Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 1.
Published in final edited form as: Semin Immunopathol. 2013 Jul 18;35(5):563–583. doi: 10.1007/s00281-013-0388-2

Macrophages in Tuberculosis: Friend or Foe

Evelyn Guirado 1, Larry S Schlesinger 1, Gilla Kaplan 2
PMCID: PMC3763202  NIHMSID: NIHMS507287  PMID: 23864058

Abstract

Tuberculosis (TB) remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis (Mtb) is acquired by the respiratory route. It is exquisitely human-adapted and a prototypic intracellular pathogen of macrophages, with alveolar macrophages (AMs) being the primary conduit of infection and disease. The outcome of primary infection is most often a latently infected healthy human host, in whom the bacteria are held in check by the host immune response. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the host immune response fails to control the growth of bacilli, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols coughed out into the environment and inhaled by new hosts. The molecular details of the Mtb-macrophage interaction continue to be elucidated. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. Macrophages demonstrate tremendous phenotypic heterogeneity and functional plasticity which, depending on the site and stage of infection, facilitate the diverse outcomes. Moreover, host responses vary depending on the specific characteristics of the infecting Mtb strain. In this chapter, we describe a contemporary view of the behavior of AMs and their interaction with various Mtb strains in generating unique immunologic lung specific responses.

Keywords: Alveolar macrophage, tissue microenvironment, Mycobacterium tuberculosis, granuloma

1. Tuberculosis

1.1 Human infection: Latency and disease

Mycobacterium tuberculosis (Mtb) is an exquisitely adapted human pathogen that infects an estimated 2 billion people [1]. Infection occurs following inhalation of aerosolized droplets containing viable bacilli. Once inhaled, the bacilli are phagocytosed in the air spaces primarily by resident alveolar macrophages (AMs) and dendritic cells (DCs). Following lung exposure, Mtb-infected phagocytes can migrate from the alveolar space into the lung interstitium and then, via the lymphatic and hematogenous routes, disseminate to other organs [2]. Subsequently, the bacilli may grow unimpeded within host macrophages, resulting in primary progressive disease or reactivation disease after a short period of latency, as seen in about 10% of immune competent individuals. Alternatively, bacillary growth may be controlled, and the bacteria may be killed or may adapt to survival within cellular granulomas in a non-replicating state, thereby establishing a latent infection, as is seen in approximately 90% of otherwise healthy hosts. Latent infection can persist for decades after exposure to Mtb before reactivating to cause active disease (primarily in the lungs), when the immune-mediated control of bacillary growth fails, as seen for example following human immunodeficiency virus (HIV) infection [3]. The ability of Mtb to establish latency, combined with the massive exposure of individuals to infectious organisms in hyperendemic areas of the world, is responsible for the huge reservoir of latently infected individuals. The Global Burden of Disease Study determined that, globally, tuberculosis (TB) is the 7th leading cause of Disability Adjusted Life Years (DALYs) and, unlike most infectious diseases, will still be among the top ten causes of DALYs in 2020 [4]. Thus, an understanding of mechanisms through which Mtb bacilli interact with host macrophages (particularly AMs), grow, persist and reactivate, is crucial to the development of new tools for improving TB control.

2. The macrophage

2.1 Macrophage heterogeneity and plasticity

Macrophages serve as the major host cell niche for the growth and survival of Mtb. However, these cells are also responsible for activation of the protective immune responses, both innate and acquired, which are necessary to control or eliminate the infection. Macrophages, derived from hematopoietic cells in the bone marrow [5,6], differentiate from promonocytic cells to mature monocytes in the peripheral blood and further into macrophages, following migration into tissue where they maintain homeostasis (low-level recruitment) or are recruited in response to inflammation/infection (high-level recruitment) [7,8]. Macrophages are present in almost all tissues throughout the body. Their pattern of differentiation is highly dependent on the local environment, including the tissue location and associated cells, as well as growth factors and cytokines present at each site. Through the expression of various cell surface receptors, the macrophage recognizes, binds and internalizes foreign particles, including Mtb. This initiates a complex process of control of intracellular growth of the bacilli via a cascade of signaling events that result in the release of soluble and cell-associated antimicrobial and innate immune mediators [9].

Early macrophage biology studies revealed heterogeneity, functional and morphologic, often based on phenotyping the diverse cell populations using antibodies [1013]. More recent advances in genetics, isolation of monocyte subsets, improved DNA microarrays and proteomics have allowed scientists to reconsider macrophage activation phenotypes in more detail [1422]. The heterogeneity observed reflects the plasticity and adaptation of the cells to different anatomical and immunologic locations. For example, high expression of a subset of pattern recognition receptors (PRRs) on AMs appears to be associated with the ability of these cells to clear particles and microbes from the lungs without causing excessive inflammation (see below). Recent studies have suggested that the initial interaction of macrophages with soluble mediators, such as cytokines, determines the functional phenotype of the cells; others have shown that macrophages can be continuously altered as the environment changes [2328]. These observations help explain the fact that Mtb interactions with macrophages can vary greatly depending upon the local microenvironment in which they occur.

For simplicity, macrophage heterogeneity has been categorized into four major groups defined primarily by in vitro cell culture under different conditions: type I and type II macrophages, alternatively activated macrophages and deactivated macrophages [15,2932]. Type I macrophages (classical activation or M1 cells) are differentiated by in vitro culture with the lymphoid cell mediator interferon gamma (IFN-γ) and lipopolysaccharide (LPS), a Gram-negative microbial trigger which induces pro-inflammatory cytokine production. Type II macrophages (innate activation) are differentiated in in vitro culture by ligation of receptors by immune complexes. Both of these macrophage phenotypes are associated with high microbicidal activity, the production of pro-inflammatory cytokines [tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β) and interleukin 6 (IL-6)] and reactive oxygen species (ROS), and the activation of inducible nitric oxide synthase (iNOS), the latter leading to the synthesis of nitric oxide (NO) [33]. Type I macrophages also have increased major histocompatibility complex (MHC) class II and cluster of differentiation (CD) 86 (CD86) expression, and increased antigen presentation. Differences between them include decreased mannose receptor (MR, CD206) expression in type I macrophages, while type II macrophages have increased production of the immunoregulatory cytokine interleukin 10 (IL-10) and decreased production of the pro-inflammatory cytokine interleukin 12 (IL-12) [30,31]. Thus, the two macrophage populations, although phenotypically similar, have distinct functional profiles.

The third major group is the alternatively activated macrophage (or M2 cells) [30,34,35]. These cells result from in vitro culture with the TH2-type cytokines, interleukin 4 (IL-4) or interleukin 13 (IL-13), which decrease cellular responsiveness to IFN-γ and inhibit the synthesis of iNOS [30]. Glucocorticoids are also able to induce an alternative macrophage activation state [32]. Alternatively activated macrophages have increased PRR expression, particularly of the MR, with decreased CD14 expression. Additionally, these cells do not produce large amounts of oxidants or pro-inflammatory cytokines, but rather secrete some anti-inflammatory cytokines (e.g. transforming growth factor beta (TGF-β)) [30,31] and decrease the TH2 response likely by regulating the stimulation of lymphocytes [36]. This macrophage population has been associated with tissue repair and humoral immunity. Finally, the deactivated macrophage phenotype is induced by in vitro culture in the presence of cytokines such as IL-10 or TGF-β, or by ligation of inhibitory receptors (i.e., CD200-CD200R, CD47- CD172a or esteroids). This macrophage phenotype has been associated with anti-inflammatory cytokine production, prostaglandin E2 (PGE2) production and reduced MHC II expression [30].

Although this classification provides a useful framework for studying and understanding macrophage heterogeneity, it is undoubtedly simplistic and lacks the impact of specialized local in vivo microenvironments on the macrophage phenotype [3741]. A more flexible classification has been suggested recently where macrophage heterogeneity, generated in response to innate or acquired immune responses, is considered to be a spectrum, wherein different cell populations, such as classically activated macrophages, wound healing macrophages and regulatory macrophages, may overlap in their functions, representing different points along a spectrum [42].

2.2 Macrophages in the Mtb granuloma

Phagocytosis of Mtb by AMs and DCs initiates a cascade of events involving the production of cytokines and chemokines, which stimulate the activation of phagocyte anti-microbial activities and recruit blood polymorphonuclear leukocytes (PMN) and additional mononuclear leukocytes into the tissue to the site of infection. The accumulation of mononuclear leukocytes around foci of infected cells leads to the formation of a macrophage-rich cell mass known as the granuloma. Macrophages play an essential role in the formation of Mtb granulomas. The macrophage population within these structures has a high turn-over rate and is diverse, including epithelioid cell, multinucleated giant cell (MNG) [43] and foamy cell [44] phenotypes. The role of the granuloma is to control the growth of intracellular Mtb and to limit bacillary dissemination. Persistence of the granuloma depends on local production of TH1 type cytokines by antigen-specific T lymphocytes, responding to the presence of Mtb antigens and leading to sustained phagocyte activation, inflammation and anti-microbial activity. If optimal, the anti-microbial granulomatous response to infection leads to complete bacillary control and latency. As the bacilli are controlled and latent infection is established, the granulomas decrease in size and cellularity, and the lungs typically show no clinical signs of disease by chest X-ray [45]. However, when the granulomatous response is not fully protective, the bacilli continue to replicate and active TB disease ensues. The lungs of such individuals contain enlarging granulomas that differentiate with time, leading to the development of cavitary pulmonary disease. A complex set of interactions between host macrophages and their response to the infecting bacilli, as well as the specific properties of the Mtb strain, are presumed to be responsible for determining the outcome during this process.

Histopathologic examination of lesions in the lungs of humans with advanced pulmonary disease reveals heterogeneous cellular architecture, most prominent in the concentrically layered cavitating and non-cavitating necrotic granulomas seen in patients with sputum-positive TB. Cross-sections of large cavitating lesions show loose cellular accumulation at the luminal surface of the cavity, consisting of numerous PMNs and macrophages, surrounded by a layer of acellular caseous necrotic material. In non-cavitating closed granulomas, the central necrotic area is fully acellular. Subtending the acellular necrotic layer in both types of lesions, there is granulomatous-fibrotic tissue with a mixed mononuclear leukocyte infiltrate consisting of Langerhans-type giant cells, epithelioid macrophages, foam cells and many scattered lymphocytes [46]. Acid-fast bacilli (AFB), apparently cell-associated, can be detected in large numbers at the cavity surface; while the granulomatous-fibrotic layer, with abundant macrophages, MNGs, and lymphocytes, is essentially devoid of visible AFB. In addition, AFBs are seldom seen in AMs residing within airspaces of the residual functional lung. In closed (non-cavitary) necrotizing granulomas, small to moderate numbers of AFB can be observed in foamy macrophages occupying the borders of the necrotic areas, most prominently where breakdown (liquefaction) is occurring. Thus, in most patients with sputum-positive disease, AFB are most numerous at the luminal surfaces of the cavities, i.e., in areas of the granulomas with a patent connection to the airways. In comparison, in sputum-negative patients, the surfaces of the cavities appear inactive, with re-epithelialization over fibrotic tissue. Despite the absence of any visible AFB and the failure to grow bacilli from many of these lesions, MNGs, epithelioid macrophages, and lymphocytes are apparent in small aggregates within the fibrotic tissue. Staining for the presence of CD3+CD4+ or CD3+CD8+ T lymphocytes reveals an abundance of these cells within the granulomatous-fibrotic layer and in lymphoid aggregates of the granuloma. Scattered T lymphocytes are also seen within the airspaces. In contrast, a striking absence of CD3+ CD4+ and CD3+CD8+ T cells is noted in the necrotic zone, as well as at the luminal surface of the cavity [46].

Taken together, these observations suggest that in the cellular granulomatous fibrotic area, adjacent to the necrotic zone, a microenvironment exists where macrophages and T cells co-localize and are free to interact directly, presumably resulting in an efficient immune response capable of inhibiting mycobacterial replication. In contrast, only millimeters away, the luminal surface of the cavity represents a microenvironment within the lung where macrophages do not co-localize with T cells, thus precluding any direct T-cell-macrophage interactions at these sites. This may result in failure to activate the macrophages, thus rendering them permissive to the growth of Mtb. Interestingly, the presence PMNs at the luminal surface of the cavity could potentially be associated with down-regulation of the local control of bacillary growth, particularly in advanced disease.

2.3 The alveolar macrophage

In addition to its central role in respiration, the lung serves as a major interface between the host and the external environment and is constantly bombarded by foreign matter, including microbes. Therefore, the lung contains an intricate pulmonary innate and adaptive immune system which serves to protect the host from inhaled foreign particulates [47,48]. Upon inhalation, small (< 5 μm) particulates, such as microbes, are able to avoid the upper airway ciliary beat, the cough reflex, and mucus clearance mechanisms to travel down the trachea and through the bronchi, where they eventually settle in the alveolus. Pulmonary innate immunity at this site is controlled by cellular and soluble components; airway and alveolar epithelial cells and leukocytes join forces with antimicrobial products (e.g. collectins, defensins, lactoferrin, and cathelicidins) secreted into the epithelial lining fluid [49]. Mtb is deposited in this environment and, thus, its interactions in this site are particularly relevant to immune pathogenesis of active TB disease.

The inflammatory response in the alveoli must be tightly regulated in order to protect the delicate gas-exchanging structures from destruction by toxic mediators of the immune system [48,5053]. AMs are closely associated with the alveolar epithelium and are continuously bathed in surfactant, which is an important immune modulator produced by type II epithelial cells [54]. AMs comprise greater than 95% of the cells found in a bronchoalveolar lavage. Only one to two AMs are found per alveolus, ranging in size from 9–40 μm in diameter [50]. These cells constitute the first line of defense against pulmonary pathogens [55]. The majority of AMs are thought to originate from peripheral blood monocytes that migrated into the airways where they differentiated [14,56]. Alternatively, and more controversial, mononuclear phagocytes present in the lung can divide in the alveolus in response to local inflammatory stimuli [50,57].

