the effects of chronic ethanol ingestion and smoke

162

Upload: others

Post on 27-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

THE EFFECTS OF CHRONIC ETHANOL INGESTION AND SMOKE

EXPOSURE ON ANTI-PNEUMOCOCCAL HOST DEFENSES

______________________________

BY

ADAM MICHAEL PITZ

______________________________

A DISSERTATION

Submitted to the Faculty of the Graduate School of Creighton University in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

in the Department of Medical Microbiology and Immunology

______________________________

Omaha, December 2008

iii

The Effects of Chronic Ethanol (EtOH) Ingestion and Smoke Exposure on Anti-Pneumococcal Host Defenses

Adam Michael Pitz

Under the supervision of Martha J. Gentry-Nielsen

from the Department of Medical Microbiology and Immunology Creighton University

Individuals who abuse alcohol and tobacco are at increased risk for pneumonia

caused by Streptococcus pneumoniae (the pneumococcus). Alcohol and cigarette smoke

are known to exert immunosuppressive effects on pulmonary defense mechanisms that

protect the host from pneumococcal pneumonia. However, the interactive, combined

immunosuppressive effects of ethanol (EtOH) ingestion and smoke exposure remain ill

defined. To study these effects, we used a rat model that mimics chronic alcohol abuse

and smoking. Rats were exposed to smoke generated by 30 reference cigarettes for one

hour twice daily for 12 weeks or to room air (sham-exposed). During the last five weeks

of exposure, the rats were pair-fed equal volumes of liquid diet containing 0% or 36%

EtOH calories.

Nasopharyngeal colonization was unaltered by EtOH ingestion and/or smoke

exposure. The movement of pneumococci from the rats’ nasopharynx into the lungs was

slightly increased by EtOH ingestion, but EtOH did not impair ciliary beating. Smoke

exposure reduced pneumococcal movement into the lungs regardless of diet, and this was

correlated with a significant increase in ciliary beat frequency.

The effect of concurrent smoke exposure and EtOH ingestion on non-neutrophil

(PMN)-mediated killing of pneumococci was investigated using an in vivo bactericidal

assay. EtOH ingestion significantly decreased the percentage of pneumococci killed

iv

within the rats’ lungs. Unexpectedly, this EtOH-induced defect was counteracted by

concurrent smoke exposure, even though smoke exposure alone did not increase

intrapulmonary pneumococcal clearance. Quantification of the bactericidal proteins

lysozyme and lactoferrin in the rats’ lungs did not explain the differences in non-PMN-

mediated killing. However, EtOH-fed rats that were sham-exposed exhibited suppressed

alveolar macrophage phagocytosis of pneumococci as well as diminished opsonization of

the bacteria within their lungs. It is uncertain if smoke exposure increased macrophage

phagocytosis due to technical problems with the assay, but concurrent smoke exposure

did not exacerbate the EtOH-related decrease in opsonic protein activity.

PMN functions also were analyzed in our rat model. Using a similar in vivo

bactericidal assay, EtOH ingestion alone was shown to abolish PMN-mediated killing of

pneumococci within the lungs. Smoke exposure alone had no effect on this activity, but

the addition of smoke exposure to EtOH ingestion restored PMN killing. The differences

in bacterial killing were not due to alterations in PMN recruitment to the lungs or PMN

phagocytosis of pneumococci. EtOH ingestion alone reduced systemic levels of

cytokine-induced neutrophil chemoattractant-1 (CINC-1), which may decrease PMN

activation and help explain the absence of pulmonary killing in the EtOH-fed animals.

These results indicate that both chronic EtOH ingestion and smoke exposure

modulate anti-pneumococcal defenses, but their effects differ significantly in hosts who

either abuse alcohol or smoke and those who practice both behaviors.

v

ACKNOWLEDGEMENTS

To my advisor, Dr. Martha Gentry-Nielsen, thank you for your commitment in

helping me with every aspect of my graduate career. I greatly appreciate how you took

me under your wing, pushed me to do my best and coached me to be a better scientist. I

am also grateful for your constructive feedback to improve my writing. All of my

success and achievements these last four years would not have been possible without you.

Huge thanks to Mary Snitily for training me on how to work with the rats and

perform the various animal procedures. It was a great joy and pleasure working along

side you during those long days for every single animal experiment. Thank you also for

your constant reminders to live a holistic life.

Thank you to the members of my committee: Dr. Laurel Preheim, Dr. Philip

Lister, Dr. Floyd Knoop, and Dr. Geoffrey Thiele. Thank you for attending my meetings,

reviewing my data, asking great questions, providing helpful suggestions, reading my

grants and dissertation, and your approval to receive my Ph.D.

Thanks to Dr. Greg Perry for your expertise in all of the flow cytometry analysis.

I appreciate your dedication to my project, specifically with those dreaded alveolar

macrophages.

I also would like to thank all of the undergraduate students Brock Hanisch, Ben

Beller, Meghan Lewandowski, Drew Minturn, Amanda Ross, Alicia Milliken, and Alan

Pitz for your time in smoke exposing and feeding the rats. Thank you also for getting

supplies ready for experiments, processing and transferring samples, and conducting

various assays. Without your help it would have been impossible to complete this

project.

vi

DEDICATION

I would like to dedicate this work to my family, who has been a constant source

of love and support. To my lovely wife Becky, thank you for your love and sacrifice

these last four years. I appreciate your bravery in helping me care for the rats. Thank

you also for all of the hours you spent listening to my seminars and reading my grants

and dissertation.

To my newborn son Noah, thanks for being such a cute baby and for going easy

on me while I finish graduate school. You provide me endless joy and love in my heart.

Thank you, Mom and Dad, for believing in me and for giving me the opportunity

to pursue my dreams. Thanks for always encouraging me to do my best. You both

sacrificed so much for me so I could receive a quality education. I believe all that you

have given me has finally paid off. Thank you also to my siblings Sara, Alan, and Lisa

for making my life fun and enjoyable.

vii

LIST OF ABBREVIATIONS APC – Allophycocyanin ATCC – American Type Culture Collection BAL – Bronchoalveolar lavage BALF – Bronchoalveolar lavage fluid CBF – Ciliary beat frequency CFDA/SE – Carboxyfluorescein diacetate succinimidyl ester CFU – Colony forming units CINC-1 – Cytokine-induced neutrophil chemoattractant COPD – Chronic obstructive pulmonary disease CRP – C-reactive protein ELISA – Enzyme-linked immunoabsorbent assay EtOH – Ethanol Gro-α – Growth-regulated protein alpha HBSS – Hanks Balanced Salt Solution HRP – Horseradish peroxidase IFN-γ – Interferon gamma IL-8 – Interleukin 8 LDH – Lactate dehydrogenase LPS – Lipopolysaccharide MIP-2 – Macrophage inflammatory protein PBS – Phosphate-buffered saline PBS-BSA – Phosphate-buffered saline and 0.5% bovine serum albumin PBS-FCS – Phosphate-buffered saline and 4% heat inactivated fetal calf serum PE – Phycoerythrin PKA – Protein kinase A PKC – Protein kinase C PMA – Phorbol myristate acetate PMN – Polymorphonuclear leukocyte PppA – Pneumococcal protective protein A PspA – Pneumococcal surface protein A PspC – Pneumococcal surface protein C SP-D – Surfactant protein D

viii

TABLE OF CONTENTS

I. INTRODUCTION…………………………………………………………………. 1

II. LITERATURE REVIEW…………………………………………………………. 3

A. Streptococcus pneumoniae and Pneumococcal Pneumonia………………. 3 B. Virulence Factors of Streptococcus pneumoniae………………………….. 17 C. Alcohol Abuse and Anti-Pneumococcal Host Defenses…………………... 21 D. Smoking and Anti-Pneumococcal Host Defenses……………………….... 28

III. SUMMARY AND RESEARCH OBJECTIVES…………………………………. 35

IV. MATERIALS AND METHODS………………………………………………… 38

A. Model of Chronic EtOH Ingestion and Smoke Exposure…………………. 38 B. Bacterial Strains…………………………………………………………… 39 C. Rat Sacrifice……………………………………………………………….. 40 D. Intranasal Infection………………………………………………………... 40 E. Culture of Nasopharynx, Trachea, and Lungs……………………………... 41 F. Intranasal Vaccination with rPppA……………………………………….... 42 G. ELISA for rPppA Antibodies…………………………………………….... 43 H. Salbutamol and Formoterol Medication…………………………………... 44 I. Ciliary Beat Frequency Analysis………………………………………….... 45 J. Transtracheal Infections……………………………………………………. 46 K. Lipopolysaccharide Instillation for PMN-mediated Assays………………. 47 L. Non-PMN-Mediated Bactericidal Assay…………………………………... 48 M. Bronchoalveolar Lavage Procedures……………………………………… 48 N. Quantification of Bactericidal Factors…………………………………….. 50 O. Quantification of Pulmonary Cell Damage………………………………... 50 P. Opsonic Deposition Assay…………………………………………………. 51 Q. C3, CRP, and SP-D ELISAs………………………………………………. 53 R. PMN-Mediated Bactericidal Assay………………………………………... 53 S. Chemokine Analysis……………………………………………………….. 54 T. PMN Phagocytosis Assay…………………………………………………. 55

U. Macrophage Phagocytosis Assay………………………………………….. 59 V. Macrophage Function Assays……………………………………………… 63 W. Intranasal Mortality Study………………………………………………… 65 X. Statistical Analysis………………………………………………………… 65

ix

V. RESULTS………………………………………………………………………… 67

A. Pneumococcal Colonization and Movement

from the Nasopharynx to the Lungs……………………………………… 67 B. PppA Vaccination Trial…………………………………………………... 68 C. Salbutamol Medication Trial…………………………………………….. 70 D. Formoterol Medication…………………………………………………... 71 E. Non-PMN-Mediated Killing……………………………………………… 76 F. Quantification of Bactericidal Factors……………………………………. 77

G. Macrophage Phagocytosis………………………………………………... 80 H. Additional Macrophage Functions……………………………………….. 84 I. Opsonic Deposition Assay………………………………………………… 87 J. PMN-Mediated Killing……………………………………………………. 90 K. Pulmonary and Systemic Chemokine Levels……………………………... 91 L. PMN Phagocytosis………………………………………………………… 92 M. Mortality Study…………………………………………………………… 93

VI. DISCUSSION……………………………………………………………………. 95

A. Pneumococcal Colonization and Movement Studies……………………… 95

B. Non-PMN Pulmonary Defenses…………………………………………… 100 C. PMN Functions……………………………………………………………. 111 D. Chemokine Production……………………………………………………. 115 E. Mortality Study……………………………………………………………. 120

VII. CONCLUSIONS………………………………………………………………… 123

VIII. BIBLIOGRAPHY………………………………………………………………. 126

x

LIST OF TABLES 1. Smoke Exposure Reduces Pneumococcal Movement…………………………….. 68

2. rPppA Specific IgG and IgA Antibody Levels Compared to Log cfu of Nasal Washes…………………………………………… 69

3. Comparison of Macrophage Phagocytosis Values………………………………... 83

4. Chemokine Production by Macrophages Stimulated with Pneumococci or PMA……………………………………………. 87

5. Chemokine Values from Lung Homogenates and Serum…………………………. 92

6. Bacteremia and Mortality Results from an Intranasal Challenge…………………. 94

7. Summary of Conclusions………………………………………………………….. 125

xi

LIST OF FIGURES

1. Progression of Pneumococcal Disease…………………………………………… 4

2. Complement Pathways and Effector Functions………………………………….. 8

3. Rat Model of Chronic EtOH Ingestion and Smoke Exposure…………………… 39

4. Intranasal Infection………………………………………………………………. 41

5. Medication Device……………………………………………………………….. 45

6. Transtracheal Infection…………………………………………………………… 47

7. Lavage……………………………………………………………………………. 49

8. Lavage Fluid Collection………………………………………………………….. 49

9. Scatter Plot of S. pneumoniae……………………………………………………. 52

10. Analysis of APC Fluorescence…………………………………………………. 52

11. CFSE-Labeled Pneumococci…………………………………………………… 56

12. Determination of PMN Phagocytosis…………………………………………… 58

13. APC-Cy7-labeled S. pneumoniae………………………………………………. 60

14. Determination of Macrophage Phagocytosis…………………………………… 62

15. Syto 9-labeled S. aureus………………………………………………………… 63

16. PppA Vaccination Fails to Reduce Colonization……………………………….. 69

17. Salbutamol Medication Decreases Pneumococcal Movement in Chow-fed Rats…………………………………………………… 70

18. Formoterol Medication Reduces Pneumococcal Movement into the Lungs of Chow-fed Rats…………………………………… 72

19. Formoterol Medication Prevents Pneumococcal Movement in Sham-EtOH Rats………………………………………………… 73

20. Smoke Exposure Increases CBF Regardless of Diet and Formoterol Medication………………………………… 75

xii

21. Formoterol Augments CBF in Sham-EtOH Rats……………………………….. 76

22. Concurrent Smoke Exposure Negates Chronic EtOH-Induced Impairment of Non-PMN-Mediated Killing……………………………………. 77

23. No Alterations of Pulmonary Lysozyme Levels by Smoke Exposure and EtOH Ingestion………………………………………. 78

24. EtOH Ingestion Drastically Increases Pulmonary Lactoferrin Levels…………. 79

25. Cellular Release of LDH Does Not Explain the EtOH-Induced Increase in Lactoferrin……………………………………… 80

26. Smoke Exposure Enhances Macrophage Autofluorescence…………………… 81

27. EtOH Ingestion Suppresses Macrophage Phagocytosis………………………... 82

28. Flow Cytometry Results Indicate EtOH Ingestion Increases Macrophage Phagocytosis of S. aureus………………………………………… 83

29. Smoke Exposure Alone Suppresses Oxidative Burst in Macrophages Stimulated by Pneumococci…………………………………. 85

30. Smoke Exposure Hinders Oxidative Burst in PMA-Stimulated Macrophages……………………………………………… 85

31. Neither EtOH nor Smoke Significantly Alters Degranulation by Macrophages Stimulated with Pneumococci………………………………... 86

32. EtOH Ingestion Reduces C3 and SP-D Deposition on Bacteria……………….. 88

33. EtOH Ingestion Decreases C3 Basal Levels…………………………………… 89

34. Concurrent Smoke Exposure Negates EtOH-Induced Defect in PMN-Mediated Killing……………………………… 91

35. Neither EtOH Ingestion nor Smoke Exposure Affects PMN Phagocytic Activity……………………………………………………… 93

1

I. INTRODUCTION

Streptococcus pneumoniae, the pneumococcus, is the major cause of community-

acquired pneumonia and bacteremia in the United States. Despite antibiotics and

vaccines, this disease still remains prevalent in several high-risk groups such as children,

the elderly, alcoholics, smokers, AIDs patients, and patients with chronic liver or lung

disease. Furthermore, the significance of this problem has risen due to the emergence of

pneumococcal resistance to multiple antibiotics. More research is needed to understand

the pathogenesis of S. pneumoniae, particularly in immunocompromised hosts, in order to

develop alternative treatments against pneumococcal infection.

Pneumococcal pneumonia begins with colonization of the nasopharynx. This

event can occur without any signs or symptoms, and colonized patients can transmit the

organism unknowingly to other hosts. Once colonization is established, pneumococci

can travel down the trachea and invade the lungs, resulting in pneumonia. The host is

equipped with several defenses to help prevent these events from happening. Mucosal

IgA antibodies aid in controlling nasopharyngeal colonization by inhibiting bacteria from

attaching to the epithelium. To prevent pulmonary invasion, the trachea is lined with the

mucociliary clearance apparatus. This consists of ciliated cells that beat in an upward

fashion to remove microorganisms trapped in mucus from the lower respiratory tract.

If S. pneumoniae breaches these upper airway defenses, it can proceed into the

lower respiratory tract. The lungs contain several extracellular and cellular defenses to

maintain a sterile environment. Among these defenses are bactericidal factors, resident

alveolar macrophages, and neutrophils (PMNs). Alveolar lining fluid that coats the lungs

2

consists of several bactericidal components such as lysozyme and lactoferrin that can

quickly kill pneumococci. Alveolar macrophages are important for detecting invading

pathogens and initiating the inflammatory response. PMNs are the primary cellular

defense against S. pneumoniae. They are essential for phagocytosing and clearing

pneumococci from the lungs in order to prevent widespread dissemination of the

organisms. Few PMNs are present in healthy lungs, but they are recruited and activated

by chemokines and cytokines produced by alveolar macrophages when an infection

occurs.

Alcohol abuse is one of the most important risk factors for pneumococcal

pneumonia. Compared to non-drinkers, alcohol abusers tend to have higher morbidity

and mortality rates related to pneumonia. Alcohol impairs mucociliary clearance, making

the lungs susceptible to pneumococcal infection. Important PMN functions are also

altered by alcohol consumption, which increases the risk of developing bacteremia.

Cigarette smoke also is associated with an increased incidence and severity of

pneumococcal pneumonia. Both the number of cigarettes smoked and years of smoking

positively correlate with the risk of developing pneumococcal disease. Cigarette smoke

reduces IgA antibodies, thus promoting bacterial colonization. Smoking also damages

the mucociliary clearance apparatus and impairs PMN activity.

This thesis describes research that was performed using a rat model of separate

and combined smoke exposure and EtOH ingestion. The focus of this project was to

study chronic smoke- and EtOH-induced alterations in innate anti-pneumococcal

defenses and evaluate alternative therapies to bolster these defenses and reduce the

development of pneumococcal pneumonia.

3

II. LITERATURE REVIEW

A. Streptococcus pneumoniae and Pneumococcal Pneumonia

Incidence of Disease

Pneumonia is among the top ten leading causes of death in the United States.

Every year 5 million cases of community-acquired pneumonia result in approximately 1.7

million hospitalizations and over 60,000 deaths [1-3]. The annual healthcare costs of

pneumonia cases are estimated to be $9 billion [4]. The most commonly identified cause

of community-acquired pneumonia is Streptococcus pneumoniae, or the pneumococcus

[5]. It was previously thought that pneumococcal infections could easily be conquered

with the discovery of antibiotics, but the pneumococcus continues to prevail. Since the

late 1970’s, the pneumococcus has become increasingly resistant to penicillin and many

other antimicrobial agents, and this trend is spreading rapidly throughout the world [6-8].

S. pneumoniae infections are among the leading causes of worldwide illness and

death for young children, the elderly, and people with underlying medical conditions such

as AIDS [9]. For several decades, the pneumococcus has been responsible for an

estimated two-thirds of the cases of lethal pneumonia in the United States [10]. The case-

fatality rate from uncomplicated pneumococcal pneumonia is 5-7% [9], but this rate can

be as high as 40% in the elderly and people suffering from illnesses such as chronic

obstructive pulmonary disease (COPD) or cirrhosis [10-13]. As described later,

pneumococcal polysaccharide vaccines are available for children and adults, but they are

limited by only protecting the host from serotypes included in the vaccine.

4

Consequently, the mortality rate attributed to pneumococcal infections is one of the

highest of any vaccine-preventable disease [14].

Pathogenesis and Anti-Pneumococcal Host Defenses

The fundamental stages of pneumococcal disease and the host defense

mechanisms involved are depicted in Figure 1 below. The left column lists the

sequenced events of the infection from colonization to the development of fatal

bacteremia. The middle column contains the primary host defense mechanisms against

each stage of infection. This research project focused on the first three stages of this

progression, as illustrated on the right side of the diagram.

Figure 1 – Progression of Pneumococcal Disease

Bacteremia and Death

The role non-PMN-mediated and PMN-mediated killing play in eradication of pneumococcal infection from the lungs.

Extracellular Bactericidal Factors, Opsonic Proteins,

Alveolar Macrophages, Neutrophils (PMNs)

Development of

Pneumonia

Stimulation of mucociliary apparatus to prevent movement of pneumococci from nasopharynx into the lungs.

Mucociliary Clearance Apparatus

Aspiration into Lungs

Production of IgA that inhibits pneumococci from colonizing nasopharynx.

Secretory IgA

Antibodies

Nasopharyngeal Colonization

Research FocusHost DefensesBacterium

5

Nasopharyngeal Colonization

Colonization of the host is the first important step in the pathogenesis of S.

pneumoniae infections. The human nasopharynx is the major reservoir for the

pneumococcus and the source of horizontal spread within the community. Pneumococcal

colonization is mainly asymptomatic with carriage rates varying from 5% to 60% among

healthy adults, depending on their living conditions [9,15,16]. Invasive disease typically

occurs when the colonizing organisms are aspirated into the lower respiratory tract, the

host is immunocompromised, or when a new or more virulent strain is acquired [17,18].

Simultaneous infection with a respiratory virus such as influenza can also greatly increase

the risk of pneumococcal disease [19,20]. For example, influenza virus infection results

in increased pneumococcal adherence to the nasal epithelium and destruction of ciliated

cells that normally help maintain lung sterility.

Mucociliary Clearance Apparatus

After nasal colonization is established, the pneumococci can be aspirated into the

airway where they can invade the lower respiratory tract. An important host defense

mechanism that protects the lungs from bacterial infection is the mucociliary clearance

apparatus. This innate defense system is dependent on mucus-producing goblet cells and

the coordinated beating of ciliated cells lining the airway lumen. The mucus entraps

aspirated microorganisms, such as pneumococci, and the cilia continuously beat in a

coordinated upward fashion to remove the mucus sheath and trapped bacteria from the

lungs. The rate at which the organisms are transported out of the airway is determined by

the frequency with which the cilia beat, also known as the ciliary beat frequency (CBF).

6

The CBF is primarily regulated by the activation of protein kinases. Ciliary beating is

increased when protein kinases A and G (PKA and PKG) are activated and decreased by

the activation of protein kinase C (PKC) [21-24].

Extracellular Bactericidal Factors

Upon entering the sterile environment of the lungs, pneumococci encounter a

variety of extracellular defenses within the alveolar lining fluid. Several of these

defenses have direct bactericidal activity that can rapidly kill the invading pneumococci

[25]. These bactericidal components include lysozyme, lactoferrin, defensins, and

surfactant.

Lysozyme and lactoferrin are the most abundant antimicrobial proteins in the

lungs [26]. Lysozyme enzymatically cleaves the peptidoglycan layer of bacterial cell

walls causing the organisms to lyse [27]. This enzyme is found in the specific granules

of neutrophils, but it is mainly produced and secreted by resident alveolar macrophages

and airway epithelial cells. Lactoferrin is an iron-binding protein that is also synthesized

by neutrophils and epithelial cells in the lungs. This extracellular protein inhibits

bacterial growth by sequestering the iron that is required for microbial metabolism. In

addition to this bacteriostatic effect, iron-free lactoferrin (apolactoferrin) exhibits direct

bactericidal activity by disrupting or possibly even penetrating the bacterial cell

membrane [28,29].

Defensins are a family of small, single-chain peptides that are secreted by airway

epithelial cells and produced by phagocytes. They have potent bactericidal activity

against both Gram-positive and Gram-negative pathogens [30]. Their antimicrobial

7

function consists of forming multimers and creating pores in the bacterial membrane

[30].

Surfactant is composed mostly of phospholipids with smaller amounts of fatty

acids and proteins. The primary function of surfactant is to prevent compression of the

lungs during respiration and reduce tension in the alveoli [31]. Besides this mechanical

function, surfactant also helps protect the lungs from infection. Surfactant long-chain

free fatty acids are the most potent anti-pneumococcal factor in the alveolar lining fluid

[32,33]. The fatty acids perform in a detergent-like manner to degrade the invading

pathogen by increasing cell membrane permeability.

Opsonic Proteins

In addition to bactericidal factors, the alveolar lining fluid also contains proteins

that opsonize or coat bacteria so they are recognized and effectively engulfed and killed

by pulmonary phagocytes. The three main pulmonary opsonic components are

complement component C3, the acute phase C-reactive protein (CRP), and surfactant

protein D (SP-D).

The complement system comprises over 30 serum and membrane proteins which,

when activated, form a cascade of reactions contributing to the elimination of invading

pathogens. Complement is a particularly important opsonin in defense against

pneumococcal infection, as people defective in complement components have a much

higher rate of developing invasive pneumococcal disease than healthy individuals [34-

36]. Complement can bind to the pneumococcus by one of three pathways as shown in

Figure 2. While these pathways are activated differently, they all converge with the

8

Figure 2 – Complement Pathways and Effector Functions

deposition of C3b, the binding portion of component C3 released after its enzymatic

cleavage, on the bacterial surface. Complement C3 is predominantly formed in the liver

by hepatocytes and secreted into the bloodstream, but bronchial epithelial cells, type II

alveolar cells, and lung fibroblasts also generate C3. Regardless of the activation

pathway, opsonization with C3b is the most important effector function of the

complement system in defense against S. pneumoniae, as this leads to phagocytosis of the

organism. The final stage of the complement cascade is the membrane attack complex,

which forms pores in the bacterial membrane, causing lysis. This complex is formed by

complement C5b, C6, C7, C8, and C9. A pore forms on the bacterial surface once C9

9

polymerizes and inserts itself through the bacterial membrane. This allows a net influx of

Na+ and water resulting in the death of the bacterial cell. However, this complex is

ineffective against Gram-positive organisms, including pneumococci, due to the thick

peptidoglycan layer of their cell wall [37].

S. pneumoniae activates complement by both the classical and alternative

pathways, but the classical pathway has been found to be the dominant complement

pathway for innate immunity to the pneumococcus [38]. This is initiated by antibodies

that bind to the surface of S. pneumoniae, primarily to the polysaccharide capsule. The

Fc portion of the antibodies binds and activates C1q, which is the first component of the

complement cascade. Teichoic acid within the pneumococcal cell wall, on the other

hand, is the main activating factor for the alternative pathway [39]. Finally, S.

pneumoniae does not efficiently activate the mannan-binding lectin pathway because

mannose binding lectins bind poorly to pneumococci [40,41].

The acute phase protein CRP also functions as an opsonin to promote

phagocytosis by interacting with CRP receptors on the surface of macrophages. CRP

binds specifically to phosphocholine, the major component of the C-polysaccharide on S.

pneumoniae [42]. CRP is produced and secreted by liver hepatocytes as well as

lymphocytes, monocytes, upper airway epithelial cells, and alveolar macrophages. The

classical pathway can also be activated by CRP by recruiting C1q to the surface of the

bacteria after it binds [43].

Besides phospholipids and free fatty acids, surfactant also consists of four distinct

proteins that have been grouped in a family called collectins. Among these is SP-D,

which is synthesized and secreted by alveolar type II cells and non-ciliated bronchial

10

cells. This protein has been shown to bind and aggregate microorganisms, activate

macrophages, and enhance phagocytosis and killing of pathogens [44-46]. SP-D knock-

out mice showed enhanced susceptibility to an intranasal inoculation of S. pneumoniae,

indicating SP-D is an important host defense against pneumococcal infection [47].

Alveolar Macrophages

Resident alveolar macrophages comprise the first line of cellular defense in the

lungs, and they are responsible for the rapid phagocytosis and killing of many bacterial

pathogens. However, several researchers have reported that alveolar macrophages do not

efficiently phagocytose and kill S. pneumoniae [48-50]. Their key defensive role during

pneumococcal infection is to coordinate the pulmonary inflammatory response. When

alveolar macrophages encounter pneumococci, they release proinflammatory cytokines

and chemokines to recruit and activate neutrophils (PMNs), which are the primary

cellular defense against pneumococci as discussed below.

In rats, the major chemokines responsible for PMN recruitment are macrophage

inflammatory protein-2 (MIP-2) and cytokine-induced PMN chemoattractant-1 (CINC-

1), the homologues to human interleukin-8 (IL-8) and growth-regulated protein alpha

(gro-α), respectively. Both chemokines act through a common receptor on PMNs called

CXCR2. These chemokines increase PMN β2-integrin adhesion molecule expression,

phagocytosis, and super oxide radical production [51-53]. Although MIP-2 and CINC-1

act on the same receptor, they have slightly different roles in PMN recruitment and

activation. MIP-2 binds with a higher affinity to CXCR2 and is capable of desensitizing

the receptor to the effects of other chemokines. Unlike MIP-2 which remains in the

11

lungs, CINC-1 can leave the alveolar space and enter the systemic circulation [54,55].

The rationale for this is CINC-1 can act upon the PMNs before they are desensitized by

MIP-2, which is the more potent chemoattractant in the lungs [54].

PMN Phagocytosis and Killing

PMNs are the major immune cells responsible for pneumococcal clearance from

the lungs. In order to effectively eradicate an infection, PMNs must be recruited to the

lungs, phagocytose the pneumococci, and kill the bacteria intracellularly. Few PMNs are

present in the uninfected lung, but following infection, proinflammatory cytokines and

chemokines, such as MIP-2 and CINC-1, are secreted by alveolar macrophages and

pulmonary epithelial cells to activate and recruit large numbers of PMNs.

Once PMNs enter the lungs, they encounter the invading pneumococci that have

been opsonized for phagocytosis. In order to bind to the opsonized bacteria, PMNs

express surface receptors that recognize either the Fc fragment of IgG or complement

component C3. Once the phagocyte binds to the pathogen, a vacuole or phagosome

forms around the bound pathogen and it becomes internalized.

Immediately after phagocytosis, PMNs destroy the engulfed organisms by

oxygen-dependent and oxygen-independent mechanisms. PMNs possess four types of

intracellular granules which contain a variety of enzymes and proteins that help kill the

phagocytosed bacteria [56]. These granules fuse with the phagosome to form the

phagolysosome and release their toxic contents into this vacuole. Along with

degranulation, the nicotinamide adenine dinucleotide phosphate oxidase system forms on

the phagolysosome membrane and converts oxygen to the superoxide radical. The

12

superoxide radical is essential for the formation of multiple bactericidal components. In

addition, its dismutation results in the production of hydrogen peroxide [57].

Myeloperoxidase and nitric oxide synthase are also involved in oxygen-dependent

killing of microorganisms. Myeloperoxidase is an enzyme that uses hydrogen peroxide

to form hypochlorous acid (HOCl), the most potent antibacterial oxidant produced by the

PMN. HOCl has been shown to impair DNA synthesis and bacterial replication [58].

Myeloperoxidase is also known to interfere with DNA synthesis and ATP synthesis

systems [58,59]. Nitric oxide synthase leads to the formation of reactive nitrogen

intermediates including nitric oxide and nitroxyl anion that destroy bacterial DNA.

Oxygen-independent killing occurs primarily through the actions of microbicidal

proteins contained within the specific granules. These non-oxidative components include

proteases, defensins, lysozyme, and lactoferrin. Their activity degrades the bacterial cell

wall and membrane, resulting in lysis and eventually death of the pathogen.

Macrophages can also undergo an oxidative burst and degranulation and execute similar

killing mechanisms as PMNs, but they lack myeloperoxidase and lactoferrin. With the

combination of reactive oxygen species and granular antimicrobial proteins, phagocytes

can effectively eradicate a wide variety of microorganisms, including S. pneumoniae

[60].

Adaptive Immune Response

Although the innate immune system is the primary host defense against

pneumococcal infections, adaptive immunity is also important against the pneumococcus.

