<<

Licentiate thesis from the Department of , Wenner-Gren Institute, Stockholm University, Sweden

Innate and adaptive immune responses in the . Contribution to protection against mycobacterial

Olga Daniela Chuquimia Flores

Stockholm 2011

SUMMARY Host defense against Mycobacterium (Mtb) is mediated by a combination of innate and adaptive . In this thesis we investigated the role of components of innate system such as TLR2 signalling and alveolar epithelial cells type II (AEC II) in the immune responses against mycobacterial infections. Since TLR2 has been shown to be important in the defense against mycobacterial infections; in paper I we investigated the role of TLR2 to generate acquired immune responses. We compared both humoral and cellular immune responses in TLR2-/- and WT (wild type) mice immunized with the mycobacterial 19kDa (TLR2 ligand) or Ag85A (non-TLR2 ligand). We did not find any differences in the humoral responses in both mouse strains. However, we found some deficiencies in the memory compartment of TLR2-/- mice immunized with 19kDa. In addition, the presenting cells (APC) compartment in TLR2-/- mice, for instance marrow derived (BMM) and pulmonary macrophages (PM) in this study, has also shown deficiencies. This effect was more evident when PM were used as APC. We next evaluated the responses in both BMM and PM upon stimulation with anti-CD40 and TLR ligands where PM were the low responders to TLR2 ligand and to anti-CD40 both in the production of different and in the up-regulation of the co-stimulatory molecules. Together, our results have demonstrated the importance of TLR2 in the generation of specific immune responses. In paper II, we investigated the role of AEC II in the defense against mycobacterial infections. AEC II have been suggested to play an important role in the local immune responses to inhaled . First, we compared murine AEC II with PM in their ability to take up and control mycobacterial growth and their capacity as APCs. AEC II were able to internalize and control bacterial growth as well as presenting antigen to memory T cells. In addition, both cells types were compared in their capacity to produce cytokines, and other factors where AEC II exhibited a different pattern of than PM. Also, a more complete profile of AEC II responses reveled that AEC II were able to secrete different factors important to generated various effects in others cells. The major finding in this study was that upon TNF, AEC II produced MCP-1 a involved in the recruitment /macrophages to the sites of . Since TNF is predominantely produced by macrophages, we speculate that both cell types may communicate and influence each other. In conclusion, our results provide more evidence of the important role of AEC II in the immune responses in the .

2

LIST OF PAPERS

This thesis is based on the following original manuscript, which will be referred to by their roman numeral in the text.

I. Muhammad J. Rahman, Olga D. Chuquimia, Dagbjort H. Petursdottir, Natalia Periolo, Mahavir Singh and Carmen Fernández. Contribution of TLR2 signalling to the specific . Deficiencies in T-cell memory and antigen presenting cell compartments in the TLR2 knockout mice. Manuscript.

II. Olga D. Chuquimia, Dagbjort H. Petursdottir, Muhammad J. Rahman, Katharina Hartl, Mahavir Singh and Carmen Fernández. The role of alveolar epithelial cell type II in initiating and shaping pulmonary immune responses. Communication between the innate and adaptive immune systems. Submitted.

3

TABLE OF CONTENTS Page

SUMMARY 2 LIST OF PAPER 3 TABLE OF CONTENTS 4 LIST OF ABBREVIATIONS 5 INTRODUCTION 6 Mucosal immunity in the respiratory tract 6 Innate and adaptive responses in the 7 Tuberculosis 10 Mycobacterium tuberculosis 10 Pathogenesis of tuberculosis 11 Immune responses against tuberculosis 12 Innate immune cells 12 Adaptive immune cells 16 Role of TLRs in mycobacterial Infection 18 Cytokines and chemokines in TB infection 19 Pro-inflammatory Cytokines 19 Anti-inflammatory cytokines 22 Chemokines 23 GM-CSF 25 PRESENT STUDY 26 Aims 26 General aim 26 Specific aims 26 Materials and Methods 26 Results and Discussion 27 Paper I 27 Paper II 31 Concluding Remarks 35 Future Plans 35 ACKNOWLEDGEMENTS 36 REFERENCES 37

4

LIST OF ABBREVIATIONS

AEC II Alveolar epithelial cells type II AM Alveolar macrophages APC Antigen-presenting cell BCG Bacillus Calmette-Guérin CT Cholera DC Dendritic cells DC-SIGN -specific intracellular adhesion molecule-3-grabbing non-integrin ELISA - linked immunosorbet assay HIV immunodefiency HK-BCG Heat- killed BCG IFN-α Interferon alpha IFN-γ IL Interleukin i.n. Intranasal IP-10 Interferon gamma-induced 10 kDa i.v. Intravenous kDa KiloDalton KC keratinocyte-derived chemokine LAM Lipoarabinomannan LM lipomannan Lys-BCG BCG lysate LPS Lipopolysaccharide MALT Mucosa-associated lymphoid MCP-1 -chemotactic protein-1 MHC Major Histocompatibility complex MIP-2 -inflammatory protein-2 MMP-9 Matrix metallopeptidase-9 Mtb Mycobacterium tuberculosis Pam3 Lipopeptide tripalmitoyl-S-glycerylcysteine PM Pulmonary macrophages s.c. Subcutaneously TB Tuberculosis TCR T-cell receptor TGF-β Transforming growth factor beta-β TLRs Toll-like receptors TNF Tumor-necrosis factor WT Wild type

5

INTRODUCTION

Mucosal immunity in the respiratory tract The mucosal surface is a major portal of entry for pathogens in the respiratory, gastrointestinal, urogenital tracks and other moist surfaces in the lining of reproductive track. The mucosal surfaces have the task of providing protection of the membrane against infection as a natural barrier, preventing the uptake of antigens (microorganisms and foreign particles). Another important task is the preservation of mucosal with the production low immune responses to harmless antigens (mucosal tolerance). In the respiratory tract, the mucosal (local) and systemic immune system are involved in the defense and protection against inhaled microorganisms (control of movement and infection levels). However, the innate branch of the mucosal immune system is critical for controlling infection in the early stages of exposure to inhaled microorganisms. Once the inhaled arrive to the mucosa surface, they are trapped by mucus and removed toward the and swallowed. Antimicrobial peptides are produced and secreted by the surface of the respiratory tracks to kill many microorganisms that have penetrated the mucous layer. Those bacteria that are resistant to antimicrobial peptides are killed by a variety of reactive species produced by . Finally, persistent bacterial infections which escape the are eliminated by the (1,2). An important function on the respiratory tract is the maintenance of local immunological homeostasis and therefore the integrity of surfaces related to the discriminatory functions of the immune system to their limits. The incoming antigens in the respiratory tract are dominated by highly immunogenic but harmless of and animal origin. The host could undergo premature from chronic airway if these harmless proteins could induce strong adaptive immune responses. For that, it is imperative that the respiratory mucosal immune system discriminates the background antigenic noise from the much rarer signals transmitted by pathogen associated antigens to produce an immunological balance in the air ways. Memory T-cell responses have to be tightly regulated to minimize damage in the local epithelial cell surfaces, particularly for the alveolar gas exchange surfaces, as these tissues contain the largest vascular bed in the body and function as a magnet for circulating memory (3).

6

Innate and adaptive responses in the lung The lung consists of two major anatomic compartments: the vascular and the airway compartment. Endothelial cells in the , , and are the cells most actively involved in an inflammatory response in the vascular system. Epithelial cells therefore could have similar function in the respiratory compartment. Distal airway-epithelial cells and alveolar epithelial cells are vital for maintenance of the pulmonary air- barrier. The alveolar epithelium is composed of Type I Alveolar epithelial cells or membranous pneumocytes and Type II Alveolar epithelial cells (AEC II) or granular pneumocytes. Type I epithelial cells are squamous, large thin cells that cover 90-95% of the alveolar surface and are essentially involved in gaseous exchange. AEC II are cuboidal cells that constitute 15% of total parenchymal lungs cells and cover about 7% of the total alveolar surface. AEC II contain characteristic lamellar inclusion bodies, the intracellular storage of pulmonary surface-active material () (4,5). They are also considered as progenitor cells capable of proliferating and differentiating into type I cells. Recent evidence suggests that airway epithelial cells might also act as immune effector cells in response to harmful exogenous stimuli. Several studies have shown that airway epithelial cells express on their surface adhesion molecules and secrete various immune molecules such as cytokines, chemokines and other factors (6-11). Through the expression and production of these inflammatory mediators, not only the vascular but also the airway epithelium is thought to play an important role in the initiation and exacerbation of an inflammatory response within the airways. Also, in the lungs the presence of biosensors as pattern recognition receptors (PRR) is important in the lung immune responses. The toll-like receptors (TLRs) are able to induce a signalling pathway with activation of kinases and nuclear factors, resulting in transcription of inflammatory mediators such as tumor necrosis factor (TNF) or members of the interleukin family (12). In addition, the leukocyte homing to sites of acute inflammation is a crucial step during an inflammatory response. Adhesion molecules play a major role in the inflammatory process by mediating adherence of leukocytes to the and initiating extravasation of these cells. The intercellular adhesion molecule-1 (ICAM-1), a member of the immunoglobulin superfamily, is a cell surface glycoprotein and a ligand for the β2-integrins CD11a/CD18 and CD11b/CD18 on leukocytes. It is up-regulated by a variety of inflammatory stimuli such as endotoxin and different cytokines. The ICAM-1 is expressed by endothelial and epithelial cells but its functional role could be different in both cells.

7

In terms of the different immunological functions, the lung has also been divided into two compartments: the conducting airways overlayed by mucosal tissue, and the lung Figure 1, which comprises thin-walled alveoli that are specialized for gas exchange.

Figure 1: Local immune cells in the two lung compartments. (Nature rev.2008; 8:142-152, reprinted with permission from NPG)

The basic morphology of the conducting airways is similar and consists of a surface epithelium composed largely of ciliated and secretory cells overlying subepithelial tissue that consists predominantly of connective tissues and glands. The proportion and type of these elements vary at different levels of the conducting system. The cells in the conducting airways with the production of locally secreted immunoglobulin (Ig)A provide mechanisms for of inhaled antigens. The cells of the immune system are present

8 within the epithelium of all conducting airways. Dendritic cells (DC) and macrophages have dense networks in the epithelium. The major population of DC is composed of myeloid DC subset but also the plasmacytoid DC can be found in the mucosa airway. Resident airway mucosal DC are specialized in immune surveillance with a high capacity for antigen uptake, but a reduced ability to stimulate T cells. They are strategically positioned for antigen uptake both within and directly under the surface epithelium and continuously sample incoming airborne antigen by extending their dendrites through the intact epithelial layer into the airway (13,14). can be found either singly or in clusters in the airway lamina propria and in the submucosa. Effector and memory CD4+ and CD8+ T cells (defined by their expression of CD45RO), as well as B cells are also present in the airway mucosa (in the intraepithelial and within the underlying lamina propria) and may play a role in the constitution of bronchial-associated lymphoid tissue (15) which has been suggested to have a significant role in local immunological homeostasis in the respiratory tract early in life. Most intraepithelial T cells express CD8+, whereas CD4+ T cells are more frequently found in the lamina propria (16,17). B cells might be contributing to local in the nodes that drain the lungs. Plasma cells are in the lamina propia and the role of these cells is mainly the production of polymeric IgA but also IgM to clear inhaled pathogens (18). Other types of cells such as mast cells, basophiles, and have also been found in the lamina propria. The lung parenchyma consists of alveoli that are separated by fine vascularised interstitial tissue. Lung parenchymal DC, macrophages, and T cells arise in the alveolar space, the alveolar-epithelial layer and the interstitium. In the steady-state conditions the alveolar space (as reflected by broncho-alveolar lavage fluid composition) consists of 80- 90% macrophages, the remainder being T cells and DCs. However, it has been found a large sequestered T-cell population in the lung parenchyma but its role in the local mucosal homeostasis is still unclear (18-24). The lung parenchyma also contains B cells and mast cells, but no plasma cells.

9

Tuberculosis Tuberculosis (TB) is an infectious caused by Mycobacterium tuberculosis (Mtb), which most commonly affects the lungs. It is transmitted from person to person via droplets from the and lungs of people with an active . In 2009, there were an estimated of 9.4 million cases of TB globally (equivalent to 137 cases per 100 000 population). Of this incident an estimated 1.1 million (12%) were HIV-positive. These numbers are slightly lower than those reported in previous years, reflecting better estimates as well as reductions in HIV prevalence in the general population. Of these HIV-positive TB cases, approximately 80% are in the African region (24). Bacillus Calmette-Guérin (BCG) is the unique available against TB, BCG is prepared from a strain of the attenuated (weakened) live bovine tuberculosis bacillus, Mycobacterium bovis. The programs with BCG in new-borns and protect and reduce risk against childhood TB meningitis and milliary TB (vaccinated near 90%). However, its efficacy diminishes with time and it affords only variable protection against pulmonary disease. Consequently, to find a more effective TB vaccine is considered a global priority (25-27). Over one hundred TB vaccine candidates (DNA and subunit ) have been developed, using different approaches to induce protective immunity (27). Until now, there is not a new vaccine able to achieve a level of protection better than BCG. However, models of vaccination using with BCG and boosting with mycobacterial antigens strategy have been used previously to achieve high levels of protection (28). We have demonstrated that BCG priming and HBHA (heparin-binding hemagglutinin, a mycobacterial antigen) boosting in neonatal mice induce protective immune responses (29).

Mycobacterium tuberculosis Mtb is a fairly large non motile rod-shaped bacterium distantly related to the Actinomycetes. Many nonpathogenic mycobacteria are components of the normal flora in , found most often in dry and oily locals. The rods are 2-4 µm in length and 0.2-0.5 µm in width. The tubercle bacilli are obligatory aerobic intracellular pathogens with predilection for the lung tissue rich in oxygen supply. Mtb has a slow generation time, 15-20 hours; this physiological characteristic may contribute to its virulence (30). The diagnostic of TB is based on the determination of TB in sputum samples, , , PCR and other types of studies such as culture of mycobacteria in the Löwenstein-Jensen (LJ), Kirchner, or Middlebrook media (7H9, 7H10, and 7H11) (30).