AMs are generally considered to be alternatively activated macrophages. While the cells effectively eradicate routinely encountered microbes, they often fail to do so for host-adapted intracellular pathogens such as Mtb. The specific innate inflammatory response produced by AMs upon recognition and uptake of pathogens influences the subsequent adaptive immune system and determines whether the microbe is successfully eliminated with minimal damage to the host [58]. In this regard, AMs have a unique phenotype that includes expression of high levels of a subset of surface PRRs such as the MR, specific Toll-like receptors (TLRs), and scavenger receptor (SR) A [59]. They also have high expression of intracellular regulators, such as nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) that respond to infectious agents [60,61] and mediate their clearance [62] and yet do not cause the same marked increase in local tissue inflammatory responses seen in other macrophage populations [58]. Thus, AMs appear to be immunoregulatory cells with high phagocytic activity but relatively poor bactericidal and antigen presentation capabilities, and with the ability to suppress lymphocyte activation [63,64]. They possess a relatively attenuated respiratory burst, increased production of the anti-inflammatory mediators PGE2 and TGF-β, as well as IL-10, and function to inhibit the amplification of signaling leading to a robust pro-inflammatory response [30,31,6569]. In this way, AMs protect the delicate lung tissues from destruction by inflammatory mediators [70] or a damaging oxidative burst [71]. In mice, this decrease in oxidative metabolites is partially due to AM production of arginase, which limits NO production [72]. AMs produce less of the intracellular signaling molecule TLR9, which is consistent with a decreased inflammatory response to pathogens [73]. AMs also have decreased production of calcium-dependent protein kinase C (PKC) isoforms and decreased activation of the transcription factor activator protein 1 (AP-1) [74]. Through the increased activity of PPARγ, they also function to inhibit the amplification of intracellular signaling leading to a robust pro-inflammatory response by repressing the transcription factors nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), AP-1, and signal transducer and activator (STAT) [60,7577].

Pulmonary collectins play an important role in the Mtb-AM interaction. Upon entry into the alveolus, Mtb encounters surfactant, and two surfactant-associated collectins, surfactant protein A (SP-A) and surfactant protein D (SP-D), which regulate the early interaction of the bacilli with resident phagocytes [53,7880]. SP-A has been shown to enhance PRR activity, increase phagocytosis, alter production of pro-inflammatory cytokines, and decrease reactive nitrogen intermediates (RNIs) and reactive oxygen intermediates (ROIs) in response to stimuli [53,81]. In contrast, SP-D decreases phagocytosis of Mtb by binding to the mannose caps of the Mtb cell wall mannosylated lipoarabinomannan (ManLAM) (see below) and inhibiting the normal interaction with the MR [82]. SP-D-opsonized Mtb that enter the macrophage have reduced intracellular growth due in part to increased phagosome-lysosome fusion [82,83].

In summary, the increased phagocytic potential, combined with a highly regulated and relatively balanced pro- and anti-inflammatory response, makes AMs ideal for preserving the alveolar structure and its essential gas exchange function. Host-adapted intracellular pathogens like Mtb appear to be able to exploit the tightly balanced activity of these cells to enhance their survival and persistence [60,84].

3. Macrophage interaction with Mtb

3.1 Macrophage recognition of Mtb

Macrophages express an array of PRRs and phagocytic receptors that play crucial roles in their recognition and response to pathogens, essential in the initiation of the innate immune response. The divers cell surface receptors are responsible for the generation of combinatorial signals that result in macrophage activation as part of the host defense machinery against invading pathogens [8587].

I. C-type lectins: Mannose receptor, DC-SIGN, Dectin-1 and Mincle

The MR (CD206) is expressed on AMs [88,89], monocyte-derived macrophages and DCs [90], but not on monocytes [9093]. It is the predominant C-type lectin expressed on non-activated human macrophages; this selective distribution is different for other mammalian macrophages. Its eight lectin-like carbohydrate recognition domains (CRDs) bind with high avidity and affinity to mannans, notably endogenous unwanted high mannose N-linked glycoproteins, to maintain homeostasis of the host [94,95]. The MR also interacts with microbial pathogen-associated molecular patterns (PAMPs) that contain mannose, found on many different pathogens [96]. The development of these mannosylated PAMPs is thought to be a form of molecular mimicry by which pathogenic microbes, such as Mtb, can evade the immune system through cloaking themselves in molecules that are similar to mannosylated glycoproteins found within the host. Mtb is recognized by the MR via its mannosylated surface structures [97]. MR recognition varies among Mtb strains [98,99], and its involvement has been postulated to be a marker of host adaptation [97]. The MR can discriminate among specific cell wall components of the bacteria due to subtle variations in the degree and nature of mannan motifs. It specifically binds to the mannose caps of ManLAM [100] and to higher order phosphatidyl-myo-inositol mannosides (PIMs) found in greater amounts in pathogenic bacteria [101]. MR ligation on non-activated macrophages produces an anti-inflammatory response by stimulating the release of anti-inflammatory cytokines [102] and inhibiting the production of pro-inflammatory IL-12 [103] and ROS [104]. Entry of Mtb through the MR leads to the development of a unique phagosome that has reduced fusion with the lysosome [101,105,106]. This phagosome does not acidify normally in part because it does not acquire all subunits of the vacuolar proton ATPase [107,108]. MR engagement by Mtb leads to the induction of PPARγ which, as noted earlier, functions to inhibit a robust pro-inflammatory response. As a prototypic PRR, the MR links innate and adaptive immune mechanisms and, with regards to Mtb, facilitates presentation of lipids and ManLAM through CD1b [90,109116]. Finally, the MR is thought to play a role in cellular adhesion and fusion, with the formation of MNG, and is thus implicated in Mtb granuloma formation [117].

DC-SIGN (dendritic-cell specific intercellular adhesion molecule 3 grabbin non-integrin) demonstrates high avidity binding to mannosylated glycoconjugates such as N-linked high mannose structures and fucose-containing glycans [101,118121]. It is present on immature and mature DCs and small sets of macrophages [89,122] and binds to a variety of microbial pathogens [123]. In general, DC-SIGN is not expressed on non-activated human macrophages; however, its expression can be up-regulated upon Mtb infection [124] and induced on macrophages stimulated with IL-4 or IL-13 and granulocyte macrophage colony stimulating factor (GM-CSF) [125]. Mtb ManLAM and PIMs serve as ligands for DC-SIGN on DCs [101,126]. Upon interaction of DC-SIGN with Mtb, the phagocytosed bacteria are targeted for phagosome-lysosome fusion in the antigen presenting cells (APC) [127129] and DC maturation is impeded [126].

Dectin 1 is a transmembrane PRR expressed on macrophages as well as DCs, monocytes and a subset of T cells [130]. It binds to β-glucan, a common component of the fungal cell wall [131]. Dectin 1 ligation triggers phagocytosis and intracellular signaling cascades, including synergistic interactions with the TLRs, leading to a pro-inflammatory response [132] and a respiratory burst [133]. The role of Dectin-1 in Mtb infection is still unclear. Although no specific mycobacterial ligands for Dectin-1 have been identified so far, this receptor interacts with mycobacteria in concert with TLR2 to produce cytokines such as TNF-α and IL-12p40 [134,135]. Recent studies have proposed additional roles for Dectin-1 in Mtb infection [136,137].

Mincle (Macrophage-inducible C-type lectin; also known as Clec4e or Clecsf9) is mainly expressed on myeloid cells. Mincle expression is very low on non-activatd leukocytes, but is up-regulated after exposure to various inflammatory stimuli, such as cytokines and TLR ligands [138]. Its ligation can induce cytokines such as TNF-α, MIP-2 (macrophage inflammatory protein 1) (CXCL2), and IL-6. Mincle can sense infection by some fungi [139] and can detect an endogenous protein, spliceosome-associated protein 130 (SAP 130), which is released from necrotic host cells. Mincle has recently been shown to serve as a PRR for trehalose dimycolate (TDM) from Mtb [140] but is not essential for controlling Mtb infection in mice [141].

II. Complement receptors and Fcγ receptors

The complement C3 receptors (CR1, CR3 and CR4) and Fcγ (FcγRI, II, III) receptors are major phagocytic receptors on monocytes and macrophages, although their expression and activities vary in a tissue-specific manner. Among the CRs, CR4 is reported to be more prominent in AMs than CR1 and CR3 [142]. In fact, the relative CR expression pattern changes during differentiation with CR3 ≫ CR4 on monocytes developing into CR4 > CR3 on AMs [143]. Thus, complement-opsonized pathogens will interact differently with AMs compared with other macrophage/monocyte populations.

CRs expressed on human monocytes and macrophages play an important role in both opsonic and non-opsonic phagocytosis of Mtb [59,144146], the latter through interaction of the receptor with surface polysaccharides [147], lower order phosphatidylinositol mannosides (PIMs; see below) and glycopeptidolipids [148]. Although CR3 mediates Mtb phagocytosis, the host response following phagocytosis through this receptor is unclear for human macrophages. In vitro analysis of peritoneal macrophages from CR3-knockout mice showed decreased uptake of Mtb with equivalent bacterial growth compared to wild type [149]. However, injecting intravenous Mtb into CR3-knockout mice failed to reduce bacterial burden or lessen pathological lesions [150].

Despite the relatively increased expression of Fcγ receptors on AMs, these receptors do not play a role in the initial phagocytosis of Mtb in the absence of opsonizing immune antibody [144] which generally requires activation of the adaptive immune response for specific antibody production, a process that takes several weeks. However, when the bacteria are opsonized with Mtb-specific antibody and phagocytosed, there is enhanced phagosome-lysosome fusion [151], facilitating an increased host macrophage protective mechanism.

III. Toll-like receptors

TLRs are a highly conserved family of transmembrane receptors with an extracellular amino-terminal leucine-rich repeat (LRR) domain that recognizes PAMPs and an intracellular carboxy-terminal tail that is homologous to the interleukin 1 receptor (IL-1R) [152]. The receptor contains a Toll-IL-1R (TIR) domain that forms a scaffold for the assembly of signaling intermediates. TLRs are present on AMs [153], neutrophils [154], lymphocytes [155] and DCs [156] as well as on alveolar epithelial cells [157]. There are at least twelve mammalian TLRs, each responding to a variety of ligands [86,158161]. After specific ligand binding, TLRs such as TLR2 and TLR4 initiate an intracellular signaling cascade, which generally leads to differential activation of NF-κB and an inflammatory response [86,162,163]. However, several negative regulators, such as interleukin-1 receptor-associated kinase M (IRAK-M), have been identified [164], and pulmonary surfactant can drive increased IRAK-M expression and IL-10 production in macrophages [165]. Signaling can also lead to alternate intracellular cascades, resulting in an anti-inflammatory response [166]. To add to the complexity, TLRs are increasingly found to interact with other cell surface receptors, leading to a modulated inflammatory response [167,168].

TLRs are critical mediators of the immune response to a variety of pathogens, including Mtb [169,170]. TLRs are either expressed on the cell surface (e.g. TLR2 and 4) or intracellularly (e.g. TLR8 and 9) [171]. Mtb and its cell wall components are recognized by several TLRs, including TLR1, TLR2, TLR4, TLR6, and TLR9 [172176]. Among them, evidence for genetic variants associated with TB susceptibility is most abundant for TLR2, which functions alone or as a heterodimer with TLR1 or TLR6. The 19kDa lipoprotein, lipomannan (LM), and lower order PIMs found on the surface of the mycobacterial cell wall have all been shown to interact with TLR2 [177179]. TLR expression and function are influenced by the local pulmonary microenvironment. For example, SP-A up-regulates the surface expression of TLR2 on human macrophages, while inhibiting the intracellular signaling of TLR2 and TLR4, which results in a dampened pro-inflammatory response [80].

IV. Scavenger receptors

There are several scavenger receptors (SRs) on AMs with reported roles in antimicrobial host defense, namely SR I and II (SR-AI/II) and the macrophage receptor with collagenous structure (MARCO) [180182]. In the context of Mtb infection, MARCO is thought to be involved in TLR signaling in response to cell wall components, resulting in increased NFκB activation [183].

V. CD14

CD14 is highly expressed in macrophages and monocytes. It binds to the plasma membrane through a glycophosphatidylinositol (GPI) anchor, although it can also be found in its soluble form in plasma [184]. CD14 recognizes peptidoglycan from Gram-positive bacteria [185] and LPS from Gram-negative bacteria [186]. Other bacterial ligands are lipoteichoic acid, lipoproteins and lipoarabinomannan (LAM) from mycobacteria, and mannuronic acid from Gram-negative bacteria [185,187,188]. CD14 has been shown to facilitate the uptake of non-opsonized Mtb by microglia (resident brain macrophages) [189], although by itself is not capable of mediating phagocytosis of the bacilli by human macrophages without the cooperation of other receptors [190] such as TLR2 and 4. Upon ligand binding, macrophage activation leads to inflammatory cytokine production.