Anti-pneumococcal antibodies provide immunity against S. pneumoniae by preventing

13

colonization and promoting pneumococcal clearance in the host. For several decades, it

was thought that the main adaptive response to pneumococcal infection was the

production of IgM and IgG antibodies that bind to the polysaccharide capsule to mediate

opsonization and enhance phagocytosis [61]. However, this idea has evolved as a result

of recent studies documenting the importance of antibodies targeting pneumococcal

proteins that are vital in pneumococcal pathogenesis [62-64]. For example, one animal

experiment showed that antibodies against pneumolysin, the multifunctional toxin

produced by S. pneumoniae, were anti-inflammatory and averted invasive disease [65].

Secretory IgA antibodies also play a role in pneumococcal resistance by providing one of

the first lines of defense against nasopharyngeal colonization [66]. These antibodies limit

colonization by binding to the surface of pneumococci and preventing attachment to the

epithelium.

Until recently, T-cells were not considered to have a major role in the adaptive

immune response against the pneumococcus. Polysaccharide capsules from many

serotypes of S. pneumoniae are considered to be T-cell independent antigens because

they express repeating epitopes that cross-link B-cell receptors and induce B-cell

activation and proliferation without T-cell help. However, T-cells are involved in

antibody responses to other pneumococcal protein antigens such as pneumolysin and

surface proteins that are important in pneumococcal immunity. T-cells also appear to

contribute to early resistance against pneumococcal infection independently of their role

in adaptive antigen-specific responses. Knock-out mice that displayed a significant

decrease in CD4+ T-cell levels had an increased susceptibility to an intranasal infection

with 100% mortality by 3 days post-infection, whereas all wild-type mice survived the

14

challenge [67]. T-cells also have been shown to migrate to infected lung tissue during

pneumococcal pneumonia by an unknown interaction with pneumolysin [68,69]. In

addition, Malley and colleagues demonstrated a crucial role for CD4+ T-cells in antibody-

independent acquired immunity to pneumococcal colonization [70]. This was shown by

immunizing antibody-deficient or CD4+ T-cell-deficient mice intranasally with an

unencapsulated pneumococcal strain followed by an intranasal challenge with type 6

pneumococci. The antibody-deficient mice had reduced pneumococcal colonization

while the CD4+ T-cell deficient mice had similar levels of pneumococcal colonization as

the unimmunized control mice. More experiments are being done to understand the exact

mechanisms of the T-cell response against S. pneumoniae.

Treatment and Prevention of Pneumococcal Disease

Since the advent of antibiotics in the 1940s, penicillin was the preferred choice for

treating pneumococcal infections until the early 1990s, when antimicrobial resistance

among clinical isolates of S. pneumoniae in the United States first emerged as a

significant problem [71]. The prevalence of resistance to penicillin has increased steadily

and is now at approximately 35% in this country [72-74]. A similar trend has been

observed for other antimicrobial classes including macrolides, clindamycin, tetracyclines,

trimethoprim-sulfamethoxazole, and chloramphenicol [75]. Multidrug resistant strains

are also on the rise; one report found that 34.6% of isolates in the U.S. are resistant to

more than one antibiotic [76]. Fluoroquinolones are the only class of antimicrobials that

has escaped the problem of emerging resistance among S. pneumoniae. However, with

the increasing use of fluoroquinolones to manage respiratory tract infections in adults and

15

the growing number of nonsusceptible isolates to these antibiotics in other countries [77],

fluoroquinolone resistance in this pathogen is expected to eventually emerge in this

country.

Despite these alarming trends, antibiotic resistance in S. pneumoniae may have

reached a plateau and started to decrease [75]. It has been speculated that several factors

may be responsible for this event. Healthcare providers are using oral antibiotics more

appropriately to treat patients with acute respiratory tract illnesses [78]. The increasing

use of the conjugated pneumococcal vaccine, which covers antibiotic-resistant serotypes,

may be impacting resistance [79-84]. Lastly, a large percentage of adults are being

vaccinated for influenza, and a lower incidence of influenza infections results in fewer

bacterial infections, including those caused by S. pneumoniae [85,86].

Vaccination is the most effective strategy in preventing pneumococcal disease.

Currently, there are two pneumococcal polysaccharide vaccines licensed in the United

States. They are a 23-valent pneumococcal capsular polysaccharide vaccine

(Pneumovax, Merk) and a 7-valent pneumococcal conjugate vaccine (Prevnar, Wyeth).

The 23-valent vaccine contains capsular material from the 23 serotypes that commonly

cause invasive disease in the United States. This vaccine is recommended for adults over

the age of 50, especially the elderly and people at high risk for infection such as patients

with cirrhosis or COPD. Several studies have found the 23-valent vaccine to be at least

partially protective and to effectively reduce the incidence and mortality of invasive

pneumococcal disease [87-90]. However, results from other studies contradict these

findings, and the extent of the vaccine’s protective effect continues to be disputed

[91,92]. A major problem of Pneumovax is that it has poor efficacy and immunogenicity

16

in certain high-risk groups, such as alcoholics, smokers, the elderly, children <2 years,

and patients with chronic liver and lung diseases [87,91,93]. The vaccine also has no

effect on pneumococcal colonization and provides protection against only 23 of the >90

pneumococcal serotypes [91,94].

In the year 2000, the pneumococcal conjugate vaccine was approved in the United

States for routine use in all children aged <5 years [95]. This vaccine contains capsular

material from 7 serotypes that cause 80% of invasive pneumococcal disease in young

children. The immunogenicity of this vaccine is effective in this age group due to the

conjugation of each polysaccharide to a non-toxic diphtheria toxin protein [96]. The

introduction of Prevnar led to substantial reductions in the incidence of invasive disease

in children [80,97-99]. Use of this vaccine reduced pneumococcal disease among

unvaccinated populations by reducing nasopharyngeal colonization and transmission of

vaccine-type pneumococci, a process known as herd immunity [80]. As mentioned

above, the conjugate vaccine also has made a strong impact in preventing infections

caused by antimicrobial-resistant strains since the included serotypes were among those

with the highest percentages of antibiotic resistance. Prevnar has even more limited

serotype coverage, and vaccinated children are now being colonized and developing

infections with non-vaccine serotypes [100-102]. Due to the drawbacks of these two

pneumococcal vaccines, more research is needed to explore other types of pneumococcal

vaccines, particularly those that target conserved proteins such as pneumolysin or

pneumococcal surface proteins [62-64,103].

17

B. Virulence Factors of Streptococcus pneumoniae

Polysaccharide Capsule

The polysaccharide capsule is considered the major virulence factor of S.

pneumoniae because unencapsulated bacteria are less invasive compared with the same

encapsulated strain [104]. For example, as little as 10 cfu of capsulated pneumococci can

cause disease in small rodents, while as many as 106 cfu of unencapsulated pneumococci

are needed to reproduce a similar disease [105]. This virulence factor constitutes the

outer most part of the pneumococcus and creates a protective shell that surrounds the

microorganism. The capsule is polymeric consisting of repeating units of

oligosaccharides. Over 90 different serotypes of S. pneumoniae have been identified

based on the structure of their capsule. Functionally, the polysaccharide capsule

modulates the interaction between the bacterium and its environment, including the

interaction and adherence to host tissues. The capsule is also known to evade

opsonization and phagocytosis [106].

Cell Wall

The pneumococcal cell wall is composed of a thick peptidoglycan layer that

surrounds the cytoplasmic membrane. Peptidoglycan is composed of repeating units of

N-acetyl muramic acid and N-acetyl glucosamine that crosslink to form glycan chains

[107]. Unlike the polysaccharide capsule, the cell wall is highly immunogenic and it has

been shown in animal models that the purified form of the pneumococcal cell wall can

reproduce symptoms of pneumococcal disease [107,108]. One of the most antigenic

components of the cell wall is teichoic acid or C-polysaccharide. Teichoic acid is

18

commonly found in Gram-positive bacteria and its immunostimulatory activity is due to

phosphocholine residues that can bind to CRP [109]. The pneumococcus utilizes teichoic

acid to anchor surface proteins such as pneumococcal surface proteins A and C [110],

which will be discussed in detail in the next section.

Pneumococcal Surface Proteins A and C

A variety of proteins expressed on the surface of S. pneumoniae contribute to its

virulence and pathogenesis within the host. The functions of these surface-exposed

proteins include interactions with host tissues or concealing the bacteria from the host’s

defense mechanisms. Some of these proteins are being used as alternative targets for

developing new vaccines. Among these proteins are pneumococcal surface proteins A

and C (PspA and PspC).

PspA is one of the major surface proteins and antigens of the pneumococcus and

is produced by all strains [111]. Antibodies raised against PspA are protective against

pneumococcal disease. In fact, mice vaccinated with PspA survived a lethal dose of S.

pneumoniae [112]. Although PspA is expressed on all pneumococcal serotypes, it is

highly variable among the different strains [113]. This allows different pneumococcal

serotypes to repeatedly infect the same host.

PspA plays a crucial role in protecting S. pneumoniae from the host complement

system. This is attributed to the electronegative charge on the surface-exposed end of the

protein which repulses binding of the complement proteins and as a consequence,

prevents complement activation [114]. By comparing a wild-type pneumococcal strain to

19

a mutant strain lacking PspA, it was demonstrated that PspA’s anti-complement activity

reduced complement-mediated clearance and phagocytosis of S. pneumoniae [115].

PspC, also known as choline binding protein A, was the first adhesion molecule to

be discovered on pneumococci. PspC aids in bacterial adherence and colonization to host

tissues and this was confirmed by studies of PspC-deficient mutant pneumococci [116].

The adhesion properties of this protein act as a physical bridge between the

pneumococcal cells and the host cells by utilizing choline of teichoic acid at one end and

the host glycoconjugates on the other end [116]. This bridging interaction seems to be

limited to cytokine-activated human cells expressing certain glycoconjugates [116]. It

has been suggested that this process might be involved in advancing the pneumococcal

disease from colonization to invasion [110].

PspC also functions in inhibiting complement. PspC has the ability to bind host

Factor H, a protein that protects host cells from complement by inhibiting the activation

of the alternative complement pathway. The binding of Factor H to a host cell promotes

degradation of C3b already deposited on the cell surface. Therefore, the ability of PspC

to bind Factor H enables the pneumococcus to evade the activation of complement by

disguising itself as a host cell [117].

The structure and sequence of PspC is very similar to PspA. Immunization

studies have reported that PspA vaccination may produce antibodies that cross-react with

PspC [115]. Mice vaccinated with PspC have been shown to be protected from

pneumococcal colonization and invasive disease [63]. However, like PspA, PspC is

highly variable among pneumococcal strains and not all pneumococci express this

surface protein [118].

20

Pneumolysin

Pneumolysin is a member of the thiol-activated cytolysins and is produced by all

clinical isolates [119]. Unlike other pneumococcal virulence factors, this protein toxin is

not surface exposed. Pneumolysin resides in the cytoplasm and is released when the

pneumococcus undergoes autolysis. However, one study has challenged this idea by

reporting that pneumolysin is also released during the exponential phase of growth [120].

Pneumolysin has several distinct functions, especially in the early stages of

pneumococcal infection. Its cytolytic activity affects all eukaryotic cells by attaching to

membrane bound cholesterol and forming pores in the lipid bilayer. The toxin damages

ciliated bronchial epithelial cells, which inhibits ciliary beating and facilitates the spread

of pneumococci into the lower respiratory tract [121]. Pneumolysin disrupts the alveolar-

capillary boundary layer providing nutrients for bacterial growth and promotes

penetration through the lung tissue into the bloodstream [122,123]. The cytotoxic effects

of pneumolysin also can directly inhibit phagocyte and immune cell function, which

leads to the suppression of the host inflammatory and immune responses. Low

concentrations of pneumolysin are able to inhibit PMN respiratory burst, chemotaxis, and

bactericidal activity [124].

In addition to cytolytic activity, pneumolysin can also activate complement.

When pneumolysin is released it can bind the Fc portion of IgG, which directly activates

the classical complement pathway [125]. This complement-activating activity of

pneumolysin consumes complement proteins and diverts complement activation away

from the bacterial surface [125]. Several studies have demonstrated that pneumolysin

21

significantly reduced opsonization of pneumococci and this subsequently results in

decreased PMN-mediated phagocytosis and killing of the organisms [126,127].

C. Alcohol Abuse and Anti-Pneumococcal Host Defenses

Incidence of Disease

Excessive use of alcohol is the third most lethal modifiable risk factor affecting

health in this country [128]. The prevalence of alcohol abuse among Americans is 17%

[129], and this has remained steady for the last fifteen years [130,131]. In the year 2000,

alcohol consumption accounted for 85,000 deaths in the U.S. [128]. Aside from its

adverse physical effects, alcohol abusers tend to have impulsive lifestyles that can lead to

risky sexual activity, injuries, and suicide [132,133]. These types of behavior also

increase the risk of developing chronic illnesses [134], such as cirrhosis of the liver, and

many types of infections [135].

Alcohol abuse is one of the most common predisposing risk factors for bacterial

pneumonia, and alcohol-abusers experience higher morbidity and mortality from

pulmonary infections than the general population [136]. Among all bacterial

pneumonias, S. pneumoniae is the most frequent pathogen in alcohol-abusing patients

[135,137-140]. These patients experience more severe clinical symptoms, require longer

hospital stays and treatment, and have slower resolution of their disease [137]. They

have an increased risk of developing pneumococcal bacteremia, which greatly increases

their mortality [141] In addition, they are more likely to have recurring episodes of

pneumonia [142]. The explanation behind these findings is that alcohol has

immunosuppressive properties that adversely affect both innate and adaptive immune

22

pulmonary host defenses. Several studies indicate ethanol (EtOH) ingestion has

detrimental effects on vital anti-pneumococcal defenses that will be further explained in

the next several sections.

Pneumococcal Colonization of Alcohol Abusers

The lifestyle of many alcohol abusers tends to increase their risk of developing

pneumococcal disease. There are no reports that link alcohol abuse to increased

pneumococcal colonization of the nasopharynx. Alcohol abuse can depress

consciousness and the cough reflex, factors that increase the risk of aspirating colonized

pneumococci into the lungs [136,143].

Impairments to Mucociliary Clearance

Chronic EtOH exposure affects CBF by blocking the activation of PKA. Studies

have shown that EtOH desensitizes the ciliary response to isoproterenol, a short-acting

β2-agonist that stimulates ciliary beating through PKA [144]. Additionally, this EtOH-

induced defect in mucociliary clearance was detected in animals that were fed EtOH for

five to six weeks [145,146]. EtOH-fed rats have decreased CBF, resulting in increased

pneumococcal movement into their lungs after an intranasal infection [145].

Defects in Extracellular Bactericidal Factors

Although no associations have been made between alcohol abuse and alterations

in levels of free pulmonary lysozyme, lactoferrin, or defensins, there is some evidence

that EtOH does affect these proteins. One study demonstrated that in vitro EtOH

23

exposure significantly reduces lysozyme secretion by human macrophages [147], and this

parallels decreased levels of lysozyme in the serum of alcoholics [148]. In another study,

acetaldehyde, the major byproduct of metabolized EtOH found in the saliva, airways and

blood, was shown to inhibit the bactericidal function of lysozyme [149]. Alcoholics

actually have higher levels of lactoferrin in their serum compared to non-drinkers [148],

because alcohol abuse increases iron concentration in the bloodstream [150,151]. This

disturbance in iron homeostasis tends to saturate lactoferrin and may limit the availability

of apolactoferrin. Chronic EtOH consumption may also modulate the defensins.

Because alcohol induces oxidative stress in the lungs, it damages pulmonary cells

including type II pneumocytes, the major cell type that generates defensins [152].

Excessive chronic alcohol ingestion suppresses the production and antimicrobial

function of surfactant. Using a rat model exposed to EtOH for six weeks, surfactant loss

was linked to EtOH-induced damage to type II pneumocytes [153], the main source of

surfactant, as well as an 80-90% reduction of glutathione in the lungs [152]. Glutathione

is a potent antioxidant synthesized primarily by the liver that is abundant in alveolar cells

and fluid. This EtOH-induced defect in surfactant production was reversed by treating

EtOH-fed rats with the glutathione precursor procysteine [153].

In contrast to chronic EtOH exposure, rats exposed to EtOH for seven to ten days

decreased the anti-pneumococcal activity of surfactant, but it was not due to a decrease in

amount of surfactant produced. Pneumococcal killing by surfactant from EtOH-fed rats

was 4 logs lower than surfactant from pair-fed control rats [33]. Unexpectedly, the robust

killing ability of free fatty acids from the surfactant of control animals was lost with the

addition of surfactant from EtOH-fed animals [33]. This suggests that EtOH ingestion

24

results in the production of an inhibitory component in the lungs that inactivates the

bactericidal function of the free fatty acids.

Alterations in Opsonic Activity

Only one published study has reported findings on the relationship between EtOH

ingestion and the opsonic activity within the lungs. Lung lavage fluid obtained from

guinea pigs after six weeks of EtOH feeding had diminished levels of surfactant with

decreased opsonic activity towards Staphylococcus aureus [154]. Serum CRP levels are

unaltered in mice that consumed EtOH for six weeks and in a sample of heavy human

drinkers [155,156], but CRP concentrations in the lungs were not reported. In contrast,

only a few reports address the effects of EtOH ingestion on host complement, and the

results are contradictory. Serum complement levels have been reported both as reduced

and increased in various studies of alcoholic patients [157,158]. Serum hemolytic

activity was decreased in acutely intoxicated patients in one study [159], but another

reported that the addition of alcohol to normal serum in vitro had no affect on this activity

[160]. A third group found no change in serum complement activity in volunteers after

8-28 days of EtOH consumption [161,162].

Meri and colleagues reported that six weeks of EtOH ingestion induced the

expression of complement components and also down-regulated the expression of

complement regulatory proteins by the liver in mice and rats [155,163]. This led to the

deposition of complement C3 and other complement components on the liver

hepatocytes. These results indicate that EtOH ingestion nonspecifically activates

25

complement, which may lead to the depletion of functional complement available for

opsonization.

Impairments in Alveolar Macrophage Function

Although alveolar macrophages inefficiently phagocytose and kill S. pneumoniae,

they still play a vital role in the pulmonary inflammatory response during pneumococcal

pneumonia. EtOH exposure has been shown to suppress a wide range of macrophage

functions. Alveolar macrophages collected from EtOH-fed rats and rat macrophages

exposed to EtOH in vitro both exhibit decreased oxidative burst when stimulated with

either Staphylococcus aureus or endotoxin [164,165]. Two weeks of EtOH feeding in

mice similarly inhibited the ex vivo production of proinflammatory cytokines by their

alveolar macrophages [166]. Release of MIP-2 and CINC-1 in the lungs of rats injected

intraperitoneally with EtOH also were inhibited after being infected with S. pneumoniae

[167]. Another published study found that peritoneal macrophages harvested from

EtOH-fed rats for 12 weeks demonstrated impaired phagocytosis of Candida albicans,

despite having larger numbers of Fc and C3b surface receptors that facilitate

phagocytosis compared to macrophages from control rats [168].

Impairments in PMN Function

PMNs are essential for effective pneumococcal clearance from the lungs. Failure

of this cellular defense can result in uncontrolled pneumococcal growth within the lungs,

leading to bacteremia and eventually death. It is well-established that EtOH exerts

various effects on the functional activity of PMNs. Circulating PMNs must first be

26

activated and recruited to the lungs by cytokines and chemokines. Rats exposed acutely

to an intraperitoneal injection of 20% EtOH and then infected intratracheally with S.

pneumoniae 30 minutes later had decreased pulmonary production of MIP-2 and CINC-1

[167,169]. This suppressed response delayed PMN recruitment, a deficiency that

persisted even when MIP-2 was administered intratracheally [169]. EtOH inhibits PMN

β2-integrin expression, which is critical for PMN adhesion to the endothelium and

migration into the infected tissue [170]. However, one week of EtOH ingestion by rats in

our laboratory consistently failed to suppress pulmonary chemokine levels or repress

PMN recruitment to their lungs [171,172].

The phagocytic activity of rat PMNs was reduced by exposure to very high EtOH

concentrations in vitro, but physiologically relevant concentrations of EtOH in the same

study failed to inhibit phagocytosis [173]. Although acute EtOH reduced the phagocytic

activity of circulating PMNs [170,174], it did not affect phagocytosis by PMNs recruited

into the lungs [171,175].

In vitro and in vivo studies in our laboratory have established that EtOH ingestion

impairs PMN-mediated killing of S. pneumoniae. PMNs isolated from the peripheral

blood of rats consuming EtOH for one week killed significantly fewer pneumococci in

vitro than PMNs isolated from the blood of pair-fed control rats [176]. This was related

to a decrease in both oxygen radical production and degranulation in PMNs from the

EtOH-fed rats [177]. These results correlate with a human study in which PMNs from

alcoholics exhibited diminished elastase activity and superoxide production [178]. The

EtOH-induced defect in PMN bactericidal activity was further confirmed in our

laboratory using an in vivo PMN killing assay in which PMNs were pre-recruited to rats’

27

lungs five hours prior to infection. Rats fed EtOH for one week had killed 20% fewer

pneumococci within their lungs than their pair-fed counterparts one hour after the

transtracheal infection [171].

Alterations in the Adaptive Immune Response

Numerous studies have shown that alcohol consumption suppresses acquired

immune defenses including both cell-mediated and humoral immunity. Chronic

alcoholics are known to be lymphopenic [179-181], and long-term alcohol feeding in

animals also resulted in decreased size and cell numbers in the thymus and spleen [182-

184]. It has been proposed that the underlying mechanism for this phenomenon is that

EtOH induces lymphocyte apoptosis [185]. In addition to the low number of lymphoid

cells, an impaired proliferation response also has been reported, signifying that EtOH-

exposed lymphocytes have a reduced capacity to undergo proliferation and differentiation

in response to an antigenic challenge [186]. This decreased response in T-cells is partly

due to impairment in antigen presentation by antigen presenting cells [187-189]. Chronic

EtOH exposure also significantly reduces the absolute number of CD4+ T-cells and

hinders T-cell recruitment to the lungs after a pulmonary infection [190].

The development of specific antibodies is important in protecting the host against

pneumococcal infection. Alcohol-induced defects of specific antibody production would

be expected to adversely affect the eradication of invading pneumococci from the airways

in patients with pneumonia [137]. Despite decreases in B-cell numbers, alcoholics with

liver disease have increased circulating levels of IgA, IgM, and IgG [158,188,191,192].

In contrast, an analysis of bronchoalveolar lavage fluid in patients with alcoholic liver

28

disease showed reduced total IgG concentrations [193]. B-cell functions were intact in

EtOH-fed animals when T-cell-independent antigens were administered [194]. This B-

cell response was also present in chronic alcoholics when vaccinated with 23-valent

pneumococcal polysaccharide vaccine [195]. However, chronic alcohol ingestion hinders

the development of specific antibodies in response to challenges with T-cell-dependent

antigens in experimental animals [186,196,197].

D. Smoking and Anti-Pneumococcal Host Defenses

Incidence of Disease

Cigarette smoking is the single most preventable cause of morbidity and mortality

in the United States [128]. One-fourth of American adults smoke cigarettes, [198] and

every year 1.5 million people in this country become daily smokers [199,200]. Although

the overall smoking rate among adults from the last four decades has decreased, there is

an alarming increase of cigarette smoking among high-school students [201,202]. In

addition, tobacco use has markedly increased worldwide since the 1980s [201]. Smoking

is responsible for 438,000 annual deaths in this country [203] and over 5 million annual

deaths worldwide [204]. The prevalence of cardiovascular diseases, lung cancer, and

microbial infections is higher in cigarette smokers than nonsmokers. Smoking is also the

strongest risk factor for developing lung carcinoma, COPD, and emphysema [205].

Clinical studies have associated smoking with an increased incidence and severity

of respiratory illnesses, including pneumococcal pneumonia [206-208]. A population-

based case-control study found smoking to be the strongest independent risk factor for

invasive pneumococcal disease among immunocompetent, non-elderly adults [209].

29

Furthermore, they found a dose response relationship between the amount of smoking

and the risk of pneumococcal pneumonia [209]. Smokers account for approximately half

of otherwise healthy adult patients with severe pneumococcal disease [210,211].

Like alcohol abuse, smoking alters both the innate and adaptive immune systems,

which is the likely cause for increased risk of pneumococcal infection and other diseases

in this population. Smoking compromises several key anti-pneumococcal defenses. It

promotes bacterial colonization of airway epithelial cells [212], damages the mucociliary

clearance apparatus [213], and increases alveolar vascular and epithelial permeability

[214,215]. The sections below will explain in more detail the alterations of host defense

mechanisms against pneumococcal pneumonia caused by the actions of cigarette smoke.

Smoking and Nasopharyngeal Colonization

Cigarette smoking increases the risk of nasopharyngeal colonization by potential

pathogens including S. pneumoniae [216-218]. Buccal epithelial cells isolated from

smokers were significantly more susceptible to pneumococcal adherence in vitro

compared to buccal cells isolated from nonsmokers [219-221]. This was also true for

cells from ex-smokers who had refrained from smoking for at least three years [212].

Furthermore, these results were reproduced when buccal cells from nonsmokers were

incubated in medium containing cigarette smoke extract before exposure to the organisms

[219]. Cigarette smokers also have lower amounts of salivary and pulmonary IgA

antibodies, which may contribute to their higher rates of pneumococcal colonization and

incidence of pneumococcal pneumonia [222,223]

30

Impairments of Mucociliary Clearance

Cigarette smoke impairs mucociliary function. When bovine bronchial epithelial

cells were exposed to cigarette smoke extract in vitro, the beat frequency of their cilia

was down-regulated in conjunction with the activation of PKC [224]. Human studies

also revealed that smoking directly damages ciliated epithelial cells, causing ciliary loss

and stasis and hindering mucociliary clearance [225-227]. These defects of the

mucociliary apparatus lead to increased pneumococcal movement from the nasopharynx

to the lungs after intranasal infection [145].

Alterations in Extracellular Bactericidal Factors

There is a paucity of studies describing the effects of smoke on the overall activity

of bactericidal factors in the lungs, but smoking has been shown to alter pulmonary levels

of antibacterial proteins. Smokers have higher lysozyme concentrations in their alveolar

lining fluid, and their alveolar macrophages have a higher lysozyme content than those of

nonsmokers [228,229]. Another study found elevated levels of lactoferrin in smokers

with chronic bronchitis, but lactoferrin was only slightly elevated in asymptomatic

smokers compared to non-smokers [230]. A proteomic analysis of human

bronchoalveolar lavage fluids (BALFs) revealed increased amounts of PMN defensins in

smokers with COPD [228]. By contrast, certain key surfactant proteins important for

lung defense such as SP-A and SP-D are significantly lower in human smokers

[231,232], and this trend is also exhibited in rats exposed to smoke for 70 weeks [233].

An in vitro study demonstrated that cigarette smoke inhibits surfactant production by type

II pneumocytes [234]. In contrast, mice smoke-exposed for 6 months have a higher

31

expression and concentration of surfactant proteins in their lungs than mice exposed to

room air [235,236].

Modifications of Opsonic Proteins

Cigarette smoke has been shown to modulate the levels of opsonic proteins. In

vitro studies reported that smoke modifies complement C3, resulting in activation of the

alternative pathway and decreased susceptibility of C3 to complement regulatory proteins

[237,238]. Smoke-induced complement activation may deplete available C3 for bacterial

opsonization, and this could account for why smokers with COPD have lower serum

levels of complement C3 [239,240]. However, similar serum concentrations of C3 were

reported for asymptomatic smokers and nonsmokers [240].

Smokers have been reported to have significantly higher levels of plasma CRP

[241-244], and there is a strong correlation between CRP levels and the number of

cigarettes smoked, pack-years of smoking, and duration of smoking [242,243]. CRP

concentrations remain elevated even after 5 years of smoking cessation, suggesting

smoke does not directly increase CRP. The elevated levels may be due to an

inflammatory stimulus such as tissue damage caused by smoking [243].

As mentioned above, smoking also decreases the production of surfactant in the

lungs. BALFs from healthy human smokers, for example, have significant reductions in

SP-D [231,232]. It therefore has been postulated that an unknown inhibitory compound

in cigarette smoke interferes with either the biosynthesis of surfactant or active transport

mechanisms involved in surfactant secretion by alveolar type II cells [234].

32

Alterations in Alveolar Macrophage Function

Several reports suggest that alveolar macrophage function may actually be

enhanced by cigarette smoke, but there are conflicting results concerning the influence of

cigarette smoke on macrophages. There is an increased number of macrophages in the

lungs of human smokers and in mice exposed to cigarette smoke for six weeks as

determined by bronchoalveolar lavage [146,245]. Macrophages from smokers also are in

a higher activation state, containing an overload of cytoplasmic material and larger

quantities of lysozyme and elastase than macrophages obtained from nonsmokers

[229,245]. However, in vitro studies have shown smoke exposure hinders human

alveolar macrophage phagocytosis and inhibits their response to endotoxin; thereby

causing them to generate fewer superoxide anions [246,247]. Yet another study reported

no smoke-induced defects in human alveolar macrophage phagocytosis [248].

Interestingly, that same study found smoking suppressed the bactericidal activity of the

macrophages even though they exhibited a robust oxidative burst. Finally, Morrison

reported no differences in IL-8 and gro-α levels in BALFs from smokers and

nonsmokers, but when alveolar macrophages from the smokers were stimulated in vitro

with endotoxin they produced more chemokines than those from nonsmokers [249].

Alterations in PMN Function

The documented effects of smoke exposure on PMN function are also

inconsistent and not well defined. Certain fractions of cigarette smoke were shown to be

potent inhibitors of PMN chemotaxis [250], but other studies report cigarette smoke

enhances this activity. This is evidenced by the fact that uninfected human smokers have

33

significantly higher concentrations of neutrophils and chemokines in their BALFs than

nonsmokers, and the increases are dose-dependent on cigarette smoke [251]. Cigarette

smoke extract stimulates human endothelial cells, lung fibroblasts, and airway epithelial

cells to release IL-8 and other PMN chemoattractants [252-254]. Furthermore, PMNs

isolated from rats that were either treated with nicotine for one week or exposed to

cigarette smoke for 15 weeks had greater chemotactic responses than those from control

rats [255]. To add to the controversy, rats exposed to smoke for 8 weeks had no

alterations in pulmonary chemokine levels or PMN recruitment to their lungs after

endotoxin was administered transtracheally [171].

The effects of smoking on PMN phagocytosis and killing of bacteria are

conflicting as well. Salivary PMNs isolated from smokers immediately after having a

cigarette had increased phagocytic activity compared to PMNs from non-smoking

controls [256]. However, this effect was specific for salivary PMNs, because there was

no difference in phagocytosis by peripheral PMNs isolated from the same subjects. Other

in vitro studies found cigarette smoke extract hinders phagocytosis by human peripheral

PMNs [257,258]. Furthermore, published studies disagree on whether cigarette smoke

inhibits [259] or stimulates [255,260] the PMN oxidative burst. One study observed a

delayed rate of bacterial clearance in smoke-exposed mice of 6-8 weeks compared to

unexposed mice infected with Pseudomonas aeruginosa, even though the smoke-exposed

animals had increased levels of proinflammatory cytokines and more PMNs in their lungs

[261]. To the contrary, smoke-exposed rats of eight weeks displayed no defects in PMN

phagocytosis and killing of S. pneumoniae within their lungs [171].