10

Pathogenesis of Tuberculosis Most frequently Mtb enter the body via the respiratory tract. Once Mtb gets in the pulmonary alveoli, the TB infection (primary TB) begins with the invasion and replication of the tubercle bacilli into the endosomes of macrophages. However, it has been described that DC can take up and transport the bacteria from the site of infection in the lungs to the local lymph nodes (31). Also, alveolar epithelial cells and other surrounding cells in the respiratory tract have been reported to be invaded by Mtb (32-34). The invasion of Mtb is the first host- pathogen interaction that decides the outcome of infection. The production and secretion of IFN-γ (which can activate macrophages to destroy the intracellular bacteria) and other cytokines and chemokines produced by CD4+ T cells are critical in the host is response to Mtb. The infected DC are observed as the primary APC responsible for activating CD4+ T cells as part of the adaptive immune response. It has been described that the modulation of DC could lead to weak immunity against Mtb allowing a latent infection (35). CD8+ T cells can also directly kill infected cells. Within 2 to 6 weeks of infection, cell-mediated immunity develops, and there is an influx of lymphocytes, fibroblasts and activated macrophages into the lesions resulting in granuloma formation. The granuloma functions are overall to prevent the dissemination of Mtb and to provide a local environment for communication between immune cells. However, the granuloma formation does not always eliminate the mycobacteria. Thus, the bacteria can become dormant giving rise to a latent infection. Another consequence of granuloma formations is the development of cell death and tissue necrosis. Dead macrophages form a caseum and there is an exponential growth of the bacilli contained in the caseous centers of the granuloma. The bacilli may remain forever within the granuloma, get re-activated later or may get discharged into the airways after an enormous increase in number, necrosis of bronchi and cavitation. The tissue destruction and necrosis produce , which represents the last-attempt defense mechanism of the host when all other mechanism failed. The secondary TB lesions start with the Mtb dissemination from the site of initial infection in the lung through the lymphatic nodes or bloodstream to other parts of the lungs and in the body, the apex of the lung and the regional lymph nodes being favored sites for the Mtb. Around 15% of TB patients develop extrapulmonary TB in the pleura, lymphatics, , genito- , meninges, peritoneum, or . On the other hand, about 90-95% of the people infected with Mtb have asymptomatic, latent TB infection, with only a 10% lifetime chance that a latent infection will progress to TB disease. (30,35,36).

11

Immune responses against Tuberculosis Although, macrophages serve as the long-term host for mycobacteria, Mtb infects and actives DC as well as other cell types in the lungs. However, activated macrophages can kill intracellular bacteria and participate in a protective type 1 (Th1) response. In mycobacterial infection, Th1-type cytokines have been shown to be essential for protective immunity (37). However, other factors are involved and can decide the outcome of the disease. Both CD4+ and CD8+ T cells provide protection against Mtb (38). However, T-cell effector function can be achieved only after priming and differentiation has occurred.

Innate immune cells Macrophages In the lungs, macrophages are considered the first line of defense against inhaled microorganism. Although, they are morphologically similar, it is possible that the function of macrophages is regulated according to their localization in the lungs. In fact, they have been considered to form several different subpopulations on the basis of their anatomic location such as the airway macrophages, situated at or under the epithelial lining of conducting airways, the alveolar interstitial macrophage, the alveolar surface macrophage, the intravascular macrophages, located adjacent to the endothelial cell and the pleural macrophages resident in the pleura space (39). Normally, the principal function of macrophages is and uptake of antigen from the immune system to defend local tissues from the development of specific immune responses. Alveolar macrophages (AM) have been shown to take up most of the particulate material that is delivered intranasally but they do not migrate to regional lymphoid nodes. In addition, AM can self-regulate their functions on request to mount an appropriate immune response, and they are not considered to have a significant role in antigen presentation (40). Mtb uses macrophages as its preferred habitat (30), which is important for its survival. Macrophage Mtb interactions and the role of macrophage in host response can be summarized under the following headings: surface binding of Mtb to macrophages; -lysosome fusion; mycobacterial growth inhibition/killing; recruitment of accessory immune cells for the local inflammatory response and presentation of antigens to T cells for the development of acquired immunity (41). Complement receptors (CR1, CR2, CR3 and CR4), mannose receptors and other cell surface receptor molecules play an important role in binding the organisms to the phagocytes. The interaction between mannose- receptors on phagocytic cells and mycobacteria seems to be mediated through the

12 mycobacterial surface glycoprotein lipoarabinomannan (LAM) (42). E2 and IL-4, a Th2-type , up-regulate complement and mannose receptors functions. IFN-γ decreases the receptor expression, resulting in diminished ability of the Mtb to adhere to macrophages. According to some studies, prevention of phagolysosomal fusion is a mechanism by which Mtb survives inside macrophages (40-43). The production of IFN-γ by T cells is a consequence of antigen presentation from infected macrophages. TNF and IL-12 (43) are secreted by activated monocyte/macrophages as shown in murine studies (44,45). A major effector function responsible for antimycobacterial activity induced by IFN-γ and TNF is nitric oxide (NO), generation of reactive oxygen intermediates (ROI) and reactive nitrogen intermediates (RNI) (46).

Dendritic cells DC are APC specialized for T-cell activation. Immature DC are -derived cells present in most non-lymphoid tissues, where they act as sentinel cells against incoming pathogens. The typical phenotype of DC in human lungs is the high expression of MHC class II and CD205 (type I C-type lectin, that has been described as a DC-specific multilectin receptor), together with low expression of CD8, CD40, CD80 and CD86. In this state, DC are able to take up and process antigens. DC can also act as a potent APC in situ in some other eg. (20,21,23,47,48). The recruitment of airway mucosal DC in response to bacterial stimuli is through the chemokine receptor (CCR)1 and CCR5. However, responses to virus or protein recall antigens use alternative chemokine-ligand receptor combinations. In addition, CCR7 is the principal molecule involved in the migration of antigen-bearing lung DC to regional lymphoid nodes. Different subpopulations of DC have been identified in the lungs (49,50). The myeloid DC subsets, in particular, have been found in the airway mucosa as important subpopulations contributing to the local immunity. The plasmacytoid DC have also been identified as another important subset in the tolerance to foreign antigens. A further indication of the potentially important functions of the plasmacytoid DC subset is their distinct pattern of TLR expression and the high capacity to produce interferon-(IFN)-α in response to microbial stimuli (50-54). However, unlike myeloid DC, human plasmacytoid DC have poor APC activity and there is no evidence for plasmacytoid DC migration out from the lung (3,20). In TB, DC can take up the Mtb through receptors such as TLR, DC-specific ICAM-3- grabbing non-integrin (DC-SIGN) binding to mycobacterial mannose residues; the ; the DEC205 (CD205), the CR3 and scavenger receptors (53-57). Mtb affects

13 through inhibition of phagosome maturation and improved survival in macrophages. The outcome of mycobacteria in DC is controversial with reports from no growth only to survival or unrestricted growth (58-60). Following endocytosis of mycobacteria, activated DCs migrate to draining lymph nodes, where they prime T cells (30). Upon Mtb exposure, engagement of TLRs on DC induces the secretion of cytokines IL-1β, IL-12, IL-18, and IFN-α, which stimulates T cells to produce IFN-γ, a cytokine essential for the bactericidal activity of DC through induction of reactive oxygen intermediates. Another important task of DC is their role as key APC cells in TB since they express MHC class I and II, CD1 (a MHC-like molecule) and co stimulatory molecules such as CD80, CD86 and CD40 needed to prime naïve T cells. The mycobacterial T-cell inducing antigens are either lipids presented on CD1 molecules, or proteins such as ESAT-6, CFP-10, Ag85, or lipoprotein p19 (61). Several studies have also demonstrated that DC can induce protection in mice infected with BCG (62)

Alveolar epithelial cells type II (AEC II) The strategic location of AEC II in the interface between the outside and pulmonary vasculature leads them to be considered as important immunologic modulators in the alveolar space. AEC II may therefore be important in the first line of defense against inhaled pathogens for instance Mtb. Ultra structural criteria used to identify AEC II are the presence of , apical microvilli and specific junctional proteins (47,48). These cells perform different functions including the ion transport, alveolar repair in response to injury and regulations of surfactant metabolism. AEC II are the source of lipid pulmonary (SP-A, SP-B, SP-C and SP-D). SP-B and SP-C enhance the biophysical properties of the lipid components of surfactant, including the lowering of , whereas SP- A and SP-D are involved in innate immune defense enhancing the clearance of a variety of lung pathogens by AM (63). In addition, AEC II secrete antimicrobial proteins, such as lysozymes, and complement components (e.g., C2,C3,C4 and C5) and a variety of cytokines, chemokines and diffusible factors that may be involved in the activation of AM and other cell types during lung inflammation (64,65). AEC II have also been implicated in the modulation of the innate and adaptive immunity due to the expression of PRRs on their surfaces such as TLR2 and TLR4 (10,66). Also, the constitutive expression of MHC II in AEC II (67) is consistent with the possible function of AEC II as antigen presenting cells in the lungs. Other studies have suggested the possible contribution of AEC II in T-cell tolerance to exogenous or innocuous antigens in the

14 lungs due to their lack of the expression of co-stimulatory molecules needed for the activation of T cells (68). Moreover, AEC II were proposed to contribute in balancing inflammatory and responses in the lung by connecting innate and adaptive immune mechanisms, and to establish peripheral T- cell tolerance to respiratory self-antigen (69). The role of AEC II is still not clear in Mtb infection. However, previous studies have demonstrated that Mtb is able to invade and replicate inside AEC II derived cell lines (70,71). Also, the production of NO and IFN-γ by Mtb infected human AEC II cell line have suggested the possible role of AEC II in the responses against Mtb (72,73). Furthermore, a contribution of AEC II in the adaptive response against Mtb has been demonstrated when AEC II present mycobacterial antigens to antigen-specific T cells (74). On the other hand, there are many suggestions about the interaction of AEC II and macrophages due to their close localization in the alveoli. One study has shown that bacterial growth was reduced in Mtb or Mycobacterium avium-infected macrophages when these cells were co-cultured with an AEC II-cell line (75). Also, AEC II presented an increase in the mitochondrial RNA expression of surfactant proteins, cytokines, chemokines and GM-CSF, indicating a crucial role of AEC II in the potentiation of macrophage-anti mycobacterial activity (10,75).

Neutrophils Neutrophils are considered to be the earliest cells recruited to sites where antigen enters in the body and/or inflammatory signals are triggered. They also have well- characterized microbicidal mechanisms such as those dependent on oxygen and the formation of extracellular traps (76). In mice, the role played by neutrophils in TB is controversial. These cells have been detected in the beginning of an infection as well as several days after infection (77,78) , and they were thought to have an important role in the control of mycobacterial growth. However, the capacity of neutrophils to kill mycobacteria is still not fully understood. It might be that the major role of these cells is in granuloma formation (79) and of the transfer of their own microbicidal molecules to infected macrophages (80).

Natural killer (NK) cells and Natural Killer T (NKT) cells NK cells are a small fraction of lymphocytes, already specialized to display a cytotoxic activity against certain types of target cells, especially, host cells that have become infected with virus and host cells that have become cancerous. NK cells lack TCRs or BCRs. The activation of NK cells is through the signals from activating and inhibitory receptors. In

15 addition, NK cells can secrete different cytokines and chemokines such as IFN-γ, IFN-α and IL-22 (81-83). NK cells improve also the function of γδ T cells, another type of lymphocytes which play a role in the immune response against Mtb due to their capacities to act as cytolytic cells and in their secretion of IFN-γ (84). In TB, NK cells become activated during the early response to pulmonary TB with large production of IFN-γ but their depletion does not markedly alter host resistance to Mtb infection (41,57). However, the major contribution of NK cells in the defense against Mtb could be the secretion of IL-22, an important cytokine involved in the promotion of phagosome-lysosome fusion in macrophages (85). In contrast to NK cells, NKT cells are T cells with αβ TCRs and with the expression of some of the cell-surface molecules also present on NK cells. NKT cells recognize glycolipid antigen presented by a MHC-like molecule called CD1d. Also, NKT cells are able to secrete large amounts of either Type 1 cytokines such as IFN-γ or Type 2 cytokines such as IL-4 and IL-13. Perhaps the function of NKT cells is to provide and early rapid help for a cell-mediated immune response (IFN-γ) and or an -mediated response (IL-4) distinct from that seen by conventional T-helper cells which required several days to be activated. If so, NKT cells would represent a link between innate and adaptive immunity. It has been found that murine CD1d-restricted NKT cells mediate protection against Mtb in vivo (86).

Adaptive immune cells CD4+ T cells CD4+ T cells and their derived cytokines are crucial in the defense and protection against Mtb. CD4+ T cells are recognising antigenic peptides in the context of products encoded by the major histocompatibility complex (MHC) class II. The frequency of IFN-γ- producing CD4+ T cells has been widely used as a correlation of protection against Mtb. The role of IFN-γ in protection against TB has been clearly shown in mice with a disrupted IFN-γ gene and in humans with in involved in the IFN-γ and IL-12 pathways (38, 87-89). Also, mice deficient in either CD4 or MHC II molecules have shown an increase in susceptibility to Mtb (90).

16

CD8+ T cells CD8+ T cells are also important for effective T-cell immunity against Mtb. CD8+ T cells are activated after interaction of the TCR with a processed antigen bound to MHC class I. Also, CD8+ T cells have effector functions, such as cytolysis and release of potent cytokines such as IFN-γ and TNF (91). Mice deficient in critical components of the MHC class I processing and presentation pathway are more susceptible to Mtb infection (92). In addition, CD1b (a human MHC-like molecule)-restricted CD8+ T cells are able to inhibit the growth of Mtb in vitro (93).

B cells Classically, B cells and are thought to offer no significant contribution in the protection against Mtb. However, the role for B cells in host immune response to Mtb, has been suggested when mycobacteria-infected polymeric-Ig-receptor deficient mice display a delayed immune response with an increase in the bacterial growth in the lungs, implicating a role for secretory IgA in an optimal TB immunity (94). In addition, activated DC were able to internalize more efficiently BCG when BCG were coated with specific antibodies (95). Furthermore, the role of B cells as antigen presenting cell in TB has been suggested (96).

γδ T cells γδ T cells may directly recognize small mycobacterial peptic antigens and non-protein ligands in the absence of antigen-presenting cells. In mice, a single contact with Mtb substantially increases the number of γδ T cells, but not the number of αβ T cells (CD4+ and CD8+ T cells) in the draining lymph nodes. In mice infected with Mtb, γδ T cells accumulate at the site of infection and seem to be necessary for early containment of mycobacterial infections (97). Like γδ T cells, CD1-restricted T cells do not react with mycobacterial protein antigens in the context of MHC class I or class II molecules. Instead, these T cells react with mycobacterial lipids or glycolipid antigens bound to CD1 on antigen-presenting cells. CD1 molecules have close structural resemblance to MHC class I but are relatively nonpolymorphic. In mycobacterial infections, several different T-cell subsets have been found to interact with CD1, including CD4- CD8- (double-negative) T cells, CD4+ or CD8+ single-positive T cells, and T cells. CD1-restricted T cells display cytotoxic activity and are able to produce IFN-γ (97-101).