VI. The NOD-like receptors

The NOD (nucleotide-binding oligomerization domain)-like receptor (NLR) family members play a major role in innate immunity through inducing an inflammatory response and regulating cell death/survival pathways. They are a second line of recognition, inducing a pro-inflammatory response to bacteria once inside the macrophage [191]. NLRs are mainly expressed in APCs, including AMs and epithelial cells. Most mammalian NOD family members contain 3 distinct functional domains: an amino terminal effector binding domain (EBD), a centrally located NOD, and a carboxy-terminal ligand recognition domain. The NLR family can be divided into four subfamilies depending on the composition of their N-terminal EBD. The differing N-terminal domains are as follows, with subfamily name: acidic transactivation domains (NLRAs), caspase activation and recognition domain (CARD) (NLRCs), pyrin domains (NLRPs), and baculovirus IPA repeat domains (NLRBs). NOD1 and NOD2 are part of the NLRC subfamily and contain a LRR domain, NOD domain, and CARD domains [192]. NOD1 protein is abundantly found in multiple cell types, whereas NOD2 protein expression is abundant in human macrophages [193,194]. NOD1 and NOD2 recognize specific muropeptides found in the peptidoglycan layer of Gram-positive and Gram-negative bacteria [195]. Upon ligation, they activate the mitogen-activated protein kinases (MAPKs) and thereby indirectly allow for NF-κB and AP-1 activation, leading to the production of pro-inflammatory cytokines [196]. NOD2 can directly bind and activate caspase-1 and interact with the NALP1/NALP3 (NACHT, LRR and PYD domain-containing proteins 1 and 3) inflammasome, which causes the activation of caspase-1, an enzyme needed to cleave pro-IL-1β into its active secreted form [197]. Furthermore, there are many reports on the synergistic crosstalk between TLR agonists and NOD2 agonists in pro- and anti-inflammatory cytokine release [198205].

NLRs have proven to be important in the recognition of a variety of bacterial pathogens, including Mtb [197,206216]. Recent studies have reported that NOD2 does not have a significant role in controlling Mtb growth during early infection in mouse macrophages [217] but may play a role during late infection [214]. A decrease in pro-inflammatory cytokine production has been observed in mouse NOD2 knockout bone marrow-derived macrophages and naive murine AMs in response to Mtb, without affecting intracellular bacterial growth [214,217]. NOD2 recognizes an N-glycolylated form of muramyl dipeptide (GMDP) found in Mtb [195] and controls the nature of the inflammatory response and subsequent fate of Mtb and M. bovis BCG in human macrophages [194].

3.2 The Mtb phagosome

During normal phagocytosis, actin-mediated membrane movements engulf the bacterium into a phagosome with the sequential recruitment of Rab GTPases to the phagosomal membrane, which then recruits the vacuolar ATPases to acidify the phagosomal contents. The membrane ultimately fuses with a lysosome to merge the contents of the acidified phagosome with the lysosomal acid hydrolases. However, during Mtb infection, the phagosome trafficking pathway is altered through multiple mechanisms to disrupt normal host cell microbicidal activities and/or phagocyte effector functions [218221]. Consequently, once inside the macrophage, Mtb resides in a unique phagosome with an abnormally high pH of ~6.2 and limited fusion of pre-formed lysosomes [222,223]. The early trafficking pattern of the Mtb phagosome includes fusion with early endosomes, since both iron [224,225] and glycosphingolipids [226] are found associated with it. However, Mtb ManLAM can inhibit normal calcium increase in the cytosol, causing a disruption in calmodulin complex formation with phosphatidylinositol 3-kinase (PI3K) [222,227,228] and preventing the recruitment of phosphatidylinositol 3-phosphate (PI3P) to the phagosomal membrane [229]. PI3P is a critical lipid intermediate in the recruitment of the vacuolar GTPases to the phagosome [230]. Mtb also inhibits sphingosine kinase which inhibits calcium signaling [231]. Inhibition of full maturation of the phagosome also involves the lack of recruitment of Rab5 effector proteins, such as early endosomal autoantigen 1 (EEA1) [59,232] and hVPs35, to the phagosomal membrane [223,233,234]. EEA1 and Syntaxin-6 are required for the delivery of lysosomal hydrolases, cathepsins and vacuolar ATPases. The Mtb phagosome lacks a specific type III PI3K, required for retention of EEA1 on the endosomal membrane [108,230,235]. Ultimately, these processes result in a failure of phagosome maturation between the Rab5 (an early endosomal marker) [236] to Rab7 (a late endosomal marker) [237] conversion.

ESAT-6/CFP-10 (early secretory antigenic target 6/culture filtrate protein 10), the SecA 1/2 proteins and the eukaryotic-like serine/threonine protein kinase G (PknG) from Mtb interfere with phagosomal maturation [238241]. Also, several of the Mtb lipoglycans discussed earlier have important effects on phagosome-lysosome fusion. For example, ManLAM modifies trafficking and phagosome-lysosome fusion, as well as decreasing MAPK activation, a critical intracellular signaling molecule [242,243]. The lower order PIMs (fewer mannose molecules), which are found more commonly in less virulent mycobacteria, can enhance phagosomal fusion with early endosomes [179]. Glycolipids such as TDM can interfere with membrane trafficking, preventing phagosome maturation [244]. Recent studies have focused on the utilization of host fatty acid stores and fatty acid metabolism for persistence of Mtb in the phagosome [245247].

Many studies over several decades have provided evidence that Mtb enters and divides within the macrophage phagosome. However, there continues to be active debate over whether Mtb can also escape from the phagosome into the cytosol [248] and why this would be advantageous to the bacterium or the host. Experiments performed by several groups during the mid 1980s and 1990s showed electron microscopy images where Mtb appears devoid of a phagosomal membrane some days after infection [249251]. More recently, Brown and colleagues [252] showed that at least some portion of intracellular M. marinum (a highly genetically related mycobacterium) escape from the phagosome into the cytosol using an actin-based propulsion system similar to Listeria; others have shown that the presence of region of difference 1 (RD1) in the bacterial genome is required for this escape [253]. Similarly, Mtb and M. marinum have been shown to be ejected from the amoeba Dictyostelium through an actin-based structure called the ejectosome, using elements of RD1 [254]. Peters and colleagues have recently reported that Mtb can escape from the phagosome of DCs and macrophages after several days in culture [255] in an RD1- and ESAT-6-dependent manner [256]. Several factors have contributed to the discordant results obtained among labs throughout the years regarding bacterial localization and escape. These include the source of cells used, bacterial strain, multiplicity of infection, length of infection, how the bacterial inoculum was prepared, and the microscopy technique used to visualize the phagosomal membrane.

Some studies have proposed that Mtb might be present in the macrophage cytosol several days after infection on its way to escape from the cells and to spread to adjacent cells. Consistent with this, there is growing evidence that virulent Mtb induces cell necrosis by activation of the cytosolic positioned inflammasome in an RD-1 dependent fashion [257,258]. In a very recent study, phagosomal rupture by Mtb and M. marinum was closely followed by necrotic cell death of the infected macrophages [259]. Thus, further studies are needed to determine whether phagosomal escape is a virulence strategy in terms of intracellular survival or just a consequence of membrane rupture during cell death. Another possibility is that early following infection of macrophages, intraphagasomal Mtb, which possesses an RD-1 dependent ESX-1 secretion system, is able to perforate but not destroy the phagosomal membrane, allowing for a mixture of phagosomal and cytosolic components [260,261]. Finally, it has been postulated that the escape of Mtb from the phagosome to the cytoplasm might be a mechanism by which mycobacterial antigens can be processed and loaded onto MHC I for presention to CD8+ T cells. While the escape of virulent Mtb from the phagosome provides a possible explanation for this form of antigen presentation, an alternative hypothesis is that the phagosome can interact directly with the endoplasmic reticulum [255,262264].

3.3 The pro-inflammatory cytokine/chemokine response

Local and systemic macrophage production of cytokines and chemokines are central to the cellular response to Mtb infection. These soluble mediators play an important role, not only in controlling early infection, but also during the chronic infection state [265,266]. Of major importance to Mtb infection control are the macrophage cytokines TNF-α, the IL-12 family, IL-6, IL-1α/β and IL-10 [267272].

TNF-α is an autocrine/paracrine cytokine produced by a variety of cells, including macrophages, DCs, lymphocytes, neutrophils, mast cells and endothelial cells. During Mtb infection, the cytokine functions to regulate the inflammatory response, stimulating the production of IL-1 and IL-6 [273]. It also contributes to the control of Mtb by inducing the production of ROIs and RNIs by macrophages and the early secretion of chemokines [274]. TNF-α plays a crucial role in maintaining granuloma structure and function [275]. Its production is highly regulated because excess TNF-α leads to tissue damage and immunopathology, along with worsened clinical symptoms.

IL-12 represents a family composed of IL-12p40 (homodimer p40 + p40), IL-12 or IL-12p70 (heterodimer p35 + p40), interleukin 23 (IL-23) (heterodimer p40 + p19) or interleukin 27 (IL-27) (heterodimer cytokine with Epstein-Barr virus induced gene 3 [EBI3 or IL-27B] + p28 [known as interleukin 30 (IL-30)]). It is produced by macrophages and DCs after activation by microbial ligands and other cytokines [276] and leads to the development of the TH1 response during Mtb infection [277,278]. Several studies have shown that IL-12 plays an important role in both innate and adaptative immune responses to Mtb infection. The level of complexity of this family of cytokines during Mtb infection has been highlighted recently [279]. The role of IL-27 during Mtb infection has been focused on during the past decade. This cytokine promotes both pro- and anti-inflammatory responses. Together with IL-12, it initiates the TH1 response, enabling the release of T cell IFN-γ during infection [280,281], although it can contribute to uncontrolled inflammatory responses over time [282,283]. A recent study has shown that IL-27 inhibits human macrophage activity during Mtb infection [284].

IL-23 is produced in response to interleukin 17 (IL-17) from TH17 CD4 T cells, and recent studies have demonstrated that the IL-17/IL-23 pathway may play a key role in controlling mycobacterial infections [285287] by enhancing the development of protective and regulatory immune responses. IL-17 promotes the development of antimicrobial responses, chemokine production and the recruitment of inflammatory cells [288,289]. IL-23 is required for the maintenance of TH17 responses against Mtb by inducing IL-17 production by memory T cells [290,291]. In general, the IL-17 family is crucial in keeping the balance between bacterial killing and minimizing tissue damage.

IL-6, produced by macrophages and a variety of other cell types [292,293], has both pro- and anti-inflammatory effects. It is produced during early Mtb infection and is critical for early protective mechanisms, such as cytotoxic T cell differentiation.

IL-1β is a product of inflammasome activation and regulates a number of processes important in controlling Mtb infection, including iNOS production [294], phagosomal acidification and maturation, up-regulation of adhesion molecules [295] and regulation of a variety of enzymatic activities, such as cyclooxygenase and phospholipase A [296,297]. It is essential in maintaining resistance against Mtb infection.

IL-10 is produced by monocytes, macrophages, DCs, and T regulatory cells (T regs) [298,299]and functions as an immunomodulatory cytokine. During Mtb infection, it is thought to suppress inflammation to limit tissue damage. It inhibits pro-inflammatory cytokines (IL-1, IL-6, IL-12, IL-8 and IFN-γ) [300305]and chemokines (CCL (C-C motif ligand) 3, CCL4 and CCL5) impacting cell recruitment, the generation of oxidants [300,306,307] and antigen processing and presentation [308]. Recent studies have shown that IL-10 may interfere with phagosome maturation in human macrophages [309].

The production of chemokines is essential for the recruitment of inflammatory cells to the site of infection. The early recruitment of macrophages is an important step in controlling the infection [310]. Mtb is a strong inducer of chemokines that participate in protective and immunopathogenic host responses during Mtb infection [311,312]. Several studies have focused on the expression of inducible chemokines after Mtb infection of macrophages in vitro [313]. The results have shown that human macrophages produce CCL2, CCL3, CCL4 and CCL5 (MCP-1 (monocyte chemoattractant protein 1), MIP-1a, MIP-1b and RANTES (regulated upon activation normal T cell expressed and secreted)) in response to virulent mycobacterial strains [314,315]. MIP-1a, MIP-1b and RANTES induce T cell activation and proliferation [316] and activation of macrophages [317], and MIP-1a promotes T cell differentiation [318]. Several studies have shown the presence of MCP-1, MIP-1a, RANTES and IP-10 in the serum and bronchoalveolar lavage of TB patients [314,319]. AMs produce more CCL2, CCL3, CCL4, IP-10 (CXCL (C-X-C motif ligand) 10) and CCL5 than monocytes after Mtb infection [314]. These results also suggest that chemokines interacting with CCR (chemokine C-C motif receptor) 1, CCR2 and CCR5 play a role in the influx of cells to the site of infection, thereby impacting granuloma formation [320]. Expression of chemokines by macrophages can influence TNF-α production by macrophages after Mtb infection, specifically CCL2, CCL3, CCL4, CCL5, CXCL10 and CXCL13 [321]. Chemokine receptor expression may also be affected by TNF-α production. Therefore, TNF-α may influence the chemokine network expression during Mtb infection, which can indirectly impact granuloma formation. Also, some mycobacterial cell wall components regulate the induction of chemokine secretion by macrophages [322].

3.4 Macrophage death during Mtb infection

The ability to regulate the death of infected host cells is important during many microbial infections. The host regulates cell death pathways to enhance the induction of immunity and to control pathogen dissemination, while the pathogen uses many strategies to manipulate host cell death pathways to enhance its survival. Apoptosis (programmed cell death) plays a critical role in homeostasis and embryonic development. Apoptosis is an energy-dependent process mediated by the caspase cascade that results in the formation of apoptotic vesicles, organelles where the cell contents are kept inside a plasma membrane. Apoptotic cells, including macrophages, are degraded by adjacent macrophages through efferocytosis, which has recently been shown to function as a host defense mechanism for Mtb [323]. Apoptosis represents primarily an anti-inflammatory and immunoregulatory process [324]. However, a recent study has shown that apoptosis may induce a pro-inflammatory response upon macrophage phagocytosis of Mtb-triggered apoptotic neutrophils [325]. In contrast to apoptosis, necrosis is a relatively energy-independent process that does not involve caspase activation. Necrosis elicits a pro-inflammatory response as the cell membrane is permeabilized and the cell contents are extruded. It has long been thought to be a non-specific response to excessive stress, tissue damage or microbial invasion of the cell. However, it may in fact follow a series of programmed steps [326]. Macrophage necrosis in the TB granulomas appears to be driven by the bacillary load and the proapoptotic cytokines produced by the infected and associated uninfected leukocytes surrounding bacilli-infected cells in the granuloma, and is a hallmark of the host granulomatous response to Mtb infection [327].