34

Impairments in Adaptive Immunity

It is well documented that chronic smoke exposure adversely affects both humoral

and cell-mediated functions of the adaptive immune response [262-264]. Cigarette

smoking in humans is known to increase the number of blood leukocytes, but at the same

time reduce their activity [265]. Nicotine has been found to be the mediating factor that

induces immunosuppression. Nicotine treatment impairs antigen receptor-mediated

signal transduction in T-cells, thus resulting in T-cell anergy [266,267]. This prevents T-

cells from responding correctly to antigens that would allow them to enter the cell cycle

and proliferate. Smoke-exposed mice therefore displayed inhibited antigen-specific T-

cell responses in their lungs [268]. Similar defects in cell signaling were also detected in

T-cells from human smokers and smoke-exposed animals [269,270].

Cigarette smoke also represses antigen-induced B-cell activation [266]. These

modulating effects on T-cells and B-cells result in defective antibody responses to both

T-cell-dependent and -independent antigens [271-274]. Several investigators have shown

that long-term smoking significantly reduces antigen-specific antibodies in humans

[222,223,275,276]. However, smokers have increased levels of autoantibodies [277,278],

which partially explains the higher incidence of certain autoimmune diseases in smokers.

35

III. SUMMARY AND RESEARCH OBJECTIVES

Pneumococcal infections still remain a significant problem throughout the world.

The majority of community-acquired pneumonia cases in the United States are caused by

S. pneumoniae. Alcohol abuse and smoking increase the incidence of respiratory tract

infections caused by the pneumococcus. Alcohol abusers have recurring bouts of

pneumococcal pneumonia, longer hospital stays, an increased risk of developing

bacteremia, and a higher mortality rate from this disease than non-alcohol abusers.

Cigarette smokers have an increased risk of being colonized by S. pneumoniae and

developing invasive pneumococcal disease. Previous research has shown EtOH ingestion

and smoke exposure deter host defenses that protect against pneumococcal pneumonia,

but few studies have evaluated the combined effects of these two insults. Between 80-

95% of alcoholics smoke cigarettes and approximately 70% of them smoke more than

one pack/day [279]. Likewise, over half of multipack/day smokers are alcohol

dependent. Therefore it is important to study the combined effects of these co-

morbidities on host immunity to pneumococcal disease. Furthermore, current therapies

are ineffective in this high risk group and new therapeutic strategies are needed to protect

this population from pneumococcal infection.

It is difficult to study the effects of alcohol and smoke in human subjects for it is

impossible to tease out the individual effects of drinking and smoking in humans that

engage in both behaviors. Additionally, it is hard to control for the amount of alcohol

that is consumed and the amount of cigarettes that are smoked as well as the duration of

each exposure. For these reasons we developed a rat model of EtOH ingestion and

36

smoke exposure that parallels a heavy human smoker and alcohol abuser [280]. The

advantages of using an animal model include controlling for the amount of alcohol

consumed, the number of cigarettes smoked, and the duration of the two insults as well as

identifying the separate and collective effects of these two behaviors.

Innate immune defenses against the pneumococcus are vital in preventing

invasive disease. Alcohol ingestion and smoke exposure impair many aspects of innate

immunity including mucociliary clearance, bactericidal factors, alveolar macrophages,

and PMNs. Little is known about the combined effects of these two behaviors on anti-

pneumococcal defenses. The purpose of this work was to utilize a rat model that

combines EtOH ingestion and smoke exposure to study their separate and combined

effects on innate defense mechanisms necessary for controlling pneumococcal disease

and then target these key defenses for therapeutic intervention. The effect of chronic

EtOH ingestion and smoke exposure on nasopharyngeal colonization and mucociliary

clearance was investigated by quantifying pneumococcal colonization and tracking

pneumococcal movement from the nasopharynx to the lungs. In addition to this study, an

intranasal vaccine containing a recently discovered pneumococcal surface protein called

pneumococcal protective protein A (PppA) was tested to reduce colonization.

Immunization with PppA was shown to be protective against several serotypes in mice

[64]. Two β2-agonists, which stimulate ciliary beating by activating PKA, also were

evaluated to up-regulate mucociliary clearance and inhibit pneumococcal invasion of the

lungs. Next, alterations of chronic EtOH consumption and smoke exposure on non-

PMN-mediated defenses were identified by an in vivo bactericidal assay, quantification of

bactericidal proteins, an opsonic protein deposition assay, and several assays measuring

37

important macrophage functions. Finally, EtOH- and smoke-induced modifications on

PMN function were analyzed by another in vivo killing assay and a phagocytosis assay.

38

IV. MATERIALS AND METHODS

A. Model of Chronic EtOH Ingestion and Smoke Exposure

Male Sprague-Dawley rats weighing 100-120 grams were used for all

experiments. Smoke exposed rats were passively exposed to smoke generated by 30

reference cigarettes (2R4F, Tobacco Health Research Institute, University of Kentucky)

in whole-body chambers (Teague Enterprises, Davis, CA) twice daily Monday through

Friday and once daily on Saturday and Sunday for 12 weeks [280]. Smoke inhalation

was quantified by measuring the total suspended particles in each chamber [281]. For the

same time periods, sham-exposed control rats housed in a separate room were placed in

similar chambers and exposed to room air.

For the first 6 weeks of smoke- and sham-exposure, the rats were housed in group

cages and fed rat chow and tap water ad libitum. After 6 weeks, the rats were placed in

single cages with their water removed and food replaced with the Lieber-Decarli liquid

control diet (Dyets, Inc., Bethlehem, PA). The rats were acclimated to the control diet for

three days. Then the rats were paired by similar weight and one rat from each pair

received an alcohol liquid diet containing 36% EtOH calories. To control for variations

of diet consumption and nutrition, the other rat was pair-fed the same amount of control

diet as his EtOH-fed mate consumed the day before. The rats consumed the indicated

diets for the last 5 weeks of concurrent smoke- and sham-exposure. This resulted in four

different treatment groups, which will be referred to as smoke-EtOHs, smoke-pairs,

sham-EtOHs, and sham-pairs. The timeline to generate rats used in this model is shown

in Figure 3.

39

Figure 3 - Rat Model of Chronic EtOH Ingestion and Smoke Exposure

36% EtOH diet ad lib Chow-feed liquid control diet (Smoke-expose) (smoke-expose) 0% EtOH pair-fed

6 weeks 3 days 5 weeks Infect

36% EtOH diet ad lib Chow-feed liquid control diet (Sham- (Sham-expose) (sham-expose) 0% EtOH pair-fed expose)

(Smoke- expose)

(Smokee-

expos ) (Smoke- expose)

B. Bacterial Strains

The serotype 3 S. pneumoniae (ATCC 6303, American Type Culture Collection,

Rockville, MD) was used for the majority of the experiments. This is a clinical isolate

that is highly encapsulated and virulent in humans and rats. In addition to S. pneumoniae

ATCC 6303, Staphylococcus aureus (ATCC 29213, American Type Culture Collection)

was used in a macrophage phagocytosis experiment. For the in vitro macrophage

function assays, an additional S. pneumoniae strain labeled as DW 3.8 was utilized. DW

3.8 is an unencapsulated serotype 3 mutant derived from a S. pneumoniae WU2 strain by

inserting transposon Tn916 into a genetic region responsible for polysaccharide capsule

formation [104].

All bacteria strains were stored at -80oC in Todd-Hewitt broth (Becton, Dickinson

and Company, Sparks, MD) containing 10% glycerol. For the colonization, bacterial

movement, vaccination, medication, mortality, opsonic deposition, PMN phagocytosis,

and macrophage studies, the pneumococcal strains were grown to stationary phase at

40

37oC in an atmosphere of 5% CO2 in air incubator in Todd-Hewitt broth containing 5%

heat-inactivated rabbit serum. For the PMN-mediated and non-PMN-mediated killing

assays, fifteen ml of a stationary phase culture was inoculated into 75 ml of fresh Todd-

Hewitt broth containing 5% heat-inactivated rabbit serum. The resulting culture then was

grown for 4 hours at 37oC in 5% CO2 in air to mid-log phase. For each experiment, the

pneumococci were collected by centrifugation at 13,776 x g for 10 minutes. The

resulting pellets were washed twice with phosphate-buffered saline (PBS) and diluted in

PBS to the appropriate optical density at 540 nm to achieve the desired inoculum. The S.

aureus strain was grown as a lawn on sheep blood agar plates (Remel, Lenexa, KS) for

16 hours at 37oC in 5% CO2 in air. The staphylococci were collected from the plates

using a sterile swab and suspended in sterile water to an optical density of 1.0 at 540 nm.

The number of colony forming units (cfu) present in each inoculum was confirmed by

serial dilution and viable counts on sheep blood agar plates.

C. Rat Sacrifice

In all experiments, the rats were euthanized by an intraperitoneal injection of 75

mg/kg body weight of pentobarbital (Nembutal, Abbott Laboratories, Abbott Park, IL).

Once the rats lost consciousness they were exsanguinated by cardiac puncture using a 21

ga. x ¾” scalp vein set (Excel, Los Angeles, CA).

D. Intranasal Infection

Immediately after their morning smoke exposure, the rats were lightly

anesthetized with isoflurane (Midrad, Inc., Bethlehem, PA) and held in an upright

41

position. The specified inoculum of type 3 S. pneumoniae ATCC 6303 in 100 μl PBS

was deposited by a dropwise injection into the rats’ nostrils through a micropipette tip

(Figure 4). Following intranasal inoculation, the rats were held in the vertical position for

a few seconds to allow for aspiration of the fluid.

Figure 4 – Intranasal Infection

Figure 4 For intranasal infections rats were lightly anesthetized and held in a vertical position. One hundred μl of the inoculum was then deposited by a dropwise injection into both nostrils.

E. Culture of Nasopharynx, Trachea, and Lungs

One week after an intranasal infection with 1 x 106 cfu of S. pneumoniae ATCC

6303, the rats were euthanized and nasal washes were performed to quantify

pneumococcal colonization. The trachea was exposed by blunt-end dissection and the

proximal end was occluded with surgical string. A 22 ga. catheter (Becton Dickinson and

Co., Sandy, UT) then was inserted into the trachea above the occlusion and a retrograde

injection of 1 ml PBS was collected from the nares into a sterile Petri dish. The number

42

of pneumococci present in the nasal wash fluids was determined by performing standard

plate counts on blood agar plates containing 4 μg/ml of gentamycin to eliminate

contaminating normal flora organisms [282]. The trachea and lungs then were removed

en bloc and placed in a sterile Petri dish. The trachea was excised from the lungs with

sterile scissors, and the proximal end (carina) and distal end were sampled separately by

inserting an ultra-fine cotton swab (Fisher Scientific, Pittsburgh, PA) 0.5 cm into the

opening. The cotton swab was vortexed for 5 sec in a tube containing 100 μl of sterile

PBS, and plate counts were performed on the resulting suspension. The lungs then were

homogenized in a sterile tissue grinder in a total volume of 10 ml of sterile PBS and the

total number of bacteria present in the lungs was quantified by plate counting.

F. Intranasal Vaccination with rPppA Unexposed chow-fed rats were immunized intranasally with recombinant

pneumococcal protective protein A (rPppA, Wyeth Vaccine Research, Pearl River, NY)

to test its ability to reduce pneumococcal colonization as shown previously in mice [64].

The rats were vaccinated under light isoflurane anesthesia with 50 μl of PBS containing

either 20 μg rPppA or 20 μg of the irrelevant antigen keyhole limpet hemocyanin

(Calbiochem, San Diego, CA). Each antigen was combined with 5 μg of native cholera

toxin subunit B adjuvant (Sigma, St. Louis, MO). The intranasal vaccines were

administered by a dropwise instillation through a micropipette tip placed at the opening

to the nares, resulting in inhalation of the proteins. The rats were immunized three times

at two week intervals. One week after the third immunization, the rats were anesthetized

with isoflurane and a cardiac puncture was performed to collect 1 ml of blood for

measuring serum antibody titers. While the rats were under anesthesia, noninvasive nasal

43

washes were performed to determine their mucosal antibody titers. To accomplish this, a

piece of 22 ga. polyethylene tubing connected to a 1cc syringe was inserted into one

nostril and 1 ml of PBS was injected and collected from the other nostril into a sterile

Petri dish. One week later (two weeks after their last vaccination), the rats were infected

intranasally as described above with 1 x 106 cfu of S. pneumoniae ATCC 6303. They

were then euthanized 1 week post-infection and pneumococcal colonization was

quantified as described above.

G. ELISA for rPppA Antibodies

Immunoglobulin G (IgG) and immunoglobulin A (IgA) antibody titers against

rPppA were measured in the serum and nasal washes from rats vaccinated with rPppA

using a previously described ELISA with some minor modifications [64]. For measuring

IgG, 96-well plates were coated overnight at 4oC with 100 μl/well of a 5 μg/ml solution

of rPppA in PBS. The plates were washed five times with PBS containing 0.1% Tween

20 and then blocked by the addition of 300 µl/well of 5% dry milk in PBS for one hour at

room temperature. After washing, 100 μl of rat IgG standards (0-320 ng/ml; Invitrogen,

Carlsbad, CA), or dilutions of the rats’ serum samples were added to each well and

incubated at room temperature for 1.5 hours. The plates were washed, 100 μl of

biotinylated anti-rat IgG (0.06 μg/ml, Jackson ImmunoResearch Laboratories, West

Grove, PA) was added to each well, and the plates were incubated at room temperature

for 1 hour. After washing, 100 μl of streptavidin-horseradish peroxidase (HRP) (0.1

μg/ml, Jackson ImmunoResearch Laboratories) was added to each well and incubated for

1 hour at room temperature. The plates were washed and 100 μl of 2,2'-Azinobis [3-

44

ethylbenzothiazoline-6-sulfonic acid]-diammonium salt (ABTS) substrate (Pierce,

Rockford, IL) was added to each well. The plates were incubated for 20 min and the

reaction was then stopped by the addition of 100 μl/well of 1% sodium dodecyl sulfate

(SDS) in sterile water. Color development was quantified at 405 nm. All samples were

tested in duplicate and the results were recorded as μg/ml of specific immunoglobulin. A

similar procedure was performed to measure IgA antibodies, in which biotinylated anti-

rat IgG was replaced with biotinylated anti-rat IgA (1 μg/ml, AbD Serotec, Raleigh, NC )

Because rat IgA standards were not available, the results were recorded as endpoint titers

consisting of the reciprocal of the highest serum dilution providing an absorbance value

of 0.1.

H. Salbutamol and Formoterol Medication For experiments to examine the efficacy of β-agonist medication to protect

against pneumococcal movement from the nasopharynx to the lungs, an apparatus was

developed to facilitate delivery of the medication to the rats (Figure 5). The rats were

restrained in DecapiCones (Braintree Scientific Corp, Braintree, MA) and placed into a

series of connected PVC pipes. A nebulizer (Super SportNeb 3050SS, Medical

Industries, Adel, IA) was used to aerosolize 5 ml of 0.5% salbutamol (Nephron

Pharmaceuticals Corporation, Orlando, FL) in PBS or 48 μg of formoterol (Foradil,

Shering Corporation, Kenilworth, NJ) dissolved in 5 ml of 0.01% acetic acid. For 20

min, one of the vaporized solutions was dispersed through tubes of identical length and

diameter inserted into each rat’s DecapiCone.

Twelve hours after rats were infected intranasally with 1 x 106 cfu of S.

pneumoniae ATCC 6303, they were medicated three times a day for 1 week with

45

salbutamol or twice daily for 1 week with formoterol. Control rats were medicated at the

same time intervals in an identical apparatus with nebulized vehicle solution alone. The

rats were rotated throughout the different chambers of the appropriate apparatus to

compensate for any slight variability in the amount of drug delivered to each chamber.

After 1 week of medication, the rats were euthanized and their nasopharynx, trachea, and

lungs were cultured as described above.

Figure 5 – Medication Device

Figure 5 A medication apparatus was used to expose the rats to salbutamol and formoterol. Rats were restrained in Decapicones and placed into the PVC pipes. A nebulizer aerolized the drug and delivered it to the rats via the plastic tubes.

I. Ciliary Beat Frequency Analysis

Ciliary beat frequency was quantified in the formoterol medication study by

computerized frequency analysis at Creighton University. Tracheal rings 1-2 mm thick

were excised from each animal’s trachea. The samples were maintained at 24 ± 0.5oC on

a thermostatically controlled heated stage and digitally analyzed using the Sisson-

46

Ammons Video Analysis (SAVA) system (Ammons Engineering, Mt. Morris, MI).

Whole field analysis software (Ammons Engineering) automatically analyzed the entire

captured image of all ciliated cells in a given field. All cilia amplitudes and frequencies

were collated, mapped to the screen image, and statistically analyzed to determine the

frequency average, standard deviation, and standard error of the entire image. The

frequency of the beating cilia was sampled in at least 6 different fields, and the data from

those fields was compiled to produce a mean ciliary beat frequency in Hertz (Hz) for

each rat.

J. Transtracheal Infections

Rats were infected transtracheally with type 3 S. pneumoniae ATCC 6303 for the

non-PMN-mediated killing, PMN-mediated killing, opsonic deposition, and phagocytosis

experiments, or with S. aureus ATCC 29213 for the macrophage phagocytosis study.

Under light anesthesia with isoflurane, the rats were laid on their backs on a surgery

table. The area over the trachea was cleaned with 70% ethanol and a small incision was

made through the skin with a scalpel. The trachea was exposed by blunt-end dissection,

and a 22 ga. catheter was inserted into the trachea toward the lungs. With the rat in a

vertical position, a 1cc syringe was used to deliver the specified incoculum of bacteria

suspended in 0.3 ml PBS into the catheter. This was followed by an injection of 0.1 ml

of air to simulate aspiration (Figure 6). The catheter was then removed and the incision

was closed with two metal clips.

47

Figure 6 – Transtracheal Infection

Figure 6 Transtracheal infections were performed on anesthetized rats by making a small incision on the neck and exposing the trachea by blunt-end dissection. A catheter then was inserted into the trachea. After placing the rat in a vertical position, the inoculum was injected into the catheter followed by an injection of air.

K. Lipopolysaccharide Instillation for PMN-mediated Assays

Five hours prior to transtracheal infection for the PMN killing and phagocytosis

assays, PMNs were recruited to the rats’ lungs by transtracheally instilling

lipopolysaccharide (LPS) from Escherichia coli O26:B6 (Sigma). The rats were

anesthetized with isoflurane and a 22 ga. catheter was inserted into the trachea as

described above. Twenty μg of LPS suspended in 0.2 ml PBS was instilled into each

rats’ lungs. The incision was then closed with two metal clips.

48

L. Non

t of two control rats sacrificed immediately after infection, using the following

equati

(Mean cfu’s at time 0 – cfu’s of test rats at 1hr) / Mean cfu’s at time 0 (x 100)

cedures

Ex vivo

-PMN-Mediated Bactericidal Assay

To measure pulmonary killing of S. pneumoniae by non-PMN-mediated defenses,

including the bactericidal activity of alveolar lining fluid and macrophages, our

laboratory developed an in vivo bactericidal assay. Rats were infected transtracheally

with 1 x 106 cfu of S. pneumoniae ATCC 6303 as described previously. At one hour

post-infection, the rats were euthanized and the trachea and lungs were removed en bloc.

After detaching the trachea, the lungs were homogenized in sterile tissue grinders in a

total volume of 10 ml of PBS. Plate counts were then performed on the homogenates to

quantify the number of viable organisms remaining in the lung tissue. The percentage of

pneumococcal killing was determined by comparing the lung counts of each test rat to the

mean coun

on:

M. Bronchoalveolar Lavage Pro

Bronchoalveolar Lavage

Bronchoalveolar lavage (BAL) was performed ex vivo to collect bacteria and

pulmonary cells from the rats’ lungs for the opsonin deposition and phagocytosis assays.

In this procedure, the rats were euthanized and their lungs were perfused with 30 ml of

ice cold PBS injected into the right ventricle of the heart and drained through a 16 ga.

needle inserted into the left ventricle for the removal of peripheral blood cells. Following

perfusion, the lungs and trachea were removed en bloc, and bacteria and pulmonary cells

were washed from the lungs as described previously [283]. Briefly, 10 ml aliquots of ice

49

cold Hanks Balanced Salt Solution (HBSS) without Mg++, Ca++, and phenol red

(Gibco/Invitrogen, Carlsbad, CA) were instilled into the lungs with a 10cc syringe

through a 22 ga. catheter inserted into the trachea (Figure 7). Following each instillation,

the lavage fluid was collected by dependent drainage until a total volume of 50 ml was

covered (Figure 8).

situ

re

Figure 7 – Lavage Figure 8 – Lavage Fluid Collection

Figures 7 and 8 Ex vivo bronchoalveolar lavage was performed by extracting the trachea and lungs en bloc. Ten ml of HBSS was injected through a catheter inserted into the trachea. The lavage fluid then was collected by dependent drainage.

In Bronchoalveolar Lavage

Bronchoalveolar lavage was performed in situ to collect BAL samples from

uninfected rats for the quantification of pulmonary lysozyme, lactoferrin, lactate

dehydrogenase (LDH), and opsonins. The rats were euthanized and the trachea was

exposed as described above. A single 10 ml aliquot of ice cold PBS was repeatedly

instilled and withdrawn 5 times via a 10cc syringe attached to a 22 ga. catheter inserted

50

into the trachea. This resulted in the collection of 7-8 ml of lavage fluid which was

centrifuged at 600 x g for 10 min at 4oC to remove cells and debris. The supernatant then

was sterile filtered using a 0.22 μm filter (Millipore, Billerica, MA) and the filtrate was

stored in 1 ml aliquots at -80oC until analyzed for individual antimicrobial and opsonic

protein levels as well as total protein concentration determined by the Quick Start

radford protein assay kit (Bio-Rad Laboratories, Hercules, CA).

N. Qu

lyzed in duplicate and results were recorded as ng of

ctoferrin/mg of total protein.

O. Qu

re run in duplicate and results were

g of total protein.

B

antification of Bactericidal Factors

Bactericidal factors were quantified in lavage samples collected by in situ

bronchoalveolar lavage as described above. Lysozyme activity in the lavage samples was

quantified using the commercially available EnzChek Lysozyme Assay Kit (Molecular

Probes, Eugene, OR). Samples were assayed in duplicate and results were recorded as

activity units/mg of total protein. Lactoferrin was measured by the commercially

available Bioxytech Lactof-EIA for human lactoferrin (Oxis International, Inc., Portland,

OR). Samples were again ana

la

antification of Pulmonary Cell Damage

Lactate dehydrogenase (LDH), a protein marker for cellular damage, was

measured in lavage fluid collected from uninfected rats as described above. The LDH-

Cytotoxicity Assay Kit from Biovision Research Products (Mountain View, CA) was

utilized to measure LDH levels. All samples we

recorded as milliunits of LDH/m

51

P. Opsonic Deposition Assay

Infection and Staining of Opsonins on Pneumococci

To quantify opsonization of S. pneumoniae, our laboratory developed an assay

that measures the in vivo deposition of three opsonins on the surface of pneumococci.

Rats were infected transtracheally with 1 x 107 cfu of live S. pneumoniae ATTC 6303.

After exactly 30 min, each rat was euthanized and ex vivo bronchoalveolar lavage was

performed as described above. The rats’ pulmonary cells were collected from the lavage

fluid by centrifugation at 450 x g for 30 min. The resulting supernatant was centrifuged

at 13,776 x g for 10 min to collect pneumococci recovered from the rats’ lungs. To

remove remaining rat pulmonary cells from the bacterial pellet, the cells were lysed by

adding 10 ml of distilled water followed 10 seconds later by the addition of an equal

volume

ria

en were washed twice again and fixed in 1% formalin for flow cytometric analysis.

of double-strength PBS.

The final bacterial pellet from each rat’s lungs was washed once in PBS and

separated into three 50 μl aliquots. Each aliquot was incubated at 37oC for 30 min with 1

ml of PBS containing 10 μg/ml of biotinylated rabbit anti-rat C3 antibody (Immunology

Consultants Laboratory, Newberg, OR), rabbit anti-rat CRP antibody (Immunology

Consultants Laboratory), or mouse anti-rat SP-D antibody (BMA Biomedicals, Augst,

Switzerland). After the bacteria were washed twice, each aliquot was incubated at room

temperature for 10 min with 1 ml of 1 μg/ml of a streptavidin-conjugated

allophycocyanin (APC) fluorochrome (BD Pharmingen, San Diego, CA). The bacte

th

52

Flow Cytometric Analysis

Pneumococci isolated from the rats’ lungs and labeled with antibodies to the

various opsonins were analyzed using a FACSAria flow cytometer (Becton Dickinson,

San Jose, CA) to quantify the percentage of pneumococci with C3, CRP, or SP-D bound

to their surface. On each day of experimentation, a forward and side scatter plot of

unlabeled pneumococci grown in culture was used to set the analysis gate for

pneumococci recovered from the rats’ lungs (Figure 9). The gated bacteria then were

analyzed in the fluorescent APC channel at 660 nm to quantify C3, CRP, and SP-D

the surface of pneumococci (Figure 10). binding to

Figure 10 – Analysis of

APC Fluorescence Figure 9 – Scatter Plot of

S. pneumoniae

Forw

ard

Scat

ter

% o

f Max

Side Scatter APC

Figures 9 and 10 Bacteria were gated based on forward and side scatter in the flow cytometer. The gated bacteria then were analyzed for fluorescence to quantify opsonic protein deposition on the surface

53

Q. C3,

esults from all three ELISAs were

corded as μg of the opsonin/mg of total protein.

R. PM

CRP, and SP-D ELISAs

C3 and CRP were quantified in lung lavage samples with commercially available

rat C3 and rat CRP ELISA kits (Immunology Consultants Laboratory). SP-D in the

lavage samples was quantified by a self-developed capture ELISA. Briefly, 96-well

plates were coated overnight at room temperature with 100 μl/well of 0.5 μg/ml of anti-

rat SP-D antibody (Hycult Biotechnology, Uden, Netherlands). The plates were washed

4 times with 0.05% Tween 20 in PBS and then blocked for 1 hour at room temperature

with 200 μl/well of 1% bovine serum albumin in PBS. After the plates were washed, 100

μl/well of rat SP-D standard (0-400 ng/ml, Hycult Biotechnology) and duplicate samples

of lavage fluid diluted 1:400 in 0.05% Tween 20 with 0.1% bovine serum albumin in

PBS were added, and the plates were incubated at room temperature for 2 hours. After

washing, 100 μl/well of biotinylated anti-rat SP-D antibody (0.1 μg/ml, BMA

Biomedicals) was added and the plates were incubated for 30 min at room temperature.

The plates then were washed, 100 μl of streptavidin-HRP (Pierce) was added to each

well, and they were incubated for 30 min at room temperature. After washing, 100 μl of

tetramethylbenzidine (TMB) substrate was added to each well. After 10-20 min of

incubation at room temperature, the reaction was stopped by the addition of 100 μl/well

of 2N H2SO4 and the plates were read at 450 nm. R

re

N-Mediated Bactericidal Assay

Similar to the non-PMN-mediated bactericidal assay, this assay measures the

pulmonary killing of pneumococci in vivo after the pre-recruitment of PMNs. Five hours

54

after LPS-induced PMN recruitment, the rats were infected transtracheally with 1 x 106

cfu of S. pneumoniae ATCC 6303. Exactly one hour post-infection, the rats were

euthanized and their lungs were lavaged once in situ with 10 ml HBSS. One ml of the

collected lavage fluid was used to prepare cytospin slides (Cytospin 2, Shandon, Chesire,

UK) to determine the percentage of PMNs recruited to the rats’ lungs. Each slide was

stained with Protocol Hema-3 (Fisher Diagnostics, Middletown, VA) and two hundred

cells from each sample were counted manually in a Nikon Labopho-2 light microscope

(Melville, NY). The lungs then were removed and homogenized in a total volume of 10

ml of PBS including the remaining lavage fluid. Plate counts were performed on the

homogenates and the percentage of bacterial killing was determined as described above

r the non-PMN-mediated killing assay.

S. Ch

homogenates and serum using commercially available kits from R&D Systems

fo

emokine Analysis

Chemokine levels were measured by ELISAs in serum and lung homogenates

from each of the rats utilized in the PMN-mediated bactericidal assay. At the time of

euthanasia, blood collected from each animal by cardiac puncture was placed in a serum

separator tube. The blood was centrifuged at 1900 x g for 15 min and the serum was

frozen at -80oC in 1 ml aliquots until analyzed. After completion of the serial dilutions

for plate counts, the crude lung homogenates were centrifuged at 13,776 x g for 15 min.

The supernatants then were sterile filtered through a 0.22 μm filter to remove any

contaminating pneumococci and 1 ml aliquots were frozen at -80oC. MIP-2 levels were

measured in the lung homogenates and CINC-1 levels were measured in the lung

55

(Minneapolis, MN). All samples were analyzed in duplicate, and results were recorded

as pg of cytokine/ml of serum or homogenate.

T. PMN Phagocytosis Assay

Bacteria Staining and Infection

To quantify the phagocytic activity of recruited PMNs, a phagocytosis assay was

developed to measure the uptake of S. pneumoniae by PMNs within the rats’ lungs [171].

This assay utilized live S. pneumoniae ATCC 6303 fluorescently stained with 5-(and 6-)

carboxyfluorescein diacetate succinimidyl ester (CFDA/SE, Molecular Probes).

CFDA/SE is a non-fluorescent diacetate form of CFSE that easily passes through the

bacterial cell wall. Once inside the organism, the acetate groups are cleaved off by

cytosolic esterases and the molecule is converted to the fluorescent form of CFSE.

Pneumococci were stained by incubating them at 37oC for 30 min in the dark with 2 μM

solution of CFDA/SE. After washing twice with PBS to remove excess dye, the bacteria

were resuspended in PBS to an optical density of 1.0. The CFSE-labeled organisms

fluoresced brightly in the flow cytometer as demonstrated in Figure 11.

56

Figure 11 – CFSE-Labeled Pneumococci

Figure 11 Pneumococci were labeled with CFSE in order to quantify PMN phagocytosis by flow cytomtery. Bacteria stained with CFSE fluoresced brightly compared to unstained bacteria.

PMNs were pre-recruited to the rats’ lungs by LPS instillation as described above.

Exactly 5 hrs later, the rats were infected transtracheally with 1 x 108 cfu of CFSE-

labeled pneumococci. Exactly 15 min post-infection, each rat was euthanized and ex vivo

bronchoalveolar lavage was performed as described above. Pulmonary cells collected

from the lavage fluid were washed once with HBSS using differential centrifugation (180

x g for 10 min) to remove unassociated bacteria. Contaminating red blood cells then

were lysed by the addition of 10 ml of deionized water followed 10 seconds later by an

equal volume of double-strength PBS. The remaining cells were centrifuged and

resuspended in PBS containing 4% heat-inactivated fetal calf serum (PBS-FCS). The

57

cells were counted in a hemacytometer and the cell suspension was adjusted to a final

concentration of 2 x 107 cells/ml for antibody staining.

