3 pulmonary disease [17]. No

patients had stage 4 disease. A computed tomogram (CT) of the chest was performed

in all these patients. Stage 1 pulmonary sarcoidosis included those with bilateral hilar

lymphadenopathy without evidence of interstitial or parenchymal disease. Patients with

Stage 2 disease had mediastinal and/or hilar lymphadenopathy with evidence of

pulmonary involvement. Stage 3 disease comprised those with interstitial or

parenchymal disease without lymphadenopathy and stage 4 was those with irreversible

pulmonary fibrosis. The diagnosis was supported by endobronchial or transbronchial

biopsy at bronchoscopy in six patients (37.5%). For a further two patients characteristic

histological features were also determined by mediastinal lymph node biopsy following

inconclusive bronchial biopsies. All BAL and biopsy specimens were sent for

mycobacterial culture and all were negative. In seven cases, the biopsies were not

helpful and a combination of high clinical suspicion, a raised serum angiotensin

converting enzyme (SACE) level and an abnormal BAL lymphocyte profile were

supportive of the diagnosis in most cases (table 5.3). Patient 15 had skin lesions from

which histology demonstrated characteristic features of sarcoidosis.

When analysed together, the BAL leukocyte differentials in the sarcoidosis patients

demonstrated a striking lymphocytosis (median 65.7%, IQR: 46.4- 77.0%). When the

patients were divided into the different stages of pulmonary disease (figure 5.1), the

median percentage lymphocytosis was higher in those with stage 1 (74.4%) than those

with stage 2 disease (51.2%). This difference was not significant (p=0.11). In the single

patient with stage 3 disease the BAL lymphocyte percentage was 35.3%.

The median BAL CD4/CD8 ratio for the whole group was 5.1 (IQR: 3.9-9.5), but as

with the BAL lymphocyte percentages, the CD4/CD8 ratio was higher in stage 1 (median

8.8) than stage 2 sarcoidosis (median 4.4), figure 5.2. The difference between the two

was not significant (p=0.15). In the single case with stage 3 disease, the BAL CD4/CD8

ratio was 4.2.

103

Page 107: Lymphocyte Responses in the Lung in Patients with ...

Table 5.3 Demographic and diagnostic features of patients with sarcoid.

Patient Sex/age Ettinicity^ H isto log / Stage® SAGE‘S

BAL leukocyte %GD4/

GD8®

Lymph Mac Neut

1 M 34 G Non-diagnostic 1 (BHL + EN) 40 78.0 19.5 2.5 8.0

2 F 29 G Non-diagnostic 1 (BHL + EN) 58 70.7 27.8 1.6 11.0

3 M 3 0 G Non-diagnostic 1 (BHL + EN) 112 83.8 15.7 0.5 15.8

4 F 42 G Supportive (med LN) 1 (BHL,ML) 89 65.7 ?1.6 0.9 4.6

5 F 41 G Non-diagnostic 1 (BHL + EN) 85 53.3 45.5 1.2 9.5

6 M 36 A Supportive (med LN) 1 (BHL) 80 81.9 17.9 0.2 4.8

7 F 49 BAG Supportive (EB) 2 (BHL + nodules) 135 44.8 53.4 1.8 8.7

8 F 71 G Supportive (EB+TB) 2 (BHL,ML + nodules) 169 82.0 17.5 0.5 2.5

9 F 40 G Supportive (EB+TB) 2 (BHL + nodules) 61 54.3 44.0 1.7 3.6

10 M 4 0 BA Supportive (LN+liver) 2 (BHL,ML + nodules) 254 68.4 24.7 6.9 9.4

11 M 33 G Non-diagnostic 2 (BHL,ML + nodules) 47 76.0 23.3 0.7 43.4

12 M 29 BA Supportive (EB) 2 (BHL,ML + nodules) 120 48.0 50.9 1.1 2.5

13 F 35 G Supportive (EB) 2 (BHL + nodules) 88 26.8 71.0 2.2 2.0

14 M 4 5 G Non-diagnostic 2 (BHL + nodules) 40 39.6 59.4 1 5.1

15 F 52 BA Non-diagnostic 3 (Reticulo-nodular) 112 35.3 64.6 0.1 4.2

Footnotes

1 Ethnicity: C= Caucasian, A= Asian, BAC= black Afro-Caribbean, BA= black

African, 0= other.

2 Supportive histology included non-caseating granulomas. The biopsy site is in

parenthesis. Med LN = mediastinal lymph node biopsy, EB = endobronchial

biopsy, TB = transbronchial biopsy. Non-diagnostic biopsies failed to

demonstrate granulomas in the lung.

3 Staging of pulmonary sarcoidosis into stage 1, 2 or 3 disease. BHL = bilateral

hilar lymphadenopathy, EN = erythema nodosum, ML = mediastinal

lymphadenopathy.

4 SACE = serum angiotensin converting enzyme. The normal range at the Royal

Free Hospital is < 50.

5 The BAL CD4/CD8 ratio determined by FCM.

104

Page 108: Lymphocyte Responses in the Lung in Patients with ...

Figure 5.1

100n

$

I75-

50-

25-

Stage 1 Stage 2 Stage 3

Figure 5.1

Percentage of BAL lymphocytes by flow cytometry in patients with sacoidosis

according to the stage of their pulmonary disease. The bars represent the median

values for each group. The differences between the groups were not statistically

significant.

Figure 5.2

20-1

o2So

o

stage 1 Stage 2 Stage 3

Figure 5.2

BAL CD4/CD8 ratio determined by FCM in patients with sarcoidosis according to

the stage of their disease. The bars represent median values.

105

Page 109: Lymphocyte Responses in the Lung in Patients with ...

5.3.4 Tuberculosis

51 patients were diagnosed with tuberculosis of which 39 were HIV seronegative

and 12 HIV seropositive. The leukocyte differentials and CD4/CD8 ratios were further

considered in HIV negative group, the majority of whom had pulmonary disease (31,

79.5%). Of the patients with non-pulmonary disease, two had tuberculous

lymphadenopathy, one spinal and one pharyngeal disease. These patients had normal

chest radiographs with failure to culture the organism from BAL. Four patients were

diagnosed with disseminated TB in which there was both pulmonary and extra-

pulmonary involvement. In patient 5 the predominant clinical manifestation was cerebral

tuberculomas. Patient 6 had miliary TB, whilst patients 7 and 8 had predominantly lymph

node disease.

Mycobacterium tuberculosis was confirmed by culture in all but three patients. In

patient 4 with pharyngeal TB, acid fast bacilli were seen within granulomas of a

pharyngeal biopsy, but the specimen was not sent for culture. The pharyngeal mass

resolved on treatment. Patients 33 and 34 in whom a clinical diagnosis of pulmonary TB

was made had suggestive respiratory symptoms and chest radiographic abnormalities

both of which resolved on anti-tuberculous therapy. M. tuberculosis was cultured from

the appropriate tissue biopsies of the patients with non-pulmonary TB, other than patient

4.

In most patients a polymerase chain reaction (PCR) test was also performed on the

BAL. The demographic and diagnostic characteristics as well as the BAL FCM findings

are detailed in table 5.3. The BAL leukocyte differentials demonstrated a lymphocytosis

in many, but not all patients with TB. The BAL lymphocyte percentages were compared

with those from the sarcoidosis patients and five healthy control subjects (figure 5.3).

The difference in the median BAL lymphocyte percentage between the patients with TB

and those with sarcoidosis was highly significant (p< 0.0001). Nevertheless, there was

still considerable overlap in the BAL lymphocyte percentages between these two groups

suggesting that a raised lymphocyte percentage alone would not be a very good

discriminating marker. When compared to the control subjects, there was no significant

difference in the BAL lymphocyte percentages in those with TB (p= 0.07).

Several patients with TB were noted to have very low BAL lymphocyte percentages,

and these were usually associated with a co-existing BAL granulocytosis. The site of the

BAL was detailed in this subgroup by referring to the chest radiographic and/or

computed tomographic findings in addition to the bronchoscopy report.

106

Page 110: Lymphocyte Responses in the Lung in Patients with ...

Table 5.4 Demographic, diagnostic and BAL FCM data of patients with TB

Patient Sex/age Ethnicity^ TB diagnosis BAL findings^ AFB PCR cuit

BAL Leukocyte % Lymph Mac Neut

BALCD4/CD8

1 F 16 BA Spinal TB 42.8 57.1 0.1 2.72 M 2 8 BA Lymph TB - - - 32.0 65.6 2.4 1.23 F 42 BA Lymph TB - - - 29.0 18.1 52.8 2.94 F 39 BUK Pharyngeal TB - - - 46.2 52.6 1.2 3.25 M 3 2 BA Disseminated TB - + + 43.6 55.3 1.1 7.36 M 21 BA Disseminated TB - + + 44.6 53.5 1.9 2.27 F 32 0 Disseminated TB - ND® + 20.2 77.9 2.1 2.78 F 41 BUK Disseminated TB - + + 27.6 70.2 2.2 4.19 F 24 C Pulmonary TB + + + 33.0 30.2 36.8 2.510 M 2 4 A Pulmonary TB + + + 62.0 37.1 0.9 0.411 F 21 BA Pulmonary TB + + + 20.8 6.3 72.9 11.712 M 18 BA Pulmonary TB - + + 33.5 63.7 2.8 2.013 M 2 4 A Pulmonary TB - - + 70.4 27.6 2.0 2.614 M 2 7 C Pulmonary TB - - + 15.7 82.4 1.9 2.415 M 3 7 A Pulmonary TB + + + 41.3 7.2 11.5 1.716 M 35 C Pulmonary TB + + + 4.6 27.9 67.5 1.217 M 31 C Pulmonary TB + + + 15.2 79.7 5.1 1.118 F 26 BA Pulmonary TB + + + 46.8 46 7.2 5.919 F 27 BA Pulmonary TB - - + 10.3 19.5 70.2 1.120 M 2 8 A Pulmonary TB - + + 7.0 70.6 22.4 1.321 M 31 C Pulmonary TB + + + 3.2 15.3 81.5 3.022 M 5 0 BUK Pulmonary TB + + + 0.7 6.0 93.3 2.923 M 19 BA Pulmonary TB + + + 18.0 14.0 68.0 4.324 M 5 5 A Pulmonary TB - + + 39.9 54.8 5.3 5.225 M 2 5 A Pulmonary TB - + + 44.9 43.9 11.2 3.426 M 3 0 A Pulmonary TB - + + 7.1 78.7 14.2 1.727 M 4 0 0 Pulmonary TB - + + 23.1 75.0 1.9 0.328 M 4 8 A Pulmonary TB - ND + 16.8 19.4 63.8 1.029 M 4 0 0 Pulmonary TB + ND + 16.9 14.1 69.0 2.830 M 3 5 0 Pulmonary TB - ND + 18.3 37.0 44.7 1.331 F 66 C Pulmonary TB + + + 1.3 18.3 80.4 0.632 F 35 A Pulmonary TB - - + 29.7 66.0 4.3 3.233 M 3 2 C Pulmonary TB - - - 43.3 52.6 4.1 4.734 M 4 0 C Pulmonary TB - - - 13.1 86.1 0.8 1.035 M 20 A Pulmonary TB - + + 14.6 78.7 6.7 2.336 M 3 3 BA Pulmonary TB + + + 28.2 35.5 36.3 4.637 F 20 A Pulmonary TB - + + 62.1 24.5 13.4 1.438 F C Pulmonary TB + - + 25.4 71.6 3 139 M A Pulmonary TB + ND + 35.8 63.3 0.9 1.7

Footnotes

1. Ethnicity. BA=black African, A=Asian, C=Caucasian, 0=other

2. Acid fast bacilli (APB), polymerase chain reaction (PCR) and culture results for

M. tuberculosis from BAL. Patients 1-3 had positive cultures from tissue biopsies.

Patients 33 and 34 were clinical diagnoses and patient 4 was a combination of

pathological and clinical diagnosis but without culture of the organism.

3. ND=not done

107

Page 111: Lymphocyte Responses in the Lung in Patients with ...

Figure 6.3

1 0 0

IS'o.cQ.E

75-

50-

25-

0-

TB Sarcoid Control

Figure 5.3

The BAL lymphocyte percentages in patients with tuberculosis, sarcoidosis and in

healthy control subjects. The bars represent median values.

Figure 5.475

I8

I>.

50

Pulmonary Pulmonary Non-pulmonaryTB TB cavity TB

Figure 5.4

The percentage of BAL lymphocytes in patients with pulmonary tuberculosis. The

first column contains the lymphocyte percentages when washings were taken

from areas of radiologically abnormal lung that were not cavities. In the second

column washings were taken from cavities and in the third column, washings were

taken from radiologically normal lung.

108

Page 112: Lymphocyte Responses in the Lung in Patients with ...

In cases where washings were performed from a pulmonary cavity, the predominant

leukocytes were granulocytes with a corresponding reduction in the lymphocyte

percentage. In several cases (patients 22 and 31), frank pus was aspirated at BAL. The

BAL lymphocyte percentages were then compared between those in whom washings

were taken from cavities, those with non-cavitatory radiographic abnormalities and those

with normal chest radiographs (figure 5.4). This latter population consisted of those with

non-pulmonary disease. A reanalysis of the BAL lymphocyte percentages following the

separation of those with tuberculosis into different groups based on the type of disease

revealed several interesting features.

First, those with non-pulmonary disease all had a BAL lymphocytosis, a finding that

is of relevance for the antigen-specific analyses detailed in chapter 8. Second, advanced

pulmonary TB with cavitation was characterized by a granulocytosis (neutrophila) with

corresponding low lymphocyte percentages. Nevertheless, even after those with

cavitation were separated from the main group with TB, two patients were noted to have

low BAL lymphocyte percentages, (patients 20 and 26, table 5.3). Interestingly, these

two individuals were entirely asymptomatic, but were referred to the infectious diseases

team at this hospital following the discovery of abnormal chest radiographs on arrival into

the United Kingdom by air. These findings are consistent with the hypothesis that the

immune response plays a primary role in the symptomatology of TB and that in the early

phase of mycobacterial proliferation, patients might be expected to have low BAL

lymphocyte percentages as these cells are actively recruited to the site of infection.

Finally, the BAL lymphocyte percentages were compared in eight patients with TB

in whom washings were simultaneously taken from radiologically normal and abnormal

lung (table 5.4). In two of these subjects (patients 19 and 21, table 4.3), washings were

taken from apical cavities as well as radiologically unaffected lobes. Taken together with

the BAL lymphocyte differentials from the patients with non-pulmonary disease, these

findings demonstrate that a generalised BAL lymphocytosis is a common feature in

patients with pulmonary and non-pulmonary TB. The two caveats are that washings

should not be taken from cavities and also that patients should be symptomatic.

A reanalysis of the BAL lymphocyte percentages following the exclusion of the two

patients with asymptomatic disease and those in whom washings were taken from

cavities demonstrated a significant difference when compared to the healthy controls

(P=0.01), figure 5.5.

109

Page 113: Lymphocyte Responses in the Lung in Patients with ...

Table 5.5 BAL lymphocyte percentages from washings taken from radiologically

abnormal and normal areas in patients with pulmonary tuberculosis

Patient Radiologically abnormal Radiologically normal

1 10.3% 20.3%

2 3.2% 17.2%

2 43.6% 34.4%

4 18.0% 26.4%

5 22.4% 46.2%

6 20.0% 16.7%

7 65.0% 68.5%

8 29.7% 16.3%

Footnotes

1. Washings were taken from apical cavities from patients 1 and 2.

Figure 5.5

7 5(0

1O 5 0 .c Q.E

< 2 5 00

All TB symptomatic Controls and non cavitatory TB

Figure 5.5

Scatter plot demonstrating the BAL lymphocyte percentages in all TB

patients, in symptomatic TB patients when washings were not taken from cavities

and in healthy controls.

110

Page 114: Lymphocyte Responses in the Lung in Patients with ...

It should be noted both that the number of normal controls was small and also that

two non-smoking subjects had rather high BAL lymphocyte percentages of 22.2% and

23.3% respectively. The lymphocyte percentages in healthy subjects generally lie within

the range of 4-18% in non-smokers and 3-8% in smokers [11]. However, it has also been

demonstrated that the lymphocyte percentages in healthy individuals may fluctuate

considerably [18]. The comparison between the BAL lymphocyte differentials in the TB

patients and the controls may reach more statistical significance with a larger control

group.

The CD4/CD8 ratios were also examined in all patients with TB and compared with

the values obtained in patients with sarcoidosis and in control subjects (figure 5.6).

When compared to the patients with sarcoidosis, the BAL CD4/CD8 ratios in patients

with TB were significantly lower (p=0.001). There was no statistical difference in the

CD4/CD8 ratios between the patients with TB and the controls (p=0.44).

Figure 5.6

20-1

o200QÜIO

TB Sarcoidosis Controls

Figure 5.6

BAL CD4/CD8 ratios derived by flow cytometry in patients with tuberculosis,

sarcoidosis and in control subjects.

I l l

Page 115: Lymphocyte Responses in the Lung in Patients with ...

5.3.5 HIV

The final major group to be considered in terms of the BAL leukocyte differentials and

CD4/CD8 ratio were the HIV+ patients. Of the 70 total HIV+ patients in whom BAL was

performed, a diagnosis was achieved in 29 (41.4%). As expected, the CD4 counts were

lower in the patients with respiratory pathogens (median 47 cells/pl IQR: 11-109) than

those in whom no pathogens were found (median 133 cells/^il (IQR: 36-261). In the HIV+

group with respiratory disease, different BAL pathogens occurred at varying levels of

immunosuppression. In patients with bacterial infections, the median blood CD4 count

was 107 cells/|il (IQR: 104-176), in those with tuberculosis it was lower (median 60

cells/pl, IQR: 51-244) whilst the HIV+ group with PCP, CMV, MAI, cryptococcus and

invasive strongyloides had the lowest CD4 counts (median 14 cells/p.1 IQR: 7-31) (figure

5.7). Since the hallmark of HIV infection is the depletion of CD4 lymphocytes, both the

leukocyte differentials, and more especially the CD4/CD8 ratios would therefore be

expected to be disturbed in HIV+ individuals.

c3OoÛ0

1CQ

500

400-

300-

200 -

100

Figure 5.7

No BAL Bacterial pathogen infection

TB Otherdiagnoses

Figure 5.7

Blood CD4 count in HIV+ patients according to pathogens determined in BAL. The

group denoted ‘other diagnoses’ had Pneumocystis carinii, Cytomegalovirus,

Mycobacterium avium intracellulari, cryptococcus or strongyloides isolated from

the BAL. All data points and median values are shown

112

Page 116: Lymphocyte Responses in the Lung in Patients with ...

In view of this a decision was taken to focus the analysis on BAL from patients in whom

no respiratory pathogens were identified since the effect of any lung pathogens would be

difficult to interpret in the context of co-infection with HIV. Although there were 41

patients in whom no BAL diagnosis was made, a number of these subjects had very high

granulocyte percentages by FCM suggestive of a bacterial infection, despite failure to

culture an organism. Seven patients had a BAL granulocyte percentage of greater than

40% and were therefore excluded, leaving 34 available for analysis.

The remaining 34 patients were categorized according to their blood CD4 count into

three groups; those with a CD4 count of <100 cells/pl, those with 101-200 cells/pl and

those with >201 cells/pl. There was a tendency for an increasing BAL lymphocytosis in

those with lower blood CD4 counts (figure 5.8), although the differences between the

BAL lymphocyte percentages in the highest and lowest CD4 groups did not reach

statistical significance (p=0.06).

Figure 5.8

Q- 30

CD40-100

fi/

CD4101-200

CD4 201 +

Figure 5.8

Comparison of BAL lymphocyte percentages in HIV+ patients without respiratory

pathogens grouped according to their blood CD4 counts. Mean values and

standard error of the mean are shown.