Finally, regarding inflammasome-related cell death types, pyroptosis and pyronecrosis can both occur with microbial infection. They are considered to be pro-inflammatory and are a way to increase the recruitment of immune cells to the site of infection, although they can be detrimental to the host if excessive inflammation is elicited. While it has been shown that Mtb is able to activate the NLRP3 inflammasome through ESAT-6 [328330], evidence is lacking for whether Mtb can induce pyroptosis and pyronecrosis in the macrophage.

In summary, cell death has been associated with both Mtb virulence and host defense. In the granulomas, where leukocyte necrosis can be extensive, macrophage death is closely associated with the pathology and tissue damage of chronic TB disease. The relative contribution of various specific cell death pathways during the course of Mtb infection in vivo (primary infection, latency and reactivation) awaits further study.

3.5 Chronic inflammation and fibrosis

The progression of Mtb infection to active disease leads to a chronic inflammatory state and eventually tissue necrosis, fibrosis and remodeling. Macrophages play a central role in these processes by secreting cytokines and inflammatory mediators, such as prostaglandins, and producing enzymes and growth factors that promote connective tissue degradation, fibrosis and angiogenesis. The matrix metalloproteases (MMPs) are important mediators of extracellular matrix proteolysis and tissue. Various types of collagenases (MMP1, MMP13), elastases (MMP12) and gelatinases (MMP2, MMP9) are members of the MMP family. Each of these enzymes degrades specific components of the underlying connective tissues associated with granulomas in the lung or other infected organs [331]. The interaction of these MMPs with their respective tissue inhibitors of metalloproteinases (TIMPs) in response to Mtb infection is an important regulatory component of immune pathogenesis at the site of Mtb infection [332].

During TB, Mtb promotes destruction of the lung extracellular matrix to cause necrosis, liquefaction and ultimately formation of cavities, where Mtb can proliferate and spread from the interstitium to the airways [46]. To generate cavities, Mtb requires the activity of MMPs [333,334]. The MMP:TIMP ratio is critical in regulating the proteolysis of tissue and controlling tissue damage. In this regard, several studies have assessed MMP expression in response to Mtb infection in vitro and in animal models. Expression of MMP-1 and MMP-9 is up-regulated in human THP-1 cells after stimulation with mycobacteria [335,336] or LM via TLR and CD14 signaling [337]. Mtb infection can up-regulate MMP-1, −3, −7 and −10 as well as the related A disintegrin and metalloproteinases (ADAMs) in primary human macrophages [338]. In addition, MMP-1 up-regulation was higher in Mtb-infected primary human cells, compared to M. bovis BCG, while MMP-7 production was equivalent. In a rabbit model of pulmonary TB, the transcript levels of MMP-1, 2, 3, 9, 1, 13 and 14 were found to be elevated during the active disease process, and characterized by tissue necrosis and cavitation [339]. Consistent with this finding, a fibrotic capsule surrounding the lung granulomas of rabbits with active disease was observed, as demonstrated by positive staining for collagen deposition [339]. In humans, the concentrations of MMP-1 and −3 were elevated in induced sputum and bronchoalveolar fluid of TB patients compared with non-TB patients [340]. A recent study has correlated the presence of MMPs, TIMPs and pro-inflammatory cytokines in human TB pleuritis patients, concluding that levels of MMP-1, −7 and −9, as well as TIMP-3, in vivo correlate with production of the pro-inflammatory cytokines IFN-γ and IL-6 [341]. Histologic studies on human TB granulomas have shown that MMP expression occurs at the site of Mtb granuloma formation; specifically, MMP-1 and −7 are expressed by epithelioid cells and giant cells [338], while MMP-9 is produced by the pulmonary epithelial cells [342]. Although the literature is robust for the importance of MMP-9 during Mtb infection, its biological relevance compared with other MMPs remains unclear.

4. Mtb strain diversity

4.1 Strain specific macrophage activation

Recent reports have indicated that different clinical Mtb strains can induce differential host immune responses, leading to variable levels of pathogenesis in animal models [343346]. Indeed, the view of pathogenic mycobacteria as a relatively homogeneous clonal population with minimal functional genetic diversity has been challenged by molecular genotyping [343,347,348] and whole genome sequencing of members of the Mtb cluster (MTC) [349,350]. An analysis of diverse Mtb and MTC strains show phylogenetically-constrained patterns of bacterial gene expression, including lineage-, genotype-, and strain-specific signatures. These observations suggest a functionally heterogeneous population of pathogenic mycobacteria, highlighting the impact of genetic diversity [351]. In the absence of significant horizontal gene transfer, lineages of Mtb may define discrete evolutionary trajectories bearing distinct phenotypic properties [352,353]. Thus, biomedically relevant traits may be non-randomly distributed in the bacterial population along clonal lines [354,355]. Recently, phylogenetically diverse Mtb strains have been shown to exhibit markedly different virulence and pathogenesis phenotypes in macrophage in vitro culture and animal infection models [344]. Moreover, a number of specific Mtb strain families have been reported to show an unusual degree of outbreak or epidemic potential, drug-resistance or marked tissue tropism in humans [356358]. Thus, there is evidence of genetic variation and associated phenotypic diversity in Mtb. However, there have been few in-depth studies to examine the mechanistic underpinnings for these clinically and epidemiologically important associations.

Recent studies suggest that the differential induction of host macrophage activation and consequently host immunity plays an important role in this Mtb strain-dependent diversity of pathogenesis [343,344]. One study has shown that a W-Beijing strain was able to replicate in cultured human macrophages at a 4–8 fold higher rate, compared to other unrelated Mtb strains [359]. In mice infected with a W-Beijing isolate (HN878), the bacillary load in the lungs was 10-fold higher than in mice infected with a non-W-Beijing isolate (CDC1551) [344]. Reduced survival of HN878-infected mice correlated with relatively weak pro-inflammatory immune cytokine production following in vitro macrophage infection (e.g. TNF-α). This differential immune response was attributable to the presence of an Mtb-specific phenolic glycolipid (PGL-tb) in HN878 [360]. The differential cytokine responses of macrophages exposed to lipid extracts prepared from HN878 or CDC1551 were similar to those stimulated by the intact bacilli, while other cellular fractions did not induce a differential response [361]. When a single gene necessary for the synthesis of PGL-tb (polyketide synthase, pks1-15) was disrupted (HN878pks1-15::hyg), a less virulent phenotype was obtained; complementation of the PGL-tb phenotype restored virulence. In another report, hypoimmunogenic strain CH, responsible for an outbreak in Leicester, was linked to a specific chromosomal deletion (Rv1519) [362]. While some of the microbial factors driving immunologic phenotypes have been shown to be strain-specific, a study describing cytokine responses between broader, phylogenetically defined “ancient” and “modern” lineages, noted reduced in vitro human macrophage responses in the latter group, possibly lending selective advantage in the context of rapidly expanding human populations [363]. A study by Lopez et al. found that genetically distinct Mtb strains (representing the four major lineages found globally) resulted in a spectrum of immunopathologies in a murine intratracheal infection model [343]. Macrophage infections with various Mtb strain types induced a differential pattern of cytokines in vitro [364]. Dormans et al. found in a murine model that 19 different Mtb complex strains from 11 major genotypes produced responses that varied widely with respect to virulence, pathology, bacterial load and delayed-type hypersensitivity [365]. In support, a study by Homolka et al noted that genetic diversity appeared to have functional consequences during intracellular infection of bone marrow-derived macrophages, where transcriptomic profiles were lineage-specific [351].

These studies and others show that subtle genetic alterations among clinical Mtb strains can lead to variance in the ability to induce immune responses in the infected host [366]. In this regard, recent studies have compared the immune response to the clinical Mtb isolates HN878 and CDC1551during infection of New Zealand White (NZW) rabbits. These studies and others demonstrate that selection of the Mtb strain used for infection determines whether the animals control the infection and establish latency (e.g. CDC1551) or fail to control infection with development of progressive cavitary disease (e.g. HN878) [367,368]. Infection of rabbits with the HN878 strain leads to the formation and maturation of all of the granuloma types discussed earlier, thereby mimicking human TB [367]. Gene arrays and RT-PCR have shown that, as granulomas progress, the nature of macrophage activation evolves as well [367,369].

4.2 The mycobacterial cell envelope

Induction of differential immune responses to Mtb strains is due in part to differences in the nature of the mycobacterial cell envelope, which plays a critical role in the survival of bacteria within macrophages [370,371]. The cell envelope consists of an innermost plasma membrane, followed by a peptidoglycan-arabinogalactan layer, a thick mycolic acid layer, and then an outer layer consisting mainly of surface carbohydrates and proteins [372]. Several of the components of the Mtb cell wall have been shown to be immunomodulatory, including the lipoglycoconjugates: LAM, LM and PIMs. These lipoglycoconjugates are biosynthetically and structurally related and are thought to be located both in the innermost layer as well as exposed on the surface of the bacteria. The surface-exposed carbohydrate moieties are, thus, available to interact with lectin components of the innate immune system, such as the MR and SP-D [83,98,100,145].

PIMs vary in their number of mannose sugars and acyl chains, which affect their interaction with cell surface receptors, such as the MR and DC-SIGN [101]. These variations confer subtle differences on the immune response. In general, LMs are thought to be pro-inflammatory molecules with variable effects on the immune system based on the species of origin. TNF-α production is robust for M. smegmatis LM-stimulated human macrophages, but not for LM from Mtb. This diversity was found to be due in part to differential microRNA regulation [373] in response to subtle variations in the structure of the molecule, such as the presence of increased succinates in M. smegmatis LM [374]. These results are consistent with the decreased TNF-α response seen in virulent mycobacteria infection compared to nonpathogenic mycobacteria [375]. Similarly, it has been shown that LM stimulates macrophages through different receptors based on the number of acyl chains with tri-acylated LM interacting through TLR2/TLR1, while tetra-acylated LM interacts through TLR4 [376].

The LAMs are the largest, most complex lipoglycans, built upon LM with branches of arabinans followed by species-specific ‘caps,’ which are the terminal molecules on the end of some of the arabinan branches. These caps can be 1-, 2- or 3- linked mannoses, known as mono-, di- or tri-mannoside caps (ManLAM), phosphatidyl-myo-inositol caps (PILAM), or uncapped (AraLAM from M.chelonae) [377]. Even within Mtb strains, there is variability in the number and type of caps in mycobacteria. For example, pathogenic Mtb laboratory strains (Erdman, H37Rv) have a higher proportion of α 1–2 linked di-mannoside-capped ManLAM, while less pathogenic strains (H37Ra) and species (M. marinum and M. avium) have relatively more mono-mannoside-capped ManLAM [378], and nonpathogenic M. smegmatis has infrequent PILAM [379]. ManLAMs are also important regulators of the immune response that assist in bacterial survival. As with the other lipoglycoconjugates, the interaction of ManLAM with the host immune system depends upon subtle variations in structure. For example, the number of mannose caps and degree of mannose capping can affect the cell receptor that interacts with the bacteria, since ManLAM from three different Mtb strains, all known to have mainly di-mannosyl groups on ManLAM, showed variations in their binding to the MR [99]. Thus, subtle variations in structures of these lipoglycans are important in determining the interaction of the bacteria with specific cell receptors and, ultimately, the nature of the host immune response [380]. We have recently shown this to be the case for a set of clinical Mtb isolates found to naturally possess a truncated ManLAM structure [99].

5. Conclusions

Our current understanding of the role of macrophages in TB, although incomplete, clearly demonstrates the central role these cells play both in the host protective immune response and control of infection, and in the maintenance of chronic infection and its associated tissue damage and pathology. As we gain more knowledge about macrophage responses in the context of organ-specific microenvironments, our understanding of the molecular details underlying the pivotal Mtb-macrophage interactions that occur during TB infection will become more defined. In future studies, it is critical that we better understand these interactions during the entire spectrum of TB from primary infection, dissemination, microbial growth within granulomas, control of infection and latency and re-activation disease. It is clear that the nature of the macrophage receptor recognition, signaling, inflammation and antigen presentation pathways differ during different stages of infection and disease, and that the nature of the infecting Mtb strain also contributes to this diversity of responses. Since recent publications have highlighted the major differences in immune response among humans and different animal models, future studies must focus on comparative biology among mammalian hosts with an eye towards the use of animal models that better recapitulate what is seen in human disease. In addition, an important future focus will be studies of human clinical samples and the use of platform technologies to maximize our understanding of human TB in order to develop rational approaches to find critically needed new biomarkers, therapies and vaccines.