17

Role of TLRs in mycobacterial infection The TLRs are probably the best studied group of PRRs. TLR as biosensors have a critical role in the host defense recognizing conserved structures in bacteria and to induce innate immune responses and to prime antigen-specific adaptive immunity. In humans, the Toll family comprises about 10 family members with a highly conserved intracellular signalling domain that resembles the signalling domain found in the mammalian IL-1 receptor. After activation of the receptor, this Toll/IL-1 receptor (TIR) domain interacts with different adaptor molecules that through activation of NF-κB and/or IFN-regulatory factors (IRF) leading to the transcription activation of a broad panel of genes. The between Toll-like family members also extends to the extracellular part of the receptor. Multiple leucine-rich repeats (between 19 and 25) and a single membrane proximal cysteine motive are involved in specific binding to a wide variety of microbial and endogenous ligands. Unclear is how such conserved domains in Toll-like members are able to recognize different ligands specifically, also given that hydrophobic interactions seem to be a prominent factor (102). In the respiratory tract, since the lung is continuously exposed to a wide variety of airborne antigens and , it is essential to have an appropriate faster and selective immune response. This response requires precise regulation of both proinflammatory and anti-inflammatory responses. Members of TLRs family in the initiate innate as well as adaptive immune responses following their binding to pathogens associated molecular patterns (PAMP). For example, the TLR2 binds to bacterial lipoproteins and lipoteichoic (LTA), TLR4 recognizes LPS from most gram-negative bacteria. TLR5 recognizes bacterial flagellin (monomer that makes up the filament of bacteria flagella), TLR7 and TLR8 recognise single stranded RNA from viruses and TLR9 mediates cellular response to DNA containing unmethylated CpG motif present in bacterial DNA (102,103). Innate immune responses after mycobacterial infection are initiated after recognition of mycobacterial components by PRRs like TLRs. The immune-costimulatory activity of mycobacterial DNA is attributed to the presence of palindromic sequences including the 5’- CG-3’ motif “CpG motif” to bind TLR9 (104,105). The mycobacterial cell wall consists of several glycolipids. Among these, lipoarabinomannan (LAM), lipomannan(LM) and phosphatidyl-myo-inositol mannoside (PIM) are recognized by TLR2. The 19kDa lipoprotein of Mtb also activates macrophages via TLR2 (102,106). The in vivo importance of the TLR- mediated signals in host defense against Mtb was emphasised in studies using mice lacking MyD88, a critical component in TLR signalling. MyD88-deficient mice are highly susceptible to airborne infection with Mtb (85,107). In contrast to mice lacking MyD88, mice

18 lacking individual TLR are not dramatically susceptible to Mtb infection. Susceptibility of TLR2-deficient mice to Mtb infection varies in different studies (108,109). BCG and Mtb infected TLR2-/- and TLR4-/- mice were more susceptible to mycobacterial infections at early stages of infections. Moreover, TLR2-/- but not TLR4-/-infected macrophages decrease the antibacterial activity (110). In addition, in an in vitro study, the implication of TLR2 but not TLR4 resulted in an impairment of IFN-γ mediated killing when macrophages were stimulated with different TLR2 and TLR4 ligands (111). TLR4-deficient mice did not show high susceptibility to Mtb infection (112,113). A report demonstrated that TLR9-deficient mice are susceptible to Mtb infection and mice lacking both TLR2 and TLR9 are more susceptible (114). These findings indicate that multiple TLRs might be involved in mycobacterial recognition. However, mice deficient in TLR2, TLR4 and TLR9 express a milder phenotype than MyD88 deficient mice in mycobacterial infection (115).

Cytokines and chemokines in Mtb infection The immune response against Mtb is complex, involving many cytokines and chemokines. Cytokines play an important role in the regulation of host immune response against the mycobacteria by controlling effector functions of immune and non-immune cells. Chemokines and chemokine receptors lead the cells to specific sites within the tissues; it is probable that these cells participate in the granuloma formation seen in TB. The severity of TB is defined by differences in the activation of immune regulatory chemokines and cytokines.

Pro-inflammatory cytokines IL-12 IL-12 is a key player in host defense against Mtb. IL-12 is produced mainly by phagocytic cells such as DC and macrophages once Mtb is taken up. IL-12 has a crucial role in the induction of IFN-γ production by T cells and NK cells (116). In TB, IL-12 has been detected in lung infiltrates, in , in granulomas, and in lymphadenitis. The expression of IL-12 receptors is also increased in cells from fluid from patients with active pulmonary tuberculosis (117). The protective role of IL-12 can be inferred from the observation that IL-12 deficient mice are highly susceptible to mycobacterial infections (118). In humans suffering from recurrent non-tuberculous mycobacterial infections, deleterious genetic mutations in the genes encoding IL-12p40 and IL-12R have been identified. These patients display a reduced capacity to produce IFN-γ (119). Apparently, IL-

19

12 is a regulatory cytokine which connects the innate and adaptive host response to mycobacteria and probably it exerts its protective effects mainly through the induction of IFN-γ.

TNF Monocytes, macrophages, and DC infected with mycobacteria or in contact with mycobacterial products induce the production of TNF, a prototype pro-inflammatory cytokine (119). TNF plays a key role in granuloma formation, induces macrophage activation, and has immunoregulatory properties in mice (120). TNF is also important for containment of latent infection in granuloma in TB patients (121). Deficient mice which are unable to make TNF or lack the TNF receptor p55 display an increased susceptibility for mycobacteria (121). In human TB, no TNF gene mutations have been found and no positive associations have yet been established between gene polymorphism for TNF and disease susceptibility (122).

IFN-γ Protective anti-mycobacterial immune responses involve mainly IFN-γ secreted by T cells to activate macrophages and do induce their microbicidal functions. However, other cells such as NK cells and DC can produce IFN-γ in response to Mtb early during infection. This will lead to the development of antigen-specific IFN-γ-producing CD4+T cells (123). Also, it has been suggested that macrophages can produce IFN-γ in Mtb-infected mice (124). However, Mtb has developed mechanisms to limit the activation of macrophage by IFN-γ (111,121). In addition, it has been found in a human study, that the production of IFN-γ not always correlates with mycobacterial inhibition (23). In line with this, it has been described in mice that IFN-γ secretion from animals immunized with mycobacterial antigens does not always correlate with protection (125). Thus, even if IFN-γ is important to generate immune responses and protection against Mtb, it is not sufficient for eliminating these mycobacteria.

IL-18 IL-18, a pro-inflammatory cytokine which shares many features with IL-1, was initially discovered as an IFN-γ-inducing factor, acting in synergy with IL-12. It has since been found that IL-18 also stimulates the production of other proinflammatory cytokines, chemokines, and transcription factors. There is evidence for a protective role of IL-18 during mycobacterial infections since IL-18 deficient mice are highly susceptible to Mtb (126,127). In mice infected with Mybobacterium leprae, resistance is correlated with a higher expression

20 of IL-18 (128). The major effect of IL-18 in this model seems to be the induction of IFN-γ. Indeed, in TB pleurisy, parallel concentrations of IL-18 and IFN-γ were found (129). Also, Mtb-mediated production of IL-18 by peripheral blood mononuclear cells is reduced in TB patients, and this reduction may be responsible for reduced IFN-γ production (129). The role of IL-18 in response against Mtb seems to be protective since lack IL-18 induces a decrease of protective Th1 response, probably leading to mycobacterial propagation.

IL-1β IL-1β is mainly produced by monocytes, macrophages and DC. In TB patients, IL-1β is expressed in excess in the granulomatous lymph nodes from patients with tuberculosis (130). Studies in IL-1α and -1β double-deficient mice suggest an important role of Il-1β in TB (131). It has been found that IL-1R type I-deficient mice (which do not respond to IL-1) display an increased mycobacterial outgrowth and also defective granuloma formation after infection with Mtb (132). The major role of IL-1β in host defense against Mtb seems to be a critical ligand to induce signalling in determining the MyD88 dependent phenotype (133). Thus IL-1β is a critical component of innate resistant against Mtb.

IL-6 IL-6 is a pleiotropic cytokine which plays a major role in hematopoiesis, T- and B- cell differentiation, and inflammation. IL-6 is secreted by T cells and macrophages as part of the immune inflammation response to trauma such as burns or other tissue damage. IL-6, which has both pro and anti-inflammatory properties, is produced early during mycobacterial infection and at the site of infection. IL-6 may be harmful in mycobacterial infections, as it inhibits the production of TNF and IL-1β and promotes in vitro growth of M. avium (134). However, it has been found that the secretion of IL-6 by infected Mtb macrophages in mice may contribute to the inability of IFN-γ to eradicate Mtb infection (135). In addition, the observations that IL-6-deficient mice display increased susceptibility to Mtb infection suggest a protective role of IL-6 (136).

21

Anti-inflammatory cytokines IL-10

This cytokine is produced by macrophages after phagocytosis of Mtb and after binding of mycobacterial LAM (137). T lymphocytes, including Mtb-reactive T cells, are also capable of producing IL-10. In patients with TB, expression of IL-10 mRNA has been found in circulating mononuclear cells, in pleural fluid, and in alveolar lavage fluid IL-10 antagonizes the proinflammatory cytokine response by down regulation of the production of IFN-γ, TNF and IL-12 (138). IL-10 would be expected to interfere with host defense against Mtb. Indeed, IL-10 transgenic mice developed a larger bacterial burden upon mycobacterial infection (138, 139). In human TB, IL-10 production was higher in anergic patients, both before and after successful treatment, suggesting that Mtb-induced IL-10 production suppresses an effective immune response (139).

Transforming growth factor (TGF)-β The principal role of TGF-β is the control of proliferation and functions in most cells. Mycobacterial products induce production of TGF-β in monocytes and DC. Interestingly, LAM from virulent mycobacteria induces TGF-β production (140). TGF-β is produced in excess during human TB (141). TGF-β seems to neutralize protective immunity in TB by suppressing T-cell mediated immunity, inhibiting proliferation and IFN-γ production; in macrophages it antagonizes antigen presentation, proinflammatory cytokine production, and cellular activation (142). Naturally, inhibitors of TGF-β eliminate the suppressive effects of TGF-β in mononuclear cells from TB patients and in macrophages infected with Mtb. In the anti-inflammatory response, TGF-β and IL-10 seem to synergize. TGF-β may also interact with IL-4. Paradoxically, in the presence of both cytokines, T cells may be directed towards a protective Th1-type profile (143). However, the role of TGF-β is maybe similar as the IL-10, probably inhibiting the activation of Mtb-reactive CD4+.

IL-4 In intracellular infections such as TB, IL-4 cytokine has been found to inhibit IFN-γ production and macrophage activation. In mice infected with Mtb, and reactivation of latent infection are both associated with increased production of IL-4 (144). However, this is not a consistent finding, and it still remains to be determined whether IL-4

22 causes or merely reflects disease activity in human TB. Thus, the role of IL-4 in the susceptibility to TB is not yet entirely resolved.

Chemokines IL-8 In humans, IL-8 attracts neutrophils, T lymphocytes, and possibly monocytes. Upon phagocytosis of Mtb or stimulation with LAM, macrophages produce IL-8. This production is substantially blocked by neutralization of TNF and IL-1β, indicating that IL-8 production is largely under the control of these cytokines (145). Human pulmonary epithelial cells also produce IL-8 in response to Mtb (146). In TB patients, IL-8 has been found in bronchoalveolar lavage fluid, lymph nodes, and plasma. However, the central role of IL-8 in the host immune response to Mtb seems to be the leukocyte recruitment to areas of granuloma formation in TB (147).

Keratinocyte-derived (KC) chemokine (CXCL1) and macrophage-inflammatory protein (MIP)-2 (CXCL2) The murine chemokines KC and MIP-2 are the major chemoattractants responsible for recruiting neutrophils. Both chemokines bind to chemokine receptor, CXCR2 (148). The two chemokines are closely related (149). They are also considered homologs to the human GRO chemokines that are functionally similar to the IL-8 CXC chemokine family (150,151). The MIP-2 mRNA expression was induced in mice infected with different Mtb strains (151). Also, Lipoarabinomannan (LAM), a cell wall component of Mtb resulted in a neutrophilic cell influx into the bronchoalveolar lavage fluid and also induced increases in the lung concentrations of MIP-2 and KC (152).

Monocyte chemotactic protein (MCP)-1 (CCL/21) MCP-1 is produced by monocytes, macrophages and epithelial cells (153,154). Mtb, preferentially induces production of MCP-1 by monocytes to the site of infection (155). In murine models, deficiency of MCP-1 inhibits granuloma formation (156). Also, C-C chemokine receptor 2-deficient mice, which fail to respond to MCP-1, display reduced granuloma formation and suppressed Th1-type cytokine production (157) and die early after infection with Mtb (158). MCP-1 was found in elevated concentrations in alveolar lavage fluid, serum, and pleural fluid from tuberculosis patients (159,160). Therefore, the role of

23

MCP-1 during Mtb infection is the recruitment of monocyte/macrophages and T cells to the site of infection maybe to help in the granuloma formation.

Matrix metallopeptidase (MMP)-9 MMP-9, also known as 92 the kDa type IV collagenase, 92 kDa gelatinase or gelatinase B (GELB), is an enzyme that in humans is encoded by the mmp9 gene. MMP-9 is produced by monocytes, macrophages, neutrophils, keratinocytes, fibroblasts, osteoclasts and endothelial cells, and is involved in inflammatory responses, tissue remodelling, wound healing, tumor growth and (161,162). of the (MMP) family play a significant role in many biological activities including many aspects of the granuloma formation (163,164). In general, MMPs are endopeptidases responsible for degrading components of the such as and proteoglycans, and as potent chemokine antagonists. They play an important role in leukocyte migration and tissue remodelling (165). Some studies suggested that MMP-9 is up-regulated by Mtb and associated with local tissue damage in TB meningitis (166). Pulmonary epithelial cells potentially produce MMP-9 and an excess of MMP-9 due Mtb infection causes tissue destruction (167). However, other studies have suggested that the early secretion of MMP-9 is required for recruitment of AM to induce tissue remodelling for allowing the formation of tight well organized granulomas (168). Thus, the role of MMP-9 could be either helping in the granuloma formation or producing tissue damage during Mtb infection.