Page 117: Lymphocyte Responses in the Lung in Patients with ...

More interesting was a comparison between the CD4/CD8 ratios in blood and BAL for

each of these CD4 categories. As expected, the CD4/CD8 ratios in both blood and BAL

declined in parallel with a decline in the blood CD4 count. However, for each blood CD4

category, the CD4/CD8 ratio in BAL was lower than that in blood (figure 5.9). The

differences between BAL and blood CD4/CD8 ratios reached statistical significance for

those with a CD4 count <100 and 101-200 cells/pl (p=0.009 and 0.01 respectively), but

not for the highest CD4 group (p=0.22).

Figure 5.9

oro

COÛOÛo

0.75-1

0.50-

0.25-

0.00

CD40-100

CD4101-200

CD4 201 +

Figure 5.9

Comparison of the CD4/CD8 ratios in blood (hatched bars) and BAL (black bars) in

HIV+ patients without respiratory pathogens according to their blood CD4 counts.

Means and standard error of the means are shown.

Taken together, the findings of a declining CD4/CD8 ratio, but an increasing

lymphocytosis in BAL in patients with increasing immunodeficiency suggests that a BAL

CD8 lymphocytosis may be a feature of advanced HIV disease.

In order to directly investigate whether this was the case, this data was also analysed

by separately determining the percentage of CD8 and CD4 cells from the total CD3+ T

cell pool in BAL and blood for each of the blood CD4 categories. CD4 lymphocytes

accounted for fewer of the total number of T cells in BAL than blood for each of the three

CD4 categories and the difference between these two sites was statistically significant

14

Page 118: Lymphocyte Responses in the Lung in Patients with ...

for each category (figure 5.10). When this analysis was performed on CDS lymphocytes,

no statistically significant difference was detected in the proportion of CDS lymphocytes

between BAL and blood for each CD4 category (figure 5.11).

40 -

° o .5 §_ 30

ÜQ +

2 0 -

O CO O

o Ü 10

+COQ Ü2 S

i sou

100n

Ô 754 oQ -

5 0 -

2 5 -

Figure 5.10

p=0.04

p=0.02

p=0.01

X

CD40-100

CD4101-200

Figure 5.11

CD4 201 +

p = 0 . 1 p = 0 . 1 2 p=0.44

CD40-100

CD4101-200

---- \—CD4 201 +

Figures 5.10 and 5.11

Box and whisker plots comparing the percentage of CD4 lymphocytes (figure 5.10)

and CDS lymphocytes (figure 5.11) from the total T cell compartment in BAL

(black) and blood (red) according to blood CD4 categories in HIV+ patients without

respiratory disease. The p values indicate the significance between the T cell

subset percentages in BAL with those in blood for each CD4 category.

15

Page 119: Lymphocyte Responses in the Lung in Patients with ...

When the CD8 proportions in BAL alone were considered, there was a significant

increase in this lymphocyte subset between those in the lowest and the highest blood

CD4 categories (p=0.002). These findings confirm that there is a more profound CD4

lymphopoenia in BAL than blood and that a CD8 lymphocytosis is a characteristic

feature of advanced HIV in both blood and BAL. Since the proportion of CD3+ events

that were neither CD4+ nor CD8+ did not change with declining CD4 count

(approximately 10% in BAL), it is reasonable to attribute the increasing total lymphocyte

percentage in BAL in HIV+ subjects to a CD8 lymphocytosis. Nevertheless, CD3-

lymphocytes such as natural killer cells and B cells could have contributed to the

lymphocytsosis and these were not measured here.

5.4 Analysis of leukocyte differentials in non-BAL fluidsLeukocyte differentials and CD4/CD8 ratios were performed by flow cytometry in tissue

fluids other than BAL in several cases. The samples investigated included pleural fluid

from five patients and ascitic and cerebrospinal fluid from one patient each.

Figure 5.12

ssc

bC D 1 5 C D 4

C D1 58

C D4

U .

S S C C D 1 5 C D4

Figure 5.12

FCM dotplots of leukocyte discrimination and lymphocyte T cell subset analysis in

pleural fluid (a), ascitic fluid (b) and cerebrospinal fluid (c).

116

Page 120: Lymphocyte Responses in the Lung in Patients with ...

As demonstrated in figure 5.12, CD45 directed panleukogating allowed the

discrimination of leukocytes from non-leukocytes and within the leukocyte gate

lymphocytes were easily distinguished in all samples by their low side scatter. The

discrimination of CD15+ granulocytes and CD4/CD8 T cell subsets was also achieved by

FCM in the same manner as with BAL.

5.5 DiscussionThis chapter has examined the BAL leukocyte differentials determined by flow

cytometry in patients with sarcoidosis, tuberculosis and HIV. Although many previous

studies have investigated these cellular features in similar patients using cytospin

technology, the published data using FCM has been limited mainly to determining BAL

CD4/CD8 ratios in sarcoidosis. The data presented here demonstrates broad agreement

with previous cytospin studies. This finding, together with fact that FCM enables the

rapid and precise enumeration of the CD4/CD8 ratios by FCM when compared to

cumbersome immunofluorescent techniques required for cytospin preparations supports

the conclusion of chapter 3 that FCM is the optimum technique for BAL leukocyte

analysis. Moreover, preliminary data presented here has demonstrated that a simple

CD45 directed gating strategy together with CD15 allows the differentiation of the

leukocyte populations in tissue fluids other than BAL. Likewise, the CD4/CD8 ratios in

these fluids can be readily determined by FCM exactly as in BAL.

Aside from this broad conclusion regarding the suitability of FCM for tissue fluid

analysis, the results from this chapter have also stimulated a critical appraisal of the

published literature on the diagnostic relevance of leukocyte differentials in different

respiratory diseases.

Sarcoidosis is the most widely examined disease in terms of BAL leukocyte

differentials. Many investigators have demonstrated that a BAL lymphocytosis and

increased CD4/CD8 ratio are supportive of the diagnosis [2-6, 19]. However, others have

questioned the diagnostic relevance of raised BAL CD4/CD8 ratios [20, 21]. One

problem with the analysis of these BAL parameters is that sarcoidosis is an evolving

disease and therefore patients presenting with early disease may have profoundly

different BAL differentials than those with late pulmonary fibrosis. Therefore studies that

have considered all stages of the disease together may be misleading. For example,

Kantrow et al analysed the CD4/CD8 ratio in 86 patients with sarcoidosis and found that

Only 42% of these had BAL CD4/CD8 ratios >4 [20]. However, in their sample 46% had

117

Page 121: Lymphocyte Responses in the Lung in Patients with ...

stage 2 and 14% stage 3 sarcoidosis. These authors did not distinguish the I cell subset

ratios with different stage of disease presentation. By contrast, the findings recorded in

this thesis support those of other investigators that stage 1 disease is more likely to be

associated with both a BAL lymphocytosis and a raised CD4/CD8 ratio [2, 4].

The conclusion, therefore is that in early sarcoidosis BAL is most likely to be a

helpful diagnostic test. This finding is of significance since in stage 1 disease

endobronchial and transbronchial biopsies are less likely to reveal characteristic non-

caseating granulomata than with stage 2 or 3 disease [22, 23]. The data presented in

this thesis supports these findings since none of the five patients with stage 1

sarcoidosis had diagnostic endobronchial or transbronchial biopsies and two of these

went on to have mediastinal lymph node biopsies. The value of undertaking

endobronchial and transbronchial biopsies may therefore be questioned in those with

presumed stage 1 sarcoidosis in whom Mycobacterium tuberculosis culture and skin

tests are negative and the BAL differentials are characteristic. In particular, it has been

suggested that the risks of performing mediastinal lymph node biopsy outweigh the

potential diagnostic benefits in this setting [24].

When patients with tuberculosis rather than sarcoidosis were examined, similar BAL

features were noted as with previous published data. In particular, a predominant BAL

lymphocytosis but with a CD4/CD8 ratio within the normal range has been described [8,

25]. The data presented in this thesis confirm that a BAL lymphocytosis is a common

feature in tuberculosis. It has also been demonstrated here that a raised BAL

lymphocyte percentage is a hallmark of non-pulmonary and disseminated TB.

When washings were performed from tuberculous cavities, the predominant cell

types were often granulocytes with corresponding decreases in the lymphocyte

percentages. Nevertheless, when washings were performed simultaneously from both

radiologically unaffected and affected areas, a lymphocytosis was noted from the

unaffected lung in all cases. This finding is contrary to that from previous investigators

where lymphocyte percentages in washings from radiologically normal lung were similar

to those seen in control subjects [9].

The demonstration of a BAL granulocytosis when washings were taken from

tuberculous cavities as compared to a lymphocytosis from radiologically normal or non-

cavitatory areas may reflect different outcomes in the battle between pathogen and host

response. It has been known from both murine and human studies that T cell responses,

in particular those involved in the production of type-1 cytokines such as IFN-y and TNF-

118

Page 122: Lymphocyte Responses in the Lung in Patients with ...

a are crucial for the formation of protective granulomas and the control of infection [26-

33]. In the light of these findings, the demonstration of large proportions of granulocytes

and corresponding low percentages of lymphocytes from tuberculous cavities may

represent failure of protective immune responses [34].

The data presented here on the BAL differentials from patients with both sarcoidosis

and tuberculosis is largely in agreement with previously published data. Only a few

healthy control subjects undenwent bronchoscopy and cell differential analysis in this

thesis. Two control subjects had relatively high BAL lymphocyte percentages (22.5 and

23%). Most large studies report the range of lymphocyte percentages to be between 3-

18% in non-smoking subjects and 3-8% in smokers [11]. Nevertheless, wide fluctuations

have been noted in the lymphocyte proportions in healthy subjects who had serial BAL’s

[18].

A large proportion of the BAL samples were obtained from HIV+ patients, reflecting

the mix of patients seen at this institution. As expected, opportunistic infections with

pathogens such as Pneumocystis carinii, cytomegalovirus, Mycobacterium avium

intracellulari and Cryptococcus occurred at low CD4 counts whereas tuberculosis and

other bacterial infections occurred with better preserved CD4 cell counts. The findings of

a relative CD4 lymphopoenia in BAL when compared to blood in patients in whom no

pathogens were determined in the BAL is of interest. Several investigators have

demonstrated that HIV is present in BAL [35-39]. The differences in CD4 lymphocyte

percentages between lung and blood could reflect differences in the HIV viral load

between the two sites. The CD4 lymphopoenia may be a function of either direct HIV

mediated cell death or alternatively of immune activated cell death. Some evidence from

simian models exists that immune activation may be the most important factor in the

local depletion of CD4 lymphocytes since macrophage tropic SIV viral strains

predominated in the lung whilst lymphotropic strains were dominant in the blood [35].

The role of immune activation in HIV pathogenesis is explored further in chapter 6.

These studies into the leukocyte differentials and lymphocyte subset ratios in BAL

demonstrate the difference between the lung and the blood in a variety of different

disease states and provide tantalizing clues to the nature of disease pathogenesis.

Nevertheless, such parameters are crude measurements of processes that are

undoubtedly subtle and complex. What is clear, however is two points: first that the lung

is the relevant investigative site in patients with respiratory disease and second that a

119

Page 123: Lymphocyte Responses in the Lung in Patients with ...

powerful and precise tool such as FCM will be instrumental in exploring the nature of

these immune responses.

5.6 References

1. Semenzato G, Chilosi M, Ossi E, et al. Bronchoalveolar lavage and lung

histology. Comparative analysis of inflammatory and immunocompetent cells in

patients with sarcoidosis and hypersensitivity pneumonitis. Am Rev RespirDls

1985;132:400-4.

2. Ward K, O'Connor C, Odium C and Fitzgerald MX. Prognostic value of

bronchoalveolar lavage in sarcoidosis: the critical influence of disease

presentation. Thorax 1989;44:6-12.

3. Costabel U, Bross KJ, Guzman J, et al. Predictive value of bronchoalveolar T cell

subsets for the course of pulmonary sarcoidosis. Ann N Y Acad Sol

1986;465:418-26.

4. Drent M, van Velzen-Blad H, Diamant M, et al. Relationship between

presentation of sarcoidosis and T lymphocyte profile. A study in bronchoalveolar

lavage fluid. Chest 1993;104:795-800.

5. Poulter LW, Rossi GA, Bjermer L, et al. The value of bronchoalveolar lavage in

the diagnosis and prognosis of sarcoidosis. EurRespirJ 1990;3:943-4.

6. Winterbauer RH, Lammert J, Selland M, et al. Bronchoalveolar lavage cell

populations in the diagnosis of sarcoidosis. Chest 1993;104:352-61.

7. Baughman RP, Dohn MN, Loudon RG and Frame PT. Bronchoscopy with

bronchoalveolar lavage in tuberculosis and fungal infections. Chest 1991;99:92-

7.

8. Hoheisel GB, Tabak L, Teschler H, et al. Bronchoalveolar lavage cytology and

immunocytology in pulmonary tuberculosis. Am J Respir Crit Care Med

1994;149:460-3.

9. Ainslie GM, Solomon JA and Bateman ED. Lymphocyte and lymphocyte subset

numbers in blood and in bronchoalveolar lavage and pleural fluid in various forms

of human pulmonary tuberculosis at presentation and during recovery. Thorax

1992;47:513-8.

120

Page 124: Lymphocyte Responses in the Lung in Patients with ...

10. Hunninghake GW, Kawanami O, Ferrans VJ, et al. Characterization of the

inflammatory and immune effector cells in the lung parenchyma of patients with

interstitial lung disease. Am Rev RespirDls 1981;123:407-12.

11. Bronchoalveolar lavage constituents in healthy individuals, idiopathic pulmonary

fibrosis, and selected comparison groups. The BAL Cooperative Group Steering

Committee. Am Rev RespirDls 1990;141:8169-202.

12. Padovan CS, Behr J, Allmeling AM, et al. Immunophenotyping of lymphocyte

subsets in bronchoalveolar lavage fluid. Comparison of flow cytometric and

immunocytochemical techniques. J Immunol Methods 1992;147:27-32.

13. Mukae H, Kohno 8, Morikawa I , et al. Two-color analysis of lymphocyte subsets

of bronchoalveolar lavage fluid and peripheral blood in Japanese patients with

sarcoidosis. Chest 1994;105:1474-80.

14. Dauber JH, Wagner M, Brunsvold 8, et al. Flow cytometric analysis of

lymphocyte phenotypes in bronchoalveolar lavage fluid: comparison of a two-

color technique with a standard immunoperoxidase assay. Am J Respir Cell Mol

B/o/1992;7:531-41.

15. Brandt B, Thomas M, von Eiff M and Assmann G. Immunophenotyping of

lymphocytes obtained by bronchoalveolar lavage: description of an all-purpose

tricolor flow cytometric application. J Immunol Methods 1996;194:95-102.

16. Drent M, Jacobs JA, de Vries J, et al. Does the cellular bronchoalveolar lavage

fluid profile reflect the severity of sarcoidosis? EurRespirJ 1999;13:1338-44.

17. Beaton A Beaton D and Leitch G. Crofton and Douglas's Respiratory Diseases. 5

ed. Oxford: Blackwell Bcience, 2000

18. Laviolette M. Lymphocyte fluctuation in bronchoalveolar lavage fluid in normal

volunteers. Thorax 1985;40:651-6.

19. Chretien J, Venet A, Danel 0, et al. Bronchoalveolar lavage in sarcoidosis.

Respiration 1985;48:222-30.

20. Kantrow BP, Meyer KG, Kidd P and Raghu G. The CD4/CD8 ratio in BAL fluid is

highly variable in sarcoidosis. EurRespirJ 1997;10:2716-21.

21. Agostini C, Trentin L, Zambello R, et al. CD8 alveolitis in sarcoidosis: incidence,

phenotypic characteristics, and clinical features. Am J Med 1993;95:466-72.

22. Descombes E, Gardiol D and Leuenberger P. Transbronchial lung biopsy: an

analysis of 530 cases with reference to the number of samples. Monaldi Arch

Chest Dis 1997;52:324-9.

121

Page 125: Lymphocyte Responses in the Lung in Patients with ...

23. Bilaceroglu S, Perim K, Gunel O, et al. Combining transbronchial aspiration with

endobronchial and transbronchial biopsy in sarcoidosis. Monaldi Arch Chest bis

1999;54:217-23.

24. Reich JM, Brouns MG, O'Connor EA and Edwards MJ. Mediastinoscopy in

patients with presumptive stage I sarcoidosis: a risk/benefit, cost/benefit analysis.

C/?esM 998; 113:147-53.

25. Drent M, Wagenaar SS, Mulder PH, et al. Bronchoalveolar lavage fluid profiles in

sarcoidosis, tuberculosis, and non-Hodgkin's and Hodgkin's disease. An

evaluation of differences. Chest 1994;105:514-9.

26. Chackerian AA, Perera TV and Behar SM. Gamma interferon-producing CD4+ I

lymphocytes in the lung correlate with resistance to infection with Mycobacterium

tuberculosis. Infect Immun 2001;69:2666-74.

27. Feng CG, Bean AG, Hooi H, et al. Increase in gamma interferon-secreting

CD8(+), as well as CD4(+), T cells in lungs following aerosol infection with

Mycobacterium tuberculosis. Infect Immun 1999;67:3242-7.

28. Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in

resistance to Mycobacterium tuberculosis infection. J Exp Med 1993; 178:2249-

54.

29. Flynn JL, Goldstein MM, Chan J, et al. Tumor necrosis factor-alpha is required in

the protective immune response against Mycobacterium tuberculosis in mice.

Immunity 1995;2:561-72.

30. Faith A, Schellenberg DM, Rees AD and Mitchell DM. Antigenic specificity and

subset analysis of T cells isolated from the bronchoalveolar lavage and pleural

effusion of patients with lung disease. Clin Exp Immunol 1992;87:272-8.

31. Schwander SK, Torres M, Sada E, et al. Enhanced responses to Mycobacterium

tuberculosis antigens by human alveolar lymphocytes during active pulmonary

tuberculosis. J Infect Dis 1998;178:1434-45.

32. Jouanguy E, Lamhamedi-Cherradi S, Altare F, et at. Partial interferon-gamma

receptor 1 deficiency in a child with tuberculoid bacillus Calmette-Guerin infection

and a sibling with clinical tuberculosis. J Clin Invest 1997;100:2658-64.

33. Newport MJ, Huxley CM, Huston S, et al. A mutation in the interferon-gamma-

receptor gene and susceptibility to mycobacterial infection. N Engl J Med

1996;335:1941-9.

122

Page 126: Lymphocyte Responses in the Lung in Patients with ...

34. Condos R, Rom WN, Liu YM and Schluger NW. Local immune responses

correlate with presentation and outcome in tuberculosis. Am J Respir Crit Care

Med 1998;157:729-35.

35. Babas T, Vieler E, Hauer DA, at al. Pathogenesis of SIV pneumonia: selective

replication of viral genotypes in the lung, y/ro/ogy 2001;287:371-81.

36. Twigg ML, Soliman DM, Day RB, etal. Lymphocytic alveolitis, bronchoalveolar

lavage viral load, and outcome in human immunodeficiency virus infection. Am J

Respir Crit Care Med 1999;159:1439-44.

37. Caufour P, Le Grand R, Cheret A, et ai. Secretion of beta-chemokines by

bronchoalveolar lavage cells during primary infection of macaques inoculated

with attenuated nef-deleted or pathogenic simian immunodeficiency virus strain

mac251. J Gen Virol 1999;80:767-76.

38. Semenzato G, Agostini 0, Chieco-Bianchi L and De Rossi A. HIV load in highly

purified CD8+ T cells retrieved from pulmonary and blood compartments. J

Leukoc Biol 1998;64:298-301.