Antibody Staining of Pulmonary Cells

Pulmonary cells from each rat’s lavage sample were stained in 96-well U-bottom

microplates with fluorochrome-tagged antibodies for flow cytometric analysis. Fifty μl

of the cell suspension were incubated for 30 min in the dark on ice with 50 μl of a

monoclonal antibody cocktail containing 0.25 μg of phycoerythrin (PE)-conjugated anti-

rat granulocyte antibody RP-1 (BD Pharmingen) and 2 μg of the biotinylated anti-rat

monocyte antibody 1C7 (BD Pharmingen). The cells then were centrifuged at 180 x g for

4 min and washed twice with PBS-FCS. One-hundred μl of PBS-FCS containing 1 μg of

streptavidin-APC was added and the cells were incubated for an additional 5 min in the

dark on ice. After two more washes in PBS-FCS, the cells were resuspended in PBS

containing 1% formalin for flow cytometric analysis. Negative control samples for cell

staining consisted of unstained cells from each rat and cells stained with streptavidin-

APC only.

Flow Cytometric Analysis

Three-color flow cytometric analysis was performed on cells from each rat using

a FACSAria flow cytometer with dual laser excitation (488 nm and 633 nm). For each

day the assay was run, a control rat infected with non-fluorescent pneumococci was

included to exclude autofluorescence in the CFSE channel of the RP-1 positive PMNs.

Based on scatter characteristics that included neutrophils and macrophages, a minimum

58

of 10,000 cells were analyzed in the PE (575 nm channel) for PMNs and the APC (660

nm channel) for macrophages. The percentage of RP-1 positive cells fluorescing more

bright in the CFSE (530 nm) channel than the PMNs from the control rat was used to

determine the percentage of PMNs with associated bacteria (Figure 12). The frequency

of RP-1 positive PMNs and the percentage of PMNs containing associated fluorescent

bacteria were determined for each sample using FlowJo Software (Tree Star, Ashland,

OR). To determine the relative number of fluorescent pneumococci engulfed by each

PMN, a phagocytic index was calculated by multiplying the percentage of PMNs that

contained fluorescent pneumococci by their mean fluorescent intensity.

Figure 12 – Determination of PMN Phagocytosis

Test Rat Infected with CFSE-Labeled Bacteria

Control Rat Infected with Unlabeled Bacteria

Figures 12 To determine bacterial uptake gated PMNs from test rats were analyzed for CFSE fluorescence and compared to PMNs from a control rat infected with unlabeled bacteria.

59

U. Macrophage Phagocytosis Assay

Bacterial Staining and Infection

To evaluate the ability of macrophages to phagocytose bacteria within the lung,

an assay was adapted from the PMN phagocytosis assay to measure the in vivo uptake of

bacteria by macrophages within the lungs. This assay used fluorescently-labeled S.

pneumoniae ATCC 6303. To override the problem of macrophage autofluorescence, S.

pneumoniae was labeled with the far-red fluorochrome APC-Cy7 (BD Pharmingen)

which fluoresced brighter than the autofluorescence of the macrophages (Figure 13). The

pneumococci were incubated for 30 min at room temperature with 0.5% rabbit antiserum

to type 3 pneumococcal polysaccharide (Statens Seruminstitut, Copenhagen, Denmark).

After the bacteria were washed twice with PBS, they were incubated for 30 min at room

temperature with 1 μg/ml of a biotinylated anti-rabbit immunoglobulin antibody (BD

Pharmingen). After washing, the pneumococci then were incubated for an additional 10

min at room temperature with 1 μg/ml of a streptavidin-APC-Cy7 conjugate, washed

again, and resuspended in PBS to an optical density of 1.0.

60

Figure 13 – APC-Cy7-labeled S. pneumoniae

% o

f Max

APC-Cy7

Macs

S. pneumoniae

Figure 13 APC-Cy7 caused bacteria to fluoresce brighter than macrophages.

Rats were infected transtracheally with 1 x 108 cfu of S. pneumoniae. At exactly

15 min post-infection, each rat was euthanized and an ex vivo bronchoalveolar lavage was

performed. Pulmonary cells were collected and washed as described above. Cells then

were resuspended in PBS with 0.5% bovine serum albumin (PBS-BSA) and the

macrophages were separated from PMNs by magnetic cell sorting as described below.

Magnetic Cell Sorting

Macrophages could not be labeled with the biotinylated 1C7 antibody due to the

possibility of streptavidin-APC binding to pneumococci since they also were labeled with

a biotinylated antibody. As an alternative, macrophages were separated from

contaminating PMNs by magnetic cell sorting [48]. For 15 min, cells were incubated on

ice with 1 ml of PBS-BSA containing 4 μg of biotinylated anti-rat granulocyte antibody

61

(22262D, BD Pharmingen). The cells were centrifuged at 180 x g for 6 min and washed

twice with PBS-BSA. The final cell pellets were resuspended in 90 μl of PBS-BSA, to

which were added 10 μl of streptavidin-coated MACS super-paramagnetic microbead

suspension (Miltenyi Biotec Inc., Sunnyvale, CA). After incubation for an additional 15

min on ice, the cells were washed and resuspended in 0.5 ml of PBS-BSA. The cell

suspension then was loaded onto a MiniMACS separation column suspended in a

magnetic field according to the manufacturer’s directions (Miltenyi Biotec Inc.).

Unlabeled macrophages that traveled through the column were collected, washed, and

resuspended in PBS containing 1% formalin for flow cytometric analysis. This method

resulted in > 95% purity.

Flow Cytometric Analysis

Similar to the PMN phagocytosis assay, the FACSAria flow cytometer was used

to analyze the fluorescence of macrophages from each rat. A control rat infected with

non-fluorescent bacteria was included each day the assay was performed to determine the

extent of the macrophage autofluorescence in the APC-Cy7 channel detecting the

bacteria. Macrophages isolated by magnetic cell sorting were gated based on forward

and side scatter and analyzed in the APC-Cy7 (780 nm) channel for phagocytosis of

pneumococci. The percentage of macrophages fluorescing more bright in the APC-Cy7

channel than macrophages from the control rat infected with unlabeled bacteria was

determined as the percentage of macrophages with associated bacteria (Figure 14).

FlowJo Software was used to analyze the flow cytometric results, and the phagocytic

index was calculated for each sample as described for the PMN phagocytosis assay.

62

Figure 14 – Determination of Macrophage Phagocytosis

Bacteria Fluorescence

Test Rat Infected with APC-Cy7-labeled Bacteria

Control Rat Infected with Unlabeled Bacteria

Figure 14 To determine macrophage phagocytosis, macrophages gated by forward and side scatter were analyzed for bacterial fluorescence and compared to macrophages from a control rat infected with unlabeled bacteria.

Bacteria Fluorescence

Adaptation of Phagocytosis Assay

The macrophage phagocytosis assay was repeated using S. aureus ATCC 29213.

S. aureus was labeled with the bright green DNA dye Syto 9 (Molecular Probes) which

also fluoresced brighter than the autofluorescent macrophages (Figure 15). S. aureus was

stained by incubating at room temperature for 15 min in the dark with 10 nM Syto 9 in

sterile water. The bacteria were washed twice to remove excess dye and resuspended in

sterile water to an optical density of 1.0. The rats were infected, euthanized, and their

pulmonary cells were collected as described above. The cells from each rat were

resuspended in PBS-FCS, stained with RP-1 and 1C7 antibodies, and analyzed in the PE

(PMNs) and APC (macrophages) channels as described in the PMN phagocytosis assay.

The APC positive cells were then analyzed in the Syto 9 (530 nm) channel for uptake of

63

staphylococci and the percentage of macrophages with associated bacteria was

determined as described in the previous section.

Figure 15 – Syto 9-labeled S. aureus

Macs S. aureus

% o

f Max

Syto 9 Figure 15 Syto 9 caused bacteria to fluoresce brighter than macrophages.

Microscopic Analysis of Macrophages

Cytospin slides prepared from lavage samples from each rat infected with S.

aureus were stained as described above and examined in a light microscope. Two

hundred cells from each sample were counted to verify the percentage of macrophages

determined by flow cytometry to have phagocytosed staphylococci.

V. Macrophage Function Assays

Three assays were performed to measure oxidative burst, degranulation, and

chemokine release by macrophages stimulated in vitro with either S. pneumoniae or

phorbol myristate acetate (PMA). Macrophages were harvested from uninfected rats’

64

lungs by ex vivo bronchoalveolar lavage as described above. After washing the

pulmonary cells and lysing any contaminating erythrocytes, macrophages from each rat

were counted in a hemacytometer.

To measure oxidative burst, a chemiluminescence assay was performed. One x

105 macrophages seeded in each well of a 96-well plate were incubated at room

temperature in 200 μl of HBSS (Gibco/Invitrogen) containing Mg++, Ca++, 5 mM

glucose, and 50 μM Luminol salt (Sigma). The macrophages in individual wells were

exposed to either 5 x 106 cfu of DW 3.8 S. pneumoniae preopsonized with normal rat

serum or 1 μg/ml phorbol myristate acetate (PMA) (Sigma) plus 0.5 μg/ml ionomycin

(Sigma). Chemiluminescence emitted from the oxidized Luminol was measured using a

Victor3 1420 Multilabel Counter (Perkin Elmer, Waltham, MA). The plate was read at

time zero and every 10 min up to 1 hour. Each sample was run in duplicate and the

results were recorded as light units and compared to unstimulated cells.

To measure degranulation and chemokine release, 5 x 105 macrophages in 1 ml of

complete RPMI media (Gibco/Invitrogen) supplemented with 50 μM 2-mercaptoethanol

were aliquoted into wells of a 24-well cell culture plate. The plate was incubated for 1

hour at 37oC to allow macrophage adherence. After removing the supernatant, 1 ml of

fresh RPMI media was added to each well that contained 0.5 μg/ml recombinant rat IFN-

γ (eBioscience, San Diego, CA) and either 2.5 x 107 cfu of preopsonized DW 3.8 S.

pneumoniae or 1 μg/ml PMA plus 0.5 μg/ml ionomycin. The cells were incubated at

37oC for 1 hour, after which 250 μl of supernatant was collected to measure lysozyme.

The remaining supernatant was collected after 5 hours for chemokine analysis. All

65

samples were stored at -80oC until analyzed for lysozyme, MIP-2, and CINC-1 using

commercially available kits as described above.

W. Intranasal Mortality Study

A mortality study was conducted following an intranasal infection with 1 x 108 of

S. pneumoniae ATCC 6303. Mortality was assessed for 9 days post-infection.

Temperature transponders (BioMedic Data Systems, Seaford, DE) were implanted

subcutaneously in the top of the rats’ heads at the time of infection. Temperatures then

were recorded twice daily and the rats were observed for abnormal movement, ruffling of

the fur, and/or bleeding from the eyes or nose. On days 2 and 5 post-infection, 50 μl of

blood was collected by aseptic puncture of the rats’ foot veins [284] and standard plate

counts were performed for quantification of bacteremia. If a rat’s temperature dropped

below 35oC, it was euthanized and was counted as having succumbed to the infection.

X. Statistical Analysis

The mortality data were analyzed using Fisher’s Exact test. All other data were

tested for normality and equal variance before statistical tests were performed. To

compare smoke-exposed vs. sham-exposed rats or vaccinated/medicated vs. control

groups in the vaccination and medication trials, the Students t-test was utilized for normal

data or the Mann-Witney Rank Sum Test for non-normal data. Paired t-tests were used to

compare EtOH-fed rats to their pair-fed controls within the same exposure group. Two

Way ANOVA was utilized with the Holm-Sidak method for pair-wise comparisons to

determine differences among exposure, diet, and the combination of the two factors. In

66

the PppA vaccination trial, Spearman’s rank order correlation was used to correlate anti-

PppA antibody levels and the log cfu recovered from each rat’s nasal passages.

Spearman’s correlation test also was used in the formoterol medication study to correlate

ciliary beat frequency with the number of organisms recovered from the lung

homogenates of each rat. For all statistical tests, a p-value <0.05 was considered

significant.

67

V. RESULTS

A. Pneumococcal Colonization and Movement from the Nasopharynx to the Lungs After 7 weeks of smoke or sham exposure plus 5 weeks of concurrent exposure

and pair-feeding, 6-8 rats per group were infected intranasally with S. pneumoniae ATCC

6303. One week post-infection, the rats were euthanized and the numbers of

pneumococci were quantified in their nasopharynx, trachea, carina, and lung tissue. The

plate count data from rats in this study are summarized in Table 1. All means of the log

include rats that were negative for pneumococci. There were no smoke- and/or EtOH-

induced alterations in colonization, since 100% of the rats remained colonized in the

nasopharynx with similar numbers of organisms one week after infection. EtOH

ingestion alone resulted in slightly more pneumococci remaining in the rats’ lungs. A

larger percentage of sham-EtOH rats had positive carina and lung cultures and with a

higher number of pneumococci than their sham-pair counterparts. Smoke exposure alone

greatly reduced the number of pneumococci remaining in the rats’ lungs, even in the

presence of EtOH ingestion. Regardless of diet, the percentage of smoke-exposed rats

with pneumococci in their tracheas and carinas was significantly lower than the sham-

exposed rats (p = 0.003 and 0.021, respectively). Smoke-exposed rats also had

significantly fewer organisms in their tracheas and carinas (p < 0.001 and p = 0.009,

respectively), as well as fewer organisms in their lungs compared to the sham-exposed

animals (p = 0.05).

68

Table 1 – Smoke Exposure Reduces Pneumococcal Movement

Smoke-EtOH n=7

Smoke-Pair n=8

Sham-EtOH n=6

Sham-Pair n=8

% Positive

Log cfu/ml

% Positive

Log cfu/ml

% Positive

Log cfu/ml

% Positive

Log cfu/ml

Nasal Wash 100 4.5 100 4.3 100 4.1 100 4.0

Trachea 29 0.8 25 0.9 83 2.6 88 3.5

Carina 14 0.6 13 0.2 67 2.2 50 1.8

Lungs 14 0.7 13 0.5 67 1.8 38 1.2

B. PppA Vaccination Trial

To determine if antibodies to PppA could reduce pneumococcal colonization of

the nasopharynx, a preliminary vaccination trial was conducted in which unexposed,

chow-fed rats were immunized intranasally with rPppA. Two weeks after their third

vaccination, the rats were infected intranasally with type 3 S. pneumoniae. At one week

post-infection, the rats were euthanized and pneumococcal colonization of the

nasopharynx was quantified. Figure 16 depicts the log cfu from nasal washes of

vaccinated and control rats. Intranasal immunizations with rPppA failed to reduce

pneumococcal colonization of the nasopharynx, as all of the rats remained colonized 1

week after infection. Although there were two vaccinated rats that had a 2-3 log lower

cfu count in their nasal washes, the mean log cfu of the vaccinated group was similar to

that of the control group.

The rPppA ELISAs showed the vaccinated rats had a higher concentration of anti-

PppA IgG antibodies in their serum and a higher anti-PppA IgA antibody titer in their

nasal washes compared to the control rats (Table 2). However, these heightened antibody

69

levels were not effective in reducing nasopharyngeal colonization since there were no

strong inverse correlations between anti-PppA IgG or anti-PppA IgA levels and the log

cfu recovered from the nasopharynx (-0.212 and -0.266, respectively).

Figure 16 – PppA Vaccination Fails to Reduce Colonization

Vaccinated Control0

1

2

3

4

5

Log

cfu/

ml N

asal

Was

h

Table 2 – rPppA Specific IgG and IgA Antibody Levels Compared to Log cfu of Nasal Washes

Serum IgG

(μg/ml) Mucosal IgA

titer Log cfu/ml nasal wash

Vaccinated Rats 182 5 3.2 Control Rats 30 <2 3.8

Figure 16 Pneumococcal colonization of the nasopharynx was quantified by performing plate counts on retrograde nasal washes from vaccinated and control rats. n=5-6

70

C. Salbutamol Medication Trial

To determine if salbutamol, a short-acting β2-agonist that increases ciliary

beating, could hinder pneumococcal movement into the lungs after an intranasal

infection, unexposed chow-fed rats were again used in a preliminary medication trial.

Twelve hours after the rats were infected intranasally with S. pneumoniae, they were

medicated three times a day for one week with salbutamol or PBS. At the end of

medication, the rats were euthanized and their nasopharynx, trachea, carina, and lungs

were cultured to quantify the number of pneumococci. Figure 17 below depicts the log

cfu at the various locations of each rat.

Figure 17 – Salbutamol Medication Decreases Pneumococcal Movement in Chow-fed Rats

0

1

2

3

4

5

Nasal Wash Trachea Carina Lungs

ControlSalbutamol

Log

cfu

Figure 17 Animals were medicated with salbutamol or PBS three times a day for one week after an intranasal infection with S. pneumoniae. Pneumococcal colonization and movement then were quantified by plate counts of nasal washes, tracheas, carinas, and lung homogenates. n=4-5

71

Medication with aerosolized salbutamol somewhat reduced pneumococcal

movement from the nasopharynx into the lower respiratory tract. As expected,

salbutamol exposure did not alter colonization, since all the animals had similar numbers

of pneumococci in their nasal washes. However, the majority of the medicated rats had

no organisms in their tracheas, and the one medicated rat with pneumococci in its trachea

had a 1.5 log lower cfu count than the positive control rats. No organisms were detected

in the carina and lungs of the medicated group, whereas one control animal remained

positive for pneumococci in those two samples.

D. Formoterol Medication

The longer-acting β2-agonist formoterol also was pretested for its ability to reduce

pneumococcal movement from the nasopharynx to the lungs of unexposed chow-fed rats.

Starting 12 hours after being infected intranasally with S. pneumoniae, the rats were

medicated with formoterol or vehicle solution alone every 12 hours for 1 week. The rats

then were euthanized and pneumococcal colonization and movement were quantified.

The log cfu from each sample for the medicated and control rats are shown in Figure 18

below.

72

Figure 18 – Formoterol Medication Reduces Pneumococcal Movement into the Lungs of Chow-fed Rats

0

1

2

3

4

5

6

Nasal Wash Trachea Carina Lungs

ControlFormoterol

Log

cfu

Figure 18 Log cfu of nasal washes, tracheas, carinas, and lung homogenates from rats medicated twice daily for one week with formoterol or vehicle solution alone after an intranasal infection with S. pneumoniae. n=5

As shown before, all the animals in this study remained colonized with a similar

number of organisms one week after infection followed by medication. Formoterol was

more effective than salbutamol in reducing pneumococcal movement into the lower

respiratory tract. All except one medicated rat were negative for pneumococci in their

trachea, carina, and lungs, while 4 of the 5 control rats had pneumococci in all three

cultures.

Due to the success of formoterol in this trial, it was chosen to test further in the

chronic EtOH ingestion and smoke exposure model. Following 12 weeks of smoke- or

sham-exposure, with the last five weeks including concurrent pair-feeding, rats were

infected and medicated with formoterol twice daily for one week as described above.

73

During their medication, the rats continued to be smoke- or sham-exposed and fed their

liquid diets. At the end of medication, the rats were euthanized and pneumococci were

quantified throughout their respiratory tracts. Figure 19 displays the log cfu of nasal

washes, trachea, carina, and lungs for the individual rats in each treatment and medication

group.

Figure 19 – Formoterol Medication Prevents Pneumococcal Movement in Sham-EtOH Rats

B. Trachea A. Nasal Wash

0

1

2

3

4

5

6

EtOH + + - - + + - -Formoterol + - + - + - + -

Sham Smoke

Log

cfu

0

1

2

3

4

5

6

EtOH + + - - + + - -Formoterol + - + - + - + -

Sham Smoke

Log

cfu

0

1

2

3

4

5

6

EtOH + + - - + + - -Formoterol + - + - + - + -

Sham Smoke

Log

cfu

C. Carina D. Lungs

0

1

2

3

4

5

6

EtOH + + - - + + - -Formoterol + - + - + - + -

Sham Smoke

Log

cfu

Figure 19 Rats were infected intranasally with S. pneumoniae and medicated twice daily for one week with formoterol or vehicle solution. Plate counts then were performed on nasal washes (A), tracheas (B), carinas (C), and lung homogenates (D). n=5-8

74

As seen with salbutamol, formoterol medication failed to alter pneumococcal

colonization of the nasopharynx in any of the treatment groups. Unexpectedly,

pneumococcal numbers in the lungs was not reduced by formoterol in the sham-pair

group as it was in chow-fed animals. The percentages of medicated and unmedicated

sham-pair rats positive for pneumococci in their trachea, carina, and lungs were alike

along with similar cfu counts. The only group in which formoterol medication appeared

to be successful in preventing pneumococcal movement into the lower respiratory tract

was the sham-EtOHs. Pneumococci were undetected in either the carina or the lungs of

each of the medicated sham-EtOH rats, whereas 66% of the unmedicated rats in that

same treatment group had positive carina and lung cultures. Interestingly, smoke

exposure itself increased the ability of the rats to maintain sterility in their lungs, but

formoterol had no effect on pneumococcal movement in smoke-exposed rats. Regardless

of medication and diet, the majority of the smoke-exposed rats had no detectable

organisms in their lower respiratory tract.

In addition to tracking pneumococcal movement into the lungs, baseline ciliary

beat frequency (CBF) was analyzed in rats from each medicated and treatment group.

Following euthanasia, tracheal rings from each rat were excised and transported to

Creighton University to be analyzed by the SAVA system. Smoke exposure significantly

augmented the CBF (Figure 20). This was true for all smoke-exposed vs. sham-exposed

rats regardless of diet and formoterol medication.

75

Figure 20 – Smoke Exposure Increases CBF Regardless of Diet and Formoterol Medication

Medicated Unmedicated All Animals0

5

10

15

SmokeSham

p < 0.001p = 0.009 p < 0.001C

BF

(Hz)

± SE

M

Figure 20 Baseline CBF of smoke-exposed vs. sham-exposed rats for formoterol medicated (n=10-14), unmedicated (n=13-15), and all animals combined (n=25-27).

There were no statistical differences in CBF between medicated and unmedicated

rats within any of the treatment groups except for the sham-EtOHs, in which formoterol

medication significantly increased baseline CBF (Figure 21). There also was a strong

inverse relationship between CBF and the log cfu recovered from the lung homogenates

of sham-EtOH rats, with a correlation coefficient of -0.642 (p = 0.029).

76

Figure 21 – Formoterol Augments CBF in Sham-EtOH Rats

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

5

10

15

FormoterolUnmedicated

p = 0.005C

BF

(Hz)

± SE

M

Figure 21 Baseline CBF of tracheal ring explants was analyzed by the SAVA system. Measurements were recorded on a thermostatically controlled microscope. n=5-8

E. Non-PMN-Mediated Killing To investigate the individual and combined effects of chronic EtOH ingestion and

smoke exposure on pre-PMN recruitment defenses in the lungs, an in vivo bactericidal

assay was conducted. At exactly one hour after rats were infected transtracheally with S.

pneumoniae, they were euthanized and their lungs homogenized. Plate counts were

performed on the lung homogenates to determine the percentage of bacteria killed within

each rat’s lungs (Figure 22). Five weeks of EtOH ingestion alone significantly

suppressed bacterial killing, such that the mean percentage of killing for the sham-EtOHs

was significantly lower than that for the sham-pairs (5% vs. 31%, p = 0.001). Smoke

exposure alone had no effect on early pre-PMN killing (26% vs. 31% for sham-pairs, p =

0.65). Unexpectedly, however, the addition of concurrent smoke exposure to EtOH

77

ingestion significantly increased, rather than decreased, this non-PMN-mediated

pulmonary killing (23% vs. 5% for sham-EtOHs, p = 0.016). These results indicate that

smoke exposure may actually negate rather than exacerbate EtOH-induced defects on

non-PMN-mediated pulmonary host defenses.

Figure 22 – Concurrent Smoke Exposure Negates Chronic EtOH-Induced Impairment of Non-PMN-Mediated Killing

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

10

20

30

40p = 0.001

p = 0.016

Perc

enta

ge o

f Kill

ing±

SEM

Figure 22 An in vivo bactericidal assay was performed to determine the effect of smoke exposure ± EtOH ingestion on non-PMN-mediated killing. Rats were infected transtracheally with live pneumococci. The lungs were removed and homogenized at one hour post-infection. Plate counts were performed to determine the percentage of pneumococci killed within the lungs during the one hour experimental infection. n=10-11

F. Quantification of Bactericidal Factors

Two key bactericidal proteins were quantified to determine whether the

modulations on non-PMN-mediated killing were due to alterations in the rats’ pulmonary

levels of those bactericidal factors. Lysozyme activity in lavage fluid collected from

uninfected rats from each treatment group is shown in Figure 23. Although smoke

78

exposure alone slightly increased this activity, there were no significant differences

among the mean levels of the four groups.

Figure 23 – No Alterations of Pulmonary Lysozyme Levels by Smoke Exposure and EtOH Ingestion

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

5000

10000

15000

20000Ly

sozy

me

(U/m

g pr

otei

n)±

SEM

Figure 23 Lysozyme concentrations were quantified in lavage fluid from uninfected rats. n=7

The second bactericidal protein examined in lavage fluid was lactoferrin.

Surprisingly, the animals receiving EtOH alone had significantly higher concentrations of

lactoferrin in their lungs compared to sham-pairs (p < 0.001) as shown in Figure 24.

Smoke exposure alone had no effect, but concurrent smoke exposure resulted in the loss

of this EtOH-induced increase in lactoferrin (p < 0.001).

79

Figure 24 – EtOH Ingestion Drastically Increases Pulmonary Lactoferrin Levels

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

20

40

60

80

100p < 0.001 p < 0.001

Lact

ofer

rin (n

g/m

g pr

otei

n)±

SEM

Figure 24 Quantification of lactoferrin in lavage fluid from uninfected rats. n=7-8

To determine whether EtOH ingestion alone so dramatically increased lactoferrin

levels due to release from intracellular pools, cellular damage was assessed in the lungs

by quantifying lactate dehydrogenase (LDH) in the lavage fluid (Figure 25). This was

not the case, as the results from the LDH assay were inversely related to the lavage fluid

lactoferrin levels. The mean concentration of LDH was significantly lower, rather than

higher, for the sham-EtOHs vs. the sham-pairs (p = 0.02) and the smoke-EtOHs (p =

0.004).

80

Figure 25 – Cellular Release of LDH Does Not Explain the EtOH-Induced Increase in Lactoferrin

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

50

100

150

200

p = 0.02

p = 0.004

LDH

(mU

/mg

prot

ein)

± SE

M

Figure 25 Quantification of LDH in lavage fluid from uninfected rats. n=7-8

G. Macrophage Phagocytosis

To evaluate the ability of macrophages to phagocytose pneumococci in the lungs

after chronic EtOH ingestion and smoke exposure, a novel flow cytometric assay was

developed to measure the in vivo uptake of bacteria by alveolar macrophages. In this

assay, macrophages collected from the rats’ lungs 15 minutes after infection with

fluorescently labeled pneumococci were analyzed by flow cytometry for uptake of the

fluorescent bacteria. Macrophages from smoke-exposed animals in both feeding groups

had a greatly increased fluorescent intensity than macrophages from sham-exposed

animals (Figure 26). However, this was shown to be due to a smoke-induced increase in

macrophage autofluorescence since the fluorescent intensity was similar to that of

macrophages collected from an uninfected smoke-exposed rat.

81

Figure 26 – Smoke Exposure Enhances Macrophage Autofluorescence

Bacterial Fluorescence

Forw

ard

Scat

ter

Infected Smoke-exposed Rat

Infected Sham-exposed Rat

Figure 26 Flow cytometry was used to compare the MFI of macrophages from a sham- and smoke-exposed rat infected with fluorescent pneumococci and an uninfected smoke-exposed rat.

Uninfected Smoke-exposed Rat

Although this shift in fluorescent intensity precluded use of the assay in smoke-

exposed rats, data from the sham-exposed animals proved interesting. Consistent with

the literature, few macrophages were positive for fluorescent bacteria in either group of

sham-exposed animals (Figure 27A). However, macrophages from the sham-EtOH rats

phagocytosed significantly fewer pneumococci than their pair-fed counterparts, as

determined by a significant increase in their mean fluorescent intensity (MFI) (Figure

27B; p < 0.001) and their phagocytic index (Figure 27C; p = 0.02).

82

Figure 27 – EtOH Ingestion Suppresses Macrophage Phagocytosis

Sham-EtOH Sham-Pair0

1

2

3

4

5

Perc

enta

ge o

f Pha

gocy

tosi

ngM

acro

phag

es±

SEM

Sham-EtOH Sham-Pair0

250

500

750

1000

1250

1500p < 0.001

Fluo

resc

ent I

nten

sity

± SE

MSham-EtOH Sham-Pair

0

1000

2000

3000

4000

5000

6000p = 0.02

Phag

ocyt

ic In

dex±

SEM

A B C

Figure 27 A phagocytosis assay was performed to determine the effect of smoke exposure and EtOH ingestion on macrophage uptake of pneumococci in vivo. Rats were infected with fluorescent pneumococci. Fifteen minutes after infection BAL was performed. Flow cytometry was used to determine the percentage of macrophages that had phagocytosed pneumococci (A) and the MFI (B). Phagocytic index was calculated by multiplying the percentage of macrophages containing bacteria by their MFI (C). n=8

To confirm that EtOH ingestion suppresses macrophage phagocytosis of a Gram-

positive bacterium more readily taken up by macrophages, the assay was repeated in

EtOH-fed and pair-fed sham-exposed rats using fluorescently labeled S. aureus. Results

of that assay indicated that EtOH ingestion significantly increased rather than reduced

macrophage phagocytosis (Figure 28). However, when the percentage of macrophages

containing bacteria was determined by manual counts of cytospin slides from each animal

by two independent observers, the means for the two feeding groups were similar.

Furthermore, the flow cytometry data did not correlate with the values determined by

light microscopy for both the sham-EtOH (-0.086, p = 0.92) and sham-pair (0.029, p =

1.0) rats (Table 3).