RANTES (CCL5) RANTES or CCL5 is a chemokine that binds CCR1, CCR3, CCR4, and CCR5 and is produced by epithelial cells, lymphocytes, and platelets, and acts as a potent chemoattractant for monocytes, NK cells and memory T cells, eosinophils, DC and . In addition, RANTES and other chemokines can selectively activate their corresponding lymphoid cell targets (169,170). RANTES has been shown to induce migration into the nasal mucosa of allergic patients (169). In TB, RANTES seems to play a role in regulating protective immune responses at the site of infection (171).

Interferon gamma-induced protein 10 kDa or IP-10 (CXCL10) IP-10 is a chemokine secreted by several cell types such as monocytes, endothelial cells and fibroblasts in response to IFN-γ. IP-10 has several roles such as chemoattraction for monocytes/macrophages, T cells, NK cells, and dendritic cells, promotion of T cell adhesion

24 to endothelial cells, antitumor activity, and inhibition of bone marrow colony formation and angiogenesis (172). It has been shown that Mtb inhibit the transcription of IP-10 however, in presence of IFN-γ there is an induction of IP-10 protein which appears to be involve in novel post-transcriptional events that incorporates non-canonical functions of NFκB and p38 (mapk) (173). Thus, the role of IP-10 during TB probably is probably similar to RANTES but still is not clear.

GM-CSF -macrophage colony-stimulating factor (GM-CSF) was first identified in mouse lung tissue-conditioned medium following LPS injection into mice by its ability to stimulate proliferation of mouse bone-marrow cells in vitro and generation of colonies of both and macrophages. GM-CSF can be produced by a wide variety of cell types, including fibroblasts, endothelial cells, T cells, macrophages, mesothelial cells, epithelial cells and many types of tumor cells (174). In these cells, bacterial endotoxins and inflammatory cytokines, such as IL-1, IL-6, and TNF, are potent inducers of GM-CSF. GM- CSF not only has the capacity to increase antigen-induced immune responses, but can also alter the Th1/Th2 cytokine balance. It has recently been shown that mice lacking GM-CSF die rapidly from severe necrosis when exposed to an aerosol delivered infection of Mtb because of their inability to mount a Th1 response (175). GM-CSF over-expression, however, failed to attract T cells and macrophages into the sites of infection, suggesting that uncontrolled expression of GM-CSF can lead to defects in cytokine and chemokine regulation. Therefore, excess GM-CSF does not induce an over Th1 response and very fine control of GM-CSF is needed to fight Mtb infections (176).

25

PRESENT STUDY

Aims General aim The overall aim of this study was to determine the role of components in the innate immune response against mycobacterial infection to maintain the mucosal response in the respiratory tract, and the generation of protective immune responses in a mouse model.

Specific aims - To determine the importance of TLR in recognition, processing and antigen presentation in mycobacterial infections. - To evaluate the interaction between pulmonary macrophages and non-hematopoietic immune cells, in particular Type II alveolar epithelial cells in response to mycobacterial responses.

Material and Methods The materials and methods for these studies are described in the separate papers. Briefly, the methods used in the papers are mentioned below, - ELISA - Mouse cytokine array panel A assay - Flow cytometry - Fluoresce microscopy - Luminescence assay

All the in vivo studies were directed in mice according to the ethical guidelines available at Stockholm University.

26

Results and Discussion Paper I

Contribution of TLR2 signalling to the specific immune response. Deficiencies in T cell memory and antigen presenting cell compartments in the TLR2 knockout mice.

TLRs have been found to be important in the host defense against invading microbial pathogens playing an important role in immunity by mediating the secretion of various pro- inflammatory cytokines along with other anti-bacterial effector molecules (106). Both, the innate and acquired responses against Mtb infection depend to a large degree of pattern recognition receptor such as TLRs and the common adapter MyD88. Many studies have shown that deficiencies in TLR2, TLR4 and/or TLR9 result in increased susceptibility to Mtb infections (110,111,114). Moreover, the absence of TLR2 in mice results in greatest susceptibility to Mtb infection after high-dose aerosol while a low-dose resulted in slightly increased bacterial growth (113,177,178). TLR2 has also been implicated in the recognition of mycobacterial antigens and modulation of phagocytic functions. A prolonged recognition of lipoproteins from Mtb by TLR2 has been found to limit the ability of macrophages to up- regulate MHC II expression in response to IFN-γ. This effect is associated with a reduced antigen presentation (179,180). In this study, we evaluated the role of TLR2 in the recognition of mycobacterial antigens. We compared the immune responses induced in wild type (WT) and TLR2 knockout (TLR2-/-) mice following with the 19kDa (TLR2 ligand) and the Ag85A (non-TLR2 ligand) mycobacterial antigens. Initially, we evaluated the humoral responses in both mouse strains. Previous reports have shown that of mice with recombinant Mycobacterium vaccae, which express the 19kDa antigen, results in induction of a strong type 1 immune response to the 19kDa antigen. This response is characterized by IgG2a antibodies and IFN-γ production by T cells (181). In addition, it has been reported that 19kDa induces the production of cytokines such as IL-12 which is involved in promoting Th1 responses in macrophages (182). In our study, we found that the antigen-specific antibody responses were comparable between WT and TLR2-/- mice for both antigens. In addition, increased levels of IgG2a were detected after 19kDa but not after Ag85A immunizations. These results confirmed and suggested that

27 immunization with 19kDa antigen induces Th1 responses while immunization with the Ag85A antigen induced a more Th2 type of response. We also evaluated cellular immune responses in both mouse strains. Our results have shown that comparable levels of IFN-γ were produced by both mouse strains when cells from Ag85A immunized mice were re-stimulated with the same antigen in vitro. In contrast, in the group of mice immunized with the 19kDa antigen, spleen cells from TLR2-/- mice secreted significantly lower amounts of IFN-γ compared to spleen cells from WT mice. We also evaluated whether immunization with the Ag85A was able to induce protection in WT and TLR2-/- mice. We challenged the mice with BCG and the bacterial load was determined in the lungs from WT and TLR2-/- mice immunized with Ag85A. We found that mice immunized with the Ag85A from both mouse strains were protected to a similar extends. These data are in line with previous reports of studying humoral and cellular protection after the Ag85A immunization in mice (183). The low levels of IFN-γ from TLR2-/- spleen cells after re-stimulation with 19kDa in vitro led us to investigate whether T cells were not properly primed in vivo or if the antigen presentation was not sufficient in vitro. To elucidate these questions, BMM from TLR2-/- and WT, previously infected with BCG or pulsed with the antigens (19kDa or Ag85A) were co-cultured with spleen cells from immunized mice with 19kDa or Ag85A from both mouse strains in vitro. (Fig.2a, manuscript I) We found that memory T cells were generated in vivo in both, TLR2-/- and WT mice when spleen cells from both mouse strains were co-cultured with BMM from WT mice pulsed with the 19kDa antigen or the Ag85A. The levels of IFN-γ were comparable in response to both antigens. These results indicated that the 19kDa lipoprotein, a ligand for TLR2, could be internalized, processed and presented in vivo to generate memory-T cells even if the mice were deficient in TLR2. We also found that the capacity of antigen presentation by TLR2-/- mice was independent of TLR2 when BMM from TLR2-/- mice pulsed with the Ag85A were co-cultured with spleen cells from WT and TLR2-/- mouse strains immunized with the Ag85A in vitro. However, BMM from TLR2-/- mice (pulsed with 19kDa) were affected in their capacity to present antigen to spleen cells from TLR2-/- but not with WT mice. These results suggested that the presentation of the antigens was not dependent on the presence or absence of TLR2 in the APC. However, it is clear that the presentation of antigens is affected if both, APC and T cells are deficient in TLR2.

28

Since Mtb uses macrophages as its preferred habitat in the lung, we asked whether the processing and antigen capacity of pulmonary macrophages (PM) could be affected by TLR2 and whether PM could also accomplish a similar pattern as BMM. It has been found that local soluble factors in the lung environment could determine the behavior and phenotype of AM and other cells (184). Therefore, we were aware that the environment in the lung could determine the behavior and phenotype of the resident macrophage population in this tissue. To answer these questions, spleen cells from TLR2-/- and WT mice immunized with the 19kDa antigen were co-cultured with PM from TLR2-/- and WT mice previously pulsed with 19kDa in vitro. We found a different pattern of PM in the capacity of antigen presentation compared to BMM. Moreover, PM from TLR2-/- were clearly affected in the antigen presentation to both mouse strains. In addition, TLR2-/- memory-T cells were not generated in TLR2-/- mice (Fig.2b, manuscript I). The different patterns in antigen presentation between BMM and PM led us to study the behavior of these two types of macrophages in response to different stimuli. LPS and

Pam3Cys-Ser-(Lys)4trihydrochloride (Pam3) were used as TLR4 and TLR2 ligands, respectively. Anti-CD40 was used to activate macrophages via CD40 ligation. PM and BMM from WT mice were stimulated with the stimuli mentioned above in vitro and the supernatants were collected. Kinetics of the IL-10, IL-6, IL-12 and TNF production were measured by ELISA. In addition, un-stimulated and stimulated PM and BMM were analyzed by FACS for measuring the expression of MHC II, co-stimulatory molecules such as CD40, CD80, CD86 and F4/80 (mouse mature macrophage marker) expression. We found that PM and BMM had similar pattern in the production of cytokines. However, LPS induced an early response (4 h) while Pam3 produced a late response (24 h) in the production of TNF and IL-12 by both cell types. Nevertheless, PM were more affected in the production of TNF upon Pam3 stimulation. The results from FACS analysis have shown that BMM were able to up-regulate the expression of MHC II, CD80, CD86 and CD40 upon TLRs and anti-CD40 stimulation. However, MCH II expression by PM was almost twofold stronger than BMM. In addition, PM exhibited a high expression of CD80. The expression of CD86 by PM was not up- regulated. We also evaluated other functional responses between PM and BMM such as mycobacterial uptake and intracellular growth control. We found that BMM displayed a stronger capacity to internalization and control of bacterial growth compared with PM (data not shown).

29

All these results together suggested that BMM might be considered as naive macrophages with a full expression of the large macrophage capacities whereas the growth, survival, phenotype and behavior of PM in the lungs might depend in part on locally produced factors (growth factors, cytokines or surfactant factors). Interestingly, the TLR2 ligand used in this study Pam3 induced low TNF response production in PM.

In summary, in this study we suggest that in the absence of TLR2, immune responses could be generated. However, our results suggest that might be PM are more restricted to produce a successful antigen presentation in the lungs due to the influence of local factors. The functional activity and phenotype of BMM and PM were also evaluated in this study. We found that BMM were more efficient than PM in the internalization and growth control of mycobacteria. In addition, we found a different phenotype of co-stimulatory molecules and MHC II expression between PM and BMM. However, both cell types, BMM and PM were able to produce similar patterns of cytokines in response to TLRs and anti-CD40 stimulation. We hypothesized that the activation, behavior and expression of molecules involved in antigen presentation are influenced by the local tissue environment, for instance the lungs.

30

Paper II

The role of alveolar epithelial cell type II in initiating and shaping pulmonary immune responses. Communication between the innate and adaptive immune systems.

The role of macrophages and DC responses to mycobacterial infection has been long studied before and these cells are considered key players in the defense against mycobacterial infections. However, other cell populations in the lungs have been proposed to play important roles in the pathogenesis and defense against Mtb. Due to their strategic localization, expression of immune markers such as TLR and MHC II and close interaction with other cells, especially with macrophages in the lungs, AEC II have been also considered to play an important role during mycobacterial infections. In addition, AEC II secrete a variety of antimicrobial products, cytokines and chemokines to induce different responses in the innate and adaptive system in the lungs. In paper II we compared AEC II with pulmonary macrophages (PM) in their ability to generate immune responses against mycobacterial products and BCG. We first compared the ability to take up and the capacity of intracellular growth control upon BCG infection in vitro in both cell types isolated from mouse lungs. Our data showed that even if PM were more efficient in both capacities, AEC II were also able to take up and control BCG growth. These results from primary cells were in line with previous reports of Mtb infection and replication inside AEC II cell lines (70,71). We also performed an in vitro experiment to compare the capacity of primary AEC II as APC with a professional APC, for instance PM. Previous reports have shown that AEC II express constitutively MHC II (67) and also that murine AEC II can present mycobacterial antigens to T cells (74). Our findings showed that AEC II pulsed with the 19kDa antigen (mycobacterial antigen) were clearly able to stimulate spleen cells from mice immunized with the 19kDa antigen. However, the magnitude of the response was low compared with pulsed PM. These results confirmed the capacities of AEC II to take up, process and present antigens. Therefore, we and others suggested a possible role of AEC II in the adaptive responses as APC to mycobacterial infection. However, the specific role as specialized APC in an in vivo situation in the lungs might be secondary. It is important to consider the localization in separated compartments of AEC II and T cells. Consequently, AEC II have to promote the migration of T cells from the peripheral blood and other compartments to the lung to generate a successful antigen

31 presentation. Also, another crucial factor to remark is expression of co-stimulatory molecules for a successful antigen presentation to T cells. In human and mouse studies AEC II have been reported to express a low grade or lack of expression of classical co-stimulatory molecules (68,185). The lack of co-stimulatory molecules in AEC II has been suggested to induce T-cell tolerance to suppress inflammatory responses in the lungs against harmless antigens (68). Moreover, one study has shown that AEC II are able to induce regulatory peripheral T cells inducing tolerance against self-antigens in the lungs through the expression of factors such as TGBβ (186). Thus, it might be more likely that the participation, in the adaptive system, of AEC II is through to the secretion of factors modulating the activation and function of different cell types present in the lungs. In the lungs, the production of cytokines, chemokines and other factors by local cells decides the outcome of inflammatory responses in this tissue. Although, immune cells such as macrophages, DC are secreting many of these factors; AEC II and other non-immune cells in the lungs are able to produce many factors constitutively or upon different stimuli (73,75). To gain a better understanding of the role of AEC II in the production of factors against mycobacteria, we compared the production of MCP-1, MIP-2, KC, TNF, MMP-9 and IL-12 in primary AEC II with that of PM upon different stimuli in vitro. We used as stimuli: heat killed (HK)-BCG and BCG lysate (Lys-BCG) as mycobacterial products, cytokines such as TNF and IFN-γ were used due to their importance in the responses to mycobacteria and LPS was used as a TLR4 ligand. We found a different pattern of cytokine and chemokine production in both cell types. MCP-1 chemokine was mostly secreted by primary AEC II and PM were the main producers of MIP-2 (homologue in mouse of human IL-8). Since macrophages secrete TNF and MCP-1 can activate macrophages, the possible influence of PM on AEC II and vice versa was suggested when primary AEC II secreted high amounts of MCP-1 upon TNF stimulus. We also found that the major ligands may be present in the BCG cell wall due to the fact that Lys-BCG was not as good stimulator compared with HK-BCG. TNF and IL-12 were only produced by PM upon LPS stimulation (data not shown). We also investigated the role of MMP-9, a molecule involved in the granuloma formation for controlling mycobacterial infections (167,168). We found that AEC II but not PM were good secretors of MMP-9 upon TNF stimulation. In this study, we found some different results compared with previous reports in human studies. However, even if the levels of MIP-2 secreted by murine primary AEC II were lower than PM our results were comparable with previous reports of IL-8 levels secreted by human epithelial cells (187). We also considered that the health status of mouse