39. Lewin SR, Kirihara J, Sonza S, et ai. HIV-1 DMA and mRNA concentrations are

similar in peripheral blood monocytes and alveolar macrophages in HIV-1-

infected individuals. Aids 1998;12:719-27.

123

Page 127: Lymphocyte Responses in the Lung in Patients with ...

Chapter 6

Increased Proportions of Activated and

Proliferating Memory CD8* T

Lymphocytes in both Blood and Lung

are Associated with Blood HIV Viral

Load.

124

Page 128: Lymphocyte Responses in the Lung in Patients with ...

6.1 IntroductionIn recent years there has been an intense debate about the nature of HIV

pathogenesis. It has been argued that CD4 depletion is primarily a result of infection and

subsequent cell death caused by HIV virions that homeostatic mechanisms are

eventually unable to correct [1, 2]. Others have emphasised that whilst HIV does infect

some CD4 lymphocytes, its primary mechanism of pathogenesis is by causing immune

activation of both CD4 and CD8 cells resulting in apoptosis of this activated population

[3-5]. Evidence in support of the latter theory has been provided by measuring the

proliferation of cells in vivo using radioactive labels such as Bromodeoxyuridine (brdU)

[5] or deuterated glucose [6]. These studies demonstrated reductions in not only CD4,

but also CDS cell proliferation in HIV-infected subjects when the HIV viral load was

reduced by drug therapy. Further support for the importance of immune activation in HIV

pathogenesis has been provided by the study of SIV in different Simian species. Sooty

Mangabeys and African Green Monkeys, which are the natural hosts of SIV tolerate high

SIV viral loads yet maintain relatively normal CD4 counts and live normal lifespans [7, 8j.

By contrast. Macaques infected with SIV undergo a course of infection similar to that in

humans with high viral loads resulting in CD4 cell depletion, illness and death. brDU

labeling studies have demonstrated that the Sooty Mangabeys have low rates of both

CD4 and CD8 cell turnover when compared to the Macaques [9], suggesting that they

have developed mechanisms to avoid immune activated cell death.

Studies in patients infected with HIV have demonstrated that immune activation,

determined by the expression of CD38 on CDS* T lymphocytes is associated with

disease progression [10-14] and that effective antiretroviral therapy results in a decline in

the expression of this marker in parallel with the fall in HIV viral load [15]. These findings

suggest that HIV drives immune activation, although some authors have stressed the

role of additional infections that might contribute to HIV-induced activation [16]

One serious drawback of previous studies has been the exclusive examination of

blood T lymphocytes. The lung was investigated in this study for two reasons: First,

previous studies have demonstrated that the lung is a site of HIV replication [17-20] and

therefore the investigation of this organ in addition to the blood may provide a more

closely associated picture between viral replicative events and immune activation than

the examination of blood alone. Second, the role of additional respiratory pathogens in

stimulating immune activation could be assessed by comparison with H iV subjects in

whom no BAL pathogen was identified.

125

Page 129: Lymphocyte Responses in the Lung in Patients with ...

In this study both the activation and proliferation of CDS'" memory T cells in lung and

blood was investigated. Activation was determined by CD38 expression on CD8+

lymphocytes using a simple, reproducible gating strategy. Proliferation was measured in

both the CD38‘’' ' ‘ and CD38^'"' CD8^ populations using Ki67, a nuclear marker

associated with dividing cells [21, 22].

6.2 Methods6.2.1 Patients

HIV-infected Patients undergoing bronchoscopy for suspected respiratory disease were

invited to take part in the study that was approved by the hospital ethics committee. 35

H iV patients were tested and a control group of 5 healthy individuals was also

examined. The HIV cohort mostly consisted of patients with advanced disease with only

five patients on antiretroviral therapy. The median CD4 count of the H lV patients was 75

cells/|il (IQR: 11-265) and median HIV viral load was 164,000 copies/ml (IQR: 49,300-

413,000) at the time of investigation. The demographic characteristics, CD4 counts, HIV

viral loads and BAL diagnoses of the study population are depicted in table 6.1.

6.2.2 Determination of HIV Viral Load

HIV viral loads were determined by the automated amplicor polymerase chain reaction

(Cobas Amplicor; Roche Diagnostics, Basel, Switzerland). The minimal level of detection

was 50 HIV copies/ml and the upper limit 750,000 copies/ml. Viral load was not

determined in BAL for the following reason. The procedure of BAL involves the

instillation of large volumes of saline (typically 180-200ml) with the bronchoscope

wedged into a subsegmental bronchus. The return of both fluid and cells is highly

variable and dependant on operator technique, patient tolerability and the pathological

state of the lungs. It is possible to control for these variable factors by measuring the

concentration of standard metabolites such as urea that are found in both BAL and blood

and adjusting the viral load measurements accordingly, but this was not performed here.

6.2.3 Standard Investigations for Respiratory Pathogens in BAL

All BAL samples from HIV+ individuals were investigated for the presence of respiratory

viruses, including influenza, parainfluenza, adenovirus and respiratory syncitial virus

126

Page 130: Lymphocyte Responses in the Lung in Patients with ...

Table 6.1. Demographic, Immunological, Viral and Diagnostic Data of the HIV+Study Population

Patient Sex/age Ethnicity^ Blood

CD4

Blood HIV

VL^

BAL

diagnosis

BAL

Lymphocyte%

BAL CD 4/CD8

Ratio

Antiretroviral

therapy

1 M 5 7 C 527 <50 No pathogens 44.7 0.89 HAART

2 M 35 C 397 <50 No pathogens 20.5 0.21 HAART

3 M 3 2 C 291 <50 No pathogens 3.7 0.71 HAART

4 F 47 BA 272 <400 No pathogens 87.3 0.05 HAART

5 M 4 3 C 185 506 No pathogens 6.4 0.02 HAART

6 M 3 6 BA 12 12,300 tuberculosis 76.3 0.15 HAART

7 M 4 3 BA 78 15,300 tuberculosis 40.1 0.17 none

8 M 2 9 C 566 21,000 No pathogens 6.8 0.4 none

9 M 4 2 C 3 33,600 tuberculosis 9.2 0.04 none

10 M 34 BA 185 65,000 tuberculosis 48.6 0.27 none

11 M 3 7 BA 2 83,200 tuberculosis 37.2 0.27 none

12 F 34 BA 285 88,600 tuberculosis 18.7 0.25 none

13 M 36 0 12 91,600 PCP^ 41.0 0.04 none

14 M 36 BA 258 92,400 No pathogens 87.7 0.04 none

15 M 4 6 C 9 110,000 PCP 81.4 0.002 none

16 M 4 3 0 1 110,000 PGP, CMV^, TB 11.7 0.02 none

17 F 46 0 7 146,000 PCP 22.1 0.14 none

18 M 3 2 0 126 164,000 PCP 61.3 0.07 none

19 M 41 C 399 217,000 No pathogens 24.6 0.16 none

20 M 35 BA 185 220,000 tuberculosis 29.9 0.13 none

21 M 3 0 C 40 229,000 pneumonia 12.8 0.44 none

22 M 3 9 BA 94 246,000 tuberculosis 56.1 0.01 none

23 M 2 7 C 720 261,000 No pathogens 20.6 0.47 none

24 M 34 C 10 306,000 No pathogens 55.3 0.01 none

25 M 4 9 C 75 357,000 No pathogens 33.1 0.01 none

26 M 3 2 0 145 403,000 No pathogens 14.9 0.02 none

Footnotes

Ethnicity; C=Caucasian, BA=black African, 0=other.

HIV viral load copies/ml

PCP= pneumocystis carinii pneumonia

CMV=cytomegalovirus

127

Page 131: Lymphocyte Responses in the Lung in Patients with ...

(RSV). Cytomegalovirus direct antigen fluorescent foci (DEAFF) test and polymerase

chain reaction (PGR) were performed in those with CD4 counts less than 100 cells/pl.

Culture of BAL for bacteria, fungi and mycobacteria was undertaken and in addition, a

stained specimen was examined by a cytopathologist for the presence of pneumocystis

carinii, acid fast bacilli and fungal pathogens.

6.2.4 Bronchoscopy and Sample Preparation

Fibreoptic bronchoscopy was performed according to established methodologies as

previously described in chapter 2. Bronchoalveolar lavage was performed from an area

of radiologically abnormal lung, otherwise standard right middle lobe lavage was

performed. The samples were kept on ice and then divided and aliquots sent to relevant

laboratories for pathological investigations. The remaining BAL was kept for

immunological analysis. The BAL samples were prepared as described in chapter 2. At

the time of bronchoscopy 5ml of EDTA peripheral blood was also taken.

6.2.5 Flow Cytometry and Gating strategies

The proportions of lymphocytes, granulocytes and alveolar macrophages in BAL in

addition to the CD4/CD8 ratios were determined as previously described in chapters 3

and 4. These procedures were carried out using a volumetric flow cytometer (cytoron

absolute, Raritan, New Jersey, USA) that enabled the absolute counts of CD4 and CDS

lymphocytes to be determined.

An aliquot of BAL containing 1x10® CDS'" lymphocytes and a sample of blood

containing the same number of cells were then fixed and permeabilised as previously

described in chapter 2. No lysis step was included as adequate decanting during the

fixation and permeabilisation stage removed nearly all red cells. Following this

procedure, the separate samples were stained at 4°C for 30 minutes, followed by a wash

step. The following monoclonal antibodies were used in pre-titrated optimal

concentrations in a single four-colour panel: KI67 FITC (Immunotech, Marseilles,

France), CD38 PE (Caltag Medsystems, Towcester, UK), CD8 PECy7 (Caltag

Medsystems) and CD45RA APC (Southern Biotechnology, clone sn130, Alabama,

USA). Stained blood and BAL specimens were run on a FACSCalibur flow cytometer

(Becton Dickinson, San Jose, California, US). 20,000 CD8+ lymphocytes were acquired

and the list mode data were analysed using Winlist 4.0 software (Verity inc. Topsham,

Virginia, USA). Primary immunological gating of CDQ* lymphocytes was performed and

128

Page 132: Lymphocyte Responses in the Lung in Patients with ...

these events that were confirmed to lie within a lymphoid scatter gate. These CD8^ cells

were further investigated in terms of their naive/memory phenotype by their CD45RA

isoform expression (Figure 6.1).

Figure 6.1

QO fc

IR1 . R 1 g a t e d

J R2C/5CO

ssc

Û j R 1 + R 2 g a t e d O &

F S C

R5 R4& 1

&

00( R 1 + R 2 + R 3 g a t e d

10* 10* 10’ io' 10’C D 4 5 R A C D 3 8

00Û

R 5 g a t e d

0 . 2 3 %

10* 10* 10 10* 10*

KÎ67 -------------------------►

COQO &

R 4 g a t e d

1 . 9 7 %

10* io

Ki67

Figure 6.1

FCM gating strategy for determining the activation and proliferation status of CD8"

memory lymphocytes. Primary immunological gating of CD8^ cells with low side

scatter (R1) is performed. These events are then confirmed to fall within a tight

lymphoid scatter gate (R2). Events fulfilling both R1 and R2 gating constraints are

then confirmed to be CD45RA (R3). These CD45- CD8^ memory lymphocytes are

then scrutinized in terms of their CD38 expression. 0038*""^^^ (R4) and CD38*""

CD8* memory lymphocytes are finally analysed in terms of their expression of the

proliferation marker, Ki67.

129

Page 133: Lymphocyte Responses in the Lung in Patients with ...

Since CD45RA' memory CD8+ lymphocytes formed the vast majority of BAL

lymphocytes with only a small percentage of CD45RA'^ naive/revertant cells, the degree

of activation in the memory pool in both blood and BAL compartments using CD38

expression was investigated here. Decisions regarding placement of the gate to

differentiate between CD38'' and CD38' CD8 cells were determined from staining of

control blood. The majority of CD8+ T cells in healthy individuals were CD38‘ " with only

a few activated cells, although Natural killer (NK) cells express this marker brightly.

Therefore, a decision was taken to draw a gate to differentiate between the

predominantly CD38' '"’ CD8+ lymphocytes and the CD38^"^^* NK cells. Using this method

on blood from healthy controls that had been fixed and permeabilised, the gate was

drawn at log 1.3 mean fluorescence intensity (MFI) CD38 expression (Figure 6.2). The

proportions of CD38^"^^' and CD38‘ ™ CD8* cells in BAL and blood from the HIV^ study

population and the normal controls was then determined by the same cut-off.

Lastly, the proportions of memory CD8^ lymphocytes that expressed the marker of

cell proliferation, Ki67 were determined in the CD38^"^^‘ and CD38'’'"' subpopulations

(Figure 6.1).

Figure 6.2

C D 3 8

Figure 6.2

Demonstration of the CD38 gating strategy by flow cytometry. CDS' CDB* '"’ natural

killer (NK) cells (R3) form a CDSB* "'® * population, whereas the CD3^, CDB* T

lymphocytes (R2) are predominantly CDSB'*'"’ In a healthy subject. The gate to

differentiate between CD3B^ and CD3B Is drawn at log 10 mean fluorescence

Intensity (MFI) between the NK and the bulk of the CDB T cell population.

Page 134: Lymphocyte Responses in the Lung in Patients with ...

6.2.6 Statistical Analysis

Median values and interquartile ranges were expressed in the text. Non-parametric

analysis by the Mann-Whitney method was used to compare the data sets.

6.3 Results6.3.1 Diagnoses in the HIV* patients with respiratory disease and BAL

lymphocyte percentages

In19 HIV" patients a respiratory pathogen was identified in BAL. The diagnoses were;

nine culture confirmed tuberculosis (TB), seven pneumocystis carinii pneumonia (PCP),

one bacterial pneumonia and one cytomegalovirus (CMV) infection. In addition, one

patient had multiple infections with TB, PCP and CMV simulataneously. In 19 HIV""

patients, no pathogens were identified in BAL. Three subjects in this group were

excluded because FCM demonstrated a marked BAL neutrophilia suggestive of a

bacterial lung infection despite failure to culture an organism. 16 HIV^ patients were

therefore included in this group. The BAL lymphocyte percentages were highly variable

(table 6.1). When compared to the control subjects, BAL from the HIV subjects without

respiratory disease contained a lymphocytosis (median 26.1% vs 10.2%, p=0.06). This

BAL lymphocytosis was more marked in the patients with respiratory disease (median

40.1%, p=0.02 compared to control values).

6.3.2 CD45 Isoform Expression of CD8* I lymphocytes in BAL and blood in

HIV* Patients and control subjects

BAL CDS'" T lymphocytes from HIV" patients were overwhelmingly of a memory

phenotype with a median of 97.5% CD45RA' (IQR: 96.1-97.9%). There was no

significant difference in the proportion of CD45RA' phenotype between those patients

with respiratory pathogens isolated in BAL (median 97.6%) and those in whom no

pathogens were isolated (median 97.1%). In the control patients, slightly fewer (median

91.8% IQR: 86.4-97.2%) of BAL lymphocytes were CD45RA-. Therefore, BAL in both the

HIV-infected patients and the control subjects contained predominantly memory CD8'" T

lymphocytes.

The expression of CD45RA'^ does not delineate a naïve CD8 population, however,

since some memory CD8 lymphocytes may switch from the CD45RA7RO'" isoform to

CD45RAVRO'. These cells can be distinguished from true naïve cells by their lack of

131

Page 135: Lymphocyte Responses in the Lung in Patients with ...

CD27 expression [23]. In BAL approximately a quarter of the CD45RA'" CD8+ T

lymphocytes in this HIV^ cohort also expressed CD27 and were therefore truly naïve

(data shown in chapter 7).

Peripheral blood from the HIV" patients comprised a median value of 56.3% (IQR:

48.5%-73.1%) of CD45RA- CD8* T lymphocytes. The corresponding value in blood from

the control patients was 36.9% (IQR: 32.1-42.0%).

6.3.3 CD38 expression In CD45RA' CD8* lymphocytes from BAL and blood

of HIV* patients and control subjects

The proportion CD38 "^^ CD8" T lymphocytes were examined in both blood and BAL.

The results were stratified according to the blood HIV viral load between the following

categories: undetectable to 1000 copies/ml (low viral load group), 1000 to 100,000

copies/ml (medium viral load) and greater than 100,000 copies/ml (high viral load, figure

6.3). In the first viral load category, three patients had an undetectable HIV viral load with

the assay limit of detection at 50 copies/ml. The remaining subjects had 400 and 506

HIV copies/ml. The upper limit of detection of the viral load assay was 750,000 copies/ml

and four patients had unspecified HIV viral loads above this level. Higher percentages of

CD38 "^^ CD8* T lymphocytes in both blood and BAL were associated with higher blood

HIV viral loads (figure 6.3). For the patients in the lowest category of viral load data the

median percentage of activated CD8 lymphocytes was 29.8% (IQR: 17.8-37.7%) in BAL

and 24.1% in blood (IQR: 21.9-28.4%). In the medium HIV viral load category, the CD8*

lymphocytes were more activated in both blood and BAL compartments with median

values of 42.4% (IQR: 35.9-63.8%) and 53.6% (IQR: 24.9-80.7%) respectively. Lastly, in

the highest viral load category, the CD8 lymphocytes were most activated, with median

values of CD38‘”’' ‘ CD8* T lymphocytes in BAL of 73.5% (IQR: 45.7-88.7%) and in

blood of 74.6% (IQR: 64.8-87.1%). In the control subjects only a minority of BAL (median

4.5%, IQR: 3.2-5.7%) and blood (median 12.3%, IQR: 4.2-14%) of the CD8^

lymphocytes were activated. When the control patients were examined, the percentages

of activated CD8 lymphocytes were much lower than in the HIV" patients in both BAL

(median 4.5% IQR: 3.2-5.7%) and blood (median 12.3% IQR: 4.2-14%).

132

Page 136: Lymphocyte Responses in the Lung in Patients with ...

Figure 6.3

m%oonCl

E>._ i00QO+

00coQO

100-

7 5 -

5 0 -

2 5 -

: :

C o n tro l H IV VL 0-1000

H IV V L 1000 -

100,000

HIV V L> 100 ,000 -

Flgure 6.3

Comparison between the percentages of 0038* "^ * CD8 + T lymphocytes in BAL

(black) and blood (red) in healthy controls and HIV patients according to HIV viral

load categories < 1 0 0 0 copies/ml, 1 0 0 0 -1 0 0 ,0 0 0 copies/ml and >1 0 0 ,0 0 0 copies/ml.

All data points are shown with median values demonstrated by bars.

There was a statistically significant difference in CD8 activation between the control

values and those in the low viral load category in both lung (p=0.03) and blood (p=0.05)

and in the CDB activation between the low and high viral load groups in BAL (p=0.03)

and blood (p=0.002). However, the CDB activation values only reached statistical

significance when comparing the medium-high viral load groups in BAL (p=0.03).

6.3.4 CD38 expression in CD45RA CD8 lymphocytes from BAL of HiV"

patients with and without Respiratory Pathogens

Since higher viral loads were associated with lower CD4 counts and increased rates

of pathogens detected in the lung, the analysis was repeated but this time comparing the

proportion of activated CDB+ lymphocytes for the same HIV viral load categories in BAL

from HIV^ patients with respiratory disease and those in whom no pathogens were

identified in BAL (figure 6.4). The aim for this analysis was to assess the relative

contributions of respiratory pathogens and of HIV viral load in inducing CDB cell

activation.

33

Page 137: Lymphocyte Responses in the Lung in Patients with ...

In the patients in whom BAL revealed no pathogens, the median values for

CD8+ T lymphocytes in BAL were 29.8%, 50.4% and 68.2% for each of the increasing

viral load categories. In the HIV" patients with respiratory pathogens, the median values

were 49.9% and 81.0% for the medium and high viral load categories, since no patients

with respiratory pathogens had a viral load in the lowest range group. There was no

significant difference in the percentage of activated CD8+ lymphocytes between the

patients with respiratory disease and those without (p=0.5 in the highest viral load

category). This data suggests that HIV viral load is the most significant factor in

stimulating CD8 lymphocyte activation.