83

Figure 28 – Flow Cytometry Results Indicate EtOH Ingestion Increases Macrophage Phagocytosis of S. aureus

Figure 28 To confirm EtOH ingestion decreases macrophage phagocytosis, the phagocytosis assay was repeated in sham-exposed rats infected with fluorescent staphylococci. n=7-8

Sham-EtOH Sham-Pair0

5

10

15

20

25

30

35p = 0.031

Perc

enta

ge o

f Pha

gocy

tosi

ngM

acro

phag

es±

SEM

Table 3 – Comparison of Macrophage Phagocytosis Values

Rat Flow

Cytometry Manual Counts Rat

Flow Cytometry

Manual Counts

1 53 4 1 3 22

2 3 20 2 5 11 3 6 15 3 29 13

4 22 16 4 9 16 5 50 28 5 18 32

6 39 36 6 7 14 Avg. 24 20 Avg. 7 18

Percentages of Macrophages from Sham-EtOHs Containing S. aureus

Percentages of Macrophages from Sham-Pairs Containing S. aureus

84

H. Additional Macrophage Functions

Macrophages isolated from rats in the four treatment groups then were stimulated

with either unencapsulated S. pneumoniae or phorbol myristate acetate (PMA) to

determine the effects of smoke and/or EtOH on several of their functions. A

chemiluminescence assay was performed to quantify oxidative burst. Regardless of

smoke or sham exposure, macrophages from EtOH-fed rats produced more oxygen

radicals than pair-fed rats at both 10 and 20 minutes after being stimulated by

pneumococci (Figure 29). The differences in oxidative burst between all EtOH-fed and

all pair-fed animals at 10 and 20 minutes after stimulation were statistically significant (p

= 0.023 and p = 0.032, respectively). After 20 minutes, the oxidative burst continued to

rise at a similar rate and then plateaued at 40-50 minutes in all treatment groups except

the smoke-pairs. Smoke exposure alone suppressed the oxidative response such that

smoke-pairs produced the lowest amounts of reactive oxygen species from 30 to 60

minutes after stimulation. PMA stimulated a more rapid and robust oxidative burst than

did pneumococci in macrophages from all groups of rats. Disregarding the effect of diet,

smoke exposure hindered the oxidative response by PMA-stimulated cells, such that

macrophages from smoke-exposed rats produced fewer oxygen radicals than those from

sham-exposed rats for all time points (Figure 30).

85

Figure 29 – Smoke Exposure Alone Suppresses Oxidative Burst

in Macrophages Stimulated by Pneumococci

0 10 20 30 40 50 600

50

100

150

200

250

300

350 Smoke-EtOH

Sham-Pair

Smoke-PairSham-EtOH

Time (min.)

Che

milu

min

esce

nce

(ligh

t uni

ts)±

SEM

Figure 29 A chemiluminescence assay was utilized to identify the effects of smoke exposure and EtOH ingestion on oxidative burst in macrophages. Macrophages incubated with unencapsulated pneumococci were analyzed in a luminometer every 10 minutes up to one hour. n=5-8

Figure 30 – Smoke Exposure Hinders Oxidative Burst in PMA-Stimulated Macrophages

0 10 20 30 40 50 600

100200300400500600700800 Smoke

Sham

Time (min.)

Che

milu

min

esce

nce

(ligh

t uni

ts)±

SEM

p = 0.039

p = 0.038p = 0.048

p = 0.042 p = 0.023

Figure 30 The same chemiluminescence assay was used to measure oxidative burst in macrophages stimulated with PMA. Macrophages from smoke-exposed animals produced significantly fewer oxygen radicals than from sham-exposed animals. n=12-15

86

Degranulation was measured by quantifying lysozyme release one hour after

macrophage stimulation. When pneumococci were used as the stimulus, there were no

statistical differences in lysozyme release among the four treatment groups, even though

the lysozyme level was twice as high for sham-EtOH rats (Figure 31). PMA stimulation

produced no detectable levels of lysozyme release by alveolar macrophages from any of

the rats (data not shown).

Figure 31 – Neither EtOH nor Smoke Significantly Alters Degranulation by Macrophages Stimulated with Pneumococci

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

1020304050607080

Lyso

zym

e (U

/ml)±

SEM

Figure 31 Lysozyme was quantified in media from macrophages stimulated in vitro with pneumococci for one hour. n=6-7

Macrophage chemokine release was evaluated by measuring CINC-1 and MIP-2

levels in the media five hours after stimulation (Table 4). When stimulated by

pneumococci, macrophages from both smoke- and sham-exposed rats that consumed the

EtOH diet released less of both chemokines than their pair-fed counterparts. Smoke

exposure made no difference in CINC-1 and MIP-2 levels when comparing smoke-pairs

and sham-pairs.

87

When macrophages were stimulated with PMA, EtOH ingestion alone resulted in

the release of significantly higher amounts of CINC-1 (p = 0.04). However, even though

smoke exposure alone did not affect CINC-1 production, the addition of smoke to EtOH

ingestion reversed the EtOH effect. The smoke-EtOH rats therefore released CINC-1

levels comparable to those of the control group receiving neither smoke nor EtOH.

Neither EtOH ingestion nor smoke exposure alone significantly modified macrophage

release of MIP-2, but when administered in combination, they resulted in a >2-fold

reduction in MIP-2 release than that for any of the other treatment groups.

Table 4 – Chemokine Production by Macrophages Stimulated with Pneumococci or PMA

Pneumococci PMA CINC-1 pg/ml

± SEMMIP-2 pg/ml

± SEMCINC-1 pg/ml

± SEMMIP-2 pg/ml

± SEMSmoke-EtOH 39 ± 20 44 ± 16 13 ± 6 57 ± 15

Smoke-Pair 119 ± 61 173 ± 89 26 ± 12 132 ± 46 Sham-EtOH 19 ± 18 12 ± 12 52 ± 16a,b 141 ± 60

Sham-Pair 92 ± 91 186 ± 186 18 ± 6 135 ± 52 n=4-8 aSham-EtOH vs. Sham-pair, p = 0.04. bSham-EtOH vs. Smoke-EtOH, p = 0.017.

I. Opsonic Deposition Assay

An in vivo assay was used to quantify the deposition of the three major opsonic

proteins on the surface of S. pneumoniae to determine if opsonization of intrapulmonary

bacteria is modified by chronic EtOH ingestion and/or smoke exposure. Pneumococci

recovered from the rats’ lungs were labeled with fluorochrome-tagged antibodies specific

for rat complement C3, CRP, or SP-D, and analyzed by flow cytometry. The results in

88

Figure 32 show that in the absence of smoke exposure, significantly fewer pneumococci

were coated with either C3 or SP-D within the lungs of rats consuming EtOH than in

their pair-fed controls (*p = 0.017 and **p = 0.01, respectively). CRP binding also was

somewhat lower in sham-EtOHs than sham-pairs, but the difference did not reach

statistical significance (p = 0.09). Smoke exposure did not alter opsonization when

administered alone, and it did not exacerbate the EtOH-induced decrease in opsonin

deposition, such that there were no differences in C3, CRP, or SP-D binding between

EtOH-fed and pair-fed rats that were smoke-exposed.

Figure 32 – EtOH Ingestion Reduces C3 and SP-D Deposition on Bacteria

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

10

20

30

40CRPSP-D

C3

*

*

**

**

Perc

enta

ge o

f Ops

oniz

edPn

eum

ococ

ci±

SEM

Figure 32 Mean percentage of bacteria with bound C3, CRP, or SP-D isolated from the lungs of rats. Values were quantified by transtracheally infecting rats and lavaging the organisms from their lungs 30 minutes later. Flow cytometry then was used to determine the percent of bacteria bound by each opsonin. C3 and SP-D values for sham-EtOH rats significantly lower than that for sham-pair rats (*p = 0.017 and **p = 0.01, respectively). n=7-8

89

To some extent the results of the opsonin deposition assay correlated well with

levels of the same three opsonic proteins measured by ELISAs in lavage fluid of

uninfected rats from each treatment group. Pulmonary C3 levels were significantly

decreased in EtOH-fed compared to pair-fed rats, whether or not they had been exposed

to smoke (Figure 33; *p = 0.026 and **p = 0.004). There was no significant EtOH-

induced decrease in the levels of SP-D. CRP levels were similar for all of the treatment

groups.

Figure 33 – EtOH Ingestion Decreases C3 Basal Levels

Smoke EtOH Smoke Pair Sham EtOH Sham Pair0

2

4

6

8

CRPSP-D

200

400

600

800 C3

**

**

**

Ops

onic

Pro

tein

Lev

els±

SEM

( μg/

mg

prot

ein)

Figure 33 Opsonic protein levels were quantified in lavage samples from

uninfected rats by ELISAs. C3 values for both EtOH-fed groups were significantly lower than their pair-fed counterparts (*p = 0.026 for smoke-exposed; **p = 0.004 for sham-exposed). n=6-8

90

J. PMN-Mediated Killing

The PMN-mediated killing assay was conducted to confirm that chronic, like

acute EtOH ingestion with or without concurrent smoke exposure impairs PMN killing

ability. At exactly five hours after PMNs were recruited to the rats’ lungs by LPS

instillation plus one hour after the rats were infected transtracheally with S. pneumoniae,

they were euthanized and plate counts of their lung homogenates were compared to those

of control rats sacrificed immediately after infection. This was done to determine the

percentage of bacteria killed when PMNs were present in each rat’s lungs (Figure 34).

Chronic EtOH ingestion dramatically impaired this PMN-mediated pneumococcal killing

(p = 0.01), such that the majority of sham-EtOHs (6 of 7 rats) had pneumococcal growth

in their lungs shown as negative killing. Smoke exposure did not exacerbate, but rather

abolished this detrimental effect of EtOH. Smoke-EtOHs killed significantly more

pneumococci in their lungs than sham-EtOHs (p = 0.026), making their PMN bactericidal

activity similar to that of rats that had not been exposed to either insult. Based on manual

counts of cytospin slides, neither smoke exposure nor EtOH ingestion significantly

altered PMN recruitment to the lungs, as all four rat groups averaged 85-90% of the

pulmonary cells in their bronchoalveolar lavage as PMNs.

91

Figure 34 – Concurrent Smoke Exposure Negates EtOH-Induced Defect in PMN-Mediated Killing

Smoke EtOH Smoke Pair Sham EtOH Sham Pair-40-30-20-10

010203040 p = 0.026 p = 0.01

Perc

enta

ge o

f Kill

ing

Figure 34 To determine the effect of smoke exposure and EtOH ingestion on PMN killing ability, an in vivo bactericidal assay was performed after LPS-induced PMN recruitment. Rats were infected transtracheally with pneumococci and the percentage of bacterial killing was determined one hour later by performing plate counts on lung homgenates. n=7-8

K. Pulmonary and Systemic Chemokine Levels

Pulmonary chemokine levels were measured by ELISA in the lung homogenates

from rats used in the PMN-mediated bactericidal assay (Table 5). Serum CINC-1, but

not MIP-2, values also were measured from these same rats because CINC-1, as opposed

to MIP-2, leaves the lung tissue where it is produced and enters into the systemic

circulation [55]. All four treatment groups had similar concentrations of MIP-2 and

CINC-1 in their lung homogenates. EtOH ingestion in the absence of smoke exposure

slightly decreased CINC-1 serum levels, but the decrease was not statistically significant

(p = 0.25). Once again, the EtOH-induced decrease was not seen when the rats were also

92

smoke-exposed even though smoke exposure alone did not increase serum CINC-1 levels

in pair-fed rats.

Table 5 – Chemokine Values from Lung Homogenates and Serum

Pulmonary MIP-2 (pg/ml)

Pulmonary CINC-1 (pg/ml)

Serum CINC-1 (pg/ml)

Smoke-EtOH 4309 6219 1558 Smoke-Pair 3813 5509 1551

Sham-EtOH 4788 5993 944 Sham-Pair 4155 6176 1334

L. PMN Phagocytosis

To determine if smoke- and/or EtOH-induced modifications in PMN phagocytosis

are responsible for the differences in intrapulmonary killing in the presence of PMNs, a

phagocytosis assay was performed to quantify the in vivo uptake of S. pneumoniae by

PMNs pre-recruited into the rats’ lungs for five hours. Rats infected with CFSE-labeled

pneumococci were sacrificed exactly 15 minutes later and PMNs in their bronchoalveolar

lavage fluid were identified with fluorescent antibodies and analyzed by flow cytometry.

The percentage of phagocytosing PMNs was unchanged by EtOH-ingestion and/or smoke

exposure (Figure 35A). However, EtOH ingestion without smoke exposure significantly

decreased the relative number of organisms being ingested by PMNs as indicated by the

decreased MFI in Figure 35B. This EtOH-induced decrease did not affect the phagocytic

index for this was not statistically different compared to the sham-pairs (Figure 35C). No

EtOH effect was detected among the smoke-exposed animals, and there were no

93

significant differences in either the MFI or the phagocytic index for the smoke-exposed

rats vs. their sham-exposed counterparts.

Figure 35 – Neither EtOH Ingestion nor Smoke Exposure Affects PMN Phagocytic Activity

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

10

20

30

40

50

60

70

80

% o

f Pha

gocy

tosi

ng P

MN

SEM

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

1000

2000

3000p = 0.004

Fluo

resc

ent

Inte

nsity

± S

EM

Smoke-EtOH Smoke-Pair Sham-EtOH Sham-Pair0

50000

100000

150000

200000

Phag

ocyt

ic In

dex±

SEM

C

A B

Figure 35 A PMN phagocytosis assay was performed to determine the effect of smoke exposure and EtOH ingestion on PMN uptake of pneumococci. After LPS-induced PMN recruitment rats were infected with fluorescent pneumococci. BAL was then performed on each rat at 15 minutes post-infection. Flow cytometry was used to determine the percentage of PMNs that had phagocytosed labeled bacteria (A) and the MFI (B). The phagocytic index was calculated by multiplying the percentage of PMNs containing bacteria by their MFI (C). n=6-8

M. Mortality Study An intranasal mortality study was conducted on 8 rats from each treatment group.

Each rat was infected intranasally with a lethal dose of S. pneumoniae and then

94

monitored for 9 days. Bacteremia was quantified by plate counts on each rats’ blood

sample taken on days 2 and 5 post-infection. Table 6 shows the bacteremia and mortality

results for each treatment group.

Table 6 – Bacteremia and Mortality Results from an Intranasal Challenge

Bacteremia Day 2 Bacteremia Day 5 Mortality Smoke-EtOH 1/8 (13%) 0/4 (0%) 4/8 (50%)

Smoke-Pair 0/8 (0%) 0/8 (0%) 0/8 (0%) Sham-EtOH 2/8 (25%) 0/6 (0%) 4/8 (50%)

Sham-Pair 3/8 (38%) 1/5 (20%) 4/8 (50%)

All rats that developed bacteremia eventually succumbed to the infection. All but

one of them that had bacteremia on day 2 died from the infection before blood samples

were taken again on day 5, and no other rats developed positive blood cultures on day 5.

Bacteremia was not detected in any of the surviving rats, but it was also undetected on

days 2 and 5 in half of the rats that died. Sham-exposed rats consuming the EtOH diet

had a similar bacteremia rate as their pair-fed controls on day 2 of infection. Only one of

the 16 smoke-exposed rats had organisms in its blood on day 2. The mortality rate was

50% for all treatment groups except for the smoke-pairs. Unexpectedly, this group never

developed bacteremia and all the rats survived the intranasal challenge.

95

VI. DISCUSSION

A. Pneumococcal Colonization and Movement Studies

The pneumococcal infection process begins with adherence of the bacteria to

epithelial cells of the nasopharynx. One potential reason smokers and alcohol abusers

have a higher incidence of developing pneumococcal pneumonia is an increase in

colonization. Smoking promotes pneumococcal binding to buccal epithelial cells and

reduces mucosal IgA levels, resulting in a higher risk of being colonized by S.

pneumoniae [219-223]. No studies have elucidated the effects of alcohol on

nasopharyngeal colonization by pneumococci, but alcohol ingestion may further increase

this occurrence. It was shown by flow cytometric analysis that mice ingesting EtOH for

two weeks had reduced numbers of CD4+ T-cells in the spleen and thymus [183] and this

was linked to EtOH inducing apoptosis by culturing murine thymocytes in the presence

of EtOH [184]. A decrease in CD4+ T-cells may enhance pneumococcal colonization of

the nasopharynx for it was recently reported that mice deficient in CD4+ T-cells had

decreased immunity to pneumococcal colonization when challenged intranasally [70].

We therefore hypothesized that smoke exposure would increase pneumococcal

colonization and chronic EtOH ingestion would exacerbate this problem.

When pneumococci were quantified in the nasal washes of rats infected

intranasally, neither smoke exposure nor EtOH ingestion increased nasopharyngeal

colonization. These results did not coincide with in vitro studies that found increased

bacterial adherence to buccal epithelial cells from smokers and cultured cells exposed to

cigarette smoke extract [219]. These results also do not correlate with several other

96

studies that detected higher rates of pneumococcal colonization in the nasopharynx of

smokers vs. nonsmokers [216-218]. The human studies are different from our current

experiment where they only determined the percentage of nasopharyngeal cultures that

were positive or negative for pneumococci. These studies did not quantify colonization

in human subjects at a certain time after inhaling S. pneumoniae. The inoculum used to

infect the rats was too concentrated to detect any smoke- and EtOH-induced increases in

colonization because all of the sham-pair control rats remained colonized. The amount of

pneumococci the rats inhaled is more than what people are normally exposed to in the

natural environment.

Despite not detecting any smoke- or EtOH-induced differences in colonization, it

was important to evaluate potential therapies to reduce nasal carriage of pneumococci

thereby reducing pneumococcal access to the lungs of all patients. Therapies to protect

adults from pneumococcal colonization are nonexistent, and the heptavalent vaccine for

children is limited by only reducing carriage rates for seven of the >90 different serotypes

[100-102]. Several pneumococcal proteins, including pneumococcal surface proteins A

and C (PspA and PspC), have been tested as potential vaccine antigens. Mice immunized

with PspA or PspC were partially protected against pneumococcal colonization, leading

to reduced development of pneumonia and bacteremia [62,63,282,285]. However, an

effective pneumococcal protein vaccine must provide protection against numerous

serotypes. The structure of PspA is highly variable among different pneumococcal

strains [113], and PspC is found on only 75% of pneumococci [118]. Therefore, like the

capsular polysaccharide vaccines, immunizing with these alternative protein targets

elicits limited protection.

97

A newly recognized surface-exposed pneumococcal protein called pneumococcal

protective protein A (PppA) may have great potential as a vaccine candidate. Intranasal

vaccination with recombinant PppA (rPppA) induced both local and systemic IgG and

IgA anti-PppA antibody responses [64]. Dr. Bruce Green, who first discovered the

protein, showed PppA is conserved among many different pneumococcal serotypes, and

immunizing mice intranasally with the rPppA reduced nasopharyngeal carriage of a

number of different strains [64]. In light of the success of Green’s experiment, we

obtained rPppA from him, predicting that intranasal immunization with the protein would

reduce pneumococcal colonization in our rats. Before the rPppA vaccine was used in

smoke- and EtOH-exposed rats, however, it was evaluated in unexposed control rats.

After three immunizations, pneumococcal colonization was measured after an intranasal

infection. Unlike what occurred in the mouse study, the rPppA vaccine failed to

significantly reduce pneumococcal colonization in rats. This could be explained by the

fact that although the majority of vaccinated rats had higher IgG and IgA titers than

unvaccinated rats, these antibody responses were not nearly as robust as what Green

reported in vaccinated mice [64]. Dr. Green suggested this might be due to instability of

the rPppA protein (personal communication). We intended to repeat the study using

freshly prepared rPppA, but Dr. Green has decided not to purify and pursue the protein

further.

Once colonization has been established, pneumococcal movement from the

nasopharynx to the lungs is required for development of pneumonia. Alcohol and

smoking both cause defects in mucociliary clearance, allowing organisms to enter the

sterile environment of the lungs. Alcohol consumption blunts the ciliary response,

98

decreases the gag reflex, and increases the risk of aspiration [136,143,144,286]. Smoking

is also harmful to mucociliary clearance by damaging ciliated cells and down-regulating

the CBF [224-227]. We therefore hypothesized both insults would aid in pneumococcal

invasion of the lungs.

To determine if chronic EtOH ingestion and/or smoke exposure increased

pneumococcal movement into our rats’ lungs, S. pneumoniae was tracked from the

nasopharynx to the lungs after an intranasal inoculation. EtOH ingestion alone slightly

increased pneumococcal movement to the lungs. This is consistent with a previous study

that quantified pneumococcal movement into the lungs of chronic EtOH-fed rats four

hours after an intranasal infection [145].

Contradictory to those previous findings and to our hypothesis, smoke exposure

alone appeared to inhibit rather than exacerbate pneumococcal entry into the lower

respiratory tract. This is evidenced by fewer positive cultures of the trachea, carina, and

lungs as well as lower mean numbers of organisms at each location in smoke-exposed

rats. Smoke exposure also reversed any EtOH-induced defect present in the sham-

exposed group, for EtOH-fed and pair-fed rats exposed to smoke had nearly identical

counts. The reason fewer pneumococci were seen in the airways and lungs of smoke-

exposed animals is smoke exposure dramatically increased their CBF regardless of EtOH

ingestion.

The results from the smoke-exposed animals are quite interesting, considering

that smoke exposure has been reported to decrease CBF through the activation of PKC

[224]. However, previous research in our laboratory suggested that smoke-exposed rats

had a higher baseline CBF than sham-exposed rats [145]. Yet this same experiment also

99

showed that concurrent smoke exposure exacerbated the effects of EtOH, causing

increased pneumococcal movement to the lungs four hours after an intranasal infection

[145]. The increased movement by both insults was not detected in the present

experiment at one week post-infection. The different concentrations of bacteria used to

infect the rats and the length of time the experiments were conducted after infection

might account for these conflicting results. The inoculum from the previous study was

two logs higher than our inoculum and bacteria in the lungs at four hours after infection

may be due to drainage from the nasopharynx. Whereas at one week post-infection this

initial drainage is no longer present and differences in pneumococcal infection of the

lungs may be due to alterations in pulmonary killing as demonstrated.

Two therapies to reduce the number of colonized pneumococci from entering the

lungs were tested. Salbutamol and formoterol are β2-agonists currently used as

bronchodilators in patients who suffer from asthma or COPD. β2-agonists are also

known to stimulate ciliary beating by activating PKA. We hypothesized that β2-agonist

medication would enhance mucociliary clearance in all treatment groups and perhaps

negate the EtOH-induced increase in pneumococcal movement into the lungs.

Salbutamol and formoterol were tested first in normal control rats infected intranasally

and medicated for one week by inhalation. Both β2-agonists were efficient in reducing

pneumococcal movement into the lower respiratory tract. Formoterol was chosen to treat

smoke- and EtOH-exposed rats, since it is a longer-acting β2-agonist that required less

frequent administration and it is now more commonly used in patients than the shorter-

acting salbutamol.

100

As hypothesized, formoterol prevented pneumococcal penetration into the lungs

of all EtOH-ingesting rats, but it did not alter the bacterial counts in smoke-exposed

animals. The majority of smoke-exposed rats had little bacterial movement through their

airways whether or not they received the β2-agonist. This is similar to the results from

the initial movement study discussed earlier. Interestingly, the beneficial effect of

formoterol in the normal control rats was not seen in the sham-pair animals. Stress

induced by pair-feeding may have interfered with the response to the medication. To

verify if this is true, further testing should be done in rats consuming unlimited quantities

of the liquid control diet.

Formoterol medication significantly increased the CBF in the sham-EtOH rats,

which correlates with the decreased pneumococcal movement to their lungs. However, a

formoterol up-regulation of CBF in EtOH-fed animals contradicts a previous study that

used the same rat model and showed chronic EtOH exposure desensitizes ciliated cells,

preventing their stimulation by the β2-agonist isoproterenol [145]. This difference may

be due to the previous study being conducted in vitro rather than in vivo. In the previous

study, ciliated cells were stimulated once in vitro and then the change in CBF was

analyzed whereas our current study exposed the ciliated cells to formoterol for one week

in vivo and then measured the baseline CBF.

B. Non-PMN Pulmonary Defenses

Before the arrival of recruited PMNs, a significant number of pneumococci that

reach the lungs are rapidly killed by various pulmonary defenses. Prominent among

these defenses are the alveolar lining fluid and resident alveolar macrophages. Alveolar

101

lining fluid is known to exhibit bactericidal properties consisting of lysozyme, lactoferrin,

defensins, and surfactant. This fluid also contains several opsonic proteins including

complement C3, CRP, and SP-D that coat the bacteria and promote phagocytosis.

Alveolar macrophages coordinate the inflammatory response by releasing cytokines and

chemokines to recruit and activate PMNs. Although not nearly as efficient as PMNs,

alveolar macrophages also can phagocytose pneumococci and eliminate the pathogen

through their production of an oxidative burst and degradative enzymes. Numerous

studies have shown that these various host defenses are affected by chronic EtOH or

smoke exposure. However, no in vivo studies have been performed previously to

determine the combined effects of smoke and EtOH on these pulmonary defenses in

relation to pneumococcal clearance from the lungs.

We hypothesized that both smoke and EtOH would impair non-PMN-mediated

killing, and that both insults together would decrease this host defense even further.

Using an in vivo bactericidal assay we demonstrated that half of the sham-EtOH rats had

bacterial growth in their lungs after one hour of a transtracheal infection. This severe

EtOH-induced decrease in pneumococcal clearance from the lungs corroborates previous

studies that showed EtOH inhibits the bactericidal activities of lysozyme and surfactant

[33,149]. In addition, it supports in vitro studies showing EtOH suppresses macrophage

phagocytosis and oxidative burst [164,165,168].

Contrary to our hypothesis, concomitant exposure of the rats to cigarette smoke

negated, rather than exacerbated, the EtOH-induced defect in pulmonary pneumococcal

killing. Smoke-exposed EtOH-fed rats killed similar numbers of pneumococci in their

lungs as their smoke-exposed pair-fed controls, and this activity was no different than

102

that in the sham-pair controls. Human smokers are known to have higher levels of

lysozyme, lactoferrin, and defensins in their lungs than nonsmokers [228-230], and this

could potentially contribute to the counteractive effect of smoke exposure on the EtOH-

induced defect in bactericidal activity within the lungs. However, smoke hinders

surfactant production [234] and free fatty acids in surfactant are thought to be its most

potent bactericidal factors against pneumococci [33]. Even though the effects of smoke

on macrophages is still being debated, some studies have shown smoke enhances

macrophage activity [146,245]. If so, the beneficial effects of smoke on macrophages

may out-weigh its negative effects on other pulmonary defenses and, therefore, overcome

the EtOH-induced defect on non-PMN-mediated killing.

Due to these initial results on the effects of EtOH ingestion and smoke exposure

on non-PMN-mediated killing, we had to reformulate our hypothesis to be: the

detrimental effects of chronic EtOH ingestion on innate pulmonary anti-pneumococcal

defenses are reduced by concurrent smoke exposure. To elucidate the reason for this

phenomenon, we next analyzed bactericidal factors, alveolar macrophage activity, and

opsonic proteins in smoke-exposed ± EtOH-fed rats. Lysozyme and lactoferrin, the two

most abundant pulmonary bactericidal proteins, were quantified in the airways of

uninfected animals. Although acetaldehyde from metabolized EtOH has been shown to

partially inhibit lysozyme activity [149] whereas smoking increases lysozyme

concentrations in the lungs [228,229], no differences in lysozyme activity were detected

among the four rat treatment groups. Lactoferrin concentrations however, were

dramatically increased in sham-exposed rats consuming the EtOH diet. As seen for the

non-PMN-mediated killing assay, concurrent smoke exposure negated this EtOH effect

103

since smoke-EtOH rats had similar concentrations of lactoferrin in their lungs as their

smoke-pair counterparts and the sham-pair controls. One caveat for this assay is the

lactoferrin kit was not able to quantify apolactoferrin separately from lactoferrin, making

it difficult to relate this data to the pneumococcal killing assay. Another limitation for

this kit is it quantifies human lactoferrin, but all the rat samples were analyzed using this

same kit. Unfortunately, no lactoferrin kits that measure rat lactoferrin exist.

To determine if EtOH ingestion causes damage to pulmonary epithelial cells,

resulting in their spontaneous release of lactoferrin, LDH levels were measured. LDH is

an intracellular enzyme found in most cells and is commonly used as a marker for

cellular damage. Surprisingly, the LDH results were actually the reverse of those from

the lactoferrin data, in that sham-EtOH rats had significantly lower rather than higher

levels of LDH than either the sham-pair or smoke-EtOH rats. While intriguing, these

results suggest that cellular damage was not responsible for the EtOH-induced increase in

lactoferrin levels. EtOH ingestion is known to interfere with iron homeostasis, leading to

increased iron concentrations that could explain the rise in lactoferrin [150,151]. The

increased availability of free iron could decrease apolactoferrin levels and therefore

reduce the bactericidal activity of this anti-pneumococcal protein in the lungs.

Furthermore, altered iron homeostasis also may contribute to the bacterial growth that

occurred in the EtOH-fed rats’ lungs. More studies need to be performed to quantify

apolactoferrin and iron concentrations in the lungs to elucidate this interesting

phenomenon. Although the results of our assays of pulmonary levels of lysozyme or

lactoferrin could not fully explain the differences in non-PMN-mediated pneumococcal

104

killing among the treatment groups, it is possible there are alterations of other bactericidal

factors such as defensins and surfactant fatty acids by smoke and alcohol.

Alveolar macrophages are the first immune cells to encounter S. pneumoniae

when it invades the lungs. In vitro studies found chronic EtOH ingestion and smoke

exposure decrease macrophage phagocytosis separately [168,246,247], although another

study reported that smoke has no effect at all on this macrophage activity [248].

Nonetheless, no studies have reported the effects of smoke and alcohol on macrophage

phagocytosis in vivo. Based on the results from the non-PMN-mediated killing assay, we

hypothesized that EtOH decreases macrophage phagocytosis and that smoke exposure

up-regulates this activity by activating the macrophages, thereby helping to negate the

EtOH defect. To measure the EtOH- and smoke-induced effects on macrophage

phagocytosis, we developed a novel assay to quantify in vivo bacterial uptake by alveolar

macrophages.

Within the sham-exposed rat groups, very little phagocytosis of pneumococci was

detected, agreeing with the scientific literature that macrophages have difficulty taking up

serotype 3 S. pneumoniae due to their heavy polysaccharide capsule. Although little

phagocytosis occurred, EtOH ingestion significantly hindered this macrophage function,

as evidenced by both a low number of bacterial organisms within each macrophage and a

low overall phagocytic index in the sham-EtOH rats. Suppression of macrophage

phagocytosis in the EtOH-fed rats no doubt contributes to the decreased non-PMN-

mediated pneumococcal killing within their lungs.

Macrophage phagocytosis in the smoke-exposed animals as measured by flow

cytometry was 16x higher than that in the sham-exposed animals and much higher than

105

we expected. Smoke exposure greatly increased the percentage of cells fluorescing

brightly in the bacteria channel to an average of 80%. However, based on the

fluorescence of macrophages from an uninfected smoke-exposed rat, this increase was

shown to be due to enhanced autofluorescence rather than actual bacterial uptake.

Alveolar macrophages from human smokers and smoke-exposed rats also have been

shown in the literature to emit augmented autofluorescence [287,288]. This result

precluded us from using our assay to accurately measure the effects of smoke exposure

on macrophage phagocytosis in the rats’ lungs. Additional experiments are needed to

overcome this problem, either by labeling the bacteria with an even brighter fluorescent

stain or quenching the macrophage autofluorescence without affecting their uptake or

quenching the fluorescence of the bacteria.

Because macrophage phagocytosis of pneumococci was so limited in the sham-

exposed rats, we elected to further confirm this EtOH-induced defect in sham-EtOH and

sham-pair rats using fluorescently labeled S. aureus, that are phagocytosed much more

efficiently than pneumococci. S. aureus was labeled with Syto 9 which is an

exceptionally bright green DNA stain that causes the bacteria to fluoresce brighter than

the autofluorescent macrophages. APC-Cy7 was replaced with Syto 9 because it does not

require a specific biotinylated antibody to effectively label bacteria, and we were unable

to find such an antibody that would bind to our staphylococcal strain.