Reference List

  • 1.World Health Organization. Global tuberculosis control WHO report 2011. 2011. [Google Scholar]
  • 2.Frieden TR, Sterling TR, Munsiff SS, Watt CJ, Dye C. Tuberculosis. Lancet. 2003;362:887–899. doi: 10.1016/S0140-6736(03)14333-4. [DOI] [PubMed] [Google Scholar]
  • 3.Nuermberger E, Bishai WR, Grosset JH. Latent tuberculosis infection. Semin Respir Crit Care Med. 2004;25:317–336. doi: 10.1055/s-2004-829504. [DOI] [PubMed] [Google Scholar]
  • 4.Murray CJL, Lopez AD. The global burden of disease: a comprehensive assessment of mortality and disability from diseases, injuries and risk factors in 1990 and projected to 2020. In: Murray CJL, Lopez AD, editors. The Harvard School of Public Health on behalf of the World Health Organization and The World Bank. 9. Harvard University Press; 1996. pp. 1–27. [Google Scholar]
  • 5.Hume DA, Ross IL, Himes SR, Sasmono RT, Wells CA, et al. The mononuclear phagocyte system revisited. J Leukoc Biol. 2002;72:621–627. [PubMed] [Google Scholar]
  • 6.Van FR, Cohn ZA. The origin and kinetics of mononuclear phagocytes. J Exp Med. 1968;128:415–435. doi: 10.1084/jem.128.3.415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ebert RH, Florey HW. The Extravascular Development of the Monocyte Observed In vivo. Br J Exp Path. 1939;20:342–356. [Google Scholar]
  • 8.Van FR, Diesselhoff-den Dulk MC, Mattie H. Quantitative study on the production and kinetics of mononuclear phagocytes during an acute inflammatory reaction. J Exp Med. 1973;138:1314–1330. doi: 10.1084/jem.138.6.1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kaisho T, Akira S. Critical roles of Toll-like receptors in host defense. Crit Rev Immunol. 2000;20:393–405. [PubMed] [Google Scholar]
  • 10.Austyn JM, Gordon S. F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol. 1981;11:805–815. doi: 10.1002/eji.1830111013. [DOI] [PubMed] [Google Scholar]
  • 11.Dijkstra CD, Van VE, Dopp EA, van der Lelij AA, Kraal G. Marginal zone macrophages identified by a monoclonal antibody: characterization of immuno- and enzyme-histochemical properties and functional capacities. Immunology. 1985;55:23–30. [PMC free article] [PubMed] [Google Scholar]
  • 12.Kraal G, Janse M. Marginal metallophilic cells of the mouse spleen identified by a monoclonal antibody. Immunology. 1986;58:665–669. [PMC free article] [PubMed] [Google Scholar]
  • 13.Kaplan G, Gaudernack G. In vitro differentiation of human monocytes. Differences in monocyte phenotypes induced by cultivation on glass or on collagen. J Exp Med. 1982;156:1101–1114. doi: 10.1084/jem.156.4.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, et al. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–964. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 16.Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–795. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Liddiard K, Rosas M, Davies LC, Jones SA, Taylor PR. Macrophage heterogeneity and acute inflammation. Eur J Immunol. 2011;41:2503–2508. doi: 10.1002/eji.201141743. [DOI] [PubMed] [Google Scholar]
  • 18.Li J, Pritchard DK, Wang X, Park DR, Bumgarner RE, et al. cDNA microarray analysis reveals fundamental differences in the expression profiles of primary human monocytes, monocyte-derived macrophages, and alveolar macrophages. J Leukoc Biol. 2007;81:328–335. doi: 10.1189/jlb.0206124. [DOI] [PubMed] [Google Scholar]
  • 19.Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82:244–252. doi: 10.1189/jlb.0307191. [DOI] [PubMed] [Google Scholar]
  • 20.Geissmann F, Gordon S, Hume DA, Mowat AM, Randolph GJ. Unravelling mononuclear phagocyte heterogeneity. Nat Rev Immunol. 2010;10:453–460. doi: 10.1038/nri2784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Grage-Griebenow E, Flad HD, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. J Leukoc Biol. 2001;69:11–20. [PubMed] [Google Scholar]
  • 22.Chow A, Brown BD, Merad M. Studying the mononuclear phagocyte system in the molecular age. Nat Rev Immunol. 2011;11:788–798. doi: 10.1038/nri3087. [DOI] [PubMed] [Google Scholar]
  • 23.Erwig LP, Kluth DC, Walsh GM, Rees AJ. Initial cytokine exposure determines function of macrophages and renders them unresponsive to other cytokines. J Immunol. 1998;161:1983–1988. [PubMed] [Google Scholar]
  • 24.Stout RD, Suttles J. Functional plasticity of macrophages: reversible adaptation to changing microenvironments. J Leukoc Biol. 2004;76:509–513. doi: 10.1189/jlb.0504272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Porcheray F, Viaud S, Rimaniol AC, Leone C, Samah B, et al. Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol. 2005;142:481–489. doi: 10.1111/j.1365-2249.2005.02934.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Biswas SK, Sica A, Lewis CE. Plasticity of macrophage function during tumor progression: regulation by distinct molecular mechanisms. J Immunol. 2008;180:2011–2017. doi: 10.4049/jimmunol.180.4.2011. [DOI] [PubMed] [Google Scholar]
  • 27.Stout RD, Watkins SK, Suttles J. Functional plasticity of macrophages: in situ reprogramming of tumor-associated macrophages. J Leukoc Biol. 2009;86:1105–1109. doi: 10.1189/jlb.0209073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–737. doi: 10.1038/nri3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, et al. Macrophage receptors and immune recognition. Annu Rev Immunol. 2005;23:901–944. doi: 10.1146/annurev.immunol.23.021704.115816. [DOI] [PubMed] [Google Scholar]
  • 30.Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • 31.Mosser DM. The many faces of macrophage activation. J Leukoc Biol. 2003;73:209–212. doi: 10.1189/jlb.0602325. [DOI] [PubMed] [Google Scholar]
  • 32.Goerdt S, Orfanos CE. Other functions, other genes: alternative activation of antigen-presenting cells. Immunity. 1999;10:137–142. doi: 10.1016/s1074-7613(00)80014-x. [DOI] [PubMed] [Google Scholar]
  • 33.Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001;2:907–916. doi: 10.1038/ni1001-907. [DOI] [PubMed] [Google Scholar]
  • 34.Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593–604. doi: 10.1016/j.immuni.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 35.Edwards JP, Zhang X, Frauwirth KA, Mosser DM. Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol. 2006;80:1298–1307. doi: 10.1189/jlb.0406249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nair MG, Du Y, Perrigoue JG, Zaph C, Taylor JJ, et al. Alternatively activated macrophage-derived RELM-{alpha} is a negative regulator of type 2 inflammation in the lung. J Exp Med. 2009;206:937–952. doi: 10.1084/jem.20082048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Garofalo RS, Orena SJ, Rafidi K, Torchia AJ, Stock JL, et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKB beta. J Clin Invest. 2003;112:197–208. doi: 10.1172/JCI16885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, et al. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med. 2008;205:1261–1268. doi: 10.1084/jem.20080108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, et al. Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J Immunol. 2000;164:762–767. doi: 10.4049/jimmunol.164.2.762. [DOI] [PubMed] [Google Scholar]
  • 40.Duluc D, Corvaisier M, Blanchard S, Catala L, Descamps P, et al. Interferon-gamma reverses the immunosuppressive and protumoral properties and prevents the generation of human tumor-associated macrophages. Int J Cancer. 2009;125:367–373. doi: 10.1002/ijc.24401. [DOI] [PubMed] [Google Scholar]
  • 41.Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–1616. doi: 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Helming L, Gordon S. The molecular basis of macrophage fusion. Immunobiology. 2007;212:785–793. doi: 10.1016/j.imbio.2007.09.012. [DOI] [PubMed] [Google Scholar]
  • 44.Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat Immunol. 2009;10:943–948. doi: 10.1038/ni.1781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mack U, Migliori GB, Sester M, Rieder HL, Ehlers S, et al. LTBI: latent tuberculosis infection or lasting immune responses to M. tuberculosis? A TBNET consensus statement. Eur Respir J. 2009;33:956–973. doi: 10.1183/09031936.00120908. [DOI] [PubMed] [Google Scholar]
  • 46.Kaplan G, Post FA, Moreira AL, Wainwright H, Kreiswirth BN, et al. Mycobacterium tuberculosis growth at the cavity surface: a microenvironment with failed immunity. Infect Immun. 2003;71:7099–7108. doi: 10.1128/IAI.71.12.7099-7108.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang P, Summer WR, Bagby GJ, Nelson S. Innate immunity and pulmonary host defense. Immunol Rev. 2000;173:39–51. doi: 10.1034/j.1600-065x.2000.917306.x. [DOI] [PubMed] [Google Scholar]
  • 48.Carlson TK, Brooks M, Meyer D, Henning L, et al. Pulmonary innnate immunity: Soluble and cellular host defenses of the lung. In: Marsh C, Tridandapani S, Piper M, editors. Regulation of Innate Immune Function. Transworld Research Network; Kerala: 2010. pp. 165–211. [Google Scholar]
  • 49.Zaas AK, Schwartz DA. Innate immunity and the lung: defense at the interface between host and environment. Trends Cardiovasc Med. 2005;15:195–202. doi: 10.1016/j.tcm.2005.07.001. [DOI] [PubMed] [Google Scholar]
  • 50.Fels A, Cohn ZA. The alveolar macrophage. J Appl Physiol. 1986;60:353–369. doi: 10.1152/jappl.1986.60.2.353. [DOI] [PubMed] [Google Scholar]
  • 51.Gardai SJ, Xiao YQ, Dickinson M, Nick JA, Voelker DR, et al. By binding SIRPalpha or calreticulin/CD91, lung collectins act as dual function surveillance molecules to suppress or enhance inflammation. Cell. 2003;115:13–23. doi: 10.1016/s0092-8674(03)00758-x. [DOI] [PubMed] [Google Scholar]
  • 52.Crouch E, Wright JR. Surfactant proteins A and D and pulmonary host defense. Annu Rev Physiol. 2001;63:521–554. doi: 10.1146/annurev.physiol.63.1.521. [DOI] [PubMed] [Google Scholar]
  • 53.Crowther JE, Kutala VK, Kuppusamy P, Ferguson JS, Beharka AA, et al. Pulmonary surfactant protein a inhibits macrophage reactive oxygen intermediate production in response to stimuli by reducing NADPH oxidase activity. J Immunol. 2004;172:6866–6874. doi: 10.4049/jimmunol.172.11.6866. [DOI] [PubMed] [Google Scholar]
  • 54.Williams MC. Alveolar type I cells: molecular phenotype and development. Annu Rev Physiol. 2003;65:669–695. doi: 10.1146/annurev.physiol.65.092101.142446. [DOI] [PubMed] [Google Scholar]
  • 55.Gordon SB, Read RC. Macrophage defences against respiratory tract infections. Br Med Bull. 2002;61:45–61. doi: 10.1093/bmb/61.1.45. [DOI] [PubMed] [Google Scholar]
  • 56.Suzuki T, Chow CW, Downey GP. Role of innate immune cells and their products in lung immunopathology. Int J Biochem Cell Biol. 2008;40:1348–1361. doi: 10.1016/j.biocel.2008.01.003. [DOI] [PubMed] [Google Scholar]
  • 57.Bitterman PB, Saltzman LE, Adelberg S, Ferrans VJ, Crystal RG. Alveolar macrophage replication. One mechanism for the expansion of the mononuclear phagocyte population in the chronically inflamed lung. J Clin Invest. 1984;74:460–469. doi: 10.1172/JCI111443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lambrecht BN. Alveolar macrophage in the driver’s seat. Immunity. 2006;24:366–368. doi: 10.1016/j.immuni.2006.03.008. [DOI] [PubMed] [Google Scholar]
  • 59.Schlesinger LS, Azad AK, Torrelles JB, Roberts E, Vergne I, et al. Determinants of Phagocytosis, Phagosome Biogenesis and Autophagy for Mycobacterium tuberculosis. In: Kaufmann SHE, Britton WJ, editors. Handbook of Tuberculosis. Immunology and Cell Biology. Wiley-VCH Verlag GmbH&Co. KGaA; Weinheim, Germany: 2008. pp. 1–22. [Google Scholar]
  • 60.Rajaram MV, Brooks MN, Morris JD, Torrelles JB, Azad AK, et al. Mycobacterium tuberculosis activates human macrophage peroxisome proliferator-activated receptor gamma linking mannose receptor recognition to regulation of immune responses. J Immunol. 2010;185:929–942. doi: 10.4049/jimmunol.1000866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Standiford TJ, Keshamouni VG, Reddy RC. Peroxisome proliferator-activated receptor-{gamma} as a regulator of lung inflammation and repair. Proc Am Thorac Soc. 2005;2:226–231. doi: 10.1513/pats.200501-010AC. [DOI] [PubMed] [Google Scholar]
  • 62.Hoidal JR, Schmeling D, Peterson PK. Phagocytosis, bacterial killing, and metabolism by purified human lung phagocytes. J Infect Dis. 1981;144:61–71. doi: 10.1093/infdis/144.1.61. [DOI] [PubMed] [Google Scholar]
  • 63.Roth MD, Golub SH. Human pulmonary macrophages utilize prostaglandins and transforming growth factor b1 to suppress lymphocyte activation. J Leukocyte Biol. 1993;53:366–371. doi: 10.1002/jlb.53.4.366. [DOI] [PubMed] [Google Scholar]