Figure 6.4

(/)

Io.c

E

COQO+CO

COQÜ

lOOn

75-

50-

25-

HIV VL 0-1000

HIV VL 1000-

100,000

HIV VL >100,000

Figure 6.4

Comparison between 0038'' ' ' * CDS'" T lymphocytes from BAL of HIV* patients in

whom no respiratory pathogens were identified (black) and those in whom

respiratory pathogens were found in BAL (blue) according to HIV viral load

categories. Median values are shown. There were no patients with respiratory

disease in the lowest HIV viral load category.

134

Page 138: Lymphocyte Responses in the Lung in Patients with ...

5.3.5 Expression of KI67 In activated and unactivated CD8" lymphocytes in

lung and blood

The relationship between CD38 activation and CD8 lymphocyte proliferation was next

investigated by comparing the proportions of Ki67^ CD8+ cells in the CD38^"^^ and

CD38' '"’ populations in both BAL and blood (figure 6.5). Activated, CD38 "9^ CD8+ T

lymphocytes were associated with higher percentages of Ki67^ cells in BAL (median

2.37, IQR: 1.65-3.98%) than in the CD38 '"^ CD8+ lymphocytes (median 1.10%, IQR:

0.38-1.535). Increased percentages of Ki67^ CD8 lymphocytes in the activated cells

were also documented in blood (median 1.48%, IQR: 0.77-3.03%) when compared to

the unactivated cells (median 0.04%, IQR: 0-0.29%). These differences between the

Ki67^ populations in the activated and unactivated CD8+ lymphocytes were highly

significant for both compartments (p=<0.0001). In the control subjects, high proportions

of Ki67^ CD8+ cells were noted amongst the rarer CD38^''^^' population in BAL (median

7.68, IQR: 1.96%-11.5%), whilst in the predominant CD38^"^ component, Ki67

expression was very low (median 0.12%, IQR: 0-0.54%). Ki67^ CD8 cells in blood from

the controls were very low both in the CD38^"^^’ (median 0.24%) and the CD38^"^

(median 0.01%) populations.

Figure 6.5

c/30)

oQ.E00QÜN

10.0 -,

7.5-

5.0-

2.5-

0.0

HIV BAL HIV BAL HIV Blood HIV BloodCD38+ CD38- CD38+ CD38-

Flgure 6.5

Box and whisker plots determining the percentage of Ki67^ CD8 * T lymphocytes in

the 0038 " * and CD38^"" populations in both BAL (black) and blood (red).

135

Page 139: Lymphocyte Responses in the Lung in Patients with ...

6.4 Discussion

In this study the relationship between blood HIV viral load and the features of

activation and proliferation of memory CDS'" T lymphocytes in both blood and lung was

investigated. Early studies have noted that CD38 expression on CD8 lymphocytes was

associated with accelerated HIV disease progression [10, 11, 24], a finding that was

most clearly documented in the CD45RO"^ memory CD8'’ pool [12]. This observation can

be explained in the light of the immune activation model of HIV pathogenesis by

postulating that activated lymphocytes proliferate more rapidly and undergo apoptosis at

a much faster rate than unactivated lymphocytes.

Whilst previous studies have demonstrated increased lymphocyte proliferation in HIV-

infected humans and SIV-infected animals using Ki67 expression or radiolabelling, this is

the first study to investigate directly the role that immune activation plays in cell

proliferation in both the blood and a relevant tissue compartment, the lung. In this study it

has been demonstrated that activated, CD38‘’"®* memory CD8* cells have significantly

higher rates of proliferation as measured by Ki67 expression than CD38‘ '"’ CD8* cells in

both blood and lung. This finding, together with the demonstration that the CD8

activation status in both compartments was correlated with the blood HIV viral load is

direct evidence for HIV in promoting increased proliferation.

Some authors have questioned the extent to which Ki67 expression accurately

reflects cell proliferation [14]. This consideration has arisen due to the observation that

nearly half of the KiGT" CD4* lymphocytes also expressed CTLA-4, a marker for

activated cells arrested at the G1 stage of prolferation [25]. However, Ki67 may be a

more reliable measure of cell proliferation for CD8 lymphocytes since only 10% of Ki67*

CD8 cells also co-expressed CTLA-4 [14].

The HIV viral load in the lung has not been measured here since the process of

bronchoalveolar lavage introduces a highly variable dilution factor rendering quantitative

HIV viral loads difficult to interpret. Nevertheless, our data supports the findings of

previous investigators that the lung is a site of HIV replication [17-20].

Since this cohort included HIV-infected patients both with and without respiratory

disease, it was possible to further investigate the relationship between HIV and CD8

lymphocyte activation and to consider whether co-infections with respiratory pathogens

could play an important role in this process. No significant difference between the

CD3 8 bnght CD8* lung lymphocytes was documented between the patients with respiratory

136

Page 140: Lymphocyte Responses in the Lung in Patients with ...

pathogens and those without pathogens in each of the three HIV viral load categories.

This finding is important additional evidence for the primary role of HIV in driving cellular

activation. It cannot be concluded, however, that co-infections may not exacerbate

immune activation, since in our cohort most patients had either TB or PCP. It is

interesting to note that the patient in whom CMV was the only respiratory pathogen

found did not have especially activated BAL CD8+ lymphocytes.

Lastly, this study raises the issue of whether the CD38 activation status of CD8*

lymphocytes could be used as a surrogate marker for HIV viral load that may be

applicable in resource-poor settings. Whilst CD38* "® * CD8 cells were significantly higher

in both blood and BAL, for each viral load category there were several outliers

suggesting that such a marker may not be reliable in routine clinical practice. Moreover,

HIV-infected subjects from resource-poor settings may have increased activation status

of their lymphocytes due to a variety of co-factors as previously demonstrated [16].

In summary, a simple, reliable gating strategy for determining the CD38 activation

statues of CD8'" lymphocytes has been developed here. Using such a system it has

been shown that CD8 cell activation is related to the blood HIV viral load in both blood

and an important tissue site, the lung. The primary role of HIV in stimulating this immune

activation is strengthened by the demonstration that respiratory co-infections did not

significantly increase the CD8 activation state of lung CD8^ lymphocytes when compared

to those with matched HIV viral loads without respiratory pathogens. Lastly, it has also

been documented that the activated, CD38‘’"®* CD8 cells proliferate more than the

CD38'*" cells. Taken together, these findings are consistent with the model that HIV

drives cell activation and proliferation and that this may be the central mechanism of HIV

pathogenesis.

6.5 References

1. Ho DD, Neumann AU, Perelson AS, et al. Rapid turnover of plasma virions and

CD4 lymphocytes in HIV-1 infection. Nature 1995;373:123-6.

2. Mohri H, Bonhoeffer S, Monard 8, et ai. Rapid turnover of T lymphocytes in SIV-

infected rhesus macaques. Science 1998;279:1223-7.

3. Hazenberg MD, Stuart JW, Otto SA, et al. T-cell division in human

immunodeficiency virus (HIV)-I infection is mainly due to immune activation: a

137

Page 141: Lymphocyte Responses in the Lung in Patients with ...

longitudinal analysis in patients before and during highly active antiretroviral

therapy (HAART). Blood 2000;95:249-55.

4. Grossman Z, Meier-Schellersheim M, Sousa AE, et ai. CD4+ T-cell depletion in

HIV infection: are we closer to understanding the cause? Nat Med 2002;8:319-

23.

5. Kovacs JA, Lempicki RA, Sidorov lA, et ai. Identification of dynamically distinct

subpopulations of T lymphocytes that are differentially affected by HIV. J Exp

Med 2001;194:1731-41.

6. Deeks SG, Hoh R, Grant RM, et al. CD4+ T cell kinetics and activation in human

immunodeficiency virus-infected patients who remain viremic despite long-term

treatment with protease inhibitor-based therapy. J Infect Dis 2002;185:315-23.

7. Kaur A, Grant RM, Means RE, et al. Diverse host responses and outcomes

following simian immunodeficiency virus SIVmac239 infection in sooty

mangabeys and rhesus macaques. J Virol 1998;72:9597-611.

8. Broussard SR, Staprans SI, White R, et al. Simian immunodeficiency virus

replicates to high levels in naturally infected African green monkeys without

inducing immunologic or neurologic disease. J Virol 2001;75:2262-75.

9. Kaur A, Barabasz, A., Rosenzweig, M., McClure, H, Feinberg, M. and Johnson,

R. Dynamics of T-Lymphocyte Turnover in Sooty Mangabeys, a Non-Pathogenic

Host of Simian Immunodeficiency Virus Infection. In: 9th Conference on

Retroviruses and Opportunisitc Infections. Seattle, 2002

10. Levacher M, Hulstaert F, Tallet S, et al. The significance of activation markers on

CD8 lymphocytes in human immunodeficiency syndrome: staging and prognostic

value. Clin Exp Immunol 1992;90:376-82.

11. Giorgi JV, Liu Z, Hultin LE, et al. Elevated levels of CD38+ CD8+ T cells in HIV

infection add to the prognostic value of low CD4+ T cell levels: results of 6 years

of follow-up. The Los Angeles Center, Multicenter AIDS Cohort Study. J Acquir

Immune Defic Syndr 1993;6:904-12.

12. Bofill M, Mocroft A, Lipman M, et al. Increased numbers of primed activated

CD8+CD38+CD45RO+ T cells predict the decline of CD4+ T cells in HIV-1-

infected patients. Aids 1996;10:827-34.

13. Liu Z, Cumberland WG, Hultin LE, et al. Elevated CD38 antigen expression on

CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression

to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count.

138

Page 142: Lymphocyte Responses in the Lung in Patients with ...

soluble immune activation markers, or combinations of HLA-DR and CD38

expression. J Acquir Immune Defic SyndrHum Retrovirol 1997;16:83-92.

14. Leng Q, Borkow G, Weisman Z, et al. Immune activation correlates better than

HIV plasma viral load with CD4 T-cell decline during HIV infection. J Acquir

Immune Defic Syndr 2001 ;27:389-97.

15. Tilling R, Kinloch S, Goh LE, et al. Parallel decline of CD8+/CD38++ T cells and

viraemia in response to quadruple highly active antiretroviral therapy in primary

HIV infection. Aids 2002;16:589-96.

16. Bentwich Z, Kalinkovich A and Weisman Z. Immune activation is a dominant

factor in the pathogenesis of African AIDS. Immunol Today 1995;16:187-91.

17. Chayt KJ, Harper ME, Marselle LM, et al. Detection of HTLV-III RNA in lungs of

patients with AIDS and pulmonary involvement. Jama 1986;256:2356-9.

18. Linnemann CC, Jr., Baughman RP, Frame PT and Floyd R. Recovery of human

immunodeficiency virus and detection of p24 antigen in bronchoalveolar lavage

fluid from adult patients with AIDS. Chest 1989;96:64-7.

19. Twigg HL, Soliman DM, Day RB, etal. Lymphocytic alveolitis, bronchoalveolar

lavage viral load, and outcome in human immunodeficiency virus infection. Am J

Respir Crit Care Med 1999; 159:1439-44.

20. Semenzato G, Agostini 0, Chieco-Bianchi L and De Rossi A. HIV load in highly

purified CD8+ T cells retrieved from pulmonary and blood compartments. J

Leukoc Biol 1998;64:298-301.

21. Gerdes J, Lemke H, Baisch H, et al. Cell cycle analysis of a cell proliferation-

associated human nuclear antigen defined by the monoclonal antibody Ki-67. J

Immunol 1984; 133:1710-5.

22. Schwarting R, Gerdes J, Niehus J, et al. Determination of the growth fraction in

cell suspensions by flow cytometry using the monoclonal antibody Ki-67. J

Immunol Methods 1986;90:65-70.

23. Hamann D, Kostense S, Wolthers KC, et al. Evidence that human

CD8+CD45RA+CD27- cells are induced by antigen and evolve through extensive

rounds of division. Int Immunol 1999;11:1027-33.

24. Liu Z, Hultin LE, Cumberland WG, et al. Elevated relative fluorescence intensity

of CD38 antigen expression on CD8+ T cells is a marker of poor prognosis in HIV

infection: results of 6 years of follow-up. Cytometry 1996;26:1-7.

139

Page 143: Lymphocyte Responses in the Lung in Patients with ...

25. Brunner MC, Chambers CA, Chan FK, et al. CTLA-4-Mediated inhibition of early

events of T cell proliferation. J Immunol 1999:162:5813-20.

140

Page 144: Lymphocyte Responses in the Lung in Patients with ...

Chapter 7

Memory Phenotype CD8+ T

Lymphocytes Including CD45RA+ CD27

Revertants Accumulate in the Lung

141

Page 145: Lymphocyte Responses in the Lung in Patients with ...

7.1 IntroductionThe pathways of differentiation of CD8+ T lymphocytes following their encounter with

antigen are understood in some detail. It is well established that changes in the

expression of the CD45RA/RO isoform is now no longer an adequate sole discriminating

marker for differentiating naïve and memory CD8 lymphocytes. Several investigators

have shown that the expression of the co-stimulatory molecules CD28 and CD27 can be

used in conjunction with CD45RA to distinguish subpopulations of memory CD8

lymphocytes [1]. More recently, this analysis has been taken a step further by perfoming

CD8 lymphocyte subset analysis on antigen-specific cells through the use of class-1

tetramers or peptide induced cytokine synthesis methods [2-6]. These studies have

produced remarkable insights into the function of subpopulations of memory CD8

lymphocytes in terms of the ability of these cells to synthesise cytotoxic effector

molecules such perforin or granzymes and cytokines such as IFN-y.

However, the vast majority of studies in this field have concentrated on examining

lymphocyte responses in blood, with a only few important studies directed towards the

responses in lymph nodes [7-9] and other tissues [10]. Thus there is a paucity of

information on the function and phenotype of CD8 lymphocytes at tissue sites such as

the lung in humans. This is an important omission since activated lymphocyte

populations are available for analysis from the lung by simple bronchoalveolar lavage.

Indeed, the lung is a crucial primary site for encountering foreign antigen including viral

infections and tuberculosis and thus the BAL samples might be expected to contain

lymphocytes able to generate responses against various pathogens.

The aim of this chapter was therefore use to investigate CD8+ T lymphocyte subsets

using the discriminatory markers CD27 and CD45RA in order to assess the relative

accumulation of memory and naïve CD8 T lymphocytes in the lung.

7.2 Materials and Methods7.2.1 Patients

Patients undergoing bronchoscopy for suspected non-malignant respiratory disease

were invited to take part in the study, which was approved by the hospital ethics

committee. The majority of patients investigated were HIV seropositive, reflecting the

patient population of the respiratory team. A total of 46 patients were investigated, of

whom 37 were HIV+. Four subjects had sarcoidosis and six were healthy controls

142

Page 146: Lymphocyte Responses in the Lung in Patients with ...

without respiratory disease. The demographic characteristics, BAL diagnoses and CD4

counts of the study population are shown in table 7.1.

Table 7.1 Demographic and diagnostic data for patients undergoing CD8

phenotypic analysis in blood and BAL

Patient Age/sex Diagnoses CD4 HIV viral load^

1 43 M HIV, P. carinii, TB 12 32 F HiV, cytomegalovirus 1 >750.0003 47 M HIV, P. carinii 2 >750,0004 37 M HiV, TB 2 83,2005 42 M HiV, TB 3 33,6006 43 F HiV 7 146,0007 46 M HiV, P.carinii 9 110,0008 37 M HiV, P.carinii 12 91,6009 45 M HiV 18 542,00010 36 M HiV, TB 18 12,30011 28 F HiV 20 423,00012 64 M HiV, P.carinii 21 503,00013 32 M HiV 35 >750,00014 40 M HiV, P.carinii 40 >750,00015 37 F HiV 43 >750,00016 47 F HiV 48 <5017 43 F HiV 55 463,00018 49 M HiV 75 357,00019 39 M HiV, TB 94 246,00020 47 F HiV, P.carinii 100 115021 31 M HiV, Bacterial infection 102 229,00022 36 F HiV, Bacterial infection 111 19723 33 M HiV, P.carinii 126 164,00024 43 M HiV 133 15,30025 34 M HiV 145 403,00026 44 M HiV 165 50627 37 M HiV 171 92,40028 37 M HiV, TB 185 220,00029 47 F HiV, TB 272 40030 34 F HiV, TB 285 88,60031 33 M HiV 291 <5032 35 M HiV 397 <5033 42 M HiV 399 217,00034 58 M HiV 527 <5035 29 M HiV 566 21,00036 27 M HiV 800 261,00037 35 M Sarcoid - -

38 36 M Sarcoid - -

39 29 M Sarcoid - -

40 45 M Sarcoid - -

41 58 M Control - -

42 38 M Control - -

43 30 M Control - -

44 54 M Control - -

45 24 M Control - -

46 36 F Control - -

Footnotes

1: HIV viral load expressed as number of copies/pl in blood. Lower limit of detection 50

copies/pl and upper limit 750,000 copies/pl.

143

Page 147: Lymphocyte Responses in the Lung in Patients with ...

7.2.2 Bronchoscopy

Fibreoptic bronchoscopy was undertaken as previously described in chapter 2.

Bronchoalveolar lavage was performed from an area of radiologically abnormal lung,

othenA/ise a standard right middle lobe lavage was performed. The samples were divided

and aliquots sent to relevant laboratories for pathological investigations, and the

remaining BAL was kept for immunological analysis. At the time of bronchoscopy 5ml of

blood was also taken.

7.2.3 Sample preparation

The BAL samples were collected into a siliconized glass container kept on ice and were

analysed within two hours of their acquisition. Aliquots were sent to the relevant

laboratories for pathological investigation and the remaining sample was filtered and

washed and the absolute CD4 and CD8 counts determined as previously described

using the CytoronAbsolute flow cytometer. A sample of BAL containing 1x10® CD8

lymphocytes and a sample of blood containing the same number of cells were then

stained for 15 minutes at room temperature with the following monoclonal antibodies in

pre-titrated optimal concentrations. CD27 FITC (Becton Dickinson), CD8 PE (Cymbus),

CD3 PECy7 (Caltag Medsystems) and CD45RA (Southern Biotechnology, Alabama,

USA). Following staining, 1ml of lysis buffer was added to the blood sample and left at

room temperature for 15 minutes to ensure red cell lysis. Both the blood and BAL

samples were then washed and the pellets resuspended in PBS. In further selected BAL

samples, staining was performed with both CD45RA and CD45RO in order to confirm

that CD8+ T lymphocytes did not co-express both markers. The antibodies used for

these samples were: CD3 FITC (Becton Dickinson), CD45RO PE (Southern

Biotechnology), CD8 PECy7 (Caltag) and CD45RA (Southern Biotechnology) and

staining was performed as described above.

7.2.4 Flow Cytometry and Gating strategies

Phenotypic analysis of the CD8 populations was performed using the FACScalibur

(Becton Dickinson). 20,000 CD8+ T lymphocytes were acquired and the list mode data

analysed using Winlist 4.0 software (Verity inc. Topsham, Virginia, USA). Primary

immunological gating of CD3+ lymphocytes was performed and these events were

confirmed to lie within a lymphoid scatter gate. Defining CD8+ T lymphocytes by their co-

144

Page 148: Lymphocyte Responses in the Lung in Patients with ...

expression of CD3 and CD8 ensured that there was no contamination with natural killer

(NK) cells that express CD8 dimly, but do not express CDS. CD8+ T cells were then

gated from this population and then further investigated in terms of their naïve/memory

phenotype by the expression of CD45RA and CD27 (figure 7.1).