The percentage of macrophages phagocytosing S. aureus was much higher in both

feeding groups than what was determined in the previous experiment for S. pneumoniae.

Contradicting our previous results, however, the flow cytometric results showed that

chronic EtOH ingestion increased instead of decreased macrophage phagocytosis of

106

staphylococci. This was not confirmed by the manual counts of cytospin slides, as they

showed no difference in phagocytosis between the two feeding groups. Flow cytometry

results from more than half of the rats in each group did not correlate with the manual

counts by light microscopy. It is not completely understood why these results occurred.

Macrophages exhibit the highest autofluorescence at the same wavelength as the Syto 9

fluorescence, so autofluorescence may have contributed to the overestimations by flow

cytometry. The underestimations by flow cytometry may be due to the sensitivity of Syto

9 to acidic conditions. Bacteria stained with Syto 9 and incubated in buffers with a pH

<5 for 15 minutes resulted in the decrease of Syto 9 fluorescence. It is known within

phagocytic cells that phagosomes become acidic (pH 3.5-4.0) upon phagosome/lysosome

fusion to help eliminate bacteria. Most of the underestimations by flow cytometry

occurred in the sham-pair rats, which suggests that EtOH ingestion may either suppress

phagosome/lysosome fusion or acidification of the phagocytic vacuole.

In addition to phagocytosis, two bactericidal mechanisms in macrophages isolated

from the lungs of smoke-exposed ± EtOH-fed rats were analyzed. In vitro assays were

performed to measure smoke- and EtOH-induced alterations in oxidative burst and

degranulation in macrophages stimulated with either unencapsulated pneumococci or

PMA. Unencapsulated pneumococci were used because macrophages can phagocytose

this mutant strain much better and produce a greater response than with type 3

pneumococci in vitro. PMA was utilized because it is known to stimulate macrophages,

but unlike bacteria, this non-particulate chemical can freely diffuse into the cell

independent of receptor-mediated phagocytosis.

107

EtOH ingestion alone up-regulated the oxidative burst in macrophages stimulated

by either S. pneumoniae or PMA. This increased response eventually leveled off and

oxygen radical production became similar to that for macrophages from sham-pair

controls. These results are contrary to those from previous studies showing alcohol

inhibits the oxidative burst in macrophages stimulated with endotoxin, heat-killed S.

aureus, or PMA [164,165]. These differences may result from the different models and

methods used to measure oxidative burst. One study gave rats a continuous intravenous

infusion of EtOH for seven hours or pair-fed rats for 12-14 weeks. Then the alveolar

macrophages were isolated at three hours after the rats received an intravenous injection

of endotoxin [164]. The other study isolated alveolar macrophages from rats that were

pair-fed for one to four weeks or exposed normal rat alveolar macrophages to various

concentrations of EtOH in culture [165]. Both studies quantified the release of certain

oxygen radical species, such as superoxide anion and nitric oxide, using specific assays to

measure each product. In our experiment we measured total oxygen radical production

within the macrophages using a chemiluminescence assay. Finally, the published studies

measured oxidative burst at one hour to 24 hours after the cells were stimulated, while

our assay measured this activity every 10 minutes up to one hour after stimulation.

A similar trend also occurred with EtOH ingestion in conjunction with smoke

exposure, where smoke-EtOH rats had a more robust early oxidative burst than smoke-

pair rats, but this only occurred when the cells were stimulated with pneumococci. PMA-

stimulated macrophages from smoke-EtOH rats, on the other hand, produced fewer

oxygen radicals than those from sham-EtOH rats. This is likely due to a smoke-induced

decrease in oxidative burst since macrophages from smoke-exposed animals in general

108

produced lower amounts of oxygen radicals than the sham-exposed animals in response

to PMA.

Smoke exposure alone delayed the oxidative burst in macrophages stimulated

with pneumococci. This was similar to another study in which macrophages isolated

from human smokers had a suppressed superoxide anion response to endotoxin [289].

However, other studies reported that alveolar macrophages from human smokers exhibit

an increased respiratory burst. Those authors utilized a 20 minute assay without

stimulation [290] and after four days of incubation with endotoxin [248]. Again, as with

the alcohol studies, comparison of these earlier smoking studies with our results is

difficult because of methodological differences.

Along with oxygen radical production, delivery of the lysosomal granule contents

to the target organisms is a major mechanism of macrophage bactericidal activity.

Lysozyme is commonly assayed as a marker for degranulation, because it is present in

the specific granules. Chronic EtOH ingestion and smoke exposure in our rat model did

not cause any significant alterations in lysozyme release in response to pneumococci.

Although macrophages from human smokers contain higher amounts of lysozyme [229],

macrophages from smoke-exposed rats released slightly less lysozyme than macrophages

from sham-exposed rats. Degranulation, as measured by lysozyme release, did not occur

in macrophages from any of the four treatment groups when stimulated by PMA. It is not

clear why this occurred, because PMA is known to cause degranulation in PMNs after 20

minutes [177]. Macrophage degranulation may not be as sensitive to non-receptor-

mediated stimulation, and this process may take longer than one hour.

109

The results from the oxidative burst and degranulation experiments do not provide

an explanation for the smoke- and EtOH-induced alterations of non-PMN-mediated

killing. Comparisons between these ex vivo studies and the in vivo bactericidal assay are

complicated, as macrophages may act differently within their native environment than in

vitro. However, it is also difficult to identify alterations in alveolar macrophages without

isolating them from the lungs. Additional experiments are required to identify the effects

of smoke and EtOH on macrophages at the sub-cellular level such as cell signaling, toll-

like receptor expression, and phagolysosome formation.

Opsonization is an important process for eliminating infection by facilitating

phagocytosis of the microorganisms. From the macrophage phagocytosis assay, EtOH

ingestion alone suppressed phagocytosis of pneumococci in the rats’ lungs. To determine

if this is due to impaired opsonization, an assay was performed to measure the in vivo

deposition of complement C3, CRP, and SP-D on pneumococci. Smoke-exposed animals

also were included in this study to determine if concurrent smoke exposure counteracts

the effect of EtOH.

No reported studies have determined the effects of EtOH ingestion on the opsonic

activity of C3 and CRP, and a review of the literature indicates no associations have been

made between smoking and this activity in the lungs. Only one study has reported that

chronic EtOH exposure decreases opsonization by surfactant [154]. In our assay, chronic

EtOH ingestion alone significantly decreased the opsonic deposition of both C3 and SP-D

on pneumococci, as well as slightly reduced in CRP opsonization. These results could

help explain the EtOH-induced suppression of macrophage phagocytosis of

pneumococci. Smoke exposure did not significantly alter the binding of any of the

110

opsonic proteins to the bacteria, but within the smoke-exposed rats the EtOH-induced

defect was not apparent. This was depicted by smoke-EtOH rats and their pair-fed

counterparts having similar percentages of pneumococci opsonized by each opsonin.

Smoke and EtOH are known to modify the levels of these opsonic proteins. To

clarify if the differences in deposition were due to altered baseline levels of opsonic

proteins in the lungs, each opsonin was quantified in uninfected rats. Significantly lower

levels of C3 were detected in the sham-EtOH rats which correlates with their reduction in

C3 opsonization. Alcohol has been shown to activate complement through the alternative

pathway, increase the deposition of C3 in the liver, and reduce C3 levels in serum

[155,157,163]. This nonspecific activation and deposition also may be occurring in the

rats’ lungs, resulting in impaired bacterial opsonization. Smoking also causes similar

activation of C3 [237-240], but smoke exposure alone did not alter baseline C3 levels in

the lungs of our rats. Within the smoke-exposed group, C3 was significantly decreased in

the EtOH-fed rats compared to their pair-fed counterparts. However, this decrease was

apparently not great enough to negatively affect C3 opsonization of pneumococci.

Differences in SP-D deposition on the pneumococci were not explained by

changes in basal levels of the protein, for sham-EtOHs, if anything, had even higher

amounts of SP-D as their sham-pair controls. These results indicate that EtOH may

suppress SP-D opsonization by another mechanism, such as altering surfactant protein

function. Finally, minimal CRP concentrations were quantified in the lungs of any of the

rats and there were no smoke- or EtOH-induced effects on these levels. This explains

why fewer pneumococci were opsonized by CRP in all treatment groups.

111

The EtOH-induced defects in C3 and SP-D deposition on pneumococci help

explain the decreased macrophage phagocytosis as well as decreased pneumococcal

clearance from the lungs of rats treated with EtOH alone. Although smoke exposure

alone did not affect opsonization, the EtOH-induced defect in opsonic activity was

undetected with concurrent smoke exposure even though EtOH ingestion still reduced the

C3 baseline levels in smoke-exposed rats. Further experiments are needed to determine if

chronic EtOH ingestion and smoke exposure alter other early complement proteins

needed for opsonic deposition such as C2, C4, or Factor B and how these results relate to

intrapulmonary clearance of pneumococci.

C. PMN Functions

PMNs are the primary cellular defense against S. pneumoniae because they can

effectively phagocytose and kill these microorganisms. Several studies have shown that

acute and chronic EtOH ingestion have different effects on PMN function. One week of

EtOH ingestion increases PMN recruitment, but decreases phagocytosis and bactericidal

activity while five weeks of EtOH ingestion does not alter PMN recruitment and

phagocytosis, but it continues to impair PMN-mediated killing. Previous work in the

laboratory showed that peripheral PMNs from short-term EtOH-fed rats were defective in

killing certain strains of pneumococci, but not others [176,177]. Using a more acute

exposure model in our laboratory, PMNs pre-recruited to the lungs of rats consuming

EtOH for one week were deficient in their ability to kill type 3 pneumococci [171]. This

same study also demonstrated that 8 weeks of concurrent smoke exposure prevented this

EtOH-induced decrease from occurring. These results from the one-week model led to

112

our hypothesis that chronic EtOH ingestion would reduce PMN-mediated killing in the

lungs and concurrent smoke exposure would negate this EtOH defect.

To examine the effects of chronic EtOH ingestion and smoke exposure on PMN-

mediated killing, we utilized the same in vivo bactericidal assay that was used in the one-

week model. This assay specifically measures PMN-mediated killing of S. pneumoniae

in the lungs because the administration of LPS to recruit the PMNs inactivates

extracellular anti-pneumococcal factors [171]. Similar to the results from the non-PMN-

mediated killing assay, EtOH ingestion alone significantly diminished PMN killing

within the lungs as compared to the sham-pair rats. The severe detrimental effect of

EtOH on PMN killing allowed pneumococci to grow within the lungs one hour after a

transtracheal infection. This result is associated with previous research that showed

PMNs from EtOH-fed rats exhibited decreased bactericidal activity in vitro against

pneumococci [176,177]. As hypothesized, the EtOH-induced defect in PMN-mediated

killing did not occur when the rats were treated by concurrent smoke exposure. The

smoke-EtOH rats killed significantly more pneumococci than their sham-exposed

counterparts. In fact, smoke exposure in conjunction with EtOH ingestion restored

killing to the level of the sham-pair rats. However, consistent with the non-PMN-

mediated killing assay, smoke exposure alone did not increase pneumococcal killing

above the level reported in the sham-pair controls.

PMN recruitment and phagocytosis then were assessed in the rats’ lungs to

determine whether a smoke-induced increase in PMN numbers or uptake of pneumococci

could explain the restoration of PMN killing in the presence of EtOH. It has been

described in the literature that chronic EtOH ingestion causes neutropenia which is

113

amplified during infection [291,292]. This was explained by EtOH down-regulating the

expression of β2-integrin which allows PMNs to bind to the endothelial wall and migrate

to the site of infection [170]. Cigarette smoking, on the other hand, is known to increase

pulmonary PMN numbers. Smoke exposure increases the production of PMNs in the

bone marrow and decreases their transit time, resulting in higher numbers of immature

PMNs circulating in the periphery [293]. Cigarette smoke stimulates alveolar

macrophages to release cytokines and chemokines leading to recruitment of PMNs to the

lungs in the absence of infection. Finally, smokers suffering from COPD are repeatedly

colonized by respiratory pathogens that result in a constant state of inflammation in the

lungs [294]. Based on these reports and the results from the PMN-mediating killing

assay, we hypothesized that EtOH ingestion would impair PMN recruitment and

concurrent smoke exposure would correct this defect by increasing the number of PMNs

recruited to the lungs.

PMN recruitment was evaluated using manual cytospin counts of lavage fluid

from the same rats used in the PMN-mediated killing assay. There were no smoke-

and/or EtOH-induced differences in the percentage of cells that were PMNs. This result

corroborates the earlier study in our laboratory that evaluated PMN recruitment in the

one-week exposure model [171]. LPS is a potent stimulator of alveolar macrophages,

causing them to produce proinflammatory cytokines and chemokines including MIP-2

and CINC-1. One possible reason no differences in PMN recruitment were measured

between the treatment groups was that the overwhelming stimulatory effects of LPS may

have masked any subtle smoke- and EtOH-induced changes in PMN recruitment.

Whether or not this is true, the decrease in PMN killing by EtOH ingestion and

114

restoration of killing by concurrent smoke exposure do not appear to be due to alterations

in PMN recruitment to the lungs.

PMNs recruited to the lungs are essentially useless if they fail to effectively

phagocytose invading pneumococci. Our laboratory showed previously that peripheral

blood PMNs isolated from rats exposed to EtOH for one week did not exhibit decreased

phagocytosis of several different pneumococcal strains [177]. Physiologically relevant

concentrations of EtOH also did not impair the phagocytic ability of cultured PMNs in

vitro [173]. In contrast, other studies reported acute EtOH exposure suppresses PMN

phagocytosis [170,174]. Cigarette smoke has been shown to have both stimulatory and

inhibitory effects on PMN phagocytic activity [256]. Based on the results from our in

vivo PMN-mediated killing assay, we hypothesized chronic EtOH ingestion would impair

PMN phagocytosis and concurrent smoke exposure would reverse this EtOH defect by

enhancing phagocytic activity.

To determine if smoke- and EtOH-induced alterations in PMN phagocytosis are

responsible for the differences in PMN-mediated killing, a PMN phagocytosis assay was

used to measure the in vivo uptake of pneumococci by pre-recruited PMNs. This is the

same assay that was developed previously in our laboratory to identify the acute effects

of smoke and EtOH exposure on PMN phagocytosis [171]. EtOH ingestion alone failed

to reduce the percentage of phagocytosing PMNs, but it significantly reduced the amount

of organisms taken up by each cell as compared to sham-pair rats. However, we believe

this decrease is not biologically relevant for it did not affect the overall amount of

phagocytosed organisms as indicated by the phagocytic index, and this is analogous to

previous results from in vitro and in vivo studies. Smoke exposure with or without EtOH

115

ingestion did not alter the phagocytic activity of PMNs. This result is not surprising since

published studies have reported no differences in phagocytosis by peripheral PMNs from

human smokers [256] and PMNs pre-recruited to the lungs of rats exposed to smoke for 8

weeks [171].

It is still unclear how the addition of smoke exposure was able to restore the

EtOH-induce impairments in killing, particularly when there was no increase in killing in

the animals exposed to smoke alone. Further experiments are needed to identify the

exact mechanisms of chronic EtOH ingestion and smoke exposure on PMN killing.

Functional assays must be performed to measure the ability of PMNs to undergo

respiratory burst and degranulation. The surface expression of the CXCR2 receptor in

PMNs also needs to be assessed due to its importance in priming and activating the cell

through the interaction with MIP-2 and CINC-1.

D. Chemokine Production

The release of chemokines by alveolar macrophages is important for the

activation and recruitment of PMNs during a pulmonary infection. Acute EtOH exposure

in rats has been shown to inhibit their ability to produce MIP-2 and CINC-1 in their lungs

after an intratracheal inoculation of S. pneumoniae [167,169]. The effect of smoke

exposure is just the opposite, where human smokers have higher concentrations of

chemokines, including IL-8 and gro-α, in their lungs [249]. Cigarette smoke is also

known to stimulate a variety of pulmonary cells to release PMN chemoattractants such as

granulocyte colony-stimulating factor and leukotrienes [252-254]. Previous research in

our laboratory found no effect of smoke or EtOH alone on pulmonary chemokine levels

116

in our short-term model after a transtracheal administration of endotoxin followed by a

transtracheal infection of pneumococci [171]. However, the combination of smoke and

EtOH significantly increased the production of MIP-2 and CINC-1 in the lungs.

Furthermore, EtOH ingestion alone reduced CINC-1 levels in the serum, but this was

normalized by the addition of smoke. The results from this previous experiment helped

formulate our hypothesis that impairments in PMN killing by chronic EtOH ingestion are

due to a reduction in serum CINC-1, a defect that is corrected by concurrent smoke

exposure through the augmentation of pulmonary chemokine production and restoration

of normal CINC-1 levels in serum.

To analyze the effects of chronic EtOH ingestion and smoke exposure on

chemokine production in response to pneumococcal infection, MIP-2 was measured in

the lung homogenates and CINC-1 was measured in both the lung homogenates and

serum from rats that were used in the PMN-mediated killing assay. Chemokine

production in the lungs was not significantly altered by smoke or EtOH alone. The

combination of the two insults also failed to increase pulmonary chemokine levels, unlike

what was detected in the short-term model [171]. EtOH ingestion alone appeared to

reduce CINC-1 in the serum and concurrent smoke exposure negated this EtOH defect,

even though smoke exposure alone did not increase the amount of CINC-1 in the serum.

Although these effects of smoke and EtOH on serum CINC-1 were not statistically

significant, they did parallel previous results from our short-term model [171] and

mirrored the results from the PMN-mediated killing assay.

The EtOH-induced defect in PMN-mediated killing was not related to decreased

pulmonary levels of MIP-2 and CINC-1. However, this defect may be associated with

117

the reduction in serum CINC-1. Pulmonary CINC-1 is known to leave the alveolar space

and enter the systemic circulation to pre-activate PMNs for phagocytosis and killing

[54,55]. The exact mechanism of CINC-1 movement out of the lungs is unknown, but it

has been shown to be an active, one-directional process [54]. The decrease in serum

CINC-1 by EtOH ingestion may be a result of impaired movement of pulmonary CINC-1

from the lungs to the circulation. Somehow, concurrent smoke exposure compensates for

this EtOH-induced impairment by allowing sufficient amounts of CINC-1 to enter the

bloodstream for PMN priming. To determine if restoration of serum CINC-1 in sham-

EtOH rats will restore PMN-mediated killing of pneumococci in the lungs, the PMN-

mediated killing assay would need to be repeated in sham-EtOH rats that receive an

intravenous injection of CINC-1.

Although no differences in pulmonary chemokine levels were detected in the rats

that received LPS, we further evaluated the ability of alveolar macrophages to release

chemokines ex vivo in the presence of pneumococci. Earlier research has reported that

macrophages from rats consuming EtOH for two weeks had decreased production of

proinflammatory cytokines in response to LPS [166]. On the other hand, macrophages

from human smokers stimulated by endotoxin secreted higher amounts of IL-8 and gro-α

than macrophages from nonsmokers. Given the results of the pulmonary and systemic

cytokine levels, we predicted chronic EtOH ingestion would decrease chemokine

production by alveolar macrophages and concurrent smoke would abolish this deficit by

up-regulating the release of chemokines.

MIP-2 and CINC-1 were quantified in media from isolated alveolar macrophages

stimulated by unencapsulated pneumococci or PMA. When incubated with

118

pneumococci, macrophages from sham-EtOH rats released reduced amounts of MIP-2

and CINC-1 than macrophages from the sham-pairs. This effect of chronic EtOH

ingestion correlates with previous studies demonstrating that EtOH-treated rats produce

decreased amounts of chemokines in their lungs when infected with S. pneumoniae

[167,169]. Smoke exposure alone did not increase chemokine production.

Unexpectedly, concurrent smoke exposure was unsuccessful in negating the EtOH-

induced impairment in chemokine production. The levels of MIP-2 and CINC-1 released

by smoke-EtOH rat macrophages were less than those from animals exposed to smoke

alone. These results are different from the chemokine levels measured in lung

homogenates from rats used in the PMN-mediated killing assay, indicating the

differences in chemokine production by alveolar macrophages are not responsible for the

results of the PMN-mediated killing assay.

No significant alterations in MIP-2 production were detected in PMA-stimulated

macrophages from each of the four treatment groups, although EtOH ingestion with

smoke exposure reduced it by greater than half. However, EtOH ingestion alone

significantly increased CINC-1 production such that macrophages from sham-EtOH rats

secreted higher amounts of CINC-1 than macrophages from their pair-fed counterparts.

Smoke exposure did not decrease CINC-1 release, but concurrent smoke exposure

reduced this EtOH-induced increase in CINC-1 back to a similar level measured in the

sham-pair controls.

The smoke- and EtOH-induced differences in chemokine production between

pneumococcal and PMA stimulation is that these two stimulators act on the macrophages

through separate pathways. Pneumococci activate macrophages through receptor-

119

mediated recognition and phagocytosis, while PMA activation is receptor-independent.

Two key receptors expressed on the surface of alveolar macrophages that recognize S.

pneumoniae are Toll-like receptors 2 and 4 (TLR2 and TLR4). The EtOH-induced

decrease in chemokine production in the presence of pneumococci may be due to

alterations in the functions of these receptors. Several studies have shown that EtOH

exposure in vitro and in vivo impairs TLR4 activation in monocytes and macrophages by

inhibiting the phosphorylation of downstream signaling proteins such as mitogen-

activated protein kinases [295-297]. Two of these same studies did not detect an EtOH

effect on TLR2 signaling [295,297], however, one study reported that acute EtOH

exposure in mice down-regulated the TLR2-mediated inflammatory response in alveolar

macrophages [298].

Cigarette smoke modulates the expression and function of TLR2 and TLR4.

Alveolar macrophages from human smokers had decreased expression of TLR2 on their

surface and failed to up-regulate TLR2 expression when stimulated with LPS in contrast

to macrophages from nonsmokers [299]. Another study found a dose-dependent down

regulation in TLR4 messenger RNA and protein expression in human airway epithelial

cells exposed to cigarette smoke extract [300]. Cell activation also was suppressed in

alveolar macrophages from human smokers stimulated with TLR2 and TLR4 agonists,

resulting in the reduction of chemokine secretion including IL-8 [301]. These smoke-

induced defects were not apparent in our assay, for smoke exposure alone did not alter

chemokine production in macrophages stimulated with pneumococci. Additional

experiments are required to further understand these results by identifying the effects of

120

smoke and EtOH on expression and function of TLR2 and TLR4 in alveolar

macrophages from our rat model.

It is unclear why differences between MIP-2 and CINC-1 production occurred in

response to PMA. Although alveolar macrophages from sham-EtOH rats secreted

significantly higher amounts of CINC-1, it is difficult to believe EtOH ingestion up-

regulated CINC-1 production when there was such a wide range of variability among the

four treatment groups. Considering these cells were isolated out of the rats’ lungs

without additional exposure to EtOH and/or smoke, the results from this assay may not

accurately represent what occurs within the lungs of the rats.

E. Mortality Study

There is a strong agreement throughout the literature that alcohol abuse and

chronic smoking increase the severity and mortality of pneumococcal pneumonia. Both

of these behaviors suppress or even damage key innate and adaptive anti-pneumococcal

defenses. However, no studies have identified the outcome of pneumococcal disease in

the chronically drinking and smoking host. We hypothesized that EtOH ingestion with

smoke exposure would further increase the mortality rate of pneumococcal pneumonia

than either insult alone.

A mortality trial was conducted to identify the effects of chronic EtOH ingestion

with or without smoke exposure on the host in response to a lethal, intranasal challenge

of S. pneumoniae. The mortality rate was unchanged by chronic EtOH ingestion where

the same number of sham-exposed rats from each diet group succumbed to the infection.

Concurrent smoke exposure did not exacerbate the mortality rate as we predicted. In fact,

121

mortality among the smoke-EtOH rats was no greater than for both sham-exposed

groups. The results from the smoke-EtOHs and sham-pairs are consistent with the trend

that is present in our previous experiments involving the host defenses of the lungs.

There were no significant differences in the bacteremia rates among these three treatment

groups, and all rats that developed bacteremia eventually died within the next 2-3 days.

Interestingly, smoke exposure alone did not compromise the rats’ immunity and actually

protected the animals from developing bacteremia and death.

Performing an intranasal mortality study includes all of the important defense

mechanisms that are involved at the different stages of pneumococcal pathogenesis.

Several studies, including our own experiments, have shown the defects of EtOH

ingestion on these key host defenses including mucociliary clearance, PMN function, and

chemokine production. Based on our results that show chronic EtOH ingestion promotes

pneumococcal invasion, decreases non-PMN- and PMN-mediated killing, impairs

opsonization, and suppresses macrophage phagocytosis and chemokine production, one

would expect the sham-EtOH rats to have a worse outcome than their pair-fed

counterparts. It is possible pair-feeding induces stress in animals which may alter their

immune response to infection. Stress increases the production of the hormone

corticosterone which is known to promote an anti-inflammatory response. Previous work

in the laboratory showed sham-pair rats have similar concentrations of corticosterone as

the sham-EtOH rats.

Smoke-EtOH rats have significantly higher corticosterone levels than sham-

EtOHs and smoke-pairs. This excessive amount of corticosterone in the smoke-EtOH

group did not increase their mortality rate to pneumococcal infection, but it may cause a

122

similar end-result as the sham-EtOH rats. Among the four different treatment groups,

smoke-pairs had the lowest concentrations of corticosterone. In addition to low

corticosterone, smoke reduced pneumococcal movement to the lungs and did not affect

any of the other host defenses that were evaluated. This could be a potential reason why

all the smoke-pair rats survived the lethal infection.

Why did smoke exposure in conjunction with EtOH ingestion not perform in the

same manner? Besides up-regulating corticosterone levels, EtOH ingestion with smoke

exposure inhibited chemokine production from alveolar macrophages when they were

exposed to pneumococci. Hindering this activity could essentially shut down PMN

recruitment and activation. Defects in PMN function were not detected in smoke-EtOH

rats when they were stimulated with LPS. However, these defects may appear during a

normal pneumococcal infection without the presence of endotoxin from Gram-negative

bacteria. Other host defenses that are susceptible to the actions of smoke and EtOH, but

have not been evaluated yet, also may have contributed to the mortality results.

Additional studies are needed to further understand these findings from the mortality

trial. Such studies include examining PMN function and chemokine production in the

lungs infected with S. pneumoniae without LPS stimulation or instilled with the major

Gram-positive cell wall component lipoteichoic acid.

123

VII. CONCLUSIONS

The studies described in this thesis have analyzed the effects of chronic EtOH

ingestion and smoke exposure on various host defenses against the pneumococcus which

work to prevent pneumococcal dissemination throughout the host. In addition,

alternative therapies have been evaluated for preventing pneumococcal pneumonia in the

smoking and drinking host. The consequences of alcohol abuse and smoke exposure on

anti-pneumococcal defenses are conflicting in the literature, and comparisons of these

studies are complicated due to the use of different models and methods. This research

was conducted using a well-established rat model of chronic EtOH ingestion and smoke

exposure, which standardizes the approach of studying these dual insults in relation to the

immune system.

The experiments were designed to mimic a natural pneumococcal infection. The

first stage of pneumococcal pathogenesis is colonization of the nasopharynx.

Nasopharyngeal colonization was unaltered by EtOH ingestion and smoke exposure.

After colonization, pneumococci must evade the mucociliary clearance apparatus to reach

the lungs. EtOH slightly increased pneumococcal movement to the lower respiratory

tract, but did not impair ciliary beating. Surprisingly, smoke exposure enhanced

mucociliary clearance resulting in a reduction of pneumococcal infection of the lungs.

Once the pneumococci reach the lungs, they encounter a variety of non-PMN defenses

including bactericidal factors and alveolar macrophages. EtOH ingestion suppressed

non-PMN-mediated killing within the lungs while concurrent smoke exposure restored

killing, even though smoke alone did not increase this activity. Although the effects of

124

EtOH and smoke on pulmonary concentrations of lysozyme and lactoferrin did not

explain the differences in non-PMN-mediated killing, the EtOH-induced defect on

pulmonary clearance was found to be associated with decreased opsonization and

macrophage phagocytosis of pneumococci. Concurrent smoke exposure did not

exacerbate the EtOH defect on opsonization, which may also be true for macrophage

phagocytosis, but this remains unknown due to the inability of our assay to accurately

measure macrophage phagocytosis in smoke-exposed animals. During a pulmonary

infection, alveolar macrophages produce cytokines and chemokines to activate PMNs and

recruit them to the lungs. Neither EtOH ingestion nor smoke exposure affected

chemokine production in the lungs, which relates to their lack of effect on PMN

recruitment and phagocytosis of pneumococci. Despite the fact that PMN recruitment

and phagocytosis were unaltered, EtOH ingestion abolished PMN-mediated killing of

pneumococci within the lungs. Once again, this EtOH-induced defect was corrected by

concurrent smoke exposure.

Pneumococci must follow a step-wise progression and evade various host

defenses in order to cause pneumonia and bacteremia. This provides several host defense

mechanisms that can be targeted for therapeutic intervention. An intranasal vaccine

containing a conserved pneumococcal surface protein was tested to reduce

nasopharyngeal colonization. The vaccine slightly increased antibody levels in the

mucosa and serum, but did not protect against pneumococcal colonization. Salbutamol

and formoterol inhalation also were evaluated to increase ciliary beating and reduce

pneumococcal invasion into the lungs. Both β2-agonists were effective in decreasing

pneumococcal movement to the lungs in unexposed, control rats. Formoterol also

125

prevented pneumococcal infection of the lungs in rats exposed to EtOH alone, but the

protection did not extend to any of the other rat treatment groups.

These studies show that EtOH ingestion and smoke exposure have differential

effects on a range of host defenses against the pneumococcus as summarized in Table 7.

More research is warranted to describe the effects of chronic EtOH ingestion and

concurrent smoke exposure on host defense strategies against the pneumococcus.

Understanding the pathogenesis of S. pneumoniae will lead to the development of

improved therapies to prevent pneumococcal infection and reduce overall mortality from

pneumococcal pneumonia. Immunity to other pathogens such as Haemophilus

influenzae, Mycobacterium tuberculosis, and influenza virus also needs to be examined in

the presence of EtOH and smoke. Our rat model would be an effective tool for

identifying the separate and combined effects of these morbidities in association with a

variety of infectious diseases.

Table 7 – Summary of Conclusions

EtOH Alone Smoke Alone EtOH with Smoke

Mucociliary Clearance —

Pulmonary Killing — Lost EtOH Defect

Bactericidal Factors — Lost EtOH Effect

Macrophage Phagocytosis ??? ???

Opsonization — Lost EtOH Defect

126

VIII. BIBLIOGRAPHY

1. Kung HC, Hoyert DL, Xu J, and Murphy SL. Deaths: final data for 2005. Natl

Vital Stat Rep 2008; 56:1-120.

2. Talwar A, Lee H, and Fein A. Community-acquired pneumonia: what is relevant and what is not? Curr Opin Pulm Med 2007; 13:177-85.

3. Colice GL, Morley MA, Asche C, and Birnbaum HG. Treatment costs of community-acquired pneumonia in an employed population. Chest 2004; 125:2140-5.