32 lung cells can be secured compared with the samples of human lung cells, which come from unhealthy lung tissues. Thus, there is not the guarantee that these lung cells are not activated or anergized. In addition, we found some contradictory results when we used a commercial AEC II cell line. We concluded that there is not a cell line that exhibit the full range of known primary AEC II functions. As we and others demonstrated that AEC II are able to produce factors such as MCP- 1 and MIP-2 chemokines, we aimed to determine a more complete profile of different factors produced by AEC II in response to stimuli such as mycobacterial products, TLR ligands and

IFNs. AEC II were stimulated with HK-BCG, Lys-BCG, LPS, Flagellin, Pam3Cys-Ser-(Lys)4 trihydrochloride (Pam3), IFN-γ and IFN-α in vitro. Supernatants were collected and were analysed with the R&D mouse proteome profile array. This protein array allowed us to determine of 40 different factors including growth factors. We found that a broad array of different factors was produced by AEC II namely: G-CSF, GM-CSF, M-CSF, KC, MCP-1, MIP-1, MIP-2, TIMP-1, IL-6, and IP-10. The analysis of unstimulated AEC II showed that these cells were able to produce constitutively some of the factors (GM-CSF, M-CSF, MCP- 1, TIMP-1, IL-6, and IP-10) while other factors (G-CSF, MIP-1, and MIP-2) were secreted only by stimulated AEC II. We also considered the importance of evaluating levels of some factors because higher levels of MCP-1 secreted by AECII upon LPS and TNF. We found that GM-CSF is probably the main growth factor produced by AEC II as a result of undetectable levels of M- CSF even after the stimulation of AEC II (data not shown). Moreover, increased levels of GM-CSF were found after LPS and Pam3 stimulation. In addition, MCP-1, KC and IL-6 were strongly produced after the induction via TLRs whereas IP-10 and RANTES were mostly induced by IFNs. Therefore, the interaction between AEC II and lymphocytes is also possible due to the IFNs (produced by lymphocytes) which were able to induce the expression chemokines (IP-10 and RANTES) assisting in the recruitment of circulating lymphocytes to areas of injury, inflammation, or viral infection. Flagellin was the weakest inducer of the three different TLRs ligands. These results suggested that AEC II are involved actively to induce different effects on other cell types in the lungs such as monocytes, macrophages, DC, and T cells. In this study, we confirmed and provided more evidence of a novel role for AEC II in the lung to generate local responses against mycobacterial infection. Our findings suggested a possible interaction of AEC II with other local cell populations in the lungs (for instance PM)

33 which is considered crucial in the responses against Mtb. Although PM were more efficient, murine AEC II were also able to internalization and the control growth of BCG. In addition, AEC II were able to process and present mycobacterial antigen to T cells. However, their role as APC appears to be secondary since AEC II and T cells belong to different compartments. The main findings in our study were the communication between AEC II and other cell populations in the lungs (PM and lymphocytes) as evidenced by the production of cytokines, chemokines and other factors by AEC II upon TLRs and IFNs stimuli.

34

Concluding Remarks Even though, the roles of components of innate immune responses against mycobacterial infection have been extensively studied, the results of this thesis provide more evidence that TLR signalling pathway, for instance TLR2, links both innate and adaptive immune responses. We have demonstrated that the antigen presentation is affected when both, APC and T cells are TLR2 deficient. We have also suggested that the functional activity and phenotype expression of the two types of macrophages used in this study (BMM and PM) are influenced by the local tissue environment. Furthermore, we provided more evidence of a novel role for AEC II in response to mycobacterial infection and TLRs and IFNs stimulation. In addition, the communication between AEC II and other cell populations in the lungs (PM and lymphocytes) was evidenced in this study through the production of cytokines, chemokines and other factors.

Future Plans The results from the paper I have suggested that immune responses in local resident macrophages in the lungs might be restricted by the local airway environment. In the paper II we suggested that AEC II are playing an important role in the lung immune responses through the secretion of different factors. Indeed, these factors produced by AEC II are important to activate resident cells such as macrophages or to induce cell migration from other compartments, for instance T cells. In order to increase the understanding of the role of AEC II and their influence in the local responses against mycobacterial infection further evaluation of the functional capacities of AEC II might be important to elucidate potential function by AEC II in the airway surfaces. There is some evidence that activated AEC II promote the migration of macrophages (188) Also, we and other have demonstrated a strong production of MCP-1 by AEC II. Thus, it is important to evaluate the influence of AEC II stimulated with mycobacterial products, TLRs and IFNs in the migration of different resident cell in the lungs. Furthermore, we will also investigate the influence of AEC II in other functional capacities such as uptake and growth control in macrophages. Moreover, it will be also important to investigate the influence of others cell types on AEC II.

35

ACKNOWLEDGEMENTS

I would like to thank my supervisor Professor Carmen Fernández for giving me the trust and the opportunity to work in her group. I sincerely grateful to you Carmen, for your large support since the time I arrived Sweden. Muchas gracias Carmen!

My special thanks to my co-authors and collaborators Katharina Hartl, Dagbjört Petursdottir, Natalia Periolo and in particular to Jubayer Rahman for being my friends and always helping me when I asked for.

To all the seniors at Immunology, Marita Troye-Blomberg, Eva Sverremark-Ekström, Klavs Berzins and Eva Severinson for all the nice discussions and advices.

To Maggan, Gelana Yadeta and Anna-Leena Jarva for all your help.

To all the staff at the animal house for being friendly and helpful.

To all the past and present students at Immunology and other Departments, in particular to Jacqueline Calla, Irene Roman, Andrea Sommer, Katarina Tiklova, Natalija Gerasimcik, and Olivia Simone for giving me your friendship and help.

To all my friends in and out of Sweden.

Finally, I would like to thank my family for being my strength and inspiration to continue my PhD studies. Papí, Mamí, Carmen, Enrique and Gustavo; your love and support are the most important pieces of my life. Todo lo que hice, hago y haré siempre será para y por ustedes!

36

REFERENCES

1. Boucher, R. C. 2003. Regulation of airway surface liquid volume by human airway epithelia. Pflugers Arch. 445: 495-498.

2. Ganz, T. 2002. Antimicrobial polypeptides in host defense of the respiratory tract. J. Clin. Invest. 109: 693-697.

3. Holt, P. G., D. H. Strickland, M. E. Wikstrom, and F. L. Jahnsen. 2008. Regulation of immunological homeostasis in the respiratory tract. Nat. Rev. Immunol. 8: 142-152.

4. Mason, R. J. and M. C. Williams. 1977. Type II alveolar cell. Defender of the alveolus. Am. Rev. Respir. Dis. 115: 81-91.

5. Mason, R. J. 1985. Pulmonary alveolar type II epithelial cells and adult respiratory distress syndrome. West. J. Med. 143: 611-615.

6. Takizawa, H. 1998. Airway epithelial cells as regulators of airway inflammation (Review). Int. J. Mol. Med. 1: 367-378.

7. Varani, J., M. K. Dame, D. F. Gibbs, C. G. Taylor, J. M. Weinberg, J. Shayevitz, and P. A. Ward. 1992. Human umbilical endothelial cell killing by activated neutrophils. Loss of sensitivity to injury is accompanied by decreased iron content during in vitro culture and is restored with exogenous iron. Lab. Invest. 66: 708-714.

8. Madjdpour, C., U. R. Jewell, S. Kneller, U. Ziegler, R. Schwendener, C. Booy, L. Klausli, T. Pasch, R. C. Schimmer, and B. Beck-Schimmer. 2003. Decreased alveolar oxygen induces lung inflammation. Am. J. Physiol. Lung Cell. Mol. Physiol. 284: L360-7.

9. Beck-Schimmer, B., R. C. Schimmer, and T. Pasch. 2004. The airway compartment: chambers of secrets. News Physiol. Sci. 19: 129-132.

10. Mayer, A. K., M. Muehmer, J. Mages, K. Gueinzius, C. Hess, K. Heeg, R. Bals, R. Lang, and A. H. Dalpke. 2007. Differential recognition of TLR-dependent microbial ligands in human bronchial epithelial cells. J. Immunol. 178: 3134-3142.

11. Pichavant, M., S. Taront, P. Jeannin, L. Breuilh, A. S. Charbonnier, C. Spriet, C. Fourneau, N. Corvaia, L. Heliot, A. Brichet, A. B. Tonnel, Y. Delneste, and P. Gosset. 2006. Impact of bronchial epithelium on dendritic cell migration and function: modulation by the bacterial motif KpOmpA. J. Immunol. 177: 5912-5919.

12. Gribar, S. C., W. M. Richardson, C. P. Sodhi, and D. J. Hackam. 2008. No longer an innocent bystander: epithelial toll-like receptor signalling in the development of mucosal inflammation. Mol. Med. 14: 645-659.

13. Nelson, D. J., C. McMenamin, A. S. McWilliam, M. Brenan, and P. G. Holt. 1994. Development of the airway intraepithelial dendritic cell network in the rat from class II major histocompatibility (Ia)-negative precursors: differential regulation of Ia expression at different levels of the respiratory tract. J. Exp. Med. 179: 203-212.

37

14. Jahnsen, F. L., E. D. Moloney, T. Hogan, J. W. Upham, C. M. Burke, and P. G. Holt. 2001. Rapid dendritic cell recruitment to the bronchial mucosa of patients with atopic asthma in response to local challenge. 56: 823-826.

15. Moyron-Quiroz, J. E., J. Rangel-Moreno, K. Kusser, L. Hartson, F. Sprague, S. Goodrich, D. L. Woodland, F. E. Lund, and T. D. Randall. 2004. Role of inducible associated lymphoid tissue (iBALT) in respiratory immunity. Nat. Med. 10: 927-934.

16. Heier, I., K. Malmstrom, A. S. Pelkonen, L. P. Malmberg, M. Kajosaari, M. Turpeinen, H. Lindahl, P. Brandtzaeg, F. L. Jahnsen, and M. J. Makela. 2008. Bronchial response pattern of antigen presenting cells and regulatory T cells in children less than 2 years of age. Thorax 63: 703-709.

17. Kocks, J. R., A. C. Davalos-Misslitz, G. Hintzen, L. Ohl, and R. Forster. 2007. Regulatory T cells interfere with the development of bronchus-associated lymphoid tissue. J. Exp. Med. 204: 723-734.

18. Lund, F. E., M. Hollifield, K. Schuer, J. L. Lines, T. D. Randall, and B. A. Garvy. 2006. B cells are required for generation of protective effector and memory CD4 cells in response to Pneumocystis lung infection. J. Immunol. 176: 6147-6154.

19. von Garnier, C., L. Filgueira, M. Wikstrom, M. Smith, J. A. Thomas, D. H. Strickland, P. G. Holt, and P. A. Stumbles. 2005. Anatomical location determines the distribution and function of dendritic cells and other APCs in the respiratory tract. J. Immunol. 175: 1609- 1618.

20. Stumbles, P. A., J. A. Thomas, C. L. Pimm, P. T. Lee, T. J. Venaille, S. Proksch, and P. G. Holt. 1998. Resting respiratory tract dendritic cells preferentially stimulate T helper cell type 2 (Th2) responses and require obligatory cytokine signals for induction of Th1 immunity. J. Exp. Med. 188: 2019-2031.

21. Jahnsen, F. L., D. H. Strickland, J. A. Thomas, I. T. Tobagus, S. Napoli, G. R. Zosky, D. J. Turner, P. D. Sly, P. A. Stumbles, and P. G. Holt. 2006. Accelerated antigen and transport by airway mucosal dendritic cells following of a bacterial stimulus. J. Immunol. 177: 5861-5867.

22. Rescigno, M., M. Urbano, B. Valzasina, M. Francolini, G. Rotta, R. Bonasio, F. Granucci, J. P. Kraehenbuhl, and P. Ricciardi-Castagnoli. 2001. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat. Immunol. 2: 361-367.

23. Hoft, D. F., S. Worku, B. Kampmann, C. C. Whalen, J. J. Ellner, C. S. Hirsch, R. B. Brown, R. Larkin, Q. Li, H. Yun, and R. F. Silver. 2002. Investigation of the relationships between immune-mediated inhibition of mycobacterial growth and other potential surrogate markers of protective Mycobacterium tuberculosis immunity. J. Infect. Dis. 186: 1448-1457.

24. Anonymous 2010. WHO global tuberculosis control report 2010. Summary. Cent. Eur. J. Public Health 18: 237.

38

25. Wu, C. Y., J. R. Kirman, M. J. Rotte, D. F. Davey, S. P. Perfetto, E. G. Rhee, B. L. Freidag, B. J. Hill, D. C. Douek, and R. A. Seder. 2002. Distinct lineages of T(H)1 cells have differential capacities for memory cell generation in vivo. Nat. Immunol. 3: 852-858.

26. Trunz, B. B., P. Fine, and C. Dye. 2006. Effect of BCG vaccination on childhood tuberculous meningitis and worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 367: 1173-1180.

27. Kaufmann, S. H. 2010. Future vaccination strategies against tuberculosis: thinking outside the box. Immunity 33: 567-577.

28. Skeiky, Y. A. and J. C. Sadoff. 2006. Advances in tuberculosis vaccine strategies. Nat. Rev. Microbiol. 4: 469-476.

29. Rahman, M. J. and C. Fernandez. 2009. Neonatal vaccination with Mycobacterium bovis BCG: potential effects as a priming agent shown in a heterologous prime-boost immunization protocol. Vaccine 27: 4038-4046.

30. Kaufmann, S. H. 2001. How can immunology contribute to the control of tuberculosis? Nat. Rev. Immunol. 1: 20-30.

31. Demangel, C. and W. J. Britton. 2000. Interaction of dendritic cells with mycobacteria: where the action starts. Immunol. Cell Biol. 78: 318-324.