Lastly, in the BAL samples in which both CD45 isoforms were stained for

simultaneously, the expression of these markers was directly determined on CD8

lymphocytes (figure 7.2).

Figure 7.1

oo

ssc

<a:in

QO

1.2 1.5

37.4 59.9

wC D 2 7

S S C C D 2 7

Figure 7.1

FCM dotplots demonstrating CDB T lymphocyte naïve and memory subsets in BAL

in a patient with HIV infection (a) and sarcoidosis (b). Memory CDB lymphocytes

comprise those cells in the bottom two quadrants and upper left quadrant.

CD45RA+ CD27+ naïve CDB T cells in the upper right quadrant are rare

populations in BAL in both patients.

145

Page 149: Lymphocyte Responses in the Lung in Patients with ...

Figure 7.2

gIf)'4-QO

§ 5.8% 0 .6%

3.1% 90.5%

SSC CD 45RO

Figure 7.2

FCM dotplots demonstrating the expression of the CD45 isoforms CD46RA and

CD45RO in CD8+ T lymphocytes in BAL. The CD45RA+ population of the CD8+

lineage is mainly represented by the CD45RA+,R0- cell type and not by the rare

transitional forms that are CD45RO+,RA+ (upper right quadrant).

7.2.5 Statistical Analysis

Median values and interquartile ranges of the expression of the CDS phenotypic markers

were noted in the text. Non-parametric analysis by the Mann-Whitney method was used

to compare the data sets.

7.3 Results7.3.1 Comparison of the proportion of memory CD8 + T lymphocytes in the

total CDB T cell pool in BAL and bloody

Memory CD8+ T lymphocytes were defined as being either CD45RA- CD27+,

CD45RA- CD27- or CD45RA+ and CD27-, the latter population being consistent with a

revertant phenotype [11]. True naive CD8+ T cells were defined as cells that were both

CD45RA+ and CD27+. In BAL CD8+ T lymphocytes from all subjects were

overwhelmingly of a memory phenotype (median 99.5% IQR: 98.8-99.8%, figure 7.3).

Peripheral blood from the same patients demonstrated a lower predominance of memory

CDS lymphocytes (median 88.0%, IQR: 78.8-93.7%). When compared to the lung, the

difference in memory CDS lymphocytes in the blood was highly significant (p=<0.001).

146

Page 150: Lymphocyte Responses in the Lung in Patients with ...

This data was then further examined to determine whether there was any difference

in the memory CD8 lymphocyte proportions between those with HIV infection and those

with sarcoidosis or the healthy control subjects.

Figure 7.3

100n

90-

70-

BloodBAL

Figure 7.3

Box and whisker plots demonstrating the percentages of CDS T lymphocytes that

expressed a memory phenotype for the whole study population in BAL and blood

In the HIV-infected patients the median BAL CDS memory proportion was 99.5% (IQR:

98.5-99.9%). In the Sarcoidosis patients the equivalent values were median 99.8%,

(IQR: 99.6-99.8%) and for the healthy control subjects they were median 98.5% (IQR:

96.2-99.6%). The proportion of memory CD8+ T cells in BAL did not differ significantly

between those with HIV and the healthy control subjects (p=0.23) or between those with

HIV and sarcoidosis (p=0.16).

When the blood was compared between these three patient groups, there was a

significant difference (p=0.02) between the CDS memory proportion in those with HIV

(median 90.2%, IQR: 83.0-93.9%) and the healthy controls (median 72.7%, IQR: 56.9-

80.2%), but not between the HIV+ patients and those with sarcoidosis.

147

Page 151: Lymphocyte Responses in the Lung in Patients with ...

7.3.2 Expression of CD27 and CD45RA in CDS lymphocyte subpopulations

in the lung and blood

Next, the relative proportions of the four subpopulations of CD8 T lymphocytes

described by the expression of CD45RA and CD27 were compared in the lung and the

blood samples from all patients together (figure 7.4). These subpopulations were

described as follows: CD45RA+ CD27+, CD45RA- CD27+, CD45RA- CD27- and

CD45RA+ CD27-. Naïve CDS lymphocytes characterized by the dual expression of

CD45RA+ and CD27+ were a rare population in the lung, accounting for a median of

0.7% of all CDS cells (IQR: 0.1-2.4%). By contrast, naïve cells were far more common in

the blood (median 13.4%, IQR: 6.0-32.S%). The difference between the proportions of

naïve CDS cells in these two compartments was highly significant (p=<0.0001).

The second population of CDS cells were CD45RA- and CD27+. In BAL these

contributed to a highly variable proportion of the total CDS pool but with a low median

proportion (4.5%, IQR: O.S-54.1%). In blood, these cells were more common (median

32.7%, IQR: 1S.5-51.S%). The difference in the proportions of these CDS cells between

the two sites was significant (p=0.02). The third population were CD45RA- CD27- and

these comprised the majority of the lung CDS lymphocytes (median 92.6%, IQR: 42.5-

97.6%). In the blood, the equivalent cells accounted for a much lower proportion of the

total CDS lymphocyte pool (median 22.7%, IQR: 11.0-40.S%). Again, the difference in

the proportions of these CDS populations between the lung and the blood was significant

(p<0.0001).

The final population was that of the CD45RA+ CD27- CDS cells. It has recently been

determined that a proportion of memory CDS lymphocytes switch their CD45 isoform

from CD45R0+/RA- to CD45RO-/RA+. This population can be distinguished from true

naïve CDS lymphocytes since they have short telomeres, indicating a replicative history

and do not express CD27 [11]. In BAL these cells were rare (median 2.2%, IQR: 0.9-

5.3%), whilst in blood they were much more common (median 31.2%, IQR: 12.4-44.0%).

The difference between the proportions of this memory pool in BAL and blood was again

significant (p<0.0001).

These findings demonstrate that lung directed CDS lymphocytes in BAL consist

ovenA/helmingly of a memory phenotype that are CD45RA- and CD27-. The new

observation is that we can find in the lung a smaller subset of CD45RA+ CDS+ cells that

are also of memory type because of their CD27 negativity. These are known to be

mature memory cells (figure 7.4).

148

Page 152: Lymphocyte Responses in the Lung in Patients with ...

Figure 7.4

BAL

2 .2%0 .7% 4 .5%

C D 4 5 R A - C D 27

92.6%

C D 45R A + C D 2 7 - C D 4 5 R A + C D 2 7 +

Blood13.4%

31.2%

32.7%

22.7%

C D 4 5 R A - C D 2 7 +

Figure 7.4

Pie charts demonstrating the percentages of the CD8 T lymphocyte

subpopulations defined by their expression of CD45RA and CD27 in BAL and

blood for the whole study population.

7.3.3 Differences in CDS lymphocyte subpopulations between patients with

HIV, sarcoidosis and healthy control subjects

Next, the same CDS lymphocyte subpopulations were examined in the three different

patient groups. This analysis was performed since chronic antigenic stimulation in

patients with untreated or inadequately treated HIV infection may result in skewed

populations of lymphocytes with increases in the memory subsets. The majority of the

149

Page 153: Lymphocyte Responses in the Lung in Patients with ...

HIV+ patients in this cohort had advanced disease with low CD4 counts (median 85

cells/pl, IQR: 18-175) and high blood HIV viral loads (median 146,000 copies/pl, IQR:

13,800-413,000). Thus HIV itself could act to stimulate the proliferation and

differentiation of lymphocytes in these patients.

CD45RA+ CD27+ naïve CD8 lymphocytes were rare populations in all three groups

in BAL. In HIV+ subjects they accounted for a median of 0.6% (IQR: 0.1-2.0%), whilst in

the sarcoidosis patients the proportions were very similar (median 0.2% (IQR: 0.1-0.5%).

In BAL from the healthy control subjects, naïve lymphocytes comprised a slightly higher

proportion of the total CD8 pool (median 1.4%, IQR: 0.3-5.4%). The difference between

the naïve lymphocyte proportions in BAL between the HIV+ and control subjects and

between the sarcoidosis and control subjects was not significant (p=0.24 and 0.11

respectively). In blood there was a significant difference (p=0.02) between the naive

CD8 population in the HIV+ subjects (median 11.6%, IQR: 5.6-29.3%) and the healthy

controls (median 33.3%, IQR: 17.0-52.8%).

The next population to be compared was that of the CD45RA- CD27+ memory

lymphocytes. In BAL from the HIV+ individuals this subset comprised a median of 3.1%

of the CD8 cells with a very wide interquartile range (0.8-61.3%). In the control group a

slightly higher proportion (median 14.3%, IQR: 1.1-27.9%) of the CD8 lymphocytes

expressed these markers, but the difference between the two was not significant

(p=0.76). In BAL from patients with sarcoidosis, similar low values of CD45RA- C027+

CD8 cells were determined (median 6.8%, IQR: 3.9-9.1%). In blood, the proportions of

this subset were very similar (p=0.27) in both the HIV+ subjects (median 33.3%, IQR:

18.8-47.9%) and in the controls (median 29.6%, IQR: 17.0-39.6%).

The third population to be compared between the different patient categories was

that of the memory CD45RA- CD27- CD8 lymphocytes. In BAL, these cells comprised

the predominant CD8 lymphoid population in the HIV+ patients (median 94.4%, IQR:

36.2-98.0%), with the corresponding proportions in the sarcoid patients being slightly

lower (median 85.7%, IQR: 80.4-91.5%) and for the controls lower again (median 76.6%,

IQR: 63.3-93.7%). The differences in the proportion of these memory lymphocytes

between all three patient groups were not siginificant (p=0.79 HIV vs control, p=0.95 HIV

vs sarcoid and p=0.61 sarcoid vs control). In the blood, the CD45RA- CD27- memory

CD8 cells were higher in the HIV patients (median 24.7%, IQR: 14.0-41.7%) than in the

healthy controls (median 8.7%, IQR: 4.7-23.2%) and this time the difference was

statistically significant (p=0.02).

150

Page 154: Lymphocyte Responses in the Lung in Patients with ...

Figure 7.5

BAL BloodCD45RA+ CD27-

1.gp/cQ6P/c 31%

HIV+ CD45RA- CD27-

333%

%4% 347%

CD45RA- CD27+ CD45RA+ CD27+

Control

7.7% 1.4%

333%

766% 296%

Figure 7.5

Pie charts demonstrating the percentages of CDS lymphocyte subsets in BAL and

blood from the HIV+ patients and control subjects.

The final population was that of the CD45RA+ CD27- ‘revertant’ memory phenotype.

In BAL from the HIV+ patients this subset accounted for very few of the total CDS pool

(median 1.9%, IQR: 0.7-4.2%), whilst in the control subjects (median 7.7%, IQR: 2.7-

10.5%) and the sarcoidosis patients (median 7.3%, IQR: 4.1-11.0%) the equivalent cells

were more common. The differences were significant between the HIV and control group

(p=0.01) and the HIV and sarcoid group (p=0.008), but not between the controls and

those with sarcoidosis (p=0.76). By contrast, the proportion of CD45RA-I- CD27-

memory CDS lymphocytes in the blood of the HIV patients (median 30.4%, IQR: 13.1-

15

Page 155: Lymphocyte Responses in the Lung in Patients with ...

42.8%) and the control subjects (median 28.4%, IQR: 10.8-45.1%) was very similar

(p=0.75). The differences in the proportions of these CD8 lymphocyte subsets in both

BAL and blood from the HIV+ patients and the healthy controls are graphically

represented in pie charts (figure 7.5).

7.4 DiscussionThis study demonstrates that CD8 lymphocytes recovered from the alveolar space are

overwhelmingly of a memory phenotype. Very few published reports have investigated

the differentiation and migration patterns of lymphocytes to tissue sites such as the lung

in humans [12, 13] or animal models [14, 15] and none of these have considered the

CD8 lymphocyte subsets in the lung using an optimum combination of discriminatory

markers such as CD45RA or RO and CD27.

Here it has been shown that this memory CD8 cell accumulation in the lung is

predominantly of a mature effector phenotype in which the cells do not express the co­

stimulatory marker CD27. These cells are CD45RA- and in a selective series of tests

their CD45RO positivity has been documented. Only very few transitional CD45RA+

R0+ doubles were found in the lungs. Therefore the CD45RA- phenotype is

synonymous with CD45RO positivity.

Several investigators have demonstrated that CD27- CD45RA- CD8 lymphocytes

express high levels of the cytotoxic molecule perforin when compared to the CD27+

CD45RA- CD8 cells in blood [2, 5, 6] and it has therefore been suggested that the

CD27+ CD45RA- phenotype may represent an intermediate stage between CD45RA+

CD27- and CD45RA- CD27- cells. It is of interest that despite the fact that low median

values for the proportion of CD45RA- CD27+ CD8 cells were obtained for all patient

groups, there were many patients in whom this population comprised a significant

minority of the total BAL CD8 pool, particularly in the HIV+ population.

This finding is of interest since it has been established that HIV results in a failure of

maturation of HIV-specific CD8 effector lymphocytes [3, 16, 17]. It would therefore be

important to investigate the antigen specificity of the BAL CD8 lymphocytes in such

patients to determine the relative proportions of HIV-specific cells within the CD27+ and

CD27- pools.

A further significant finding in this study is the discovery of small percentages of

CD45RA revertants among the CD27- memory population. This population of memory

CD8 lymphocytes has been demonstrated to contain perforin and granzyme, to

152

Page 156: Lymphocyte Responses in the Lung in Patients with ...

synthesise both interferon-y (IFN-y) and tumour necrosis factor-a (TNF-a) [1] and also to

express high quantities of the anti-apoptotic molecules Bcl-2 and B c I-X l [11, 18].

Although initially thought to be terminally differentiated [19], it has recently been

demonstrated that these cells can proliferate in response to antigens and show strong

cytotoxic activity [4, 11, 18]. In view of these findings, this subset of CD8 lymphocytes

has been termed an effector memory population and has been implicated in the control

of chronic viral illness [20]. In the control subjects, these CD45RA+ revertants comprised

7.7% of the total BAL CD8 lymphocytes, whilst in the HIV+ subjects the same population

accounted for only 1.9%. Failure of differentiation of CD8 lymphocytes in BAL to this

mature effector phenotype may represent a specific defect induced by the HIV virus. It

would be interesting to determine whether this defect is restricted to the HIV-specific

CD8 lymphocytes, or whether it occurs across a broad range of different antigen-specific

responses, a factor that would help to account for the markedly increased rates of

respiratory infections in this population.

Nevertheless, these findings do indicate that the lung is equipped to maintain long­

term immunity. Indeed, such specific responses to challenge are well documented in the

fast stimulation induced by purified protein derivative (PPD) in the BAL of patients with

tuberculosis (explored in chapter 8).

Lastly, it has been demonstrated here that naïve CD8+ T lymphocytes, characterized

by their co-expression of CD45RA and CD27 are also found in BAL. This subset

accounted for only approximately 1% of the total CD8 pool in BAL in all three patient

groups, in contrast to the higher proportions found in the blood. A small percentage of

these cells designated as memory lymphocytes may not be true CD45RA+ CD27+

lymphocytes since it has been demonstrated that approximately 10% of the CD45RA+

lymphocytes in BAL are transitional forms that co-express the CD45 isoform R0+.

Nevertheless, the conclusion can still be made that naïve CD8 lymphocytes can be

found in the alveolar space at low frequencies.

In summary, this study has demonstrated that a focused investigation of lymphocytes

using good discriminatory markers reveals important differences between the lung and

the blood, in direct conflict to the findings of previous investigators [21]. Here it has been

shown that there is a preferential accumulation of memory CD8 lymphocytes in the lung,

the majority of which displayed a mature effector phenotype characterized by the lack of

expression of the co-stimulatory molecule CD27. Importantly, it has also been

demonstrated that revertant effector memory CD8 cells are also detected in BAL and it is

153

Page 157: Lymphocyte Responses in the Lung in Patients with ...

suggested that these may play a vital role in the immune response against pathogens.

Lastly, differences in the relative proportions of these lymphocytes have been shown

between the HIV infected and normal control subjects. Specifically, the reduction in the

proportion of revertant CD8 cells in BAL in addition to the demonstration of significant

numbers of CD27+ memory cells may indicate a specific defect in the maturation of CD8

lymphocytes induced by HIV.

7.5 References1. Hamann D, Baars PA, Rep MH, et al. Phenotypic and functional separation of

memory and effector human CD8+ T cells. J Exp Med 1997;186:1407-18.

2. Kern F, Khatamzas E, Surel I, et al. Distribution of human CMV-specific memory

T cells among the CD8pos. subsets defined by CD57, CD27, and CD45 isoforms.

Eur J Immunol 1999;29:2908-15.

3. Appay V, Dunbar PR, Callan M, et al. Memory CD8+ T cells vary in differentiation

phenotype in different persistent virus infections. Nat Med 2002;8:379-85

4. Wills MR, Okecha G, Weekes MP, et al. Identification of naive or antigen-

experienced human CD8(+) T cells by expression of costimulation and

chemokine receptors: analysis of the human cytomegalovirus-specific CD8(+) T

cell response. J/mmuno/2002;168:5455-64

5. Tomiyama H, Matsuda T and Takiguchi M. Differentiation of human CD8(+) T

cells from a memory to memory/effector phenotype. J Immunol 2002; 168:5538-

50

6. van Baade D, Hovenkamp E, Callan MF, ef a/. Dysfunctional Epstein-Barr virus

(EBV)-specific CD8(+) T lymphocytes and increased EBV load in HIV-1 infected

individuals progressing to AIDS-related non-Hodgkin lymphoma. Blood

2001;98:146-55.

7. Brodie SJ, Patterson BK, Lewinsohn DA, et a i HIV-specific cytotoxic T

lymphocytes traffic to lymph nodes and localize at sites of HIV replication and cell

death. J Clin Invest 2000;105:1407-17.

8. Goodall JO, Bledsoe P and Gaston JS. Tracking antigen-specific human I

lymphocytes in rheumatoid arthritis by I cell receptor analysis. Hum Immunol

1999;60:798-805.

154

Page 158: Lymphocyte Responses in the Lung in Patients with ...

9. Labarriere N, Pandolfino MC, Raingeard D, et al. Frequency and relative fraction

of tumor antigen-specific T cells among lymphocytes from melanoma-invaded

lymph nodes. /nfJ Cancer 1998;78:209-15.

10. Tan LG, Mowat AG, Fazou 0, at al. Specificity of I cells in synovial fluid: high

frequencies of CD8(+) I cells that are specific for certain viral epitopes. Arthritis

Res 2000;2:154-64.

11. Faint JM, Annels NE, Curnow SJ, et ai. Memory T cells constitute a subset of the

human CD8+CD45RA+ pool with distinct phenotypic and migratory

characteristics. J/mmuno/2001;167:212-20.

12. Agostini C, Trentin L, Zambello R, et ai. CD8 alveolitis in sarcoidosis: incidence,

phenotypic characteristics, and clinical features. Am J Med 1993;95:466-72.

13. Albera 0, Ohio P, Solidoro P, et al. Activated and memory alveolar T-

lymphocytes in idiopathic eosinophilic pneumonia. EurRespirJ 1995;8:1281-5

14. Cerwenka A, Morgan TM and Dutton RW. Naive, effector, and memory CD8 I

cells in protection against pulmonary influenza virus infection: homing properties

rather than initial frequencies are crucial. J Immunol 1999;163:5535-43

15. Mathy NL, Walker J and Lee RP. Characterization of cytokine profiles and

double-positive lymphocyte subpopulations in normal bovine lungs. Am J Vet Res

1997;58:969-75

16. Shankar P, Russo M, Harnisch B, et al. Impaired function of circulating HIV-

specific CD8(+) T cells in chronic human immunodeficiency virus infection. Blood

2000;96:3094-101

17. Van Baarle D, Kostense 8, Hovenkamp E, et al. Lack of Epstein-Barr virus- and

HIV-specific CD27- CD8+ I cells is associated with progression to viral disease

in HIV-infection. A/ds 2002;16:2001-2011

18. Dunne PJ, Faint JM, Gudgeon NH, et al. Epstein-Barr virus-specific CD8(+) I

cells that re-express CD45RA are apoptosis-resistant memory cells that retain

replicative potential. Blood 2002;100:933-40.