4. Halm EA and Teirstein AS. Clinical practice. Management of community-acquired pneumonia. N Engl J Med 2002; %19;347:2039-45.

5. Ruiz M, Ewig S, Marcos MA et al. Etiology of community-acquired pneumonia: impact of age, comorbidity, and severity. Am J Respir Crit Care Med 1999; 160:397-405.

6. Heffelfinger JD, Dowell SF, Jorgensen JH et al. Management of community-acquired pneumonia in the era of pneumococcal resistance: a report from the Drug-Resistant Streptococcus pneumoniae Therapeutic Working Group. Arch Intern Med 2000 May 22 ;160 (10 ):1399 -408 160:1399-408.

7. Breiman RF, Butler JC, Tenover FC, Elliott JA, and Facklam RR. Emergence of drug-resistant pneumococcal infections in the United States [see comments]. JAMA 1994; 271:1831-5.

8. Butler JC, Hofmann J, Cetron MS, Elliott JA, Facklam RR, and Breiman RF. The continued emergence of drug-resistant Streptococcus pneumoniae in the United States: an update from the Centers for Disease Control and Prevention's Pneumococcal Sentinel Surveillance System. J Infect Dis 1996; 174:986-93.

9. Hawley LA, Walker FJ, and Whitney CG. Pneumococcal Disease. 3. 2002:9-1-9-14.

10. Fine MJ, Smith MA, Carson CA et al. Prognosis and outcomes of patients with community-acquired pneumonia. A meta-analysis. JAMA 1996; 275:134-41.

11. Watanakunakorn C and Bailey TA. Adult bacteremic pneumococcal pneumonia in a community teaching hospital, 1992-1996. A detailed analysis of 108 cases. Arch Intern Med 1997; 157:1965-71.

12. Robinson KA, Baughman W, Rothrock G et al. Epidemiology of invasive Streptococcus pneumoniae infections in the United States, 1995-1998: Opportunities for prevention in the conjugate vaccine era. JAMA 2001 Apr 4 ;285 (13 ):1729 -35 285:1729-35.

127

13. Marston BJ, Plouffe JF, File TM, Jr. et al. Incidence of community-acquired pneumonia requiring hospitalization. Results of a population-based active surveillance Study in Ohio. The Community-Based Pneumonia Incidence Study Group. Arch Intern Med 1997; 157:1709-18.

14. Gardner P and Schaffner W. Immunization of adults. N Engl J Med 1993; 328:1252-8.

15. Regev-Yochay G, Raz M, Dagan R et al. Nasopharyngeal carriage of Streptococcus pneumoniae by adults and children in community and family settings. Clin Infect Dis 2004; 38:632-9.

16. McCullers JA and Tuomanen EI. Molecular pathogenesis of pneumococcal pneumonia. Front Biosci 2001 Aug 1 ;6 :D877 -89 6:D877-89.:D877-D889.

17. Gray BM and Dillon HCJ. Clinical and epidemiologic studies of pneumococcal infection in children. Pediatr Infect Dis 1986; 5:201-7.

18. Johnston RBJ. Pathogenesis of pneumococcal pneumonia. Rev Infect Dis 1991; 13 Suppl 6:S509-17:S509-S517.

19. Peltola VT, Murti KG, and McCullers JA. Influenza virus neuraminidase contributes to secondary bacterial pneumonia. J Infect Dis 2005; 192:249-57.

20. Peltola VT and McCullers JA. Respiratory viruses predisposing to bacterial infections: role of neuraminidase. Pediatr Infect Dis J 2004; 23:S87-S97.

21. Levin R, Braiman A, and Priel Z. Protein kinase C induced calcium influx and sustained enhancement of ciliary beating by extracellular ATP. Cell Calcium 1997; 21:103-13.

22. Salathe M, Pratt MM, and Wanner A. Protein kinase C-dependent phosphorylation of a ciliary membrane protein and inhibition of ciliary beating. J Cell Sci 1993; 106:1211-20.

23. Wong LB, Park CL, and Yeates DB. Neuropeptide Y inhibits ciliary beat frequency in human ciliated cells via nPKC, independently of PKA. Am J Physiol 1998; 275:C440-C448.

24. Wyatt TA, Spurzem JR, May K, and Sisson JH. Regulation of ciliary beat frequency by both PKA and PKG in bovine airway epithelial cells. Am J Physiol 1998; 275:L827-L835.

25. Coonrod JD. The role of extracellular bactericidal factors in pulmonary host defense. Semin Respir Infect 1986; 1:118-29.

26. Travis SM, Conway BA, Zabner J et al. Activity of abundant antimicrobials of the human airway. Am J Respir Cell Mol Biol 1999; 20:872-9.

128

27. Coonrod JD, Varble R, and Yoneda K. Mechanism of killing of pneumococci by lysozyme. J Infect Dis 1991; 164:527-32.

28. Shaper M, Hollingshead SK, Benjamin WH, Jr., and Briles DE. PspA Protects Streptococcus pneumoniae from Killing by Apolactoferrin, and Antibody to PspA Enhances Killing of Pneumococci by Apolactoferrin. Infect Immun 2004; 72:5031-40.

29. Brock JH. The physiology of lactoferrin. Biochem Cell Biol 2002; 80:1-6.

30. Zhang P, Summer WR, Bagby GJ, and Nelson S. Innate immunity and pulmonary host defense. Immunol Rev 2000 Feb ;173 :39 -51 173:39-51.:39-51.

31. Ferrara A, Dos SC, and Lupi A. Effect of different antibacterial agents and surfactant protein-A (SP-A) on adherence of some respiratory pathogens to bronchial epithelial cells. Int J Antimicrob Agents 2001; 17:401-5.

32. Coonrod JD and Yoneda K. Detection and partial characterization of antibacterial factor(s) in alveolar lining material of rats. J Clin Invest 1983; 71:129-41.

33. Rubins JB, Charboneau D, Prigge W, and Mellencamp MA. Ethanol ingestion reduces antipneumococcal activity of rat pulmonary surfactant. J Infect Dis 1996; 174:507-12.

34. Janoff EN and Rubins JB. Invasive pneumococcal disease in the immunocompromised host. Microb Drug Resist 1997; 3:215-32.

35. Picard C, Puel A, Bustamante J, Ku CL, and Casanova JL. Primary immunodeficiencies associated with pneumococcal disease. Curr Opin Allergy Clin Immunol 2003; 3:451-9.

36. Figueroa JE and Densen P. Infectious diseases associated with complement deficiencies. Clin Microbiol Rev 1991; 4:359-95.

37. Winkelstein JA. Complement and the host's defense against the pneumococcus. Crit Rev Microbiol 1984; 11:187-208.

38. Brown JS, Hussell T, Gilliland SM et al. The classical pathway is the dominant complement pathway required for innate immunity to Streptococcus pneumoniae infection in mice. Proc Natl Acad Sci U S A 2002; 99:16969-74.

39. Winkelstein JA and Tomasz A. Activation of the alternative complement pathway by pneumococcal cell wall teichoic acid. J Immunol 1978; 120:174-8.

40. Krarup A, Sorensen UB, Matsushita M, Jensenius JC, and Thiel S. Effect of capsulation of opportunistic pathogenic bacteria on binding of the pattern recognition molecules mannan-binding lectin, L-ficolin, and H-ficolin. Infect Immun 2005; 73:1052-60.

129

41. Aderem A. Phagocytosis and the inflammatory response. J Infect Dis 2003; 187 Suppl 2:S340-5.:S340-S345.

42. Volanakis JE and Kaplan MH. Specificity of C-reactive protein for choline phosphate residues of pneumococcal C-polysaccharide. Proc Soc Exp Biol Med 1971; 136:612-4.

43. Kaplan MH and Volanakis JE. Interaction of C-reactive protein complexes with the complement system. I. Consumption of human complement associated with the reaction of C-reactive protein with pneumococcal C-polysaccharide and with the choline phosphatides, lecithin and sphingomyelin. J Immunol 1974; 112:2135-47.

44. Hickling TP, Clark H, Malhotra R, and Sim RB. Collectins and their role in lung immunity. J Leukoc Biol 2004; 75:27-33.

45. Wright JR. Host defense functions of pulmonary surfactant. Biol Neonate 2004; 85:326-32.

46. Whitsett JA. Surfactant proteins in innate host defense of the lung. Biol Neonate 2005; 88:175-80.

47. Jounblat R, Clark H, Eggleton P, Hawgood S, Andrew PW, and Kadioglu A. The role of surfactant protein D in the colonisation of the respiratory tract and onset of bacteraemia during pneumococcal pneumonia. Respir Res 2005; 6:126.:126.

48. Gentry MJ, Snitily MU, and Preheim LC. Decreased uptake and killing of Streptococcus pneumoniae within the lungs of cirrhotic rats. Immunology & Infectious Diseases 1996; 6:43-7.

49. Coonrod JD, Marple S, Holmes GP, and Rehm SR. Extracellular killing of inhaled pneumococci in rats. J Lab Clin Med 1987; 110:753-66.

50. Jonsson S, Musher DM, Chapman A, Goree A, and Lawrence EC. Phagocytosis and killing of common bacterial pathogens of the lung by human alveolar macrophages. J Infect Dis 1985; 152:4-13.

51. al-Mokdad M, Shibata F, and Nakagawa H. Effects of cytokine-induced neutrophil chemoattractants (CINCs) on shape change, adhesiveness and phagocytosis of rat neutrophils. Biol Pharm Bull 1997; 20:920-3.

52. Frevert CW, Farone A, Danaee H, Paulauskis JD, and Kobzik L. Functional characterization of rat chemokine macrophage inflammatory protein-2. Inflammation 1995; 19:133-42.

53. Frevert CW, Huang S, Danaee H, Paulauskis JD, and Kobzik L. Functional characterization of the rat chemokine KC and its importance in neutrophil

130

recruitment in a rat model of pulmonary inflammation. J Immunol 1995; 154:335-44.

54. Quinton LJ, Nelson S, Zhang P et al. Selective transport of cytokine-induced neutrophil chemoattractant from the lung to the blood facilitates pulmonary neutrophil recruitment. Am J Physiol Lung Cell Mol Physiol 2004; 286:L465-L472.

55. Zhang P, Nelson S, Holmes MC, Summer WR, and Bagby GJ. Compartmentalization of macrophage inflammatory protein-2, but not cytokine-induced neutrophil chemoattractant, in rats challenged with intratracheal endotoxin. Shock 2002; 17:104-8.

56. Sibille Y and Marchandise FX. Pulmonary immune cells in health and disease: polymorphonuclear neutrophils. Eur Respir J 1993; 6:1529-43.

57. Hampton MB, Kettle AJ, and Winterbourn CC. Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing. 1998.

58. McKenna SM and Davies KJ. The inhibition of bacterial growth by hypochlorous acid. Possible role in the bactericidal activity of phagocytes. Biochem J 1988; 254:685-92.

59. Rosen H, Michel BR, vanDevanter DR, and Hughes JP. Differential effects of myeloperoxidase-derived oxidants on Escherichia coli DNA replication. Infect Immun 1998; 66:2655-9.

60. Kobayashi SD, Voyich JM, Burlak C, and DeLeo FR. Neutrophils in the innate immune response. Arch Immunol Ther Exp (Warsz ) 2005; 53:505-17.

61. MacLeod CM, Hodges RG, Heidelberger M, and Bernhard WG. Prevention of Pneumococcal Pneumonia by immunization with specific capsular polysaccharides. J Exp Med 1945; 82:445-65.

62. Arulanandam BP, Lynch JM, Briles DE, Hollingshead S, and Metzger DW. Intranasal Vaccination with Pneumococcal Surface Protein A and Interleukin-12 Augments Antibody-Mediated Opsonization and Protective Immunity against Streptococcus pneumoniae Infection. Infect Immun 2001 Nov ;69 (11 ):6718 -24 69:6718-24.

63. Balachandran P, Brooks-Walter A, Virolainen-Julkunen A, Hollingshead SK, and Briles DE. Role of Pneumococcal Surface Protein C in Nasopharyngeal Carriage and Pneumonia and Its Ability To Elicit Protection against Carriage of Streptococcus pneumoniae. Infect Immun 2002 May ;70 (5 ):2526 -34 70:2526-34.

64. Green BA, Zhang Y, Masi AW et al. PppA, a surface-exposed protein of Streptococcus pneumoniae, elicits cross-reactive antibodies that reduce

131

colonization in a murine intranasal immunization and challenge model. Infect Immun 2005; 73:981-9.

65. Alexander JE, Lock RA, Peeters CC et al. Immunization of mice with pneumolysin toxoid confers a significant degree of protection against at least nine serotypes of Streptococcus pneumoniae. Infect Immun 1994; 62:5683-8.

66. Pilette C, Durham SR, Vaerman JP, and Sibille Y. Mucosal immunity in asthma and chronic obstructive pulmonary disease: a role for immunoglobulin A? Proc Am Thorac Soc 2004; 1:125-35.

67. Kadioglu A, Coward W, Colston MJ, Hewitt CR, and Andrew PW. CD4-T-lymphocyte interactions with pneumolysin and pneumococci suggest a crucial protective role in the host response to pneumococcal infection. Infect Immun 2004; 72:2689-97.

68. Kadioglu A, Gingles NA, Grattan K, Kerr A, Mitchell TJ, and Andrew PW. Host cellular immune response to pneumococcal lung infection in mice [In Process Citation]. Infect Immun 2000 Feb ;68 (2 ):492 -501 68:492-501.

69. Jounblat R, Kadioglu A, Mitchell TJ, and Andrew PW. Pneumococcal Behavior and Host Responses during Bronchopneumonia Are Affected Differently by the Cytolytic and Complement-Activating Activities of Pneumolysin. Infect Immun 2003 Apr ;71 (4 ):1813 -9 71:1813-9.

70. Malley R, Trzcinski K, Srivastava A, Thompson CM, Anderson PW, and Lipsitch M. CD4+ T cells mediate antibody-independent acquired immunity to pneumococcal colonization. Proc Natl Acad Sci U S A 2005; 102:4848-53.

71. Yee YC, Thornsberry C, Brown SD, Bouchillon SK, Marler JK, and Rich T. A comparative study of the in-vitro activity of cefepime and other antimicrobial agents against penicillin-susceptible and penicillin-resistant Streptococcus pneumoniae. J Antimicrob Chemother 1993; 32 Suppl B:13-9.:13-9.

72. Doern GV and Brown SD. Antimicrobial susceptibility among community-acquired respiratory tract pathogens in the USA: data from PROTEKT US 2000-01. J Infect 2004; 48:56-65.

73. Karlowsky JA, Thornsberry C, Jones ME, Evangelista AT, Critchley IA, and Sahm DF. Factors associated with relative rates of antimicrobial resistance among Streptococcus pneumoniae in the United States: results from the TRUST Surveillance Program (1998-2002). Clin Infect Dis 2003 Apr 15 ;36 (8 ):963 -70 36:963-70.

74. Doern GV, Heilmann KP, Huynh HK, Rhomberg PR, Coffman SL, and Brueggemann AB. Antimicrobial resistance among clinical isolates of Streptococcus pneumoniae in the United States during 1999--2000, including a

132

comparison of resistance rates since 1994--1995. Antimicrob Agents Chemother 2001; 45:1721-9.

75. Doern GV, Richter SS, Miller A et al. Antimicrobial resistance among Streptococcus pneumoniae in the United States: have we begun to turn the corner on resistance to certain antimicrobial classes? Clin Infect Dis 2005; 41:139-48.

76. Reinert RR. Resistance phenotypes and multi-drug resistance in Streptococcus pneumoniae (PROTEKT years 1-3 [1999-20021). J Chemother 2004; 16 Suppl 6:35-48.:35-48.

77. Van Bambeke F, Reinert RR, Appelbaum PC, Tulkens PM, and Peetermans WE. Multidrug-resistant Streptococcus pneumoniae infections: current and future therapeutic options. Drugs 2007; 67:2355-82.

78. Finkelstein JA, Stille C, Nordin J et al. Reduction in antibiotic use among US children, 1996-2000. Pediatrics 2003; 112:620-7.

79. Moore MR, Hyde TB, Hennessy TW et al. Impact of a conjugate vaccine on community-wide carriage of nonsusceptible Streptococcus pneumoniae in Alaska. J Infect Dis 2004; 190:2031-8.

80. Whitney CG, Farley MM, Hadler J et al. Decline in invasive pneumococcal disease after the introduction of protein-polysaccharide conjugate vaccine. N Engl J Med 2003; 348:1737-46.

81. Talbot TR, Poehling KA, Hartert TV et al. Reduction in high rates of antibiotic-nonsusceptible invasive pneumococcal disease in tennessee after introduction of the pneumococcal conjugate vaccine. Clin Infect Dis 2004; 39:641-8.

82. Klugman KP. Efficacy of pneumococcal conjugate vaccines and their effect on carriage and antimicrobial resistance. Lancet Infect Dis 2001; 1:85-91.

83. Klugman KP. Vaccination: a novel approach to reduce antibiotic resistance. Clin Infect Dis 2004; 39:649-51.

84. Dagan R. Conjugate vaccines: potential impact on antibiotic use? Int J Clin Pract Suppl 2001;27-8.

85. Update: Influenza activity--United States and worldwide, 2001-02 season, and composition of the 2002-03 influenza vaccine. MMWR Morb Mortal Wkly Rep 2002; 51:503-6.

86. Public health and aging: influenza vaccination coverage among adults aged > or =50 years and pneumococcal vaccination coverage among adults aged > or =65 years--United States, 2002. MMWR Morb Mortal Wkly Rep 2003; 52:987-92.

133

87. Vila-Corcoles A, Ochoa-Gondar O, Hospital I et al. Protective effects of the 23-valent pneumococcal polysaccharide vaccine in the elderly population: the EVAN-65 study. Clin Infect Dis 2006; 43:860-8.

88. Mangtani P, Cutts F, and Hall AJ. Efficacy of polysaccharide pneumococcal vaccine in adults in more developed countries: the state of the evidence. Lancet Infect Dis 2003; 3:71-8.

89. Dear K, Holden J, Andrews R, and Tatham D. Vaccines for preventing pneumococcal infection in adults. Cochrane Database Syst Rev 2003;CD000422.

90. Melegaro A and Edmunds WJ. The 23-valent pneumococcal polysaccharide vaccine. Part I. Efficacy of PPV in the elderly: a comparison of meta-analyses. Eur J Epidemiol 2004; 19:353-63.

91. Musher DM, Rueda-Jaimes AM, Graviss EA, and Rodriguez-Barradas MC. Effect of pneumococcal vaccination: a comparison of vaccination rates in patients with bacteremic and nonbacteremic pneumococcal pneumonia. Clin Infect Dis 2006; 43:1004-8.

92. Lipsky BA and Hirschmann JV. Pneumococcal polysaccharide vaccines do not protect the elderly from pneumococcal infections. Neth J Med 2004; 62:33-5.

93. Butler JC, Breiman RF, Campbell JF, Lipman HB, Broome CV, and Facklam RR. Pneumococcal polysaccharide vaccine efficacy. An evaluation of current recommendations. JAMA 1993; 270:1826-31.

94. Veenhoven RH, Bogaert D, Schilder AG et al. Nasopharyngeal pneumococcal carriage after combined pneumococcal conjugate and polysaccharide vaccination in children with a history of recurrent acute otitis media. Clin Infect Dis 2004; 39:911-9.

95. Preventing pneumococcal disease among infants and young children. Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2000; 49:1-35.

96. Casal J and Tarrago D. Immunity to Streptococcus pneumoniae: Factors affecting production and efficacy. Curr Opin Infect Dis 2003; 16:219-24.

97. Invasive pneumococcal disease in children 5 years after conjugate vaccine introduction--eight states, 1998-2005. MMWR Morb Mortal Wkly Rep 2008; 57:144-8.

98. Direct and indirect effects of routine vaccination of children with 7-valent pneumococcal conjugate vaccine on incidence of invasive pneumococcal disease--United States, 1998-2003. MMWR Morb Mortal Wkly Rep 2005; 54:893-7.

134

99. Flannery B, Schrag S, Bennett NM et al. Impact of childhood vaccination on racial disparities in invasive Streptococcus pneumoniae infections. JAMA 2004; 291:2197-203.

100. Huang SS, Platt R, Rifas-Shiman SL, Pelton SI, Goldmann D, and Finkelstein JA. Post-PCV7 changes in colonizing pneumococcal serotypes in 16 Massachusetts communities, 2001 and 2004. Pediatrics 2005; 116:e408-e413.

101. Spratt BG and Greenwood BM. Prevention of pneumococcal disease by vaccination: does serotype replacement matter? Lancet 2000; 356:1210-1.

102. Pelton SI and Klein JO. The future of pneumococcal conjugate vaccines for prevention of pneumococcal diseases in infants and children. Pediatrics 2002; 110:805-14.

103. Briles DE, Hollingshead SK, Paton JC et al. Immunizations with pneumococcal surface protein A and pneumolysin are protective against pneumonia in a murine model of pulmonary infection with Streptococcus pneumoniae. J Infect Dis 2003; 188:339-48.

104. Watson DA and Musher DM. Interruption of capsule production in Streptococcus pneumonia serotype 3 by insertion of transposon Tn916. Infect Immun 1990; 58:3135-8.

105. Austrian R. Some observations on the pneumococcus and on the current status of pneumococcal disease and its prevention. Rev Infect Dis 1981; 3 Suppl:S1-17.:S1-17.

106. Jedrzejas MJ. Extracellular Virulence Factors of Streptococcus pneumoniae. Front Biosci 4 A.D.; 9:891-914.

107. AlonsoDeVelasco E, Verheul AF, Verhoef J, and Snippe H. Streptococcus pneumoniae: virulence factors, pathogenesis, and vaccines. Microbiol Rev 1995; 59:591-603.

108. Carlsen BD, Kawana M, Kawana C, Tomasz A, and Giebink GS. Role of the bacterial cell wall in middle ear inflammation caused by Streptococcus pneumoniae. Infect Immun 1992; 60:2850-4.

109. Gould JM and Weiser JN. The inhibitory effect of C-reactive protein on bacterial phosphorylcholine platelet-activating factor receptor-mediated adherence is blocked by surfactant. J Infect Dis 2002 Aug 1 ;186 (3 ):361 -71 186:361-71.

110. Jedrzejas MJ. Pneumococcal virulence factors: structure and function. Microbiol Mol Biol Rev 2001 Jun ;65 (2 ):187 -207 ; first page , table of contents 65:187-207.

135

111. Crain MJ, Waltman WD, Turner JS et al. Pneumococcal surface protein A (PspA) is serologically highly variable and is expressed by all clinically important capsular serotypes of Streptococcus pneumoniae. Infect Immun 1990; 58:3293-9.

112. McDaniel LS, Yother J, Vijayakumar M, McGarry L, Guild WR, and Briles DE. Use of insertional inactivation to facilitate studies of biological properties of pneumococcal surface protein A (PspA). J Exp Med 1987; 165:381-94.

113. Hollingshead SK, Becker R, and Briles DE. Diversity of PspA: mosaic genes and evidence for past recombination in Streptococcus pneumoniae. Infect Immun 2000 Oct ;68 (10 ):5889 -900 68:5889-900.

114. Jedrzejas MJ, Lamani E, and Becker RS. Characterization of selected strains of pneumococcal surface protein A. J Biol Chem 2001 Aug 31 ;276 (35 ):33121 -8 276:33121-8.

115. Briles DE, Hollingshead SK, Swiatlo E et al. PspA and PspC: their potential for use as pneumococcal vaccines. Microb Drug Resist 1997; 3:401-8.

116. Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, and Tuomanen EI. Streptococcus pneumoniae anchor to activated human cells by the receptor for platelet-activating factor. Nature 1995; 377:435-8.

117. Jarva H, Janulczyk R, Hellwage J, Zipfel PF, Bjorck L, and Meri S. Streptococcus pneumoniae evades complement attack and opsonophagocytosis by expressing the pspC locus-encoded Hic protein that binds to short consensus repeats 8-11 of factor H. J Immunol 2002 Feb 15 ;168 (4 ):1886 -94 168:1886-94.

118. Dave S, Brooks-Walter A, Pangburn MK, and McDaniel LS. Pspc, a pneumococcal surface protein, binds human factor h. Infect Immun 2001 May;69 (5 ):3435 -7 69:3435-7.

119. Paton JC, Berry AM, and Lock RA. Molecular analysis of putative pneumococcal virulence proteins. Microb Drug Resist 1997; 3:1-10.

120. Balachandran P, Hollingshead SK, Paton JC, and Briles DE. The autolytic enzyme LytA of Streptococcus pneumoniae is not responsible for releasing pneumolysin. J Bacteriol 2001 May;183 (10 ):3108 -16 183:3108-16.

121. Hirst RA, Kadioglu A, O'Callaghan C, and Andrew PW. The role of pneumolysin in pneumococcal pneumonia and meningitis. Clin Exp Immunol 2004; 138:195-201.

122. Orihuela CJ, Gao G, Francis KP, Yu J, and Tuomanen EI. Tissue-specific contributions of pneumococcal virulence factors to pathogenesis. J Infect Dis 2004; 190:1661-9.

136

123. Rubins JB and Janoff EN. Pneumolysin: a multifunctional pneumococcal virulence factor. J Lab Clin Med 1998; 131:21-7.

124. Paton JC and Ferrante A. Inhibition of human polymorphonuclear leukocyte respiratory burst, bactericidal activity, and migration by pneumolysin. Infect Immun 1983; 41:1212-6.

125. Mitchell TJ, Andrew PW, Saunders FK, Smith AN, and Boulnois GJ. Complement activation and antibody binding by pneumolysin via a region of the toxin homologous to a human acute-phase protein. Mol Microbiol 1991; 5:1883-8.

126. Paton JC, Rowan-Kelly B, and Ferrante A. Activation of human complement by the pneumococcal toxin pneumolysin. Infect Immun 1984; 43:1085-7.

127. Rubins JB, Charboneau D, Fasching C et al. Distinct roles for pneumolysin's cytotoxic and complement activities in the pathogenesis of pneumococcal pneumonia. Am J Respir Crit Care Med 1996; 153:1339-46.

128. Mokdad AH, Marks JS, Stroup DF, and Gerberding JL. Actual causes of death in the United States, 2000. JAMA 2004; 291:1238-45.

129. Kilmer G, Roberts H, Hughes E et al. Surveillance of certain health behaviors and conditions among states and selected local areas--Behavioral Risk Factor Surveillance System (BRFSS), United States, 2006. MMWR Surveill Summ 2008; 57:1-188.

130. Hughes E, McCracken M, Roberts H et al. Surveillance for certain health behaviors among states and selected local areas--behavioral risk factor surveillance system, United States, 2004. MMWR Surveill Summ 2006; 55:1-124.

131. Alcohol use among adolescents and adults--New Hampshire, 1991-2003. MMWR Morb Mortal Wkly Rep 2004; 53:174-5.

132. Marczinski CA, Combs SW, and Fillmore MT. Increased sensitivity to the disinhibiting effects of alcohol in binge drinkers. Psychol Addict Behav 2007; 21:346-54.

133. Mukamal KJ, Kawachi I, Miller M, and Rimm EB. Drinking frequency and quantity and risk of suicide among men. Soc Psychiatry Psychiatr Epidemiol 2007; 42:153-60.

134. Zakhari S and Li TK. Determinants of alcohol use and abuse: Impact of quantity and frequency patterns on liver disease. Hepatology 2007; 46:2032-9.

135. MacGregor RR and Louria DB. Alcohol and infection. Curr Clin Top Infect Dis 1997; 17:291-315.:291-315.

137

136. Happel KI and Nelson S. Alcohol, immunosuppression, and the lung. Proc Am Thorac Soc 2005; 2:428-32.

137. Zhang P, Bagby GJ, Happel KI, Summer WR, and Nelson S. Pulmonary host defenses and alcohol. Front Biosci 2002; 7:d1314-30.:d1314-d1330.

138. Adams HG and Jordan C. Infections in the alcoholic. Med Clin North Am 1984; 68:179-200.

139. Ruiz M, Ewig S, Torres A et al. Severe community-acquired pneumonia. Risk factors and follow-up epidemiology. Am J Respir Crit Care Med 1999; 160:923-9.

140. Marik PE. The clinical features of severe community-acquired pneumonia presenting as septic shock. Norasept II Study Investigators. J Crit Care 2000; 15:85-90.

141. Musher DM, Alexandraki I, Graviss EA et al. Bacteremic and nonbacteremic pneumococcal pneumonia. A prospective study. Medicine (Baltimore) 2000; 79:210-21.

142. Winterbauer RH, Bedon GA, and Ball WCJ. Recurrent Pneumonia. Predisposing Illness and Clinical Patterns in 158 Patients. Annals of Internal Medicine 1969; 70:689-700.

143. Johnson WDJ. Impaired defense mechanisms associated with acute alcoholism. Ann N Y Acad Sci 1975; 252:343-7:343-7.

144. Wyatt TA, Gentry-Nielsen MJ, Pavlik JA, and Sisson JH. Desensitization of PKA-stimulated ciliary beat frequency in an ethanol-fed rat model of cigarette smoke exposure. Alcohol Clin Exp Res 2004; 28:998-1004.

145. Vander Top EA, Wyatt TA, and Gentry-Nielsen MJ. Smoke exposure exacerbates an ethanol-induced defect in mucociliary clearance of Streptococcus pneumoniae. Alcohol Clin Exp Res 2005; 29:882-7.

146. Elliott MK, Sisson JH, and Wyatt TA. Effects of cigarette smoke and alcohol on ciliated tracheal epithelium and inflammatory cell recruitment. Am J Respir Cell Mol Biol 2007; 36:452-9.

147. McCarthy SP, Lewis CE, and McGee JO. Effects of ethanol on human monocyte/macrophage lysozyme storage and release. Implications for the pathobiology of alcoholic liver disease. J Hepatol 1990; 10:90-8.

148. Lundin L, Hallgren R, and Venge P. Sequential studies on serum-levels of lysozyme, lactoferrin and eosinophil cationic protein in alcoholics after alcohol withdrawal. Scand J Haematol 1980; 25:431-8.

138

149. Brecher AS, Riley C, and Basista MH. Acetaldehyde-modified lysozyme function: its potential implication in the promotion of infection in alcoholics. Alcohol 1995; 12:169-72.

150. Potter BJ and Wang F. Molecular regulation of iron homeostasis and resistance to infection in alcoholics. Front Biosci 2002; 7:d1396-409.:d1396-d1409.

151. Gentry-Nielsen MJ, Preheim LC, Lyman KN, McDonough KH, and Potter BJ. Use of rat models to mimic alterations in iron homeostasis during human alcohol abuse and cirrhosis. Alcohol 2001; 23:71-81.

152. Holguin F, Moss I, Brown LA, and Guidot DM. Chronic ethanol ingestion impairs alveolar type II cell glutathione homeostasis and function and predisposes to endotoxin-mediated acute edematous lung injury in rats. J Clin Invest 1998; 101:761-8.

153. Velasquez A, Bechara RI, Lewis JF et al. Glutathione replacement preserves the functional surfactant phospholipid pool size and decreases sepsis-mediated lung dysfunction in ethanol-fed rats. Alcohol Clin Exp Res 2002 Aug ;26 (8 ):1245 -51 26:1245-51.