  • 64.Lyons CR, Ball EJ, Toews GB, Weissler JC, Stastny P, et al. Inability of human alveolar macrophages to stimulate resting T cells correlates with decreased antigen-specific T cell-macrophage binding. J Immunol. 1986;137:1173–1180. [PubMed] [Google Scholar]
  • 65.Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009;27:451–483. doi: 10.1146/annurev.immunol.021908.132532. [DOI] [PubMed] [Google Scholar]
  • 66.Holt PG. Alveolar macrophages. III. Studies on the mechanisms of inhibition of T-cell proliferation. Immunology. 1979;37:437–445. [PMC free article] [PubMed] [Google Scholar]
  • 67.Lipscomb MF, Lyons CR, Nunez G, Ball EJ, Stastny P, et al. Human alveolar macrophages: HLA-DR-positive macrophages that are poor stimulators of a primary mixed leukocyte reaction. J Immunol. 1986;136:497–504. [PubMed] [Google Scholar]
  • 68.Nguyen BY, Peterson PK, Verbrugh HA, Quie PG, Hoidal JR. Differences in phagocytosis and killing by alveolar macrophages from humans, rabbits, rats, and hamsters. Infect Immun. 1982;36:504–509. doi: 10.1128/iai.36.2.504-509.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wolter NJ, Kunkel SL, Lynch JP, III, Ward PA. Production of cyclooxygenase products by alveolar macrophages in pulmonary sarcoidosis. Chest. 1983;83:79S–81S. doi: 10.1378/chest.83.5_supplement.79s. [DOI] [PubMed] [Google Scholar]
  • 70.Wewers MD, Rennard SI, Hance AJ, Bitterman PB, Crystal RG. Normal human alveolar macrophages obtained by bronchoalveolar lavage have a limited capacity to release interleukin-1. J Clin Invest. 1984;74:2208–2218. doi: 10.1172/JCI111647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Oren R, Farnham AE, Saito K, Milofsky E, Karnovsky ML. Metabolic patterns in three types of phagocytizing cells. J Cell Biol. 1963;17:487–501. doi: 10.1083/jcb.17.3.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Munder M, Eichmann K, Modolell M. Alternative metabolic states in murine macrophages reflected by the nitric oxide synthase/arginase balance: competitive regulation by CD4+ T cells correlates with Th1/Th2 phenotype. J Immunol. 1998;160:5347–5354. [PubMed] [Google Scholar]
  • 73.Suzuki K, Suda T, Naito T, Ide K, Chida K, et al. Impaired toll-like receptor 9 expression in alveolar macrophages with no sensitivity to CpG DNA. Am J Respir Crit Care Med. 2005;171:707–713. doi: 10.1164/rccm.200408-1078OC. [DOI] [PubMed] [Google Scholar]
  • 74.Monick MM, Carter AB, Gudmundsson G, Geist LJ, Hunninghake GW. Changes in PKC isoforms in human alveolar macrophages compared with blood monocytes. Am J Physiol Lung Cell Mol Physiol. 1998;19:L389–L397. doi: 10.1152/ajplung.1998.275.2.L389. [DOI] [PubMed] [Google Scholar]
  • 75.Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998;391:82–86. doi: 10.1038/34184. [DOI] [PubMed] [Google Scholar]
  • 76.Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998;391:79–82. doi: 10.1038/34178. [DOI] [PubMed] [Google Scholar]
  • 77.von Knethen A, Brune B. Activation of peroxisome proliferator-activated receptor gamma by nitric oxide in monocytes/macrophages down-regulates p47phox and attenuates the respiratory burst. J Immunol. 2002;169:2619–2626. doi: 10.4049/jimmunol.169.5.2619. [DOI] [PubMed] [Google Scholar]
  • 78.Beharka AA, Gaynor CD, Kang BK, Voelker DR, McCormack FX, et al. Pulmonary surfactant protein A up-regulates activity of the mannose receptor, a pattern recognition receptor expressed on human macrophages. J Immunol. 2002;169:3565–3573. doi: 10.4049/jimmunol.169.7.3565. [DOI] [PubMed] [Google Scholar]
  • 79.Kuronuma K, Sano H, Kato K, Kudo K, Hyakushima N, et al. Pulmonary surfactant protein A augments the phagocytosis of Streptococcus pneumoniae by alveolar macrophages through a casein kinase 2-dependent increase of cell surface localization of scavenger receptor A. J Biol Chem. 2004;279:21421–21430. doi: 10.1074/jbc.M312490200. [DOI] [PubMed] [Google Scholar]
  • 80.Henning LN, Azad AK, Parsa KV, Crowther JE, Tridandapani S, et al. Pulmonary surfactant protein A regulates TLR expression and activity in human macrophages. J Immunol. 2008;180:7847–7858. doi: 10.4049/jimmunol.180.12.7847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wright JR. Immunoregulatory functions of surfactant proteins. Nat Rev Immunol. 2005;5:58–68. doi: 10.1038/nri1528. [DOI] [PubMed] [Google Scholar]
  • 82.Ferguson JS, Martin JL, Azad AK, McCarthy TR, Kang PB, et al. Surfactant protein D increases fusion of Mycobacterium tuberculosis-containing phagosomes with lysosomes in human macrophages. Infect Immun. 2006;74:7005–7009. doi: 10.1128/IAI.01402-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ferguson JS, Voelker DR, McCormack FX, Schlesinger LS. Surfactant protein D binds to Mycobacterium tuberculosis bacili and lipoarrabinomannan via carbohydratelectin interactions resulting in reduced phagocytosis of the bacteria by macrophages. J Immunol. 1999;163:312–321. [PubMed] [Google Scholar]
  • 84.Kahnert A, Seiler P, Stein M, Bandermann S, Hahnke K, et al. Alternative activation deprives macrophages of a coordinated defense program to Mycobacterium tuberculosis. Eur J Immunol. 2006;36:631–647. doi: 10.1002/eji.200535496. [DOI] [PubMed] [Google Scholar]
  • 85.Schafer G, Jacobs M, Wilkinson RJ, Brown GD. Non-opsonic recognition of Mycobacterium tuberculosis by phagocytes. J Innate Immun. 2009;1:231–243. doi: 10.1159/000173703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 87.Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022. [DOI] [PubMed] [Google Scholar]
  • 88.Wileman TE, Lennartz MR, Stahl PD. Identification of the macrophage mannose receptor as a 175-kDa membrane protein. Proc Natl Acad Sci USA. 1986;83:2501–2505. doi: 10.1073/pnas.83.8.2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.McGreal EP, Miller JL, Gordon S. Ligand recognition by antigen-presenting cell C-type lectin receptors. Curr Opin Immunol. 2005;17:18–24. doi: 10.1016/j.coi.2004.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Stahl PD, Ezekowitz RA. The mannose receptor is a pattern recognition receptor involved in host defense. Curr Opin Immunol. 1998;10:50–55. doi: 10.1016/s0952-7915(98)80031-9. [DOI] [PubMed] [Google Scholar]
  • 91.Stahl PD. The macrophage mannose receptor: current status. Am J Respir Cell Mol Biol. 1990;2:317–318. doi: 10.1165/ajrcmb/2.4.317. [DOI] [PubMed] [Google Scholar]
  • 92.Speert DP, Silverstein SC. Phagocytosis of unopsonized zymosan by human monocyte-derived macrophages: Maturation and inhibition by mannan. J Leukocyte Biol. 1985;38:655–658. doi: 10.1002/jlb.38.5.655. [DOI] [PubMed] [Google Scholar]
  • 93.Stahl PD. The mannose receptor and other macrophage lectins. Curr Opin Immunol. 1992;4:49–52. doi: 10.1016/0952-7915(92)90123-v. [DOI] [PubMed] [Google Scholar]
  • 94.Martinez-Pomares L, Linehan SA, Taylor PR, Gordon S. Binding properties of the mannose receptor. Immunobiology. 2001;204:527–535. doi: 10.1078/0171-2985-00089. [DOI] [PubMed] [Google Scholar]
  • 95.Lee SJ, Evers S, Roeder D, Parlow AF, Risteli J, et al. Mannose receptor-mediated regulation of serum glycoprotein homeostasis. Science. 2002;295:1901. doi: 10.1126/science.1069540. [DOI] [PubMed] [Google Scholar]
  • 96.Medzhihtov R, Janeway C., Jr Innate Immunity. N Engl J Med. 2000;343:338–344. doi: 10.1056/NEJM200008033430506. [DOI] [PubMed] [Google Scholar]
  • 97.Torrelles JB, Schlesinger LS. Diversity in Mycobacterium tuberculosis mannosylated cell wall determinants impacts adaptation to the host. Tuberculosis (Edinb) 2010;90:84–93. doi: 10.1016/j.tube.2010.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Schlesinger LS, Kaufman TM, Iyer S, Hull SR, Marciando LK. Differences in mannose receptor-mediated uptake of lipoarabinomannan from virulent and attenuated strains of Mycobacterium tuberculosis by human macrophages. J Immunol. 1996;157:4568–4575. [PubMed] [Google Scholar]
  • 99.Torrelles JB, Knaup R, Kolareth A, Slepushkina T, Kaufman TM, et al. Identification of Mycobacterium tuberculosis clinical isolates with altered phagocytosis by human macrophages due to a truncated lipoarabinomannan. J Biol Chem. 2008;283:31417–31428. doi: 10.1074/jbc.M806350200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schlesinger LS, Hull SR, Kaufman TM. Binding of the terminal mannosyl units of lipoarabinomannan from a virulent strain of Mycobacterium tuberculosis to human macrophages. J Immunol. 1994;152:4070–4079. [PubMed] [Google Scholar]
  • 101.Torrelles JB, Azad AK, Schlesinger LS. Fine discrimination in the recognition of individual species of phosphatidyl-myo-inositol mannosides from Mycobacterium tuberculosis by C-type lectin pattern recognition receptors. J Immunol. 2006;177:1805–1816. doi: 10.4049/jimmunol.177.3.1805. [DOI] [PubMed] [Google Scholar]
  • 102.Chieppa M, Bianchi G, Doni A, Del Prete A, Sironi M, et al. Cross-linking of the mannose receptor on monocyte-derived dendritic cells activates an anti-inflammatory immunosuppressive program. J Immunol. 2003;171:4552–4560. doi: 10.4049/jimmunol.171.9.4552. [DOI] [PubMed] [Google Scholar]
  • 103.Nigou J, Zelle-Rieser C, Gilleron M, Thurnher M, Puzo G. Mannosylated liparabinomannans inhibit IL-12 production by human dendritic cells: Evidence for a negative signal delivered through the mannose receptor. J Immunol. 2001;166:7477–7485. doi: 10.4049/jimmunol.166.12.7477. [DOI] [PubMed] [Google Scholar]
  • 104.Astarie-Dequeker C, N’Diaye EN, Le Cabec V, Rittig MG, Prandi J, et al. The mannose receptor mediates uptake of pathogenic and nonpathogenic mycobacteria and bypasses bactericidal responses in human macrophages. Infect Immun. 1999;67:469–477. doi: 10.1128/iai.67.2.469-477.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Aderem A, Underhill DM. Mechanisms of phagocytosis in macrophages. Annu Rev Immunol. 1999;17:593–623. doi: 10.1146/annurev.immunol.17.1.593. [DOI] [PubMed] [Google Scholar]
  • 106.Kang BK, Azad AK, Torrelles JB, Kaufman TM, Beharka AA, et al. The human macrophage mannose receptor directs Mycobacterium tuberculosis lipoarabinomannan-mediated phagosome biogenesis. J Exp Med. 2005;202:987–999. doi: 10.1084/jem.20051239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Singh CR, Moulton RA, Armitige LY, Bidani A, Snuggs M, et al. Processing and presentation of a mycobacterial antigen 85B epitope by murine macrophages is dependent on the phagosomal acquisition of vacuolar proton ATPase and in situ activation of cathepsin D. J Immunol. 2006;177:3250–3259. doi: 10.4049/jimmunol.177.5.3250. [DOI] [PubMed] [Google Scholar]
  • 108.Sturgill-Koszycki S, Schlesinger PH, Chakraborty P, Haddix PL, Collins HL, et al. Lack of acidification in Mycobacterium phagosomes produced by exclusion of the vesicular proton-ATPase. Science. 1994;263:678–681. doi: 10.1126/science.8303277. [DOI] [PubMed] [Google Scholar]
  • 109.Prigozy TI, Sieling PA, Clemens D, Stewart PL, Behar SM, et al. The mannose receptor delivers lipoglycan antigens to endosomes for presentation to T cells by CD1b molecules. Immunity. 1997;6:187–197. doi: 10.1016/s1074-7613(00)80425-2. [DOI] [PubMed] [Google Scholar]
  • 110.Van D, V, Marijnissen RJ, Kullberg BJ, Koenen HJ, Cheng SC, et al. The macrophage mannose receptor induces IL-17 in response to Candida albicans. Cell Host Microbe. 2009;5:329–340. doi: 10.1016/j.chom.2009.02.006. [DOI] [PubMed] [Google Scholar]
  • 111.Martinez-Pomares L, Kosco-Vilbois M, Darley E, Tree P, Herren S, et al. Fc chimeric protein containing the cysteine-rich domain of the murine mannose receptor binds to macrophages from splenic marginal zone and lymph node subcapsular sinus and to germinal centers. J Exp Med. 1996;184:1927–1937. doi: 10.1084/jem.184.5.1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Martinez-Pomares L, Mahoney JA, Kaposzta R, Linehan SA, Stahl PD, et al. A functional soluble form of the murine mannose receptor is produced by macrophages in vitro and is present in mouse serum. J Biol Chem. 1998;273:23376–23380. doi: 10.1074/jbc.273.36.23376. [DOI] [PubMed] [Google Scholar]
  • 113.Linehan SA, Martiniz-Pomares L, Stahl PD, Gordon S. Mannose receptor and its putative ligands in normal murine lymphoid and nonlymphoid organs: In situ expression of mannose receptor by selected macrophages, endothelial cells, perivascular microglia, and mesangial cells, but not dendritic cells. J Exp Med. 1999;189:1961–1972. doi: 10.1084/jem.189.12.1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Engering AJ, Cella M, Fluitsma DM, Hoefsmit EC, Lanzavecchia A, et al. Mannose receptor mediated antigen uptake and presentation in human dendritic cells. Adv Exp Med Biol. 1997;417:183–187. doi: 10.1007/978-1-4757-9966-8_31. [DOI] [PubMed] [Google Scholar]