32. Sato, K., T. Akaki, T. Shimizu, C. Sano, K. Ogasawara, and H. Tomioka. 2001. Invasion and intracellular growth of Mycobacterium tuberculosis and Mycobacterium avium complex adapted to intramacrophagic environment within macrophages and type II alveolar epithelial cells]. Kekkaku 76: 53-57.

33. Garcia-Perez, B. E., J. C. Hernandez-Gonzalez, S. Garcia-Nieto, and J. Luna-Herrera. 2008. Internalization of a non-pathogenic mycobacteria by macropinocytosis in human alveolar epithelial A549 cells. Microb. Pathog. 45: 1-6.

34. Lee, H. M., J. M. Yuk, D. M. Shin, and E. K. Jo. 2009. Dectin-1 is inducible and plays an essential role for mycobacteria-induced innate immune responses in airway epithelial cells. J. Clin. Immunol. 29: 795-805.

35. Marino, S., S. Pawar, C. L. Fuller, T. A. Reinhart, J. L. Flynn, and D. E. Kirschner. 2004. Dendritic cell trafficking and antigen presentation in the human immune response to Mycobacterium tuberculosis. J. Immunol. 173: 494-506.

36. Mueller, P. and J. Pieters. 2006. Modulation of macrophage antimicrobial mechanisms by pathogenic mycobacteria. Immunobiology 211: 549-556.

37. Flynn, J. L. and J. Chan. 2001. Tuberculosis: latency and reactivation. Infect. Immun. 69: 4195-4201.

38. Tascon, R. E., E. Stavropoulos, K. V. Lukacs, and M. J. Colston. 1998. Protection against Mycobacterium tuberculosis infection by CD8+ T cells requires the production of gamma interferon. Infect. Immun. 66: 830-834.

39

39. Lohmann-Matthes, M. L., C. Steinmuller, and G. Franke-Ullmann. 1994. Pulmonary macrophages. Eur. Respir. J. 7: 1678-1689.

40. Lambrecht, B. N. 2006. in the driver's seat. Immunity 24: 366-368.

41. Cooper, A. M. 2009. Cell-mediated immune responses in tuberculosis. Annu. Rev. Immunol. 27: 393-422.

42. Pitarque, S., G. Larrouy-Maumus, B. Payre, M. Jackson, G. Puzo, and J. Nigou. 2008. The immunomodulatory lipoglycans, lipoarabinomannan and lipomannan, are exposed at the mycobacterial cell surface. Tuberculosis (Edinb) 88: 560-565.

43. Taha, R. A., T. C. Kotsimbos, Y. L. Song, D. Menzies, and Q. Hamid. 1997. IFN-gamma and IL-12 are increased in active compared with inactive tuberculosis. Am. J. Respir. Crit. Care Med. 155: 1135-1139.

44. Bekker, L. G., S. Freeman, P. J. Murray, B. Ryffel, and G. Kaplan. 2001. TNF-alpha controls intracellular mycobacterial growth by both inducible nitric oxide synthase-dependent and inducible nitric oxide synthase-independent pathways. J. Immunol. 166: 6728-6734.

45. Flynn, J. L., M. M. Goldstein, K. J. Triebold, J. Sypek, S. , and B. R. Bloom. 1995. IL-12 increases resistance of BALB/c mice to Mycobacterium tuberculosis infection. J. Immunol. 155: 2515-2524.

46. Bilyk, N. and P. G. Holt. 1993. Inhibition of the immunosuppressive activity of resident pulmonary alveolar macrophages by granulocyte/macrophage colony-stimulating factor. J. Exp. Med. 177: 1773-1777.

47. Fehrenbach, H. 2001. Alveolar epithelial type II cell: defender of the alveolus revisited. Respir. Res. 2: 33-46.

48. Wark, P. A., S. L. Johnston, F. Bucchieri, R. Powell, S. Puddicombe, V. Laza-Stanca, S. T. Holgate, and D. E. Davies. 2005. Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with . J. Exp. Med. 201: 937-947.

49. de Heer, H. J., H. Hammad, T. Soullie, D. Hijdra, N. Vos, M. A. Willart, H. C. Hoogsteden, and B. N. Lambrecht. 2004. Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen. J. Exp. Med. 200: 89-98.

50. Hintzen, G., L. Ohl, M. L. del Rio, J. I. Rodriguez-Barbosa, O. Pabst, J. R. Kocks, J. Krege, S. Hardtke, and R. Forster. 2006. Induction of tolerance to innocuous inhaled antigen relies on a CCR7-dependent dendritic cell-mediated antigen transport to the bronchial . J. Immunol. 177: 7346-7354.

51. Demedts, I. K., K. R. Bracke, T. Maes, G. F. Joos, and G. G. Brusselle. 2006. Different roles for human lung dendritic cell subsets in pulmonary immune defense mechanisms. Am. J. Respir. Cell Mol. Biol. 35: 387-393.

40

52. Masten, B. J., G. K. Olson, C. A. Tarleton, C. Rund, M. Schuyler, R. Mehran, T. Archibeque, and M. F. Lipscomb. 2006. Characterization of myeloid and plasmacytoid dendritic cells in human lung. J. Immunol. 177: 7784-7793.

53. Tailleux, L., O. Schwartz, J. L. Herrmann, E. Pivert, M. Jackson, A. Amara, L. Legres, D. Dreher, L. P. Nicod, J. C. Gluckman, P. H. Lagrange, B. Gicquel, and O. Neyrolles. 2003. DC-SIGN is the major Mycobacterium tuberculosis receptor on human dendritic cells. J. Exp. Med. 197: 121-127.

54. Geijtenbeek, T. B., S. J. Van Vliet, E. A. Koppel, M. Sanchez-Hernandez, C. M. Vandenbroucke-Grauls, B. Appelmelk, and Y. Van Kooyk. 2003. Mycobacteria target DC- SIGN to suppress dendritic cell function. J. Exp. Med. 197: 7-17.

55. Puig-Kroger, A., D. Serrano-Gomez, E. Caparros, A. Dominguez-Soto, M. Relloso, M. Colmenares, L. Martinez-Munoz, N. Longo, N. Sanchez-Sanchez, M. Rincon, L. Rivas, P. Sanchez-Mateos, E. Fernandez-Ruiz, and A. L. Corbi. 2004. Regulated expression of the pathogen receptor dendritic cell-specific intercellular adhesion molecule 3 (ICAM-3)- grabbing nonintegrin in THP-1 human leukemic cells, monocytes, and macrophages. J. Biol. Chem. 279: 25680-25688.

56. Villeneuve, C., M. Gilleron, I. Maridonneau-Parini, M. Daffe, C. Astarie-Dequeker, and G. Etienne. 2005. Mycobacteria use their surface-exposed glycolipids to infect human macrophages through a receptor-dependent process. J. Lipid Res. 46: 475-483.

57. Junqueira-Kipnis, A. P., A. Kipnis, A. Jamieson, M. G. Juarrero, A. Diefenbach, D. H. Raulet, J. Turner, and I. M. Orme. 2003. NK cells respond to pulmonary infection with Mycobacterium tuberculosis, but play a minimal role in protection. J. Immunol. 171: 6039- 6045.

58. Bodnar, K. A., N. V. Serbina, and J. L. Flynn. 2001. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect. Immun. 69: 800-809.

59. Jiao, X., R. Lo-Man, P. Guermonprez, L. Fiette, E. Deriaud, S. Burgaud, B. Gicquel, N. Winter, and C. Leclerc. 2002. Dendritic cells are host cells for mycobacteria in vivo that trigger innate and acquired immunity. J. Immunol. 168: 1294-1301.

60. Vergne, I., J. Chua, S. B. Singh, and V. Deretic. 2004. Cell of mycobacterium tuberculosis phagosome. Annu. Rev. Cell Dev. Biol. 20: 367-394.

61. Beatty, W. L. and D. G. Russell. 2000. Identification of mycobacterial surface proteins released into subcellular compartments of infected macrophages. Infect. Immun. 68: 6997- 7002.

62. Demangel, C., A. G. Bean, E. Martin, C. G. Feng, A. T. Kamath, and W. J. Britton. 1999. Protection against aerosol Mycobacterium tuberculosis infection using Mycobacterium bovis Bacillus Calmette Guerin-infected dendritic cells. Eur. J. Immunol. 29: 1972-1979.

63. Wright, J. R. 2005. Immunoregulatory functions of surfactant proteins. Nat. Rev. Immunol. 5: 58-68.

41

64. Strunk, R. C., D. M. Eidlen, and R. J. Mason. 1988. Pulmonary alveolar type II epithelial cells synthesize and secrete proteins of the classical and alternative complement pathways. J. Clin. Invest. 81: 1419-1426.

65. Cox, G., J. Gauldie, and M. Jordana. 1992. Bronchial epithelial cell-derived cytokines (G- CSF and GM-CSF) promote the survival of peripheral blood neutrophils in vitro. Am. J. Respir. Cell Mol. Biol. 7: 507-513.

66. Armstrong, L., A. R. Medford, K. M. Uppington, J. Robertson, I. R. Witherden, T. D. Tetley, and A. B. Millar. 2004. Expression of functional toll-like receptor-2 and -4 on alveolar epithelial cells. Am. J. Respir. Cell Mol. Biol. 31: 241-245.

67. Cunningham, A. C., D. S. Milne, J. Wilkes, J. H. Dark, T. D. Tetley, and J. A. Kirby. 1994. Constitutive expression of MHC and adhesion molecules by alveolar epithelial cells (type II pneumocytes) isolated from human lung and comparison with immunocytochemical findings. J. Cell. Sci. 107 ( Pt 2): 443-449.

68. Lo, B., S. Hansen, K. Evans, J. K. Heath, and J. R. Wright. 2008. Alveolar epithelial type II cells induce T cell tolerance to specific antigen. J. Immunol. 180: 881-888.

69. Gereke, M., S. Jung, J. Buer, and D. Bruder. 2009. Alveolar type II epithelial cells present antigen to CD4(+) T cells and induce Foxp3(+) regulatory T cells. Am. J. Respir. Crit. Care Med. 179: 344-355.

70. Bermudez, L. E. and J. Goodman. 1996. Mycobacterium tuberculosis invades and replicates within type II alveolar cells. Infect. Immun. 64: 1400-1406.

71. Castro-Garza, J., C. H. King, W. E. Swords, and F. D. Quinn. 2002. Demonstration of spread by Mycobacterium tuberculosis bacilli in A549 epithelial cell monolayers. FEMS Microbiol. Lett. 212: 145-149.

72. Roy, S., S. Sharma, M. Sharma, R. Aggarwal, and M. Bose. 2004. Induction of nitric oxide release from the human alveolar epithelial cell line A549: an in vitro correlate of innate immune response to Mycobacterium tuberculosis. Immunology 112: 471-480.

73. Sharma, M., S. Sharma, S. Roy, S. Varma, and M. Bose. 2007. Pulmonary epithelial cells are a source of interferon-gamma in response to Mycobacterium tuberculosis infection. Immunol. Cell Biol. 85: 229-237.

74. Debbabi, H., S. Ghosh, A. B. Kamath, J. Alt, D. E. Demello, S. Dunsmore, and S. M. Behar. 2005. Primary type II alveolar epithelial cells present microbial antigens to antigen- specific CD4+ T cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 289: L274-9.

75. Sato, K., H. Tomioka, T. Shimizu, T. Gonda, F. Ota, and C. Sano. 2002. Type II alveolar cells play roles in macrophage-mediated host innate resistance to pulmonary mycobacterial infections by producing proinflammatory cytokines. J. Infect. Dis. 185: 1139-1147.

76. Urban, C. F., S. Lourido, and A. Zychlinsky. 2006. How do microbes evade neutrophil killing? Cell. Microbiol. 8: 1687-1696.

42

77. Pedrosa, J., B. M. Saunders, R. Appelberg, I. M. Orme, M. T. Silva, and A. M. Cooper. 2000. Neutrophils play a protective nonphagocytic role in systemic Mycobacterium tuberculosis infection of mice. Infect. Immun. 68: 577-583.

78. Fulton, S. A., S. M. Reba, T. D. Martin, and W. H. Boom. 2002. Neutrophil-mediated mycobacteriocidal immunity in the lung during Mycobacterium bovis BCG infection in C57BL/6 mice. Infect. Immun. 70: 5322-5327.

79. Seiler, P., P. Aichele, S. Bandermann, A. E. Hauser, B. Lu, N. P. Gerard, C. Gerard, S. Ehlers, H. J. Mollenkopf, and S. H. Kaufmann. 2003. Early granuloma formation after aerosol Mycobacterium tuberculosis infection is regulated by neutrophils via CXCR3- signalling chemokines. Eur. J. Immunol. 33: 2676-2686.

80. Tang, M. L., L. S. Kong, S. K. Law, and S. M. Tan. 2006. Down-regulation of integrin alpha M beta 2 ligand-binding function by the urokinase-type plasminogen activator receptor. Biochem. Biophys. Res. Commun. 348: 1184-1193.

81. Leong, J. W. and T. A. Fehniger. 2011. Human NK cells: SET to kill. Blood 117: 2297- 2298.

82. Wolk, K. and R. Sabat. 2006. Interleukin-22: a novel T- and NK-cell derived cytokine that regulates the biology of tissue cells. Cytokine Growth Factor Rev. 17: 367-380.

83. Goto, M., M. Murakawa, K. Kadoshima-Yamaoka, Y. Tanaka, K. Nagahira, Y. Fukuda, and T. Nishimura. 2009. Murine NKT cells produce Th17 cytokine interleukin-22. Cell. Immunol. 254: 81-84.

84. Zhang, R., X. Zheng, B. Li, H. Wei, and Z. Tian. 2006. Human NK cells positively regulate gammadelta T cells in response to Mycobacterium tuberculosis. J. Immunol. 176: 2610-2616.

85. Dhiman, R., M. Indramohan, P. F. Barnes, R. C. Nayak, P. Paidipally, L. V. Rao, and R. Vankayalapati. 2009. IL-22 produced by human NK cells inhibits growth of Mycobacterium tuberculosis by enhancing phagolysosomal fusion. J. Immunol. 183: 6639-6645.

86. Sada-Ovalle, I., A. Chiba, A. Gonzales, M. B. Brenner, and S. M. Behar. 2008. Innate invariant NKT cells recognize Mycobacterium tuberculosis-infected macrophages, produce interferon-gamma, and kill intracellular bacteria. PLoS Pathog. 4: e1000239.

87. Cooper, A. M., D. K. Dalton, T. A. Stewart, J. P. Griffin, D. G. Russell, and I. M. Orme. 1993. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J. Exp. Med. 178: 2243-2247.