154. Baughman RP and Roselle GA. Surfactant deficiency with decreased opsonic activity in a guinea pig model of alcoholism. Alcohol Clin Exp Res 1987; 11:261-4.

155. Bykov I, Junnikkala S, Pekna M, Lindros KO, and Meri S. Effect of chronic ethanol consumption on the expression of complement components and acute-phase proteins in liver. Clin Immunol 2007; 124:213-20.

156. Imhof A, Froehlich M, Brenner H, Boeing H, Pepys MB, and Koenig W. Effect of alcohol consumption on systemic markers of inflammation. Lancet 2001; 357:763-7.

157. Grieco MH, Capra JD, and Paderon H. Reduced serum beta 1c/1a globulin levels in extrarenal disease. Am J Med 1971; 51:340-5.

158. MacGregor RR, Gluckman SJ, and Senior JR. Granulocyte function and levels of immunoglobulins and complement in patients admitted for withdrawal from alcohol. J Infect Dis 1978; 138:747-55.

159. Johnson WD, Stokes P, and Kaye D. The effect of intravenous ethanol on the bactericidal activity of human serum. Yale J Biol Med 1969; 42:71-85.

160. Marr JJ and Spilberg I. A mechanism for decreased resistance to infection by gram-negative organisms during acute alcoholic intoxication. J Lab Clin Med 1975; 86:253-8.

139

161. Spagnuolo PJ and MacGregor RR. Acute thanol effect on chemotaxis and other components of host defense. J Lab Clin Med 1975; 86:24-31.

162. Gluckman SJ, Dvorak VC, and MacGregor RR. Host defenses during prolonged alcohol consumption in a controlled environment. Arch Intern Med 1977; 137:1539-43.

163. Jarvelainen HA, Vakeva A, Lindros KO, and Meri S. Activation of complement components and reduced regulator expression in alcohol-induced liver injury in the rat. Clin Immunol 2002; 105:57-63.

164. D'Souza NB, Nelson S, Summer WR, and Deaciuc IV. Alcohol modulates alveolar macrophage tumor necrosis factor-alpha, superoxide anion, and nitric oxide secretion in the rat. Alcohol Clin Exp Res 1996; 20:156-63.

165. Antony VB, Godbey SW, Hott JW, and Queener SF. Alcohol-induced inhibition of alveolar macrophage oxidant release in vivo and in vitro.

166. Standiford TJ and Danforth JM. Ethanol feeding inhibits proinflammatory cytokine expression from murine alveolar macrophages ex vivo. Alcohol Clin Exp Res 1997; 21:1212-7.

167. Boe DM, Nelson S, Zhang P, and Bagby GJ. Acute ethanol intoxication suppresses lung chemokine production following infection with Streptococcus pneumoniae. J Infect Dis 2001 Nov 1 ;184 (9 ):1134 -42 184:1134-42.

168. Bagasra O, Howeedy A, and Kajdacsy-Balla A. Macrophage function in chronic experimental alcoholism. I. Modulation of surface receptors and phagocytosis. Immunology 1988; 65:405-9.

169. Boe DM, Nelson S, Zhang P, Quinton L, and Bagby GJ. Alcohol-induced suppression of lung chemokine production and the host defense response to Streptococcus pneumoniae. Alcohol Clin Exp Res 2003; 27:1838-45.

170. Zhang P, Bagby GJ, Xie M, Stoltz DA, Summer WR, and Nelson S. Acute ethanol intoxication inhibits neutrophil beta2-integrin expression in rats during endotoxemia [In Process Citation]. Alcohol Clin Exp Res 1998; 22:135-41.

171. Vander Top EA, Perry GA, Snitily MU, and Gentry-Nielsen MJ. Smoke exposure and ethanol ingestion modulate intrapulmonary polymorphonuclear leukocyte killing, but not recruitment or phagocytosis. Alcohol Clin Exp Res 2006; 30:1599-607.

172. Lister PD, Gentry MJ, and Preheim LC. Ethanol impairs neutrophil chemotaxis in vitro but not adherence or recruitment to lungs of rats with experimental pneumococcal pneumonia [published erratum appears in J Infect Dis 1993 Jul;168(1):262]. J Infect Dis 1993; 167:1131-7.

140

173. Mellencamp MA. Effects of ethanol on phagocytosis and bactericidal activity of polymorphonuclear leukocytes for streptococcus pneumoniae. Advances in the Biosciences 1993; 86:473-9.

174. Stoltz DA, Zhang P, Nelson S, Bohm RPJ, Murphey-Corb M, and Bagby GJ. Ethanol suppression of the functional state of polymorphonuclear leukocytes obtained from uninfected and simian immunodeficiency virus infected rhesus macaques [In Process Citation]. Alcohol Clin Exp Res 1999; 23:878-84.

175. Zhang P, Bagby GJ, Stoltz DA, Summer WR, and Nelson S. Granulocyte colony-stimulating factor modulates the pulmonary host response to endotoxin in the absence and presence of acute ethanol intoxication. J Infect Dis 1999; 179:1441-8.

176. Jareo PW, Preheim LC, Lister PD, and Gentry MJ. The effect of ethanol ingestion on killing of Streptococcus pneumoniae, Staphylococcus aureus and Staphylococcus epidermidis by rat neutrophils. Alcohol Alcohol 1995; 30:311-8.

177. Jareo PW, Preheim LC, and Gentry MJ. Ethanol ingestion impairs neutrophil bactericidal mechanisms against Streptococcus pneumoniae. Alcohol Clin Exp Res 1996; 20:1646-52.

178. Sachs CW, Christensen RH, Pratt PC, and Lynn WS. Neutrophil elastase activity and superoxide production are diminished in neutrophils of alcoholics. Am Rev Respir Dis 1990; 141:1249-55.

179. Liu YK. Effects of alcohol on granulocytes and lymphocytes. Semin Hematol 1980; 17:130-6.

180. Bernstein IM, Webster KH, Williams RC, Jr., and Strickland RG. Reduction in circulating T lymphocytes in alcoholic liver disease. Lancet 1974; 2:488-90.

181. Eichner ER, Buchanan B, Smith JW, and Hillman RS. Variations in the hematologic and medical status of alcoholics. Am J Med Sci 1972; 263:35-42.

182. Jerrells TR, Perritt D, Eckardt MJ, and Marietta C. Alterations in interleukin-2 utilization by T-cells from rats treated with an ethanol-containing diet. Alcohol Clin Exp Res 1990; 14:245-9.

183. Saad AJ and Jerrells TR. Flow cytometric and immunohistochemical evaluation of ethanol-induced changes in splenic and thymic lymphoid cell populations. Alcohol Clin Exp Res 1991; 15:796-803.

184. Ewald SJ and Shao H. Ethanol increases apoptotic cell death of thymocytes in vitro. Alcohol Clin Exp Res 1993; 17:359-65.

185. Shao H, Zhou J, and Ewald SJ. Regulation of signal transduction and DNA fragmentation in thymocytes by ethanol. Cell Immunol 1995; 164:11-9.

141

186. Szabo G. Consequences of alcohol consumption on host defence. Alcohol Alcohol 1999; 34:830-41.

187. Berenyi MR, Straus B, and Cruz D. In vitro and in vivo studies of cellular immunity in alcoholic cirrhosis. Am J Dig Dis 1974; 19:199-205.

188. Bjorkholm M. Immunological and hematological abnormalities in chronic alcoholism. Acta Med Scand 1980; 207:197-200.

189. Mikszta JA, Waltenbaugh C, and Kim BS. Impaired antigen presentation by splenocytes of ethanol-consuming C57BL/6 mice. 1995.

190. Shellito JE and Olariu R. Alcohol decreases T-lymphocyte migration into lung tissue in response to Pneumocystis carinii and depletes T-lymphocyte numbers in the spleens of mice. Alcohol Clin Exp Res 1998; 22:658-63.

191. Wilson ID, Onstad G, and Williams RCJ. Serum immunoglobulin concentrations in patients with alcoholic liver disease. Gastroenterology 1969; 57:59-67.

192. Smith WIJ, Van Thiel DH, Whiteside T et al. Altered immunity in male patients with alcoholic liver disease: evidence for defective immune regulation. Alcohol Clin Exp Res 1980; 4:199-206.

193. Spinozzi F, Cimignoli E, Gerli R et al. IgG subclass deficiency and sinopulmonary bacterial infections in patients with alcoholic liver disease. Arch Intern Med 1992; 152:99-104.

194. Jerrells TR, Smith W, and Eckardt MJ. Murine model of ethanol-induced immunosuppression. Alcohol Clin Exp Res 1990; 14:546-50.

195. Jerrells TR, Saad AJ, and Kruger TE. Ethanol-induced suppression of in vivo host defense mechanisms to bacterial infection. Adv Exp Med Biol 1993; 335:153-7.:153-7.

196. Loose LD, Stege T, and Dr Luzio NR. The Influence of Acute and Chronic Ethanol or Bourbon Administration on Phagocytic and Immune Responses in Rats. Exp Mol Pathol 1975; 23:459-72.

197. Chang MP, Wang Q, and Norman DC. Diminished proliferation of B blast cell in response to cytokines in ethanol-consuming mice. Immunopharmacol Immunotoxicol 2002; 24:69-82.

198. Cigarette smoking among adults--United States, 2004. MMWR Morb Mortal Wkly Rep 2005; 54:1121-4.

199. Chen K and Kandel DB. The natural history of drug use from adolescence to the mid-thirties in a general population sample. Am J Public Health 1995; 85:41-7.

142

200. Substance Abuse and Mental Health Services Administration. 2003 National Survey on Drug Use & Health: Overview. 2004. Rockville, MD, (Office of Applied Studies, NSDUH Series H-24, DHHS Publication No. SMA 04-3963).

Ref Type: Generic

201. Sopori M. Effects of cigarette smoke on the immune system. Nat Rev Immunol 2002; 2:372-7.

202. Achievements in Public Health, 1900-1999: Tobacco Use--United States, 1900-1999. Morbidity and Mortality Weekly Report 48[43], 986-993. 11-5-1999. Centers for Disease Control and Prevention.

Ref Type: Magazine Article

203. Annual smoking-attributable mortality, years of potential life lost, and productivity losses--United States, 1997-2001. MMWR Morb Mortal Wkly Rep 2005; 54:625-8.

204. Sundaram R, Shulman L, and Fein AM. Trends in tobacco use. Med Clin North Am 2004; 88:1391-7, ix.

205. Ezzati M and Lopez AD. Estimates of global mortality attributable to smoking in 2000. Lancet 2003; 362:847-52.

206. Lipsky BA, Boyko EJ, Inui TS, and Koepsell TD. Risk factors for acquiring pneumococcal infections. Arch Intern Med 1986; 146:2179-85.

207. Almirall J, Bolibar I, Balanzo X, and Gonzalez CA. Risk factors for community-acquired pneumonia in adults: a population-based case-control study. Eur Respir J 1999; 13:349-55.

208. Murin S and Bilello KS. Respiratory tract infections: another reason not to smoke. Cleve Clin J Med 2005; 72:916-20.

209. Nuorti JP, Butler JC, Farley MM et al. Cigarette smoking and invasive pneumococcal disease. Active Bacterial Core Surveillance Team. N Engl J Med 2000 Mar 9 ;342 (10 ):681 -9 342:681-9.

210. Plouffe JF, Breiman RF, and Facklam RR. Bacteremia with Streptococcus pneumoniae. Implications for therapy and prevention. Franklin County Pneumonia Study Group. JAMA 1996; 275:194-8.

211. Pastor P, Medley F, and Murphy TV. Invasive pneumococcal disease in Dallas County, Texas: results from population-based surveillance in 1995 [In Process Citation]. Clin Infect Dis 1998; 26:590-5.

212. Raman AS, Swinburne AJ, and Fedullo AJ. Pneumococcal adherence to the buccal epithelial cells of cigarette smokers. Chest 1983; 83:23-7.

143

213. Foster WM, Langenback EG, and Bergofsky EH. Disassociation in the mucociliary function of central and peripheral airways of asymptomatic smokers. Am Rev Respir Dis 1985; 132:633-9.

214. Roberts CM, Cairns D, Bryant DH et al. Changes in epithelial lining fluid albumin associated with smoking and interstitial lung disease. Eur Respir J 1993; 6:110-5.

215. Sundram FX. Clinical studies of alveolar-capillary permeability using technetium-99m DTPA aerosol. Ann Nucl Med 1995; 9:171-8.

216. Greenberg D, Givon-Lavi N, Broides A, Blancovich I, Peled N, and Dagan R. The contribution of smoking and exposure to tobacco smoke to Streptococcus pneumoniae and Haemophilus influenzae carriage in children and their mothers. Clin Infect Dis 2006; 42:897-903.

217. Brook I and Gober AE. Recovery of potential pathogens and interfering bacteria in the nasopharynx of otitis media-prone children and their smoking and nonsmoking parents. Arch Otolaryngol Head Neck Surg 2005; 131:509-12.

218. Brook I and Gober AE. Recovery of potential pathogens and interfering bacteria in the nasopharynx of smokers and nonsmokers. Chest 2005; 127:2072-5.

219. El Ahmer OR, Essery SD, Saadi AT et al. The effect of cigarette smoke on adherence of respiratory pathogens to buccal epithelial cells [In Process Citation]. FEMS Immunol Med Microbiol 1999; 23:27-36.

220. Piatti G, Gazzola T, and Allegra L. Bacterial Adherence in Smokers and Non-Smokers. 36. Vol 6. 1997:481-484.

221. Fainstein V and Musher DM. Bacterial adherence to pharyngeal cells in smokers, nonsmokers, and chronic bronchitics. Infect Immun 1979; 26:178-82.

222. Barton JR, Riad MA, Gaze MN, Maran AG, and Ferguson A. Mucosal immunodeficiency in smokers, and in patients with epithelial head and neck tumours. Gut 1990; 31:378-82.

223. Gotoh T, Ueda S, Nakayama T, Takishita Y, Yasuoka S, and Tsubura E. Protein components of bronchoalveolar lavage fluids from non-smokers and smokers. Eur J Respir Dis 1983; 64:369-77.

224. Wyatt TA, Schmidt SC, Rennard SI, Tuma DJ, and Sisson JH. Acetaldehyde-stimulated PKC activity in airway epithelial cells treated with smoke extract from normal and smokeless cigarettes. Proc Soc Exp Biol Med 2000; 225:91-7.

225. Sherman CB. The health consequences of cigarette smoking. Pulmonary diseases. Med Clin North Am 1992; 76:355-75.

144

226. Verra F, Escudier E, Lebargy F, Bernaudin JF, De Cremoux H, and Bignon J. Ciliary abnormalities in bronchial epithelium of smokers, ex-smokers, and nonsmokers. Am J Respir Crit Care Med 1995; 151:630-4.

227. Stanley PJ, Wilson R, Greenstone MA, MacWilliam L, and Cole PJ. Effect of cigarette smoking on nasal mucociliary clearance and ciliary beat frequency. Thorax 1986; 41:519-23.

228. Merkel D, Rist W, Seither P, Weith A, and Lenter MC. Proteomic study of human bronchoalveolar lavage fluids from smokers with chronic obstructive pulmonary disease by combining surface-enhanced laser desorption/ionization-mass spectrometry profiling with mass spectrometric protein identification. Proteomics 2005; 5:2972-80.

229. Hinman LM, Stevens CA, Matthay RA, and Gee JB. Elastase and lysozyme activities in human alveolar macrophages. Effects of cigarette smoking. Am Rev Respir Dis 1980; 121:263-71.

230. Thompson AB, Bohling T, Payvandi F, and Rennard SI. Lower respiratory tract lactoferrin and lysozyme arise primarily in the airways and are elevated in association with chronic bronchitis. J Lab Clin Med 1990; 115:148-58.

231. Betsuyaku T, Kuroki Y, Nagai K, Nasuhara Y, and Nishimura M. Effects of ageing and smoking on SP-A and SP-D levels in bronchoalveolar lavage fluid. Eur Respir J 2004; 24:964-70.

232. Honda Y, Takahashi H, Kuroki Y, Akino T, and Abe S. Decreased contents of surfactant proteins A and D in BAL fluids of healthy smokers. Chest 1996; 109:1006-9.

233. Subramaniam S, Whitsett JA, Hull W, and Gairola CG. Alteration of pulmonary surfactant proteins in rats chronically exposed to cigarette smoke. Toxicol Appl Pharmacol 1996; 140:274-80.

234. Wirtz HR and Schmidt M. Acute influence of cigarette smoke on secretion of pulmonary surfactant in rat alveolar type II cells in culture. Eur Respir J 1996; 9:24-32.

235. Hirama N, Shibata Y, Otake K et al. Increased surfactant protein-D and foamy macrophages in smoking-induced mouse emphysema. Respirology 2007; 12:191-201.

236. Shibata Y, Abe S, Inoue S et al. Altered expression of antimicrobial molecules in cigarette smoke-exposed emphysematous mice lungs. Respirology 2008.

237. Kew RR, Ghebrehiwet B, and Janoff A. Cigarette smoke can activate the alternative pathway of complement in vitro by modifying the third component of complement. J Clin Invest 1985; 75:1000-7.

145

238. Kew RR, Ghebrehiwet B, and Janoff A. Characterization of the third component of complement (C3) after activation by cigarette smoke. Clin Immunol Immunopathol 1987; 44:248-58.

239. Miller RD, Kueppers F, and Offord KP. Serum concentrations of C3 and C4 of the complement system in patients with chronic obstructive pulmonary disease. J Lab Clin Med 1980; 95:266-71.

240. Chauhan S, Gupta MK, Goyal A, and Dasgupta DJ. Alterations in immunoglobulin & complement levels in chronic obstructive pulmonary disease. Indian J Med Res 1990; 92:241-5.:241-5.

241. Levitzky YS, Guo CY, Rong J et al. Relation of smoking status to a panel of inflammatory markers: The Framingham offspring. Atherosclerosis 2008.

242. Frohlich M, Sund M, Lowel H, Imhof A, Hoffmeister A, and Koenig W. Independent association of various smoking characteristics with markers of systemic inflammation in men. Results from a representative sample of the general population (MONICA Augsburg Survey 1994/95). Eur Heart J 2003; 24:1365-72.

243. Hastie CE, Haw S, and Pell JP. Impact of smoking cessation and lifetime exposure on C-reactive protein. Nicotine Tob Res 2008; 10:637-42.

244. Gan WQ, Man SF, and Sin DD. The interactions between cigarette smoking and reduced lung function on systemic inflammation. Chest 2005; 127:558-64.

245. Mancini NM, Bene MC, Gerard H et al. Early effects of short-time cigarette smoking on the human lung: a study of bronchoalveolar lavage fluids. Lung 1993; 171:277-91.

246. Thomassen MJ, Barna BP, Wiedemann HP, Farmer M, and Ahmad M. Human alveolar macrophage function: differences between smokers and nonsmokers. J Leukoc Biol 1988; 44:313-8.

247. Herlihy JP, Vermeulen MW, Joseph PM, and Hales CA. Impaired alveolar macrophage function in smoke inhalation injury. J Cell Physiol 1995; 163:1-8.

248. Sherman MP, Campbell LA, Gong HJ, Roth MD, and Tashkin DP. Antimicrobial and respiratory burst characteristics of pulmonary alveolar macrophages recovered from smokers of marijuana alone, smokers of tobacco alone, smokers of marijuana and tobacco, and nonsmokers. Am Rev Respir Dis 1991; 144:1351-6.

249. Morrison D, Strieter RM, Donnelly SC, Burdick MD, Kunkel SL, and MacNee W. Neutrophil chemokines in bronchoalveolar lavage fluid and leukocyte- conditioned medium from nonsmokers and smokers. Eur Respir J 1998; 12:1067-72.

146

250. Bridges RB and Hsieh L. Effects of cigarette smoke fractions on the chemotaxis of polymorphonuclear leukocytes. J Leukoc Biol 1986; 40:73-85.

251. Kuschner WG, D'Alessandro A, Wong H, and Blanc PD. Dose-dependent cigarette smoking-related inflammatory responses in healthy adults. Eur Respir J 1996; 9:1989-94.

252. Wang H, Ye Y, Zhu M, and Cho C. Increased interleukin-8 expression by cigarette smoke extract in endothelial cells. Environ Toxicol Pharmacol 2000; 9:19-23.

253. Sato E, Koyama S, Takamizawa A et al. Smoke extract stimulates lung fibroblasts to release neutrophil and monocyte chemotactic activities. Am J Physiol 1999; 277:L1149-L1157.

254. Masubuchi T, Koyama S, Sato E et al. Smoke extract stimulates lung epithelial cells to release neutrophil and monocyte chemotactic activity. Am J Pathol 1998; 153:1903-12.

255. Gillespie MN, Owasoyo JO, Kojima S, and Jay M. Enhanced chemotaxis and superoxide anion production by polymorphonuclear leukocytes from nicotine-treated and smoke-exposed rats. Toxicology 1987; 45:45-52.

256. Numabe Y, Ogawa T, Kamoi H et al. Phagocytic function of salivary PMN after smoking or secondary smoking. Ann Periodontol 1998; 3:102-7.

257. Stringer KA, Tobias M, O'Neill HC, and Franklin CC. Cigarette smoke extract-induced suppression of caspase-3-like activity impairs human neutrophil phagocytosis. Am J Physiol Lung Cell Mol Physiol 2007; 292:L1572-L1579.

258. Zappacosta B, Persichilli S, Minucci A et al. Effect of aqueous cigarette smoke extract on the chemiluminescence kinetics of polymorphonuclear leukocytes and on their glycolytic and phagocytic activity. Luminescence 2001; 16:315-9.

259. Ryder MI, Fujitaki R, Johnson G, and Hyun W. Alterations of neutrophil oxidative burst by in vitro smoke exposure: implications for oral and systemic diseases. Ann Periodontol 1998; 3:76-87.

260. Koethe SM, Kuhnmuench JR, and Becker CG. Neutrophil priming by cigarette smoke condensate and a tobacco anti-idiotypic antibody. Am J Pathol 2000 Nov ;157 (5 ):1735 -43 157:1735-43.

261. Drannik AG, Pouladi MA, Robbins CS, Goncharova SI, Kianpour S, and Stampfli MR. Impact of cigarette smoke on clearance and inflammation after Pseudomonas aeruginosa infection. Am J Respir Crit Care Med 2004; 170:1164-71.

262. Holt PG and Keast D. Environmentally induced changes in immunological function: acute and chronic effects of inhalation of tobacco smoke and other

147

atmospheric contaminants in man and experimental animals. Bacteriol Rev 1977; 41:205-16.

263. Holt PG. Immune and inflammatory function in cigarette smokers. Thorax 1987; 42:241-9.

264. Johnson JD, Houchens DP, Kluwe WM, Craig DK, and Fisher GL. Effects of mainstream and environmental tobacco smoke on the immune system in animals and humans: a review. Crit Rev Toxicol 1990; 20:369-95.

265. Hughes DA, Haslam PL, Townsend PJ, and Turner-Warwick M. Numerical and functional alterations in circulatory lymphocytes in cigarette smokers. Clin Exp Immunol 1985; 61:459-66.

266. Geng Y, Savage SM, Johnson LJ, Seagrave J, and Sopori ML. Effects of nicotine on the immune response. I. Chronic exposure to nicotine impairs antigen receptor-mediated signal transduction in lymphocytes. Toxicol Appl Pharmacol 1995; 135:268-78.

267. Geng Y, Savage SM, Razani-Boroujerdi S, and Sopori ML. Effects of nicotine on the immune response. II. Chronic nicotine treatment induces T cell anergy. J Immunol 1996; 156:2384-90.

268. Chang JC, Distler SG, and Kaplan AM. Tobacco smoke suppresses T cells but not antigen-presenting cells in the lung-associated lymph nodes. Toxicol Appl Pharmacol 1990; 102:514-23.

269. Suzuki N, Wakisaka S, Takeba Y, Mihara S, and Sakane T. Effects of cigarette smoking on Fas/Fas ligand expression of human lymphocytes. Cell Immunol 1999; 192:48-53.

270. Kalra R, Singh SP, Savage SM, Finch GL, and Sopori ML. Effects of cigarette smoke on immune response: chronic exposure to cigarette smoke impairs antigen-mediated signaling in T cells and depletes IP3-sensitive Ca(2+) stores. J Pharmacol Exp Ther 2000; 293:166-71.

271. Sopori ML, Cherian S, Chilukuri R, and Shopp GM. Cigarette smoke causes inhibition of the immune response to intratracheally administered antigens. Toxicol Appl Pharmacol 1989; 97:489-99.

272. Savage SM, Donaldson LA, Cherian S, Chilukuri R, White VA, and Sopori ML. Effects of cigarette smoke on the immune response. II. Chronic exposure to cigarette smoke inhibits surface immunoglobulin-mediated responses in B cells. Toxicol Appl Pharmacol 1991; 111:523-9.

273. Thomas WR, Holt PG, and Keast D. Humoral immune response of mice with long-term exposure to cigarette smoke. Arch Environ Health 1975; 30:78-80.

148

274. Goud SN, Kaplan AM, and Subbarao B. Effects of cigarette smoke on the antibody responses to thymic independent antigens from different lymphoid tissues of mice. Arch Toxicol 1992; 66:164-9.

275. Ferson M, Edwards A, Lind A, Milton GW, and Hersey P. Low natural killer-cell activity and immunoglobulin levels associated with smoking in human subjects. Int J Cancer 1979; 23:603-9.

276. Moszczynski P, Zabinski Z, Moszczynski P, Jr., Rutowski J, Slowinski S, and Tabarowski Z. Immunological findings in cigarette smokers. Toxicol Lett 2001; 118:121-7.

277. Mathews JD, Whittingham S, Hooper BM, Mackay IR, and Stenhouse NS. Association of autoantibodies with smoking, cardiovascular morbidity, and death in the Busselton population. Lancet 1973; 2:754-8.

278. Masdottir B, Jonsson T, Manfredsdottir V, Vikingsson A, Brekkan A, and Valdimarsson H. Smoking, rheumatoid factor isotypes and severity of rheumatoid arthritis. Rheumatology (Oxford) 2000; 39:1202-5.

279. Bien TH and Burge R. Smoking and drinking: a review of the literature. Int J Addict 1990; 25:1429-54.

280. Gentry-Nielsen MJ, Top EV, Snitily MU, Casey CA, and Preheim LC. A rat model to determine the biomedical consequences of concurrent ethanol ingestion and cigarette smoke exposure. Alcohol Clin Exp Res 2004; 28:1120-8.

281. Teague SV, Pinkerton KE, Goldsmith M et al. Sidestream Cigarette Smoke Generation and Exposure System for Environmental Tobacco Smoke Studies. Inhalation Toxicology 1994; 6:79-93.

282. Wu HY, Nahm MH, Guo Y, Russell MW, and Briles DE. Intranasal immunization of mice with PspA (pneumococcal surface protein A) can prevent intranasal carriage, pulmonary infection, and sepsis with Streptococcus pneumoniae. J Infect Dis 1997; 175:839-46.

283. Preheim LC, Gentry MJ, and Snitily MU. Pulmonary recruitment, adherence, and chemotaxis of neutrophils in a rat model of cirrhosis and pneumococcal pneumonia. J Infect Dis 1991; 164:1203-6.

284. Snitily MU, Gentry MJ, Mellencamp MA, and Preheim LC. A simple method for collection of blood from the rat foot. Lab Anim Sci 1991; 41:285-7.

285. Brooks-Walter A, Briles DE, and Hollingshead SK. The pspC gene of Streptococcus pneumoniae encodes a polymorphic protein, PspC, which elicits cross-reactive antibodies to PspA and provides immunity to pneumococcal bacteremia. Infect Immun 1999; 67:6533-42.

149

286. Burnham EL, Brown LA, Halls L, and Moss M. Effects of chronic alcohol abuse on alveolar epithelial barrier function and glutathione homeostasis. Alcohol Clin Exp Res 2003; 27:1167-72.

287. Skold CM, Andersson K, Hed J, and Eklund A. Short-term in vivo exposure to cigarette-smoke increases the fluorescence in rat alveolar macrophages. Eur Respir J 1993; 6:1169-72.

288. Skold CM, Hed J, and Eklund A. Smoking cessation rapidly reduces cell recovery in bronchoalveolar lavage fluid, while alveolar macrophage fluorescence remains high. Chest 1992; 101:989-95.

289. Higashimoto Y, Fukuchi Y, Ishida K et al. Effect of chronic tobacco smoke exposure on the function of alveolar macrophages in mice. Respiration 1994; 61:23-7.

290. Hoidal JR and Niewoehner DE. Lung phagocyte recruitment and metabolic alterations induced by cigarette smoke in humans and in hamsters. Am Rev Respir Dis 1982; 126:548-52.

291. Perlino CA and Rimland D. Alcoholism, leukopenia, and pneumococcal sepsis. Am Rev Respir Dis 1985; 132:757-60.

292. McFarland W and Libre EP. Abnormal Leukocyte Response in Alcoholism. Annals of Internal Medicine 1963; 59:865-77.

293. Terashima T, Wiggs B, English D, Hogg JC, and van Eeden SF. The effect of cigarette smoking on the bone marrow. Am J Respir Crit Care Med 1997; 155:1021-6.

294. Matheson M, Rynell AC, McClean M, and Berend N. Cigarette smoking increases neutrophil formyl methionyl leucyl phenylalanine receptor numbers. Chest 2003; 123:1642-6.

295. Oak S, Mandrekar P, Catalano D, Kodys K, and Szabo G. TLR2- and TLR4-mediated signals determine attenuation or augmentation of inflammation by acute alcohol in monocytes. J Immunol 2006; 176:7628-35.

296. Dai Q and Pruett SB. Ethanol suppresses LPS-induced Toll-like receptor 4 clustering, reorganization of the actin cytoskeleton, and associated TNF-alpha production. Alcohol Clin Exp Res 2006; 30:1436-44.

297. Dolganiuc A, Bakis G, Kodys K, Mandrekar P, and Szabo G. Acute ethanol treatment modulates Toll-like receptor-4 association with lipid rafts. Alcohol Clin Exp Res 2006; 30:76-85.

150

298. Goral J and Kovacs EJ. In vivo ethanol exposure down-regulates TLR2-, TLR4-, and TLR9-mediated macrophage inflammatory response by limiting p38 and ERK1/2 activation. J Immunol 2005; 174:456-63.

299. Droemann D, Goldmann T, Tiedje T, Zabel P, Dalhoff K, and Schaaf B. Toll-like receptor 2 expression is decreased on alveolar macrophages in cigarette smokers and COPD patients. Respir Res 2005; 6:68.:68.

300. MacRedmond RE, Greene CM, Dorscheid DR, McElvaney NG, and O'Neill SJ. Epithelial expression of TLR4 is modulated in COPD and by steroids, salmeterol and cigarette smoke. Respir Res 2007; 8:84.:84.

301. Laskin DL, Robertson FM, Pilaro AM, and Laskin JD. Activation of liver macrophages following phenobarbital treatment of rats. Hepatology 1988; 8:1051-5.