  • 115.Tan MC, Mommaas AM, Drijfhout JW, Jordens R, Onderwater JJ, et al. Mannose receptor mediated uptake of antigens strongly enhances HLA-class II restricted antigen presentation by cultured dendritic cells. Adv Exp Med Biol. 1997;417:171–174. doi: 10.1007/978-1-4757-9966-8_28. [DOI] [PubMed] [Google Scholar]
  • 116.Berney C, Herren S, Power CA, Gordon S, Martinez-Pomares L, et al. A member of the dendritic cell family that enters B cell follicles and stimulates primary antibody responses identified by a mannose receptor fusion protein. J Exp Med. 1999;190:851–860. doi: 10.1084/jem.190.6.851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.McNally AK, DeFife KM, Anderson JM. Interleukin-4-induced macrophage fusion is prevented by inhibitors of mannose receptor activity. Am J Pathol. 1996;149:975–985. [PMC free article] [PubMed] [Google Scholar]
  • 118.Mitchell DA, Fadden AJ, Drickamer K. A novel mechanism of carbohydrate recognition by the C-type lectins DC-SIGN and DC-SIGNR. Subunit organization and binding to multivalent ligands. J Biol Chem. 2001;276:28939–28945. doi: 10.1074/jbc.M104565200. [DOI] [PubMed] [Google Scholar]
  • 119.Feinberg H, Mitchell DA, Drickamer K, Weis WI. Structural basis for selective recognition of oligosaccharides by DC-SIGN and DC-SIGNR. Science. 2001;294:2163–2166. doi: 10.1126/science.1066371. [DOI] [PubMed] [Google Scholar]
  • 120.Figdor CG, Van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002;2:77–84. doi: 10.1038/nri723. [DOI] [PubMed] [Google Scholar]
  • 121.Geijtenbeek TB, Torensma R, Van Vliet SJ, van Duijnhoven GC, Adema GJ, et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell. 2000;100:575–585. doi: 10.1016/s0092-8674(00)80693-5. [DOI] [PubMed] [Google Scholar]
  • 122.Bleijs DA, Geijtenbeek TB, Figdor CG, van KY. DC-SIGN and LFA-1: a battle for ligand. Trends Immunol. 2001;22:457–463. doi: 10.1016/s1471-4906(01)01974-3. [DOI] [PubMed] [Google Scholar]
  • 123.van KY, Geijtenbeek TB. DC-SIGN: escape mechanism for pathogens. Nat Rev Immunol. 2003;3:697–709. doi: 10.1038/nri1182. [DOI] [PubMed] [Google Scholar]
  • 124.Tailleux L, Pham-Thi N, Bergeron-Lafaurie A, Herrmann JL, Charles P, et al. DC-SIGN induction in alveolar macrophages defines privileged target host cells for mycobacteria in patients with tuberculosis. PLoS Med. 2005;2:e381. doi: 10.1371/journal.pmed.0020381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Puig-Kroger A, Serrano-Gomez D, Caparros E, Dominguez-Soto A, Relloso M, et al. 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. 2004;279:25680–25688. doi: 10.1074/jbc.M311516200. [DOI] [PubMed] [Google Scholar]
  • 126.Geijtenbeek TB, Van Vliet SJ, Koppel EA, Sanchez-Hernandez M, Vandenbroucke-Grauls CM, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med. 2003;197:7–17. doi: 10.1084/jem.20021229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Bodnar KA, Serbina NV, Flynn JL. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect Immun. 2001;69:800–809. doi: 10.1128/IAI.69.2.800-809.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Tailleux L, Schwartz O, Herrmann JL, Pivert E, Jackson M, et al. DC-SIGN is the major Mycobacterium tuberculosis receptor on human dendritic cells. J Exp Med. 2003;197:121–127. doi: 10.1084/jem.20021468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Engering A, Geijtenbeek TB, Van Vliet SJ, Wijers M, van Liempt E, et al. The dendritic cell-specific adhesion receptor DC-SIGN internalizes antigen for presentation to T cells. J Immunol. 2002;168:2118–2126. doi: 10.4049/jimmunol.168.5.2118. [DOI] [PubMed] [Google Scholar]
  • 130.Taylor PR, Brown GD, Reid DM, Willment JA, Martinez-Pomares L, et al. The beta-glucan receptor, dectin-1, is predominantly expressed on the surface of cells of the monocyte/macrophage and neutrophil lineages. J Immunol. 2002;169:3876–3882. doi: 10.4049/jimmunol.169.7.3876. [DOI] [PubMed] [Google Scholar]
  • 131.Abbas AK, Lichtman AH. Cellular and Molecular Immunology. 5 2005. [Google Scholar]
  • 132.Lee MS, Kim YJ. Signaling pathways downstream of pattern-recognition receptors and their cross talk. Annu Rev Biochem. 2007;76:447–480. doi: 10.1146/annurev.biochem.76.060605.122847. [DOI] [PubMed] [Google Scholar]
  • 133.Zhang P, Summer WR, Bagby GJ, Nelson S. Innate immunity and pulmonary host defense. Immunol Rev. 2000;173:39–51. doi: 10.1034/j.1600-065x.2000.917306.x. [DOI] [PubMed] [Google Scholar]
  • 134.Yadav M, Schorey JS. The {beta}-glucan receptor Dectin-1 fucntions together with TLR2 to mediate macrophage activation by mycobacteria. Blood. 2006;108:3168–3175. doi: 10.1182/blood-2006-05-024406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Rothfuchs AG, Bafica A, Feng CG, Egen JG, Williams DL, et al. Dectin-1 Interaction with Mycobacterium tuberculosis Leads to Enhanced IL-12p40 Production by Splenic Dendritic Cells. J Immunol. 2007;179:3463–3471. doi: 10.4049/jimmunol.179.6.3463. [DOI] [PubMed] [Google Scholar]
  • 136.Van D, V, Teirlinck AC, Kleinnijenhuis J, Kullberg BJ, van CR, et al. Mycobacterium tuberculosis induces IL-17A responses through TLR4 and dectin-1 and is critically dependent on endogenous IL-1. J Leukoc Biol. 2010;88:227–232. doi: 10.1189/jlb.0809550. [DOI] [PubMed] [Google Scholar]
  • 137.Zenaro E, Donini M, Dusi S. Induction of Th1/Th17 immune response by Mycobacterium tuberculosis: role of dectin-1, Mannose Receptor, and DC-SIGN. J Leukoc Biol. 2009;86:1393–1401. doi: 10.1189/jlb.0409242. [DOI] [PubMed] [Google Scholar]
  • 138.Yamasaki S, Ishikawa E, Sakuma M, Hara H, Ogata K, et al. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008;9:1179–1188. doi: 10.1038/ni.1651. [DOI] [PubMed] [Google Scholar]
  • 139.Yamasaki S, Matsumoto M, Takeuchi O, Matsuzawa T, Ishikawa E, et al. C-type lectin Mincle is an activating receptor for pathogenic fungus, Malassezia. Proc Natl Acad Sci U S A. 2009;106:1897–1902. doi: 10.1073/pnas.0805177106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ishikawa E, Ishikawa T, Morita YS, Toyonaga K, Yamada H, et al. Direct recognition of the mycobacterial glycolipid, trehalose dimycolate, by C-type lectin Mincle. J Exp Med. 2009;206:2879–2888. doi: 10.1084/jem.20091750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Heitmann L, Schoenen H, Ehlers S, Lang R, Holscher C. Mincle is not essential for controlling Mycobacterium tuberculosis infection. Immunobiology. 2012 doi: 10.1016/j.imbio.2012.06.005. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 142.Myones BL, Dalzell JG, Hogg N, Ross GD. Neutrophil and monocyte cell surface p150,955 has iC3b-receptor (CR4) activity resembling CR3. J Clin Invest. 1988;82:640–651. doi: 10.1172/JCI113643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Arnaout MA. Structure and function of the leukocyte adhesion molecules CD11/CD18. Blood. 1990;75:1037–1050. [PubMed] [Google Scholar]
  • 144.Schlesinger LS, Bellinger-Kawahara CG, Payne NR, Horwitz MA. Phagocytosis of Mycobacterium tuberculosis is mediated by human monocyte complement receptors and complement component C3. J Immunol. 1990;144:2771–2780. [PubMed] [Google Scholar]
  • 145.Schlesinger LS. Macrophage phagocytosis of virulent but not attenuated strains of Mycobacterium tuberculosis is mediated by mannose receptors in addition to complement receptors. J Immunol. 1993;150:2920–2930. [PubMed] [Google Scholar]
  • 146.Ferguson JS, Weis JJ, Martin JL, Schlesinger LS. Complement protein C3 binding to Mycobacterium tuberculosis is initiated by the classical pathway in human bronchoalveolar lavage fluid. Infect Immun. 2004;72:2564–2573. doi: 10.1128/IAI.72.5.2564-2573.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Cywes C, Hoppe HC, Daffe M, Ehlers MRW. Nonopsonic binding of Mycobacterium tuberculosis to complement receptor type 3 is mediated by capsular polysaccharides and is strain dependent. Infect Immun. 1997;65:4258–4266. doi: 10.1128/iai.65.10.4258-4266.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Villeneuve C, Gilleron M, Maridonneau-Parini I, Daffe M, Astarie-Dequeker C, et al. Mycobacteria use their surface-exposed glycolipids to infect human macrophages through a receptor-dependent process. J Lipid Res. 2005;46:475–483. doi: 10.1194/jlr.M400308-JLR200. [DOI] [PubMed] [Google Scholar]
  • 149.Melo MD, Catchpole IR, Haggar G, Stokes RW. Utilization of CD11b knockout mice to characterize the role of complement receptor 3 (CR3, CD11b/CD18) in the growth of Mycobacterium tuberculosis in macrophages. Cell Immunol. 2000;205:13–23. doi: 10.1006/cimm.2000.1710. [DOI] [PubMed] [Google Scholar]
  • 150.Hu C, Mayadas-Norton T, Tanaka K, Chan J, Salgame P. Mycobacterium tuberculosis infection in complement receptor 3-deficient mice. J Immunol. 2000;165:2596–2602. doi: 10.4049/jimmunol.165.5.2596. [DOI] [PubMed] [Google Scholar]
  • 151.Armstrong JA, Hart PD. Phagosome-lysosome interactions in cultured macrophages infected with virulent tubercle bacilli: Reversal of the usual nonfusion pattern and observations on bacterial survial. J Exp Med. 1975;142:1–16. doi: 10.1084/jem.142.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Basu S, Fenton MJ. Toll-like receptors: function and roles in lung disease. Am J Physiol Lung Cell Mol Physiol. 2004;286:L887–L892. doi: 10.1152/ajplung.00323.2003. [DOI] [PubMed] [Google Scholar]
  • 153.Krutzik SR, Modlin RL. The role of Toll-like receptors in combating mycobacteria. Semin Immunol. 2004;16:35–41. doi: 10.1016/j.smim.2003.10.005. [DOI] [PubMed] [Google Scholar]
  • 154.Hayashi F, Means TK, Luster AD. Toll-like receptors stimulate human neutrophil function. Blood. 2003;102:2660–2669. doi: 10.1182/blood-2003-04-1078. [DOI] [PubMed] [Google Scholar]
  • 155.Dasari P, Nicholson IC, Hodge G, Dandie GW, Zola H. Expression of toll-like receptors on B lymphocytes. Cell Immunol. 2005;236:140–145. doi: 10.1016/j.cellimm.2005.08.020. [DOI] [PubMed] [Google Scholar]
  • 156.Kodowaki N, Ho S, Antonenko S, de Waal Malfyt R, Kastelein RA, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869. doi: 10.1084/jem.194.6.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Armstrong L, Medford AR, Uppington KM, Robertson J, Witherden IR, et al. Expression of functional toll-like receptor-2 and −4 on alveolar epithelial cells. Am J Respir Cell Mol Biol. 2004;31:241–245. doi: 10.1165/rcmb.2004-0078OC. [DOI] [PubMed] [Google Scholar]
  • 158.Medzhitov R, Preston-Hurlburt P, Janeway CA., Jr A human homologue of the Drosophia Toll protein signals activation of adaptive immunity. Nature. 1997;388:394–397. doi: 10.1038/41131. [DOI] [PubMed] [Google Scholar]
  • 159.Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF. A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci U S A. 1998;95:588–593. doi: 10.1073/pnas.95.2.588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Brightbill HD, Libraty DH, Krutzik SR, Yang R-B, Belisle JT, et al. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science. 1999;285:732–736. doi: 10.1126/science.285.5428.732. [DOI] [PubMed] [Google Scholar]
  • 161.Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007;7:179–190. doi: 10.1038/nri2038. [DOI] [PubMed] [Google Scholar]
  • 162.Poltorak A, He X, Smirnova I, Liu MY, Van HC, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282:2085–2088. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 163.Malhotra R, Thiel S, Reid KBM, Sim RB. Human leukocyte Clq receptor binds other soluble proteins with collagen domains. J Exp Med. 1990;172:955–959. doi: 10.1084/jem.172.3.955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kobayashi K, Hernandez LD, Galan JE, Janeway CA, Jr, Medzhitov R, et al. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell. 2002;110:191–202. doi: 10.1016/s0092-8674(02)00827-9. [DOI] [PubMed] [Google Scholar]
  • 165.Nguyen HA, Rajaram MV, Meyer DA, Schlesinger LS. Pulmonary surfactant protein A and surfactant lipids upregulate IRAK-M, a negative regulator of TLR-mediated inflammation in human macrophages. Am J Physiol Lung Cell Mol Physiol. 2012;303:L608–L616. doi: 10.1152/ajplung.00067.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Yamamoto M, Takeda K, Akira S. TIR domain-containing adaptors define the specificity of TLR signaling. Mol Immunol. 2004;40:861–868. doi: 10.1016/j.molimm.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 167.Cambi A, Koopman M, Figdor CG. How C-type lectins detect pathogens. Cell Microbiol. 2005;7:481–488. doi: 10.1111/j.1462-5822.2005.00506.x. [DOI] [PubMed] [Google Scholar]