88. Dorman, S. E. and S. M. Holland. 2000. Interferon-gamma and interleukin-12 pathway defects and human disease. Cytokine Growth Factor Rev. 11: 321-333.

89. Flynn, J. L., J. Chan, K. J. Triebold, D. K. Dalton, T. A. Stewart, and B. R. Bloom. 1993. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J. Exp. Med. 178: 2249-2254.

43

90. Caruso, A. M., N. Serbina, E. Klein, K. Triebold, B. R. Bloom, and J. L. Flynn. 1999. Mice deficient in CD4 T cells have only transiently diminished levels of IFN-gamma, yet succumb to tuberculosis. J. Immunol. 162: 5407-5416.

91. Lalvani, A., R. Brookes, R. J. Wilkinson, A. S. Malin, A. A. Pathan, P. Andersen, H. Dockrell, G. Pasvol, and A. V. Hill. 1998. Human cytolytic and interferon gamma-secreting CD8+ T lymphocytes specific for Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. U. S. A. 95: 270-275.

92. Sousa, A. O., R. J. Mazzaccaro, R. G. Russell, F. K. Lee, O. C. Turner, S. Hong, L. Van Kaer, and B. R. Bloom. 2000. Relative contributions of distinct MHC class I-dependent cell populations in protection to tuberculosis infection in mice. Proc. Natl. Acad. Sci. U. S. A. 97: 4204-4208.

93. Stenger, S., R. J. Mazzaccaro, K. Uyemura, S. Cho, P. F. Barnes, J. P. Rosat, A. Sette, M. B. Brenner, S. A. Porcelli, B. R. Bloom, and R. L. Modlin. 1997. Differential effects of cytolytic T cell subsets on intracellular infection. Science 276: 1684-1687.

94. Rodriguez, A., A. Tjarnlund, J. Ivanji, M. Singh, I. Garcia, A. Williams, P. D. Marsh, M. Troye-Blomberg, and C. Fernandez. 2005. Role of IgA in the defense against respiratory infections IgA deficient mice exhibited increased susceptibility to intranasal infection with Mycobacterium bovis BCG. Vaccine 23: 2565-2572.

95. de Valliere, S., G. Abate, A. Blazevic, R. M. Heuertz, and D. F. Hoft. 2005. Enhancement of innate and cell-mediated immunity by antimycobacterial antibodies. Infect. Immun. 73: 6711-6720.

96. Vordermeier, H. M., N. Venkataprasad, D. P. Harris, and J. Ivanyi. 1996. Increase of tuberculous infection in the organs of -deficient mice. Clin. Exp. Immunol. 106: 312- 316.

97. Griffin, J. P., K. V. Harshan, W. K. Born, and I. M. Orme. 1991. Kinetics of accumulation of gamma delta receptor-bearing T lymphocytes in mice infected with live mycobacteria. Infect. Immun. 59: 4263-4265.

98. Rosat, J. P., E. P. Grant, E. M. Beckman, C. C. Dascher, P. A. Sieling, D. Frederique, R. L. Modlin, S. A. Porcelli, S. T. Furlong, and M. B. Brenner. 1999. CD1-restricted microbial lipid antigen-specific recognition found in the CD8+ alpha beta T cell pool. J. Immunol. 162: 366-371.

99. Porcelli, S., C. T. Morita, and M. B. Brenner. 1992. CD1b restricts the response of human CD4-8- T lymphocytes to a microbial antigen. Nature 360: 593-597.

100. Sieling, P. A., M. T. Ochoa, D. Jullien, D. S. Leslie, S. Sabet, J. P. Rosat, A. E. Burdick, T. H. Rea, M. B. Brenner, S. A. Porcelli, and R. L. Modlin. 2000. Evidence for human CD4+ T cells in the CD1-restricted repertoire: derivation of mycobacteria-reactive T cells from leprosy lesions. J. Immunol. 164: 4790-4796.

44

101. Stenger, S., R. J. Mazzaccaro, K. Uyemura, S. Cho, P. F. Barnes, J. P. Rosat, A. Sette, M. B. Brenner, S. A. Porcelli, B. R. Bloom, and R. L. Modlin. 1997. Differential effects of cytolytic T cell subsets on intracellular infection. Science 276: 1684-1687.

102. Aliprantis, A. O., R. B. Yang, M. R. Mark, S. Suggett, B. Devaux, J. D. Radolf, G. R. Klimpel, P. Godowski, and A. Zychlinsky. 1999. Cell activation and by bacterial lipoproteins through toll-like receptor-2. Science 285: 736-739.

103. Fremond, C. M., V. Yeremeev, D. M. Nicolle, M. Jacobs, V. F. Quesniaux, and B. Ryffel. 2004. Fatal Mycobacterium tuberculosis infection despite adaptive immune response in the absence of MyD88. J. Clin. Invest. 114: 1790-1799.

104. Kuramoto, E., O. Yano, Y. Kimura, M. Baba, T. Makino, S. Yamamoto, T. Yamamoto, T. Kataoka, and T. Tokunaga. 1992. Oligonucleotide sequences required for activation. Jpn. J. Res. 83: 1128-1131.

105. Hemmi, H., O. Takeuchi, T. Kawai, T. Kaisho, S. Sato, H. Sanjo, M. Matsumoto, K. Hoshino, H. Wagner, K. Takeda, and S. Akira. 2000. A Toll-like receptor recognizes bacterial DNA. Nature 408: 740-745.

106. Brightbill, H. D., D. H. Libraty, S. R. Krutzik, R. B. Yang, J. T. Belisle, J. R. Bleharski, M. Maitland, M. V. Norgard, S. E. Plevy, S. T. Smale, P. J. Brennan, B. R. Bloom, P. J. Godowski, and R. L. Modlin. 1999. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science 285: 732-736.

107. Junqueira-Kipnis, A. P., A. Kipnis, A. Jamieson, M. G. Juarrero, A. Diefenbach, D. H. Raulet, J. Turner, and I. M. Orme. 2003. NK cells respond to pulmonary infection with Mycobacterium tuberculosis, but play a minimal role in protection. J. Immunol. 171: 6039- 6045.

108. Scanga, C. A., A. Bafica, C. G. Feng, A. W. Cheever, S. Hieny, and A. Sher. 2004. MyD88-deficient mice display a profound loss in resistance to Mycobacterium tuberculosis associated with partially impaired Th1 cytokine and nitric oxide synthase 2 expression. Infect. Immun. 72: 2400-2404.

109. Drennan, M. B., D. Nicolle, V. J. Quesniaux, M. Jacobs, N. Allie, J. Mpagi, C. Fremond, H. Wagner, C. Kirschning, and B. Ryffel. 2004. Toll-like receptor 2-deficient mice succumb to Mycobacterium tuberculosis infection. Am. J. Pathol. 164: 49-57.

110. Tjarnlund, A., E. Guirado, E. Julian, P. J. Cardona, and C. Fernandez. 2006. Determinant role for Toll-like receptor signalling in acute mycobacterial infection in the respiratory tract. Microbes Infect. 8: 1790-1800.

111. Arko-Mensah, J., E. Julian, M. Singh, and C. Fernandez. 2007. TLR2 but not TLR4 signalling is critically involved in the inhibition of IFN-gamma-induced killing of mycobacteria by murine macrophages. Scand. J. Immunol. 65: 148-157.

112. Heldwein, K. A., M. D. Liang, T. K. Andresen, K. E. Thomas, A. M. Marty, N. Cuesta, S. N. Vogel, and M. J. Fenton. 2003. TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J. Leukoc. Biol. 74: 277-286.

45

113. Reiling, N., C. Holscher, A. Fehrenbach, S. Kroger, C. J. Kirschning, S. Goyert, and S. Ehlers. 2002. Cutting edge: Toll-like receptor (TLR)2- and TLR4-mediated pathogen recognition in resistance to airborne infection with Mycobacterium tuberculosis. J. Immunol. 169: 3480-3484.

114. Bafica, A., C. A. Scanga, C. G. Feng, C. Leifer, A. Cheever, and A. Sher. 2005. TLR9 regulates Th1 responses and cooperates with TLR2 in mediating optimal resistance to Mycobacterium tuberculosis. J. Exp. Med. 202: 1715-1724.

115. Holscher, C., N. Reiling, U. E. Schaible, A. Holscher, C. Bathmann, D. Korbel, I. Lenz, T. Sonntag, S. Kroger, S. Akira, H. Mossmann, C. J. Kirschning, H. Wagner, M. Freudenberg, and S. Ehlers. 2008. Containment of aerogenic Mycobacterium tuberculosis infection in mice does not require MyD88 adaptor function for TLR2, -4 and -9. Eur. J. Immunol. 38: 680-694.

116. Cooper, A. M. and S. A. Khader. 2008. The role of cytokines in the initiation, expansion, and control of cellular immunity to tuberculosis. Immunol. Rev. 226: 191-204.

117. Taha, R. A., E. M. Minshall, R. Olivenstein, D. Ihaku, B. Wallaert, A. Tsicopoulos, A. B. Tonnel, R. Damia, D. Menzies, and Q. A. Hamid. 1999. Increased expression of IL-12 receptor mRNA in active pulmonary tuberculosis and . Am. J. Respir. Crit. Care Med. 160: 1119-1123.

118. Cooper, A. M., J. Magram, J. Ferrante, and I. M. Orme. 1997. Interleukin 12 (IL-12) is crucial to the development of protective immunity in mice intravenously infected with mycobacterium tuberculosis. J. Exp. Med. 186: 39-45.

119. Ottenhoff, T. H., D. Kumararatne, and J. L. Casanova. 1998. Novel human reveal the essential role of type-I cytokines in immunity to intracellular bacteria. Immunol. Today 19: 491-494.

120. Flynn, J. L., M. M. Goldstein, J. Chan, K. J. Triebold, K. Pfeffer, C. J. Lowenstein, R. Schreiber, T. W. Mak, and B. R. Bloom. 1995. Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity 2: 561- 572.

121. Algood, H. M., P. L. Lin, and J. L. Flynn. 2005. Tumor necrosis factor and chemokine interactions in the formation and maintenance of granulomas in tuberculosis. Clin. Infect. Dis. 41 Suppl 3: S189-93.

122. Dinarello, C. A. 2003. Anti-cytokine therapeutics and infections. Vaccine 21 Suppl 2: S24-34.

123. Fricke, I., D. Mitchell, J. Mittelstadt, N. Lehan, H. Heine, T. Goldmann, A. Bohle, and S. Brandau. 2006. Mycobacteria induce IFN-gamma production in human dendritic cells via triggering of TLR2. J. Immunol. 176: 5173-5182.

124. Wang, J., J. Wakeham, R. Harkness, and Z. Xing. 1999. Macrophages are a significant source of type 1 cytokines during mycobacterial infection. J. Clin. Invest. 103: 1023-1029.

46

125. Abou-Zeid, C., M. P. Gares, J. Inwald, R. Janssen, Y. Zhang, D. B. Young, C. Hetzel, J. R. Lamb, S. L. Baldwin, I. M. Orme, V. Yeremeev, B. V. Nikonenko, and A. S. Apt. 1997. Induction of a type 1 immune response to a recombinant antigen from Mycobacterium tuberculosis expressed in Mycobacterium vaccae. Infect. Immun. 65: 1856-1862.

126. Sugawara, I., H. Yamada, H. Kaneko, S. Mizuno, K. Takeda, and S. Akira. 1999. Role of interleukin-18 (IL-18) in mycobacterial infection in IL-18-gene-disrupted mice. Infect. Immun. 67: 2585-2589.

127. Schneider, B. E., D. Korbel, K. Hagens, M. Koch, B. Raupach, J. Enders, S. H. Kaufmann, H. W. Mittrucker, and U. E. Schaible. 2010. A role for IL-18 in protective immunity against Mycobacterium tuberculosis. Eur. J. Immunol. 40: 396-405.

128. Surcel, H. M., M. Troye-Blomberg, S. Paulie, G. Andersson, C. Moreno, G. Pasvol, and J. Ivanyi. 1994. Th1/Th2 profiles in tuberculosis, based on the proliferation and cytokine response of blood lymphocytes to mycobacterial antigens. Immunology 81: 171-176.

129. Vankayalapati, R., B. Wizel, S. E. Weis, B. Samten, W. M. Girard, and P. F. Barnes. 2000. Production of interleukin-18 in human tuberculosis. J. Infect. Dis. 182: 234-239.

130. Bergeron, A., M. Bonay, M. Kambouchner, D. Lecossier, M. Riquet, P. Soler, A. Hance, and A. Tazi. 1997. Cytokine patterns in tuberculous and sarcoid granulomas: correlations with histopathologic features of the granulomatous response. J. Immunol. 159: 3034-3043.

131. Sieling, P. A., D. Chatterjee, S. A. Porcelli, T. I. Prigozy, R. J. Mazzaccaro, T. Soriano, B. R. Bloom, M. B. Brenner, M. Kronenberg, and P. J. Brennan. 1995. CD1-restricted T cell recognition of microbial lipoglycan antigens. Science 269: 227-230.

132. Juffermans, N. P., S. Florquin, L. Camoglio, A. Verbon, A. H. Kolk, P. Speelman, S. J. van Deventer, and T. van Der Poll. 2000. Interleukin-1 signalling is essential for host defense during murine pulmonary tuberculosis. J. Infect. Dis. 182: 902-908.

133. Mayer-Barber, K. D., D. L. Barber, K. Shenderov, S. D. White, M. S. Wilson, A. Cheever, D. Kugler, S. Hieny, P. Caspar, G. Nunez, D. Schlueter, R. A. Flavell, F. S. Sutterwala, and A. Sher. 2010. Caspase-1 independent IL-1beta production is critical for host resistance to mycobacterium tuberculosis and does not require TLR signalling in vivo. J. Immunol. 184: 3326-3330.

134. Shiratsuchi, H., J. L. Johnson, and J. J. Ellner. 1991. Bidirectional effects of cytokines on the growth of Mycobacterium avium within human monocytes. J. Immunol. 146: 3165- 3170.

135. Nagabhushanam, V., A. Solache, L. M. Ting, C. J. Escaron, J. Y. Zhang, and J. D. Ernst. 2003. Innate inhibition of adaptive immunity: Mycobacterium tuberculosis-induced IL-6 inhibits macrophage responses to IFN-gamma. J. Immunol. 171: 4750-4757.

136. Ladel, C. H., C. Blum, A. Dreher, K. Reifenberg, M. Kopf, and S. H. Kaufmann. 1997. Lethal tuberculosis in interleukin-6-deficient mutant mice. Infect. Immun. 65: 4843-4849.