19. Champagne P, Ogg GS, King AS, et al. Skewed maturation of memory HIV-

specific CD8 T lymphocytes. Nature 2001;410:106-11

20. Sallusto F, Lenig D, Forster R, et al. Two subsets of memory T lymphocytes with

distinct homing potentials and effector functions. Nature 1999;401:708-12.

155

Page 159: Lymphocyte Responses in the Lung in Patients with ...

21. Bofill M, Lipman M, McLaughlin JE, et al. Changes in lung lymphocyte

populations reflect those seen in peripheral blood in HIV-1 positive individuals.

Eur Respir J 1998; 11:548-53

156

Page 160: Lymphocyte Responses in the Lung in Patients with ...

Chapter 8

Antigen-Specific Responses in the Lung

in Patients with Pulmonary and Non-

Pulmonary Tuberculosis

157

Page 161: Lymphocyte Responses in the Lung in Patients with ...

8.1 IntroductionIt is estimated that one third of the world’s population is infected with mycobacterium

tuberculosis and that there are eight million new cases of TB and nearly three million

deaths each year [1]. The most common route of infection is through the inhalation of

droplets carrying the mycobacterium. This results in a local lung immune response that

generally contains the infection. However, re-infection, or reactivation may occur

resulting predominantly in apical lung disease [2].

The gold standard diagnostic test for tuberculosis remains the visualisation of acid-

alcohol fast bacilli (AFB) by Ziehl-Neelsen or auramine staining, with confirmatory culture

of the organism. This can take up to eight weeks using solid culture medium. However,

only 54% of all cases of TB and 61% of those with pulmonary TB were culture positive in

1999 in the UK [3]. Although the true figures for culture positive TB may be higher due to

underreporting, there still remains a large proportion of TB diagnoses that are made on

clinical grounds alone. More recently, DMA amplification techniques have been

employed as a rapid diagnostic test in suspected TB cases [4-6]. However, the

sensitivity of DMA amplification tests may be reduced in those with smear negative

disease [7, 8].

An alternative diagnostic strategy is suggested by the discovery that antigen-

specific cells can be identified by a variety of different techniques [9-11]. Nevertheless,

these studies have so far been almost exclusively directed towards examining responses

in the blood. We reasoned that in infectious lung diseases antigen-specific responses of

lymphocytes recovered from BAL might prove to be clinically more relevant. We used

flow cytometry (FCM) to detect CD4 lymphocyte cytokine production in response to PPD

in short-term cultures of blood and BAL in patients with suspected TB.

8.2 Methods8.2.1 Patients

The hospital ethics committee approved this study to obtain BAL and blood from patients

with suspected or proven TB. Of the 60 patients included in this study, 34 were

diagnosed with mycobacterium tuberculosis infection (TB). Of these, 29 were confirmed

by culture (table 8.1), including from aspirates of bone (patient 1) and lymph node

(patients 3 and 4). TB was not cultured from the remaining five patients, but acid fast

bacilli were identified in caseating granulomas from tissue specimens in two patients (2

158

Page 162: Lymphocyte Responses in the Lung in Patients with ...

Table 8.1 Demographic and diagnostic results in patients with TB

Patient Sexandage

Ethnicity

AndBCG’

Diagnosis BAL TB diagnosis^

AFB PCR Culture

BAL Lymph %

BALCD4/CD8

BAL CD4IF N Y

Blood CD 4 IFN-y

1 F 16 BA + Spinal TB 42.8 2.7 23.5 0.052 F 36 BUK+ Pharyngeal TB - - - 22.4 3.2 31.1 0.353 M 2 8 BA + Lymph node TB - - - 32.0 1.2 22.7 0.044 F 42 BA + Lymph node TB - - - 29.0 2.9 17.7 0.565 F 33 BA+ Lymph node TB - - - 65.0 5.0 51.6 1.086 F 41 BUK+ Disseminated TB - + + 27.6 4.1 24.4 0.067 F 32 0 + Disseminated TB - - + 16.7 2.7 25.2 0.078 M 3 2 BA + Disseminated TB - + + 43.6 7.3 34.0 0.029 M 7 7 A - Disseminated TB - + - 57.0 1.6 60.p -

10 M 21 BA+ Disseminated TB - + + 44.6 2.2 25.5 0.0111 F 24 C + Pulmonary TB + + + 33.0 2.5 12.4 0.0312 M 2 4 A + Pulmonary TB + + + 62.0 0.4 11.3 1.4313 F 21 BA + Pulmonary TB + + + 20.8 11.7 17.0 0.1314 M 18 BA + Pulmonary TB - + + 33.5 2.0 37.2 0.3915 M 2 4 A + Pulmonary TB - - + 70.4 2.6 17.2 0.0216 M 2 7 C - Pulmonary TB - - + 15.7 2.4 31.7 0.1517 M 3 7 A + Pulmonary TB + + + 32.0 1.7 24.9 0.2318 M 3 5 C + Pulmonary TB + + + 4.60 1.2 25.6 0.0619 M 31 C - Pulmonary TB + + + 15.2 1.1 31.3 0.2120 F 26 BA + Pulmonary TB + + + 46.8 5.9 21.8 0.0221 F 27 BA + Pulmonary TB - - + 20.3 1.3 5.71 0.9222 M 28 A + Pulmonary TB - + + 7.0 1.3 1.98 0.0823 M 31 C + Pulmonary TB + + + 3.2 3.0 31.6 0.0724 M 50 B U K - Pulmonary TB + + + 0.7 2.9 3.01 0.0325 M 19 BA + Pulmonary TB + + + 18.0 4.3 28.4 0.2626 F 25 A+ Pulmonary TB - - + 29.7 3.2 42.5 0.5427 M 2 0 BA+ Pulmonary TB - + + 14.6 2.3 61.3 0.6928 M 33 B A - Pulmonary TB + + + 28.2 4.6 32.8 0.4129 F 20 A - Pulmonary TB - + + 62.1 1.4 26.5 0.8930 M 30 A - Pulmonary TB - + + 7.1 1.7 20.9 0.1031 M 55 0 - Pulmonary TB - + + 39.9 5.2 3.51 0.0632 M 39 C+ Pulmonary TB - - - 13.1 1.0 36.5 0.3033 M 32 c- Pulmonary TB - - - 43.3 4.7 34.0 0.0234 M 37 0 + Pulmonary TB - - + 40.6 11.3 2.07 0.03

Footnotes

1. Ethnicity; BA= black African, BUK= black UK born, A=Asian, C= Caucasian,

0=other. BCG vaccinated (+) or not (-).

2. Diagnosis of TB from BAL by smear (AFB), PCR and culture. In patients 1, 3 and

4 tissue culture confirmed TB. In patients 2 and 5 caseating granulomas and AFB

were seen on biopsy.

3. Percentage of BAL CD4 lymphocytes synthesizing IFN-y.

and 5, table 8.1). Three further subjects had clinical diagnoses of TB of whom two had

resolution of chest radiographic abnormalities on therapy and the final patient had a

159

Page 163: Lymphocyte Responses in the Lung in Patients with ...

positive tuberculin skin test and cerebrospinal fluid changes supportive of tuberculosis

meningitis. All of these five patients responded to anti-tuberculous therapy.

25 of the patients with tuberculosis had pulmonary disease, three had tuberculous

lymphadenopathy, five disseminated disease and one each had spinal and pharyngeal

TB. The patients with disseminated TB had predominantly cerebro-spinal disease

(patients 8 and 9) and lymph node disease (patients 6 and 7). The remaining patient had

classical miliary chest radiographic changes. All patients with TB with the exception of

patient 9 were HIV tested and were seronegative.

The control group comprised of 26 patients with a variety of conditions requiring a

bronchoscopy in which TB was considered in the differential diagnosis (table 8.2). Of

these, eight were diagnosed with sarcoidosis. This diagnosis was made on the basis of

computed tomograms (CT) of the chest demonstrating bilateral hilar lymphadenopathy in

all cases, with or without lung parenchymal nodules in addition to a failure to culture TB.

Supportive lung histology was present in five of the eight patients and additional

diagnostic clues were provided by the BAL lymphocyte percentage and CD4/CD8 ratios,

the serum angiotensin converting enzyme (SAGE) levels and gallium scanning.

Seven patients presenting with respiratory symptoms were confirmed on culture to

have mycobacterial infection other than TB (MOTT), including three with M. fortuitum,

and two with M kansasii and one each with M. chelonei, and M avium intracellulari. Five

of these patients responded to appropriate anti-mycobacterial therapy. The patient with

M. avium intracellulari (MAI) had previously been given a course of anti-mycobacterial

drugs for culture proven MAI that had failed to sterilise the infection and she was not

treated again and a patient with M. Kansasii was noted treated. Two of these patients

were confirmed to be HIV negative and the others were not tested. The remaining 11

patients with non-tuberculous respiratory disease had a variety of different diagnoses

(table 8.2). Two patients were confirmed to have cytomegalovirus (CMV) infection of

whom one had chronic renal failure and the other was a bone marrow recipient. In four

patients no diagnosis was determined from the bronchoscopy, but their symptoms

resolved.

Patients 1,2,7,14,17,18,19 and 20, (table 8.2) were HIV negative and the remainder

were not tested. These patients were generally older and felt not to be in a high HIV risk

group. Moreover, absolute blood CD4 counts measured from these patients as part of

the study methods were all greater than 500 cells/pl). In addition, blood was taken from

20 BCG-vaccinated control subjects who were nursing, medical and laboratory staff at

160

Page 164: Lymphocyte Responses in the Lung in Patients with ...

the Royal Free Hospital. Nine of this control group were females and the median age

was 36 years.

Table 8.2 Demographic and diagnostic results of patients with non-tuberculous

respiratory disease

Patient Sex and age

Ethnicity’

And BCG

Diagnosis BAL Lymph %

BALCD4/CD8

BAL CD4 IFN-y

BloodCD4IFN-y

1 M 4 0 B A - Sarcoidosis 68.4 9.4 15.2 0.082 M 3 3 BA+ Sarcoidosis 31.4 3.0 13.7 0.113 F 35 c+ Sarcoidosis 26.8 2.0 13.9 0.014 F 71 A - Sarcoidosis 81.7 2.5 0.19 0.035 F 41 c + Sarcoidosis 53.3 9.5 0.05 0.026 M 3 3 c + Sarcoidosis 76.0 43.4 0.14 -

7 M 4 5 0 - Sarcoidosis 39.6 5.1 2.99 0.078 F 42 c+ Sarcoidosis 67.5 4.6 4.78 0.079 F 56 c- M fortuitum 48.1 0.7 0.55 0.0110 M 3 8 A+ M fortuitum 13.7 0.7 0.39 0.011 M 6 6 A - M fortuitum 9.2 0.3 8.21 0.1612 F 60 c- MAI 34.5 4.7 2.70 0.0313 M 7 2 C - M kansasii 3.3 21.5 3.08 0.1714 M 4 3 C+ M chelonei 74.4 0.7 0.41 0.0115 M 5 8 C - M Kansasii 13.1 8.5 2.87 0.0516 F 71 0 - Carcinoma 74.5 6.2 0.0 0.0117 M 26 A + Cytomegaiovirus 21.0 0.7 1.12 0.1518 M 19 0 - Cytomegalovirus 13.0 0.5 0.35 0.0619 F 38 BUK + Pneumonia 24.2 2.7 0.57 0.0620 M 6 6 O - Lymphoma 21.4 1.7 0.06 0.0221 F 50 C + Bronchiectasis 4.1 0.4 0.31 0.0622 F 78 C - Lung fibrosis 8.4 1.0 0.45 0.023 F 66 A - No diagnosis 17.9 1.7 0.16 0.0224 F 71 0 - Carcinoma 74.5 6.2 0.0 0.0125 F 61 A - No diagnosis 3.2 1.7 0.83 0.0726 F 70 C - No diagnosis 32.6 7.2 0.83 0.0

Footnotes

1. Ethnicity and BCG staus as defined in table 8.1 above.

8.2.2 Bronchoalveolar lavage

BAL was undertaken by standard technique as described in chapter 2. An area of

radiologically affected lung was washed otherwise the right middle lobe was used. In five

patients that already had a prior microbiological diagnosis of TB, bronchoscopies were

performed within two weeks of commencing TB therapy. In the remaining cases, TB

therapy was commenced following bronchoscopy.

1 6 1

Page 165: Lymphocyte Responses in the Lung in Patients with ...

8.2.3 Sample preparation

The fresh BAL samples were prepared as described in chapter 2 and the washed and

filtered BAL was resuspended in culture medium before FCM analysis to assess the BAL

leukocyte components, CD4/CD8 ratio and the absolute CD4 count as described

previously.

8.2.4 PPD stimulation and FCM analysis

Aliquots of the BAL suspension containing 1X10® CD4+ lymphocytes in 1ml of

culture medium were placed into two sterile 5ml polypropylene tubes (Thermo Life

Sciences, UK). In addition, 1ml of peripheral blood from the same patient collected into

lithium heparin tubes was also placed into two polypropylene tubes. To one of the BAL

and blood samples, lOpg of PPD (Statens Serum Institute, Copenhagen, Denmark) was

added. The other tubes were unstimulated control samples. In selected cases, 0.08 lU of

tetanus toxoid (Pasteur Merieux, Lyon, France) was added to a third BAL sample as a

control antigen. The samples were incubated for two hours at 37°C and 5% CO2, after

which time 5pg of Brefeldin A (Epicentre Technologies, Cambridge, UK) was added and

the samples incubated for a further 14 hours.

Following incubation, the samples were vortexed vigorously to detach cells from the

walls of the tube. First, lymphocyte surface markers were stained using CD4-FITC

(Royal Free Hospital) and CDS-PerCP (Becton Dickinson) for 15 minutes in the dark at

room temperature followed by a wash step. Fixation and permeabilisation of the cells

was performed as previously described using Fix-and-Perm (An-Der-Grub, Kaumberg,

Austria) [12]. Following this, IFN-y-PE (Caltag Laboratories, Towcester, UK) and TNF-a-

APC (Becton Dickinson) were added and the samples stained at 4°C for 30 minutes,

followed by a final wash step. Analysis of the stained preparations was performed on

FACSCalibur (Becton Dickinson). 40,000 CD4+ events were acquired and the proportion

of IFN-y and TNF-a staining cells within the total lymphocyte and CD4+ T cell

populations were analysed using WINMDI software (Version 4.2a; M Trotter) in both the

PPD activated and control cultures. The responses attributable to specific PPD effects

were calculated by subtracting the responses in the control tubes from those in the PPD

activated tubes (figure 8.1).

162

Page 166: Lymphocyte Responses in the Lung in Patients with ...

8.2.5 Statistics

Median values and interquartile ranges were recorded in the text and non-parametric

statistical analysis was performed with the Mann-W hitney test to determine any

difference between data sets.

Figure 8.1

24 .2%

2.3 1%

8 ^

BALNo Ag 0.78%

■ . ■

.. 1.13%

B A LP P D 35 . 1 %

2 . 4 8 %10“ 10' io“ io ‘ 1

T N F - a

d

BloodP P D 0.09%

0.02%

IFN-y IFN-y

Figure 8.1

FCM dotplots demonstrating the proportion of CD3+ lymphocytes producing IFN-y

in response to PPD in BAL (a) and blood (d) from a patient with TB. The TNF-a

response to PPD in BAL (b) and the IFN-y response in BAL when no antigen is

added (c) are also shown. BAL CD4 responses are high (upper left quadrants) with

limited responses in the CD3+ CD4- CD8 cells (lower left quadrants). The

responses in the BAL control (c) and blood sample (d) are low.

163

Page 167: Lymphocyte Responses in the Lung in Patients with ...

8.3 Results

8.3.1 Comparison of IFN ^ and TNF-a responses to PPD In BAL between TB-

Infected and uninfected individuals

The median percentage of BAL CD4 lymphocytes producing IFN-y in response to

PPD in patients with TB was high (25.2%; IQR 17.1-33.2% ). Even higher BAL TN F-a

CD4 responses were noted in these patients (median response 34.5%; IQR: 18.6-

37.1%). FCM analysis determined that both cytokines were mostly produced by the

same activated CD4 lymphocyte population, with the higher mean fluorescence intensity

of TN Fa expression indicating synthesis of more molecules of this cytokine in the

antigen-stimulated CD4 lymphocytes than IFN-y (figure 8.1). When compared to BAL,

the most striking feature in the TB patients was the very low IFN-y and TN F-a synthetic

responses in the blood CD4 lymphocytes (median 0.11% and 0.22% respectively).

By contrast, the BAL CD4 IFN-y production in patients with non-TB respiratory

disease was low in most subjects (median 0.55%; IQR 0.31-2.99%; figure 8.2). The

differences in the CD4 IFN-y responses between the patients with TB and those with

non-tuberculous respiratory disease were highly statistically significantly (p<0.0001).

Figure 8.2

70-1

6 0 -

± 50-

ây 304

2 0 -

10 -

0

TB Sarcoid Controls MOTTFigure 8.2

Scatter plot demonstrating the percentage of BAL CD4 lymphocytes synthesising

IFN^ following incubation with PPD in the TB patients and those with sarcoidosis,

mycobacterium other than TB (MOTT) and a heterogenous group with a variety of

respiratory diagnoses other than TB (controls).

164

Page 168: Lymphocyte Responses in the Lung in Patients with ...

CD4 TNF-a responses to PPD in BAL were also low in the non-TB control patients

(median 2.08% IQR: 0.72-4.07%). The BAL cytokine synthetic responses were low in the

majority of patients regardless of whether they had been BCG-vaccinated (n=10) or not

(n=15). However, when this group were examined more carefully, high type-1 cytokine

responses were noted in several, but not all patients with sarcoidosis (figure 8.2). These

were confirmed to be genuine PPD-specific responses since BAL from each of these

patients was also stimulated with a control antigen, tetanus toxoid, which failed to elicit a

response.

Moderate cytokine responses were also demonstrated in several of the patients with

MOTT. Nevertheless, PPD stimulated lower IFN-y and TN F-a synthesis in these patients

than in the majority of the TB patients (p=0.0007).

8.3.3 Type-1 cytokine responses in PPD-stimulated CD4 lymphocytes in

BAL in patients with pulmonary and non-pulmonary TB

Intriguingly, high CD4 IFN-y and TN F-a responses to PPD in BAL were

demonstrated in TB patients with non-pulmonary disease (median 25.4% and 41.7%

respectively). The median values for the CD4 cytokine responses in the patients with

pulmonary and non-pulmonary disease were virtually indistinguishable (figure 8.3).

Figure 8.3

70-1

6 0 - ■

é*

a " 0-

" 3 0 - ■ i :

1 0 -

0 -

PulmonaryTB

Non-pulmonary/ disseminated TB

Figure 8.3

Percentage of BAL CD4 lymphocytes synthesising IFN^ following incubation with

PPD in patients with pulmonary and non-pulmonary TB.

165

Page 169: Lymphocyte Responses in the Lung in Patients with ...