47

137. Shaw, T. C., L. H. Thomas, and J. S. Friedland. 2000. Regulation of IL-10 secretion after phagocytosis of Mycobacterium tuberculosis by human monocytic cells. Cytokine 12: 483-486.

138. van Crevel, R., T. H. Ottenhoff, and J. W. van der Meer. 2002. Innate immunity to Mycobacterium tuberculosis. Clin. Microbiol. Rev. 15: 294-309.

139. Boussiotis, V. A., E. Y. Tsai, E. J. Yunis, S. Thim, J. C. Delgado, C. C. Dascher, A. Berezovskaya, D. Rousset, J. M. Reynes, and A. E. Goldfeld. 2000. IL-10-producing T cells suppress immune responses in anergic tuberculosis patients. J. Clin. Invest. 105: 1317-1325.

140. Dahl, K. E., H. Shiratsuchi, B. D. Hamilton, J. J. Ellner, and Z. Toossi. 1996. Selective induction of transforming growth factor beta in human monocytes by lipoarabinomannan of Mycobacterium tuberculosis. Infect. Immun. 64: 399-405.

141. Fulton, S. A., J. V. Cross, Z. T. Toossi, and W. H. Boom. 1998. Regulation of interleukin-12 by interleukin-10, transforming growth factor-beta, tumor necrosis factor- alpha, and interferon-gamma in human monocytes infected with Mycobacterium tuberculosis H37Ra. J. Infect. Dis. 178: 1105-1114.

142. Toossi, Z. and J. J. Ellner. 1998. The role of TGF beta in the pathogenesis of human tuberculosis. Clin. Immunol. Immunopathol. 87: 107-114.

143. Erard, F., J. A. Garcia-Sanz, R. Moriggl, and M. T. Wild. 1999. Presence or absence of TGF-beta determines IL-4-induced generation of type 1 or type 2 CD8 T cell subsets. J. Immunol. 162: 209-214.

144. Howard, A. D. and B. S. Zwilling. 1999. Reactivation of tuberculosis is associated with a shift from type 1 to type 2 cytokines. Clin. Exp. Immunol. 115: 428-434.

145. Zhang, Y., M. Broser, H. Cohen, M. Bodkin, K. Law, J. Reibman, and W. N. Rom. 1995. Enhanced interleukin-8 release and gene expression in macrophages after exposure to Mycobacterium tuberculosis and its components. J. Clin. Invest. 95: 586-592.

146. Wickremasinghe, M. I., L. H. Thomas, and J. S. Friedland. 1999. Pulmonary epithelial cells are a source of IL-8 in the response to Mycobacterium tuberculosis: essential role of IL- 1 from infected monocytes in a NF-kappa B-dependent network. J. Immunol. 163: 3936- 3947.

147. Gerszten, R. E., E. A. Garcia-Zepeda, Y. C. Lim, M. Yoshida, H. A. Ding, M. A. Gimbrone Jr, A. D. Luster, F. W. Luscinskas, and A. Rosenzweig. 1999. MCP-1 and IL-8 trigger firm adhesion of monocytes to vascular endothelium under flow conditions. Nature 398: 718-723.

148. Lee, J., G. Cacalano, T. Camerato, K. Toy, M. W. Moore, and W. I. Wood. 1995. Chemokine binding and activities mediated by the mouse IL-8 receptor. J. Immunol. 155: 2158-2164.

48

149. Tekamp-Olson, P., C. Gallegos, D. Bauer, J. McClain, B. Sherry, M. Fabre, S. van Deventer, and A. Cerami. 1990. Cloning and characterization of cDNAs for murine macrophage inflammatory protein 2 and its human homologues. J. Exp. Med. 172: 911-919.

150. Zlotnik, A. and O. Yoshie. 2000. Chemokines: a new classification system and their role in immunity. Immunity 12: 121-127.

151. Rhoades, E. R., A. M. Cooper, and I. M. Orme. 1995. Chemokine response in mice infected with Mycobacterium tuberculosis. Infect. Immun. 63: 3871-3877.

152. Juffermans, N. P., A. Verbon, J. T. Belisle, P. J. Hill, P. Speelman, S. J. van Deventer, and T. van der Poll. 2000. Mycobacterial lipoarabinomannan induces an inflammatory response in the mouse lung. A role for interleukin-1. Am. J. Respir. Crit. Care Med. 162: 486-489.

153. Lundien, M. C., K. A. Mohammed, N. Nasreen, R. S. Tepper, J. A. Hardwick, K. L. Sanders, R. D. Van Horn, and V. B. Antony. 2002. Induction of MCP-1 expression in airway epithelial cells: role of CCR2 receptor in airway epithelial injury. J. Clin. Immunol. 22: 144- 152.

154. Buettner, M., B. Meyer, S. Schreck, and G. Niedobitek. 2007. Expression of RANTES and MCP-1 in epithelial cells is regulated via LMP1 and CD40. Int. J. Cancer 121: 2703- 2710.

155. Sadek, M. I., E. Sada, Z. Toossi, S. K. Schwander, and E. A. Rich. 1998. Chemokines induced by infection of mononuclear phagocytes with mycobacteria and present in lung alveoli during active pulmonary tuberculosis. Am. J. Respir. Cell Mol. Biol. 19: 513-521.

156. Lu, B., B. J. Rutledge, L. Gu, J. Fiorillo, N. W. Lukacs, S. L. Kunkel, R. North, C. Gerard, and B. J. Rollins. 1998. Abnormalities in monocyte recruitment and cytokine expression in monocyte chemoattractant protein 1-deficient mice. J. Exp. Med. 187: 601-608.

157. Boring, L., J. Gosling, S. W. Chensue, S. L. Kunkel, R. V. Farese Jr, H. E. Broxmeyer, and I. F. Charo. 1997. Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J. Clin. Invest. 100: 2552-2561.

158. Peters, W., H. M. Scott, H. F. Chambers, J. L. Flynn, I. F. Charo, and J. D. Ernst. 2001. Chemokine receptor 2 serves an early and essential role in resistance to Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. U. S. A. 98: 7958-7963.

159. Kurashima, K., N. Mukaida, M. Fujimura, M. Yasui, Y. Nakazumi, T. Matsuda, and K. Matsushima. 1997. Elevated chemokine levels in bronchoalveolar lavage fluid of tuberculosis patients. Am. J. Respir. Crit. Care Med. 155: 1474-1477.

160. Mohammed, K. A., N. Nasreen, M. J. Ward, K. K. Mubarak, F. Rodriguez-Panadero, and V. B. Antony. 1998. Mycobacterium-mediated chemokine expression in pleural mesothelial cells: role of C-C chemokines in tuberculous pleurisy. J. Infect. Dis. 178: 1450- 1456.

49

161. Bergers, G., R. Brekken, G. McMahon, T. H. Vu, T. Itoh, K. Tamaki, K. Tanzawa, P. Thorpe, S. Itohara, Z. Werb, and D. Hanahan. 2000. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2: 737-744.

162. Hiratsuka, S., K. Nakamura, S. Iwai, M. Murakami, T. Itoh, H. Kijima, J. M. Shipley, R. M. Senior, and M. Shibuya. 2002. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer. Cell. 2: 289-300.

163. Galboiz, Y., S. Shapiro, N. Lahat, and A. Miller. 2002. Modulation of monocytes matrix metalloproteinase-2, MT1-MMP and TIMP-2 by interferon-gamma and -beta: implications to multiple sclerosis. J. Neuroimmunol. 131: 191-200.

164. Parks, W. C., C. L. Wilson, and Y. S. Lopez-Boado. 2004. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat. Rev. Immunol. 4: 617-629.

165. Woessner, J. F.,Jr. 1991. Matrix metalloproteinases and their inhibitors in remodeling. FASEB J. 5: 2145-2154.

166. Green, J. A., P. T. Elkington, C. J. Pennington, F. Roncaroli, S. Dholakia, R. C. Moores, A. Bullen, J. C. Porter, D. Agranoff, D. R. Edwards, and J. S. Friedland. 2010. Mycobacterium tuberculosis upregulates microglial matrix metalloproteinase-1 and -3 expression and secretion via NF-kappaB- and Activator Protein-1-dependent monocyte networks. J. Immunol. 184: 6492-6503.

167. Elkington, P. T., J. A. Green, J. E. Emerson, L. D. Lopez-Pascua, J. J. Boyle, C. M. O'Kane, and J. S. Friedland. 2007. Synergistic up-regulation of epithelial cell matrix metalloproteinase-9 secretion in tuberculosis. Am. J. Respir. Cell Mol. Biol. 37: 431-437.

168. Taylor, J. L., J. M. Hattle, S. A. Dreitz, J. M. Troudt, L. S. Izzo, R. J. Basaraba, I. M. Orme, L. M. Matrisian, and A. A. Izzo. 2006. Role for matrix metalloproteinase 9 in granuloma formation during pulmonary Mycobacterium tuberculosis infection. Infect. Immun. 74: 6135-6144.

169. Kuna, P., R. Alam, U. Ruta, and P. Gorski. 1998. RANTES induces nasal mucosal inflammation rich in eosinophils, basophils, and lymphocytes in vivo. Am. J. Respir. Crit. Care Med. 157: 873-879.

170. Lillard, J. W.,Jr, P. N. Boyaka, D. D. Taub, and J. R. McGhee. 2001. RANTES potentiates antigen-specific mucosal immune responses. J. Immunol. 166: 162-169.

171. Salam, N., S. Gupta, S. Sharma, S. Pahujani, A. Sinha, R. K. Saxena, and K. Natarajan. 2008. Protective immunity to Mycobacterium tuberculosis infection by chemokine and cytokine conditioned CFP-10 differentiated dendritic cells. PLoS One 3: e2869.

172. Dufour, J. H., M. Dziejman, M. T. Liu, J. H. Leung, T. E. Lane, and A. D. Luster. 2002. IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J. Immunol. 168: 3195-3204.

173. Bai, X., K. Chmura, A. R. Ovrutsky, R. P. Bowler, R. I. Scheinman, R. E. Oberley- Deegan, H. Liu, S. Shang, D. Ordway, and E. D. Chan. 2011. Mycobacterium tuberculosis

50 increases IP-10 and MIG protein despite inhibition of IP-10 and MIG transcription. Tuberculosis (Edinb) 91: 26-35.

174. Griffin, J. D., S. A. Cannistra, R. Sullivan, G. D. Demetri, T. J. Ernst, and Y. Kanakura. 1990. The biology of GM-CSF: regulation of production and interaction with its receptor. Int. J. Cell Cloning 8 Suppl 1: 35-44; discussion 44-5.

175. Quill, H., A. Gaur, and R. P. Phipps. 1989. Prostaglandin E2-dependent induction of granulocyte-macrophage colony-stimulating factor secretion by cloned murine helper T cells. J. Immunol. 142: 813-818.

176. Shi, Y., C. H. Liu, A. I. Roberts, J. Das, G. Xu, G. Ren, Y. Zhang, L. Zhang, Z. R. Yuan, H. S. Tan, G. Das, and S. Devadas. 2006. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and T-cell responses: what we do and don't know. Cell Res. 16: 126-133.

177. Sugawara, I., H. Yamada, C. Li, S. Mizuno, O. Takeuchi, and S. Akira. 2003. Mycobacterial infection in TLR2 and TLR6 knockout mice. Microbiol. Immunol. 47: 327- 336.

178. Heldwein, K. A., M. D. Liang, T. K. Andresen, K. E. Thomas, A. M. Marty, N. Cuesta, S. N. Vogel, and M. J. Fenton. 2003. TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. J. Leukoc. Biol. 74: 277-286.

179. Pai, R. K., M. E. Pennini, A. A. Tobian, D. H. Canaday, W. H. Boom, and C. V. Harding. 2004. Prolonged toll-like receptor signalling by Mycobacterium tuberculosis and its 19-kilodalton lipoprotein inhibits gamma interferon-induced regulation of selected genes in macrophages. Infect. Immun. 72: 6603-6614.

180. Harding, C. V. and W. H. Boom. 2010. Regulation of antigen presentation by Mycobacterium tuberculosis: a role for Toll-like receptors. Nat. Rev. Microbiol. 8: 296-307.

181. Rao, V., N. Dhar, H. Shakila, R. Singh, A. Khera, R. Jain, M. Naseema, C. N. Paramasivan, P. R. Narayanan, V. D. Ramanathan, and A. K. Tyagi. 2005. Increased expression of Mycobacterium tuberculosis 19 kDa lipoprotein obliterates the protective efficacy of BCG by polarizing host immune responses to the Th2 subtype. Scand. J. Immunol. 61: 410-417.

182. Post, F. A., C. Manca, O. Neyrolles, B. Ryffel, D. B. Young, and G. Kaplan. 2001. Mycobacterium tuberculosis 19-kilodalton lipoprotein inhibits Mycobacterium smegmatis- induced cytokine production by human macrophages in vitro. Infect. Immun. 69: 1433-1439.

183. Tanghe, A., O. Denis, B. Lambrecht, V. Motte, T. van den Berg, and K. Huygen. 2000. Tuberculosis DNA vaccine encoding Ag85A is immunogenic and protective when administered by intramuscular needle injection but not by epidermal gene gun bombardment. Infect. Immun. 68: 3854-3860.

184. Guth, A. M., W. J. Janssen, C. M. Bosio, E. C. Crouch, P. M. Henson, and S. W. Dow. 2009. Lung environment determines unique phenotype of alveolar macrophages. Am. J. Physiol. Lung Cell. Mol. Physiol. 296: L936-46.

51

185. Corbiere, V., V. Dirix, S. Norrenberg, M. Cappello, M. Remmelink, and F. Mascart. 2011. Phenotypic characteristics of human type II alveolar epithelial cells suitable for antigen presentation to T lymphocytes. Respir. Res. 12: 15.

186. Gereke, M., S. Jung, J. Buer, and D. Bruder. 2009. Alveolar type II epithelial cells present antigen to CD4(+) T cells and induce Foxp3(+) regulatory T cells. Am. J. Respir. Crit. Care Med. 179: 344-355.

187. Mio, T., D. J. Romberger, A. B. Thompson, R. A. Robbins, A. Heires, and S. I. Rennard. 1997. Cigarette induces interleukin-8 release from human bronchial epithelial cells. Am. J. Respir. Crit. Care Med. 155: 1770-1776.

188. Kannan, S., H. Huang, D. Seeger, A. Audet, Y. Chen, C. Huang, H. Gao, S. Li, and M. Wu. 2009. Alveolar epithelial type II cells activate alveolar macrophages and mitigate P. Aeruginosa infection. PLoS One 4: e4891.

52