Importantly, every patient with non-pulmonary TB had high CD4 cytokine responses in

BAL, suggesting that such a test may be of particular diagnostic benefit in this group

8.3.4 Type-1 cytokine synthetic responses to PPD in BAL CD4 and CDS

lymphocytes in patients with TB

In the short incubation period of this assay, PPD was demonstrated to mainly activate

CD4 lymphocytes (median IFN-y 25.2%, IQR: 17.1-33.2%) and not CD8 I lymphocytes

(median IFN-y 4.09%, IQR: 1.57-7.25%). Similarly, the TN F-a responses were low in the

CD8 lymphocytes (median 3.87%, IQR:2.26-6.81%) when compared to the CD4 cells

(median 34.5% , IQR: 18.6-37.1%). In this analysis, CD8 lymphocytes were identified as

being CD3+ and CD4-. This approach to defining CD8 lymphocytes rather than primary

immunological gating of CD8+ events was performed since it ensured that NK cells, that

express CD8 dimly, but do not express CD3 were excluded. However, it is possible that

yô T lymphocytes that also express CD3 may have been erroneously included as

responding CD8 lymphocyte in this analysis. The characterisation of CD4 lymphocyte

cytokine synthesis was the primary objective of this study since it was expected that a

complex antigen such as PPD would primarily activate CD4, rather than CD8

lymphocytes. The use of CD4 alone to determine CD4 cells was not sufficient as this

marker was variably downregulated in culture (figure 2.7), whereas this was not a feature

of CD3 staining. A more precise definition of CD4 and CD8 lymphocytes and their

synthesised cytokines will be possible with five or more colour flow cytometry.

8.3.5 Persistence of type-1 cytokine synthetic responses to PPD in BAL

following initiation of treatment for TB

The PPD-activated CD4 populations in the lung remained high in patients who had

been on anti-tuberculous therapy for up to two weeks (median seven days) prior to BAL

(patients 1,3,8,9, 13,19 and 23, table 1). In addition, steroid therapy did not significantly

attenuate the response in patient 8 who was treated with corticosteroids for ten days

before BAL. Two patients undenA/ent a repeat BAL and PPD stimulation assay within two

months of completion of their TB therapy (table 8.3). In one subject, the responses

returned to low values, but in the other, they did not. More extensive investigation will be

166

Page 170: Lymphocyte Responses in the Lung in Patients with ...

required to determine the duration of persistence of type-1 cytokine responses following

episodes of treated TB.

Table 8.3 BAL lymphocyte percentages and CD4 type-1 cytokine responses In two

patients at diagnosis of TB and following completion of TB therapy.

Patient BAL lymph % BAL CD4 IFN-y BAL CD4 TNF-a

At diagnosis 33.0 15.4 21.3

After TB therapy 35.7 0.17 0.3

At diagnosis 22.4 31.1 35.5

After TB therapy 28.1 25.9 21.8

8.3.6 Type-1 cytokine synthetic responses to PPD in BAL from

radiologically normal and abnormal areas of lung in patients with TB

High cytokine responses to PPD were present throughout the lung and not just

localised to radiologically abnormal areas in patients with TB. In patients with non-

pulmonary TB, BAL was performed from the radiologically unaffected right middle lobe

and in each case high PPD responses were noted (table 8.1). In addition, comparative

washings from areas of radiologically affected and unaffected lung were performed on

selected patients with pulmonary TB. In each case, the CD4 cytokine responses elicited

in BAL taken from radiologically normal lung (usually the right middle lobe) were

equivalent to, and in several cases, higher than the responses in BAL taken from

radiologically affected areas (table 8.4). The conclusion from these findings is that

powerful type-1 cytokine responses in patients with TB are pan-pulmonary and not

specifically directed against areas of pathology such as cavitation defined by chest

radiography.

8.3.7 Comparison of IFN^ and TNF-a responses In the blood of TB patients

with BCG-vacclnated controls

Finally, the low level CD4 responses to PPD detected in the blood of the patients with

TB were compared with those in healthy, BCG-vaccinated control subjects and with non-

BCG vaccinated subjects with respiratory disease other than TB (figure 8.4). Patients

167

Page 171: Lymphocyte Responses in the Lung in Patients with ...

with sarcoidosis and MOTT were excluded from this latter group control since the BAL

analysis suggested that both of these conditions could result in a cytokine synthetic

response to PPD.

Table 8.4 Comparison of IFN^ synthesis following stimulation with PPD in BAL

from radiologically affected and unaffected lung in patients with TB

Patient Chest radiograph % CD4 IFN-y response from abnormal lung

% CD4 IFN-y response from normal lung

1 Upper zone changes 15.4 13.3

2 Upper zone changes 34.0 18.4

3 Upper zone cavity 31.6 56.2

4 Upper zone cavity 1.56 5.71

5 Mid/upper zone changes 28.4 32.9

6 Upper zone changes 21.1 25.2

7 Upper zone changes 42.5 26.7

8 Upper zone changes 51.6 55.9

Figure 8.4

0.0

TB blood

Figure 8.4

BCGvaccinatedblood

Non-BCGnon-TBblood

Percentage of blood CD4 lymphocytes synthesising IFN^ following incubation

with PPD in patients with TB, BCG-vaccinated healthy controls and non-BCG

vaccinated patients with respiratory disease other than TB, MOTT or sarcoidosis.

168

Page 172: Lymphocyte Responses in the Lung in Patients with ...

The median frequency of IFN-y producing CD4+ T lymphocytes in response to PPD in

the TB patients was only 0.11% (IQR: 0.04-0.38%). This value was lower than in the

BCG vaccinated controls (median 0.14%; IQR: 0.08-0.27%). Similarly, the blood CD4

TN F-a responses in the TB patients were low (median 0.22, IQR: 0.10-0.57%) as were

those from the BCG vaccinated controls (median 0.32%, IQR: 0.18-1.23%). The blood

IFN-y responses in the non-BCG vaccinated patients with respiratory disease other than

TB were very low (median 0.02%, IQR: 0.001-0.03%).

8.4 DiscussionThe demonstration of high CD4 IFN-y and TN F-a synthetic responses to PPD in BAL

when compared to the low proportions in peripheral blood is powerful evidence for the

dominance of lung immune responses and for the active recruitment of TB-specific

CD4+ T lymphocytes to the lung during TB infection. Previous human [13-15] and

murine [16, 17] studies have also indicated that lung immune responses predominate

during TB infection in different experimental systems. In two of these human studies [13,

14], T cells were separated from the BAL leukocytes and incubated with peripheral blood

mononuclear cells (PBMC) and TB antigens, before investigating [^H] thymidine

incorporation. These studies used complex experimental techniques due to concerns

regarding the possible suppressive effects of alveolar macrophages and their

unsuitability as antigen presenting cells (APC). The impressive antigen-specific CD4

lymphocyte cytokine responses demonstrated in our simplified system supports the

findings of previous investigators that BAL contains effective APC [18].

The main conclusion of our paper is that the PPD activated CD4+ T cell type-1

cytokine response, measured by IFN-y or TN F-a synthesis is a useful diagnostic test for

active TB in HIV uninfected individuals. Importantly, high CD4 type-1 cytokine responses

were present not only in those with pulmonary disease, but also in patients with non-

pulmonary and disseminated TB in whom extra-pulmonary manifestations dominated the

clinical picture. This finding has particularly significant diagnostic implications as BAL

may be both an easier and safer procedure than the biopsy of other tissues in patients

investigated for occult TB.

The other significant development inherent in this assay is that the results were

available within 24 hours of acquisition of the BAL sample. It is relevant that in this TB

cohort 63% of patients were smear negative, 46% both smear and PCR negative and in

169

Page 173: Lymphocyte Responses in the Lung in Patients with ...

14% M. tuberculosis was not cultured. The rapid results achieved by the

immunodiagnostic test were therefore of considerable diagnostic interest.

The BAL type-1 cytokine synthetic responses following PPD stimulation in several

patients with sarcoidosis is another significant finding and resurrects the fascinating and

enduring question of the pathogenesis of this disease. The predominance of lung

disease, together with the pathological findings of granulomas have long been held to be

evidence that this disease may be a response to mycobacterial antigens [19]. However,

attempts to convincingly document the presence of mycobacteria, or mycobacterial

nucleic acid from tissue specimens of patients with sarcoidosis has resulted in conflicting

evidence [20-23]. Here, it has been demonstrated that some, but not all patients with this

disease have significant BAL CD4 responses following incubation with PPD, but not with

a control antigen. Further investigation in this field is certainly warranted and it will be

interesting to determine which specific mycobacterial antigens optimally stimulate these

responses.

Several cases with TB had a low (<10%) BAL CD4 cytokine responses to PPD. It is

interesting that all but one of these patients were both smear and PCR negative, and

that TB was only confirmed by culture. Three patients were investigated on the basis of

abnormal chest radiographs, but without any of the clinical features of TB such as weight

loss, fever and prolonged cough. The remaining case with a low BAL CD4 cytokine

response had advanced TB with cavitation (patient 24, table 8.1). The BAL from an

apical cavity in this subject revealed pus with only 0.7% of the recovered cells being

lymphocytes. These findings suggest some important limitations of this

immunodiagnostic test. First, BAL CD4 responses may be low in patients with

asymptomatic disease, prior to the recruitment of TB-specific lymphocytes to the site of

disease and the initiation of the potent inflammatory responses. Second, that in

advanced cavitatory TB, characterised by a neutrophilia, lymphocytes may be a scarce

population. It is likely that the development of cavitation reflects a failure of the protective

lymphocyteresponse to control mycobacterial proliferation. Nevertheless, this limitation

may be circumvented as it has been demonstrated here that high CD4 cyokine

responses to PPD are present in BAL taken from radiologically unaffected lung even in

patients with cavitatory disease.

A corollary of these observations is that testing the CD4 responses to PPD in blood

alone is inadequate to diagnose active TB. The blood lymphocyte responses from TB

patients remained indistinguishable from the responses seen in BCG vaccinated control

170

Page 174: Lymphocyte Responses in the Lung in Patients with ...

subjects (figure 8.4). The most likely explanation for the low blood responses seen in the

TB patients is that these antigen-specific lymphocytes are actively recruited to the site of

infection in the lung resulting in their relative depletion in the blood. Furthermore, it has

been demonstrated that BCG vaccinated subjects develop cytokine responses to PPD in

an ELISPOT system [24]. Therefore, PPD which shares epitopes with both

mycobacterium tuberculosis and BCG is not a discriminating stimulatory antigen when

measuring blood lymphocyte responses. Interestingly, BCG vaccination did not provoke

a significant BAL cytokine response to PPD as no difference was demonstrated between

the BCG vaccinated and unvaccinated control patients (table 1).

One could argue that TB infection and BCG vaccination can be better

discriminated by using antigens specific for TB, such as the TB early secretory antigen,

ESAT-6. Overlapping peptides of this antigen have been demonstrated to elicit IFN-y

responses in peripheral blood mononuclear cells (PBMC) in patients with TB, but not in

BCG vaccinated subjects when measured by a sensitive ELISPOT technique [11].

However, this test did not distinguish between active and latent disease. Furthermore,

the use of ESAT-6 does not solve the problem of impaired cytokine responses in blood

from patients with severe TB [25, 26]. These pitfalls are avoided by examining lung

immune responses where high cytokine responses by TB antigen-recruited cells in

addition to a generalised lymphocytosis are supportive of active TB.

The flow cytometric assay used in our study has a considerable advantage over

other methods to determine antigen-specific responses such as the ELISPOT system for

three reasons. First, FCM provides a rapid and precise quantification of the total BAL

lymphocyte percentage together with the proportions and absolute counts of the

responding cell types, CD4 or CDS. Second, different cytokine responses, in our study

IFN-y and TN F-a can be investigated from the same activated lymphocytes. Third, the

new generation of cheap red diode laser flow cytometers will render this FCM technology

an accessible and affordable option [27].

From an immunological perspective this study highlights several issues. The

importance of the type 1 cytokine axis in the control of TB has been documented in both

murine experiments [17, 28, 29] and in patients with inherited defects in the interleukin-

12 and IFN-y receptors [30-32]. Other investigators using murine TB models have

demonstrated that TN F-a is essential for the generation of protective granulomas,

without which TB control is insufficient [29, 33]. Our study gives direct support for both

IFN-y and TN F-a in mediating anti-TB responses in the lung. It is intriguing that powerful

171

Page 175: Lymphocyte Responses in the Lung in Patients with ...

cytokine responses to PPD in BAL are generated in patients with non-pulmonary TB.

These findings are suggestive of a lymphocyte recirculation pathway to the lung,

presumably reflecting the fact that even in these patients the origin of post-primary

disease was the lung.

Finally, the data presented here demonstrate the limitations of exclusively

measuring CD4 lymphocyte responses. In patients co-infected with HIV, CD4

lymphopenia may limit the applicability of this CD4 IFN-y test. In order to develop a

reliable immunodiagnostic TB test in this important group of patients, antigen-specific

CD8 responses to TB antigens will need to be explored in the lung. Furthermore, The

specificity of this method for identifying active TB will also need to be confirmed by

examining the responses to PPD in BAL from patients with treated disease.

In conclusion, a novel method for diagnosing TB by measuring intracellular cytokine

responses to PPD in BAL by a simple and rapid flow cytometric technique has been

described here. High responses were demonstrated in BAL from patients with both

pulmonary and non-pulmonary TB. In patients with non-tuberculous respiratory disease,

low responses were recorded except in some patients with sarcoidosis, perhaps

reflecting a mycobacterial origin of this disease. This test therefore appears to be a

promising diagnostic resource, particularly in those with non-pulmonary TB in whom

achieving a culture diagnosis may be both difficult and hazardous.

8.5 References1. Dye C, Scheele S, Dolin P, et al. Consensus statement. Global burden of

tuberculosis: estimated incidence, prevalence, and mortality by country. WHO

Global Surveillance and Monitoring Project. Jama 1999;282:677-86.

2. Balasubramanian V, Wiegeshaus EH, Taylor BT and Smith DW. Pathogenesis of

tuberculosis: pathway to apical localization. Tuber Lung Dis 1994;75:168-78.

3. Gatto A. Report on Cases of Tuberculosis Reported in 1999: PHLS

Communicable Diseases Surveillance Centre.

www.phls.co.uk/facts/TB/lndex/htm

4. Brisson-Noel A, Aznar C, Chureau C, et ai. Diagnosis of tuberculosis by DMA

amplification in clinical practice evaluation. Lancet 1991;338:364-6.

5. Wong CF, Yew WW, Chan CY, et ai. Rapid diagnosis of smear-negative

pulmonary tuberculosis via fibreoptic bronchoscopy: utility of polymerase chain

172

Page 176: Lymphocyte Responses in the Lung in Patients with ...

reaction in bronchial aspirates as an adjunct to transbronchial biopsies. Respir

Med 1998;92:815-9.

6. Yuen KY, Chan KS, Chan CM, et al. Use of PCR in routine diagnosis of treated

and untreated pulmonary tuberculosis. J Clin Pathol 1993;46:318-22.

7. AI Zahrani K, AI Jahdali H, Poirier L, at a/. Accuracy and utility of commercially

available amplification and serologic tests for the diagnosis of minimal pulmonary

tuberculosis. Am J Respir Grit Care Med 2000;162:1323-9.

8. Soini H, Musser JM. Molecular diagnosis of mycobacteria. Clin Chem

2001;47:809-14.

9. Maino VC, Picker LJ. Identification of functional subsets by flow cytometry:

intracellular detection of cytokine expression. Cytometry 1998;34:207-15

10. Callan MF, Tan L, Annels N, et al. Direct visualization of antigen-specific CD8+ T

cells during the primary immune response to Epstein-Barr virus In vivo. J Exp

Mecf 1998;187:1395-402

11. Lalvani A, Pathan AA, Durkan H, et al. Enhanced contact tracing and spatial

tracking of Mycobacterium tuberculosis infection by enumeration of antigen-

specific I cells. Lancet 2001;357:2017-21.

12. Kappelmayer J, Gratama JW, Karaszi E, et al. Flow cytometric detection of

intracellular myeloperoxidase, CD3 and CD79a. Interaction between monoclonal

antibody clones, fluorochromes and sample preparation protocols. J Immunol

Methods 2000;242:53-65.

13. Schwander SK, Torres M, Sada E, et al. Enhanced responses to Mycobacterium

tuberculosis antigens by human alveolar lymphocytes during active pulmonary

tuberculosis. J Infect Dis 1998;178:1434-45.

14. Faith A, Schellenberg DM, Rees AD and Mitchell DM. Antigenic specificity and

subset analysis of T cells isolated from the bronchoalveolar lavage and pleural

effusion of patients with lung disease. Clin Exp Immunol 1992;87:272-8.

15. Condos R, Rom WN, Liu YM and Schluger NW. Local immune responses

correlate with presentation and outcome in tuberculosis. Am J Respir Crit Care

Med 1998;157:729-35.

16. Chackerian AA, Perera TV and Behar SM. Gamma interferon-producing CD4+ T

lymphocytes in the lung correlate with resistance to infection with Mycobacterium

tuberculosis. Infect Immun 2001;69:2666-74.

173

Page 177: Lymphocyte Responses in the Lung in Patients with ...

17. Demangel C, Bean AG, Martin E, et al. Protection against aerosol

Mycobacterium tuberculosis infection using Mycobacterium bovis Bacillus

Calmette Guerin-infected dendritic cells. EurJ Immunol 1999;29:1972-9.

18. Havenith CE, van Haarst JM, Breedijk AJ, et al. Enrichment and characterization

of dendritic cells from human bronchoalveolar lavages. Clin Exp Immunol

1994;96:339-43.

19. Mitchell DN, Rees RJ. A transmissible agent from sarcoid tissue. Lancet

1969;2:81-4

20. Saboor SA, Johnson NM and McFadden J. Detection of mycobacterial DMA in

sarcoidosis and tuberculosis with polymerase chain reaction. Lancet

1992;339:1012-5

21. Mitchell 10, Turk JL and Mitchell DN. Detection of mycobacterial rRNA in

sarcoidosis with liquid-phase hybridisation. Lancet 1992;339:1015-7

22. Almenoff PL, Johnson A, Lesser M and Mattman LH. Growth of acid fast L forms

from the blood of patients with sarcoidosis. Thorax 1996;51:530-3

23. Bocart D, Lecossier D, De Lassence A, et al. A search for mycobacterial DMA in

granulomatous tissues from patients with sarcoidosis using the polymerase chain

reaction. Am Rev Respir Dis 1992;146:1142-8

24. Barry SM, Lipman MC, Johnson MA and Prentice HG. Respiratory infections in

immunocompromised patients. CurrOpin Pulm Med 1999;5:168-73.

25. Johnson PD, Stuart RL, Grayson ML, et al. Tuberculin-purified protein derivative-,

MPT-64-, and ES AT-6-stimulated gamma interferon responses in medical

students before and after Mycobacterium bovis BCG vaccination and in patients

with tuberculosis. Clin Diagn Lab Immunol 1999;6:934-7.

26. Sodhi A, Gong J, Silva C, et al. Clinical correlates of interferon gamma

production in patients with tuberculosis. Clin Infect Dis 1997;25:617-20.

27. Barry S, Condez, A, Johnson, MA and Janossy, G. Determination of

Bronchoalveolar Lavage Leukocyte Populations by Flow Cytometry in Patients

Investigated for Respiratory Disease. Clinical Cytometry 2002,50 in press.

28. Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in

resistance to Mycobacterium tuberculosis infection. J Exp Med 1993;178:2249-

54.

174

Page 178: Lymphocyte Responses in the Lung in Patients with ...

29. Flynn JL, Goldstein MM, Chan J, et al. Tumor necrosis factor-alpha is required in

the protective immune response against Mycobacterium tuberculosis in mice,

/mmun/fy 1995;2:561-72.

30. Altare F, Lammas D, Revy P, etal. Inherited interleukin 12 deficiency in a child

with bacille Calmette-Guerin and Salmonella enteritidis disseminated infection. J

Clin Invest 1998;102:2035-40.

31. Jouanguy E, Lamhamedi-Cherradi 8, Altare F, etal. Partial interferon-gamma

receptor 1 deficiency in a child with tuberculoid bacillus Calmette-Guerin infection

and a sibling with clinical tuberculosis. J Clin Invest 1997;100:2658-64.

32. Newport MJ, Huxley CM, Huston 8, et al. A mutation in the interferon-gamma-

receptor gene and susceptibility to mycobacterial infection. N Engl J Med

1996;335:1941-9.

33. Bean AG, Roach DR, Briscoe H, et al. Structural deficiencies in granuloma

formation in IN F gene-targeted mice underlie the heightened susceptibility to

aerosol Mycobacterium tuberculosis infection, which is not compensated for by

lymphotoxin. J Immunol 1999;162:3504-11.

175

Page 179: Lymphocyte Responses in the Lung in Patients with ...

Chapter 9

Discussion

176

Page 180: Lymphocyte Responses in the Lung in Patients with ...

9.1 DiscussionAmongst many researchers there is an idea that FCM is an ‘aristocratic’ technique. Its

use, primarily by immunologists has been to develop ever more intricate ways of

investigating the extraordinary complexity of immune responses, culminating in the

impressive, but bewildering 11-colour, 13-parameter FCM analysis of lymphocytes [1] .

Although FCM has the capability to determine not only cluster differentiation (CD)

antigens on the surface of cells, it can also analyse intracellular antigens, such as

cytokines, or nuclear markers of cellular proliferation to provide a highly complex

analysis of cellular functions. Therefore, this technology has been widely perceived as

being appropriate only for complex research applications, primarily in the field of

immunology. This view is unjustified since it is the very precision of this technology that

lends itself equally well to the simple differentiation of leukocytes as to more complex

leukocyte phenotyping studies.

The precision of FCM stems from a number of factors. First, the development of

highly specific monoclonal antibodies has enabled subsets of cells or other analytes to

be labelled. Second the discovery of different fluorochromes with which the antibodies

can be conjugated and which offer minimal spectral overlap following laser excitation has

made differentiation feasible by FCM. Third, the fact that large numbers of events are

analysed during flow cytometric acquisition of samples has ensured that statistical

variability is minimised.

These benefits offered by flow cytometry have long been recognised as an important

solution to the inherent limitations provided by manual counting of leukocyte differentials

using a microscope [2] . At the present time, automated haematology analysers

generally perform leukocyte differential counting using several techniques. However,

there is a growing acceptance that FCM is an alternative technique that is precise and

offers the advantage of being able to provide absolute cell counts. Recently, the

acceptance of CD45 panleukogating in simplifying the protocols for leukocyte and

lymphocyte differential analysis has been acknowledged [3, 4] . These developments

have heralded a new era for FCM and widened its application from complex

immunological research tool to that of a ‘workhorse’ capable of the routine evaluation of

white blood cell differentials. New flow cytometers that work on volumetric sample

acquisition and therefore provide absolute cell counts without the need for expensive

bead systems are eminently suited to this task [5] . It is particularly exciting that a

volumetric cytometer may be able to perform absolute cell counts, simple leukocyte

177

Page 181: Lymphocyte Responses in the Lung in Patients with ...

differentials and also to undertake multiplex analysis of samples to detect the presence

of either antigens, or antibodies to a wide variety of different pathogens, th is

multiplexing facility offers enormous potential for the differential diagnosis of infectious

disease [6] and is the sort of technology that could revolutionise laboratory diagnosis in

both resource rich and poor settings.

Unfortunately, there has been a reluctance to adopt these exciting new technologies

for the analysis of tissue fluid samples such as BAL. Although several investigators have

demonstrated that FCM is better suited to performing CD4/CD8 differentials than the

time consuming and cumbersome immunofluoresence or immunoperoxidase techniques

[7, 8], it has yet to be adopted as a routine diagnostic tool.

This study has therefore attempted to redress this imbalance by demonstrating that

FCM is the appropriate technology for BAL analysis. The first problem was to

demonstrate that BAL leukocyte differentials and lymphocyte subset ratios could be

reliably performed by FCM. Although previous investigators had already recognised the

value of CD45 directed gating [8, 9] , their interest was primarily in determining the

lymphocyte component in BAL. Such a focus on the lymphocytes neglected the clinically

relevant granulocyte populations in BAL and therefore did not provide adequate

evidence of the benefits of FCM over conventional cytospin methods. Here, the value of

CD45 panleukogating for distinguishing leukocytes from epithelial cells and debris in

BAL has been confirmed. In addition, it has been demonstrated that BAL lymphocyte

gating using a combination of CD45 expression and low side scatter is an adequate

method for determining the lymphocyte percentages in BAL. When this method was

compared with a lymphosum method that gated the individual lymphocyte components

(T cells, B cells and NK cells), the results were virtually indistinguishable.

The more difficult problem was to distinguish macrophages from granulocytes.

Rather than attempt to directly distinguish macrophages by FCM, a notoriously difficult

project due to their autofluorescence, heterogenous intrinsic characterisitcs and the lack

of an adequate surface marker, a different approach was adopted here. Characterisation

of granulocytes using CD15 allowed the macrophage pool to be derived as those CD45+

events remaining after subtraction of the lymohocytes and granulocytes. When

compared with cytospin preparations in which 500 BAL leukocytes were counted by a

highly experienced cytologist, this flow cytometric method showed good agreement for

determining the BAL leukocyte differentials. As expected, the coefficient of variation for

differential analysis by FCM was considerably lower than by cytospin. The conclusion of

178

Page 182: Lymphocyte Responses in the Lung in Patients with ...

this initial study was that the use of only two monoclonal antibodies, CD45 and CD15

was adequate to demonstrate the major clinically relevant leukocyte populations in BAL

by FCM. Additional discrimination of granulocytes into eosinophils and neutrophils was

achieved by the addition of an antibody against the IgE receptor, CD23.

The value of BAL lymphocyte subset analysis, in particular the CD4/CD8 ratio has

also been demonstrated to be of diagnostic value in diseases such as sarcoidosis by

many investigators and therefore it was felt to be important to include such discrimination

in a single four-colour panel together with CD45 and CD15. A simplified method by

which lymphocytes were gated on the basis of CD45 and side scatter and then directly

differentiated into CD4 and CD8 components offered no loss of precision when

compared to a more sophisticated panel including the I cell marker, CD3. The final

conclusion of this initial investigation is that a single four-colour panel combining

CD45/CD15/CD4 and CD8 can rapidly provide most of the clinically relevant information

required for the differential diagnosis of various respiratory diseases. Moreover, it has

also been demonstrated that such a system is equally applicable for the analysis of other

tissue fluid samples such as pleural fluid, cerebrospinal fluid and ascitic fluid.

One of the most significant findings of early studies investigating the lung as a site

of pathological and diagnostic interest was the discovery that the responses

demonstrated at the site of disease activity in the lung were not reflected by the

responses demonstrated in the blood. This has held true not only for the simple

lymphocyte differentials in patients with sarcoidosis, but also for the demonstration of

cytokine responses in the lung in patients with TB [10] and sarcoidosis [11] . These

findings have emphasised the importance of focusing on the immune responses at the

site of infection, rather than in the blood. Such an approach has also been informative in

patients presenting with respiratory illness in unusual circumstances. Here, an HIV-

infected man was identified who developed a pneumonic illness following the institution

of highly active antiretroviral therapy (HAART) after an episode of treated PCP. Analysis

of the CD8 lymphocyte phenotype demonstrated the predominance of proliferating

(Ki67+), perforin producing cells in BAL, that declined considerably following steroid

therapy and were not seen in the blood [12] . In this case it was argued that the highly

atypical proliferating CD8 lymphocytes may have represented an immune reconstitution

disorder to P. carinii antigen after vigorous immune restoration on HAART. Nevertheless,

although interesting and informative, studies such as these are not proof of the concept

179

Page 183: Lymphocyte Responses in the Lung in Patients with ...

since the antigen-specific responses were not measured. The most informative

investigations of tissue fluid analysis will therefore be those that focus on determining the

pathogen-specific lymphocyte responses.

In the light of its clinical importance, together with an emphasis on pulmonary

presentation, TB has emerged as the most obvious candidate for the investigation of

tissue immune responses. Several investigators have sought to determine the TB-

specific responses in the lung in both human infection and animal models, but these

early studies have generally been hampered by complex techniques that are not

appropriate for routine clinical investigation.

Several of the early studies used lymphocytes that were separated from BAL and

then incubated with peripheral blood mononuclear cells (PBMC’s) from the same

patients Before measuring proliferative responses to TB antigens [10,13]. In these two

studies the investigators presumably felt that BAL either did not contain adequate

numbers of dendritic cells (DC) or that alveolar macrophages may suppress this process

and so consequently PBMC’s were used as a source of antigen presenting cells.

Nevertheless, DC s have been identified in BAL and more importantly, these cells have

been demonstrated to be increased following TB infection in both humans [14] and

animals [15, 16]. Therefore, unmanipulated BAL from patients with TB should contain

sufficient dendritic cells for antigen presentation. This assumption has opened the way

for simple, rapid analysis of antigen-specific responses either using ELISPOT or FCM.

IFN-y synthesis in BAL has been reported both in patients with TB [10], and also in

their household contacts [17] in response to TB antigens when measured by ELISPOT.

However, it has been argued here that ELISPOT is not the optimum method for

determining antigen-specific responses in BAL since the proportion of lymphocytes and

the CD4 and CD8 subset ratios are highly variable in the lung during infectious episodes.

These facts are relevant because the TB antigens that are used in these assays, such

as PPD, Ag85, and ESAT-6 are generally large and predominantly presented to CD4

lymphocytes. By contrast, CD8 responses may be preferentially stimulated by smaller

peptides of 9 amino acids.

Therefore, flow cytometry, which can rapidly determine the cytokine synthetic

responses specifically in either the CD4 or the CD8 lymphocyte subset is likely to be far

more sensitive than the ELISPOT system. For example, in one paper in which ELISPOT

was used to determine the IFN-y secretion following incubation with PPD [10], the

median number of spot forming colonies/10® BAL cells was 400 in a small subset of

180

Page 184: Lymphocyte Responses in the Lung in Patients with ...

patients with pulmonary TB, giving a response rate of 0.4%. In the study presented in

this thesis, the median IFN-y response rate in the BAL CD4 lymphocytes was 25.2% by

flow cytometry. This huge discrepancy may explain why these authors did not find IFN-y

secretion in BAL taken from the unaffected lung in these patients in contrast to the data

presented here.

Another important advantage of FCM over ELISPOT is that multiple cytokine

responses can be determined in the same cognate T lymphocytes. The demonstration of

greater TNF-a synthesis than IFN-y in BAL CD4 cells from patients with TB is especially

interesting. The analysis of multiple cytokine synthetic capabilities by antigen-specific

cells may be particularly important in certain situations such as HIV infection, where

there may be a relative defect in one of the responses. It would be interesting to note

whether the increased presentation of non-pulmonary TB in HIV infected individuals is

not only explained by CD4 lymphopoenia, but also by a loss of TNF-a synthesis, which

has been demonstrated to be vital for protective granuloma formation in animal models.

Although PPD was used as the stimulatory antigen in this thesis, it would be

interesting to continue this work by examining the responses to other antigens that are

specific for TB and that do not share epitopes with BCG, such as ESAT-6. Since it has

been demonstrated here that prior BCG vaccination does not result in type-1 cytokine

synthesis following incubation with PPD in the lung, in contrast to the findings in

peripheral blood, it is unlikely that ESAT-6, or similar peptides will prove more sensitive

for TB diagnosis in BAL. The problem with using specific peptides will be that the

responses demonstrated will be almost certainly much smaller than with PPD. However,

it will be extremely interesting to investigate which antigens stimulate the optimum

cytokine responses in BAL from patients with sarcoidosis. The tantalising discovery of

responses to PPD in these patients has opened the old debate as to whether sarcoidosis

is indeed a response to mycobacterial antigens, although these may be environmental

mycobacteria rather than M. tuberculosis.

This simple, rapid flow cytometric method is also applicable for the study of other

clinically relevant antigen-specific responses in BAL. A significant CD4 IFN-y synthetic

response to cytomegalovirus (CMV) viral lysate was demonstrated in a bone marrow

transplant patient who developed a febrile respiratory illness. Analysis of BAL confirmed

the presence of CMV nucleic acid by PCR that was not present in the blood where the

CD4 cytokine response was correspondingly low (submitted for publication).

181

Page 185: Lymphocyte Responses in the Lung in Patients with ...

In summary, the work presented in this thesis has demonstrated the value of a

focused investigation of BAL and highlighted the role of flow cytometry in this pocess.

FCM has been shown to be applicable both for the routine analysis of simple variables

such as the leukocyte differentials and CD4/CD8 ratios in BAL as well as demonstrating

the presence of antigen-specific responses to clinically relevant pathogens such as M

tuberculosis. These findings have therefore brought flow cytometry firmly into the realms

of diagnostic investigation in the fields of respiratory medicine and infectious disease.

9.2 References

1. De Rosa SC, Herzenberg LA, Roederer M. 11-color, 13-parameter flow,

cytometry: identification of human naive I cells by phenotype, function, and T-cell

receptor diversity. Nat Med 2001 ;7:245-8.

2. Koepke JA, Dotson MA, Shifman MA. A critical evaluation of the manual/visual

differential leukocyte counting method. Blood Cells 1985;11:173-86

3. Glencross DK SL, Jani IV, Barnett D and Janossy G. CD45-Assisted

Panleukogating for Accurate, Cost-Effective Dual-Platform CD4+ T-Cell

Enumeration. Cytometry (Clinical Cytometry) 2002;50:69-78

4. Janossy G, Jani IV, Bradley NJ, et al. Affordable CD4(+)-T-cell counting by flow

cytometry: CD45 gating for volumetric analysis. Clin Diagn Lab Immunol

2002;9:1085-94.

5. Janossy G, Jani IV, Kahan M, et al. Precise CD4 T-cell counting using red diode

laser excitation: For richer, for poorer. Cytometry 2002;50:78-85

6. Jani IV, Janossy G, Brown DW, etal. Multiplexed immunoassays by flow

cytometry for diagnosis and surveillance of infectious diseases in resource-poor

settings. Lancet Infect Dis 2002;2:243-50

7. Padovan CS, Behr J, Allmeling AM, et al. Immunophenotyping of lymphocyte

subsets in bronchoalveolar lavage fluid. Comparison of flow cytometric and

immunocytochemical techniques. J Immunol Methods 1992;147:27-32.

8. Brandt B, Thomas M, von Eiff M, et al. Immunophenotyping of lymphocytes

obtained by bronchoalveolar lavage: description of an all-purpose tricolor flow

cytometric application. J Immunol Methods 1996;194:95-102.

182

Page 186: Lymphocyte Responses in the Lung in Patients with ...

9. Dauber JH, Wagner M, Brunsvold S, et aJ. Flow cytometric analysis of

lymphocyte phenotypes in bronchoalveolar lavage fluid: comparison of a two-

color technique with a standard immunoperoxidase assay. Am J Respir Cell Mol

8/0/1992:7:531-41.

10. Schwander SK, Torres M, Sada E, et al. Enhanced responses to Mycobacterium

tuberculosis antigens by human alveolar lymphocytes during active pulmonary

tuberculosis. J Infect Dis 1998;178:1434-45.

11. Wahlstrom J, Katchar K, Wigzell H, et al. Analysis of intracellular cytokines in

CD4+ and CD8+ lung and blood T cells in sarcoidosis. Am J Respir Crit Care

/Wed 2001;163:115-21.

12. Barry SM, Lipman MC, Deery AR, et al. Immune reconstitution pneumonitis

following Pneumocystis carinii pneumonia in HIV-infected subjects. HIV Med

2002;3:207-11

13. Faith A, Schellenberg DM, Rees AD, et al. Antigenic specificity and subset

analysis of I cells isolated from the bronchoalveolar lavage and pleural effusion

of patients with lung disease. Clin Exp Immunol 1992;87:272-8.

14. Henderson RA, Watkins SC, Flynn JL. Activation of human dendritic cells

following infection with Mycobacterium tuberculosis. J Immunol 1997;159:635-43.

15. Demangel C, Bean AG, Martin E, etal. Protection against aerosol

Mycobacterium tuberculosis infection using Mycobacterium bovis Bacillus

Calmette Guerin-infected dendritic cells. Bur J Immunol 1999;29:1972-9.

16. Havenith CE, Breedijk AJ, Hoefsmit EC. Effect of Bacillus Calmette-Guerin

inoculation on numbers of dendritic cells in bronchoalveolar lavages of rats.

Immunobiology 1992;184:336-47.

17. Schwander SK, Torres M, Carranza CC, et al. Pulmonary mononuclear cell

responses to antigens of Mycobacterium tuberculosis in healthy household

contacts of patients with active tuberculosis and healthy controls from the

community. J Immunol 2000;165:1479-85

183

Page 187: Lymphocyte Responses in the Lung in Patients with ...

Glossary of Abréviations

AFB acid fast bacillus

APC antigen presenting cell

BAL bronchoalveolar lavage

BMT bone marrow transplantation

CD cluster differentiation

CMV cytomegalovirus

DEAFF direct early antigen fluorescent foci

DNA deoxyribonucleic acid

EDTA ethylenediamine tetracetate

ELISA enzyme-linked immunoabsorbant assay

ESAT-6 early secretory antigen of tuberculosis-6

FCM flow cytometry

HAART highly active antiretroviral therapy

HIV human immunodeficiency virus

IFN-y Interferon-y

MFI mean fluorescence intensity

MAI mycobacterium avium intracellulari

MOTT mycobacterium other than TB

NK natural killer cell

PBMC peripheral blood mononuclear cell

PBS phosphate buffered saline

PCP Pneumocystis carinii pneumonia

PCR polymerase chain reaction

PPD purified protein derivative

RSV respiratory syncitial virus

SACE serum angiotensin converting enzyme

SEB staphylococcal enterotoxin B

SIV simian immunodeficiency virus

TB tuberculosis

TNF-a tumour necrosis factor-a

ZN Ziehl neelson

184

Page 188: Lymphocyte Responses in the Lung in Patients with ...

Appendix 1

Presentation of results of flow cytometric analysis of BAL leukocyte differentials

and CD4/CD8 ratios given to clinicians

Royal Free and University College Medical SchoolDept Immunology and Molecular Pathology, Royal free Campus, Rowland H ill St London NW3 2PF

H IV Immunology UnitProf G Janossy,MD, PhD, PRC Path, DSc RFH ex 3745BAL Leukocyte Differential Analysis by FCM

ssc

00

QU

CD 15

Lymphocytes (R2)= 83.8% Granulocytes (R3)= 0.5% Macrophages = 15.7%

CD4/CD8 ratio: 15.8

CD 4

ConclusionMassive lymphocytosis w ith greatly increased CD4/CD8 Ratio strongly supportive o f sarcoidosis.

Dr S Barry

185

Page 189: Lymphocyte Responses in the Lung in Patients with ...

Appendix 2

Example of results of cytokine synthesis IN BAL following PPD stimulation in

patients with suspected TB presented to clinicians.

R o y a l Free and Uni vers i ty Col lege M e d i c a l S c hoo lDe pt Im m u n o l o g y and M o l e c u l a r Pathology, Royal free C ampus, R ow la nd Hi l l StLondon N W 3 2P F

H I V I m m u n o l o g y Un i tP ro f G Ja nos s y , M D , P hD , F RC P at h , DSc

RFH ex 3745

Diag n ost ic cy to k ine p ro d u c t i o n assay in pat ients with suspected tuberculos i s .

Blood PPD0.62%

BAL PPD

43.5%

wmm10° ID' 1 0 ' 1 0° 10*

Blood No Ag

0.08%

BAL No Ag

1.95%

IF N -yConclus ion: There is a very large B A L C D 4 response to P PD This is support ive o f active T B .

D r S B a rry B leep 425

186