  • 168.Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34:637–650. doi: 10.1016/j.immuni.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 169.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
  • 170.Jo EK. Mycobacterial interaction with innate receptors: TLRs, C-type lectins, and NLRs. Curr Opin Infect Dis. 2008;21:279–286. doi: 10.1097/QCO.0b013e3282f88b5d. [DOI] [PubMed] [Google Scholar]
  • 171.Rosenberg PS, Che A, Chen BE. Multiple hypothesis testing strategies for genetic case-control association studies. Stat Med. 2006;25:3134–3149. doi: 10.1002/sim.2407. [DOI] [PubMed] [Google Scholar]
  • 172.Heldwein KA, Fenton MJ. The role of toll-like receptors in immunity against mycobacterial infection. Microbes Infect. 2002;4:937–944. doi: 10.1016/s1286-4579(02)01611-8. [DOI] [PubMed] [Google Scholar]
  • 173.Quesniaux V, Fremond C, Jacobs M, Parida S, Nicolle D, et al. Toll-like receptor pathways in the immune responses to mycobacteria. Microbes Infect. 2004;6:946–959. doi: 10.1016/j.micinf.2004.04.016. [DOI] [PubMed] [Google Scholar]
  • 174.Jo EK, Yang CS, Choi CH, Harding CV. Intracellular signalling cascades regulating innate immune responses to Mycobacteria: branching out from Toll-like receptors. Cell Microbiol. 2007;9:1087–1098. doi: 10.1111/j.1462-5822.2007.00914.x. [DOI] [PubMed] [Google Scholar]
  • 175.Takeuchi O, Sato S, Horiuchi T, Hoshino K, Takeda K, et al. Cutting Edge: Role of toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol. 2002;169:10–14. doi: 10.4049/jimmunol.169.1.10. [DOI] [PubMed] [Google Scholar]
  • 176.Means TK, Wang S, Lien E, Yoshimura A, Golenbock DT, et al. Human toll-like receptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol. 1999;163:3920–3927. [PubMed] [Google Scholar]
  • 177.Lopez M, Sly LM, Luu Y, Young D, Cooper H, et al. The 19-kDa Mycobacterium tuberculosis protein induces macrophage apoptosis through Toll-like receptor-2. J Immunol. 2003;170:2409–2416. doi: 10.4049/jimmunol.170.5.2409. [DOI] [PubMed] [Google Scholar]
  • 178.Dao DN, Kremer L, Guerardel Y, Molano A, Jacobs WR, Jr, et al. Mycobacterium tuberculosis lipomannan induces apoptosis and interleukin-12 production in macrophages. Infect Immun. 2004;72:2067–2074. doi: 10.1128/IAI.72.4.2067-2074.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Vergne I, Fratti RA, Hill PJ, Chua J, Belisle J, et al. Mycobacterium tuberculosis phagosome maturation arrest: mycobacterial phosphatidylinositol analog phosphatidylinositol mannoside stimulates early endosomal fusion. Mol Biol Cell. 2004;15:751–760. doi: 10.1091/mbc.E03-05-0307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Palecanda A, Kobzik L. Receptors for unopsonized particles: the role of alveolar macrophage scavenger receptors. Curr Mol Med. 2001;1:589–595. doi: 10.2174/1566524013363384. [DOI] [PubMed] [Google Scholar]
  • 181.Krieger M. Molecular flypaper and atherosclerosis: Structure of the macrophage scavenger receptor. Trends Biochem Sci. 1992;17:141–146. doi: 10.1016/0968-0004(92)90322-z. [DOI] [PubMed] [Google Scholar]
  • 182.Postlethwait EM. Scavenger receptors clear the air. J Clin Invest. 2007;117:601–604. doi: 10.1172/JCI31549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Bowdish DM, Sakamoto K, Kim MJ, Kroos M, Mukhopadhyay S, et al. MARCO, TLR2, and CD14 are required for macrophage cytokine responses to mycobacterial trehalose dimycolate and Mycobacterium tuberculosis. PLoS Pathog. 2009;5:e1000474. doi: 10.1371/journal.ppat.1000474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Pugin J, Heumann D, Tomasz A, Kravchenko VV, Akamatsu Y, et al. CD14 is a pattern recognition receptor. Immunity. 1994;1:509–516. doi: 10.1016/1074-7613(94)90093-0. [DOI] [PubMed] [Google Scholar]
  • 185.Dziarski R. Recognition of bacterial peptidoglycan by the innate immune system. Cell Mol Life Sci. 2003;60:1793–1804. doi: 10.1007/s00018-003-3019-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Fujihara M, Muroi M, Tanamoto K, Suzuki T, Azuma H, et al. Molecular mechanisms of macrophage activation and deactivation by lipopolysaccharide: roles of the receptor complex. Pharmacol Ther. 2003;100:171–194. doi: 10.1016/j.pharmthera.2003.08.003. [DOI] [PubMed] [Google Scholar]
  • 187.Flo TH, Ryan L, Kilaas L, Skjak-Braek G, Ingalls RR, et al. Involvement of CD14 and beta2-integrins in activating cells with soluble and particulate lipopolysaccharides and mannuronic acid polymers. Infect Immun. 2000;68:6770–6776. doi: 10.1128/iai.68.12.6770-6776.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Bernardo J, Billingslea AM, Blumenthal RL, Seetoo KF, Simons ER, et al. Differential responses of human mononuclear phagocytes to mycobacterial lipoarabinomannans: role of CD14 and the mannose receptor. Infect Immun. 1998;66:28–35. doi: 10.1128/iai.66.1.28-35.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Peterson PK, Gekker G, Hu S, Sheng WS, Anderson WR, et al. CD14 receptor-mediated uptake of nonopsonized Mycobacterium tuberculosis by human microglia. Infect Immun. 1995;63:1598–1602. doi: 10.1128/iai.63.4.1598-1602.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Shams H, Wizel B, Lakey DL, Samten B, Vankayalapati R, et al. The CD14 receptor does not mediate entry of Mycobacterium tuberculosis into human mononuclear phagocytes. FEMS Immunol Med Microbiol. 2003;36:63–69. doi: 10.1016/S0928-8244(03)00039-7. [DOI] [PubMed] [Google Scholar]
  • 191.Inohara, Chamaillard, McDonald C, Nunez G. NOD-LRR proteins: role in host-microbial interactions and inflammatory disease. Annu Rev Biochem. 2005;74:355–383. doi: 10.1146/annurev.biochem.74.082803.133347. [DOI] [PubMed] [Google Scholar]
  • 192.Ting JP, Lovering RC, Alnemri ES, Bertin J, Boss JM, et al. The NLR gene family: a standard nomenclature. Immunity. 2008;28:285–287. doi: 10.1016/j.immuni.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Ogura Y, Inohara N, Benito A, Chen FF, Yamaoka S, et al. Nod2, a Nod1/Apaf-1 family member that is restricted to monocytes and activates NF-kappaB. J Biol Chem. 2001;276:4812–4818. doi: 10.1074/jbc.M008072200. [DOI] [PubMed] [Google Scholar]
  • 194.Brooks MN, Rajaram MV, Azad AK, Amer AO, Valdivia-Arenas MA, et al. NOD2 controls the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis and M. bovis BCG in human macrophages. Cell Microbiol. 2011;13:402–418. doi: 10.1111/j.1462-5822.2010.01544.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J Biol Chem. 2003;278:5509–5512. doi: 10.1074/jbc.C200673200. [DOI] [PubMed] [Google Scholar]
  • 196.Chen CM, Gong Y, Zhang M, Chen JJ. Reciprocal cross-talk between Nod2 and TAK1 signaling pathways. J Biol Chem. 2004;279:25876–25882. doi: 10.1074/jbc.M400682200. [DOI] [PubMed] [Google Scholar]
  • 197.Hsu LC, Ali SR, McGillivray S, Tseng PH, Mariathasan S, et al. A NOD2-NALP1 complex mediates caspase-1-dependent IL-1beta secretion in response to Bacillus anthracis infection and muramyl dipeptide. Proc Natl Acad Sci U S A. 2008;105:7803–7808. doi: 10.1073/pnas.0802726105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Netea MG, Sutmuller R, Hermann C, Van der Graaf CA, Van der Meer JW, et al. Toll-like receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T cells. J Immunol. 2004;172:3712–3718. doi: 10.4049/jimmunol.172.6.3712. [DOI] [PubMed] [Google Scholar]
  • 199.Netea MG, Ferwerda G, de Jong DJ, Jansen T, Jacobs L, et al. Nucleotide-binding oligomerization domain-2 modulates specific TLR pathways for the induction of cytokine release. J Immunol. 2005;174:6518–6523. doi: 10.4049/jimmunol.174.10.6518. [DOI] [PubMed] [Google Scholar]
  • 200.Uehara A, Sugawara Y, Kurata S, Fujimoto Y, Fukase K, et al. Chemically synthesized pathogen-associated molecular patterns increase the expression of peptidoglycan recognition proteins via toll-like receptors, NOD1 and NOD2 in human oral epithelial cells. Cell Microbiol. 2005;7:675–686. doi: 10.1111/j.1462-5822.2004.00500.x. [DOI] [PubMed] [Google Scholar]
  • 201.van Heel DA, Ghosh S, Hunt KA, Mathew CG, Forbes A, et al. Synergy between TLR9 and NOD2 innate immune responses is lost in genetic Crohn’s disease. Gut. 2005;54:1553–1557. doi: 10.1136/gut.2005.065888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 2004;5:800–808. doi: 10.1038/ni1092. [DOI] [PubMed] [Google Scholar]
  • 203.Watanabe T, Kitani A, Murray PJ, Wakatsuki Y, Fuss IJ, et al. Nucleotide binding oligomerization domain 2 deficiency leads to dysregulated TLR2 signaling and induction of antigen-specific colitis. Immunity. 2006;25:473–485. doi: 10.1016/j.immuni.2006.06.018. [DOI] [PubMed] [Google Scholar]
  • 204.Wolfert MA, Murray TF, Boons GJ, Moore JN. The origin of the synergistic effect of muramyl dipeptide with endotoxin and peptidoglycan. J Biol Chem. 2002;277:39179–39186. doi: 10.1074/jbc.M204885200. [DOI] [PubMed] [Google Scholar]
  • 205.Yang S, Tamai R, Akashi S, Takeuchi O, Akira S, et al. Synergistic effect of muramyldipeptide with lipopolysaccharide or lipoteichoic acid to induce inflammatory cytokines in human monocytic cells in culture. Infect Immun. 2001;69:2045–2053. doi: 10.1128/IAI.69.4.2045-2053.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Girardin SE, Tournebize R, Mavris M, Page AL, Li X, et al. CARD4/Nod1 mediates NF-kappaB and JNK activation by invasive Shigella flexneri. EMBO Rep. 2001;2:736–742. doi: 10.1093/embo-reports/kve155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Marriott I, Rati DM, McCall SH, Tranguch SL. Induction of Nod1 and Nod2 intracellular pattern recognition receptors in murine osteoblasts following bacterial challenge. Infect Immun. 2005;73:2967–2973. doi: 10.1128/IAI.73.5.2967-2973.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Opitz B, Forster S, Hocke AC, Maass M, Schmeck B, et al. Nod1-mediated endothelial cell activation by Chlamydophila pneumoniae. Circ Res. 2005;96:319–326. doi: 10.1161/01.RES.0000155721.83594.2c. [DOI] [PubMed] [Google Scholar]
  • 209.Travassos LH, Carneiro LA, Girardin SE, Boneca IG, Lemos R, et al. Nod1 participates in the innate immune response to Pseudomonas aeruginosa. J Biol Chem. 2005;280:36714–36718. doi: 10.1074/jbc.M501649200. [DOI] [PubMed] [Google Scholar]
  • 210.Viala J, Chaput C, Boneca IG, Cardona A, Girardin SE, et al. Nod1 responds to peptidoglycan delivered by the Helicobacter pylori cag pathogenicity island. Nat Immunol. 2004;5:1166–1174. doi: 10.1038/ni1131. [DOI] [PubMed] [Google Scholar]
  • 211.Opitz B, Puschel A, Beermann W, Hocke AC, Forster S, et al. Listeria monocytogenes activated p38 MAPK and induced IL-8 secretion in a nucleotide-binding oligomerization domain 1-dependent manner in endothelial cells. J Immunol. 2006;176:484–490. doi: 10.4049/jimmunol.176.1.484. [DOI] [PubMed] [Google Scholar]
  • 212.Opitz B, Puschel A, Schmeck B, Hocke AC, Rosseau S, et al. Nucleotide-binding oligomerization domain proteins are innate immune receptors for internalized Streptococcus pneumoniae. J Biol Chem. 2004;279:36426–36432. doi: 10.1074/jbc.M403861200. [DOI] [PubMed] [Google Scholar]
  • 213.Ferwerda G, Girardin SE, Kullberg BJ, Le Bourhis L, de Jong DJ, et al. NOD2 and toll-like receptors are nonredundant recognition systems of Mycobacterium tuberculosis. PLoS Pathog. 2005;1:279–285. doi: 10.1371/journal.ppat.0010034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Divangahi M, Mostowy S, Coulombe F, Kozak R, Guillot L, et al. NOD2-deficient mice have impaired resistance to Mycobacterium tuberculosis infection through defective innate and adaptive immunity. J Immunol. 2008;181:7157–7165. doi: 10.4049/jimmunol.181.10.7157. [DOI] [PubMed] [Google Scholar]
  • 215.Deshmukh HS, Hamburger JB, Ahn SH, McCafferty DG, Yang SR, et al. Critical role of NOD2 in regulating the immune response to Staphylococcus aureus. Infect Immun. 2009;77:1376–1382. doi: 10.1128/IAI.00940-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Amer A, Franchi L, Kanneganti TD, Body-Malapel M, Ozoren N, et al. Regulation of Legionella phagosome maturation and infection through flagellin and host Ipaf. J Biol Chem. 2006;281:35217–35223. doi: 10.1074/jbc.M604933200. [DOI] [PubMed] [Google Scholar]
  • 217.Gandotra S, Jang S, Murray PJ, Salgame P, Ehrt S. Nucleotide-binding oligomerization domain protein 2-deficient mice control infection with Mycobacterium tuberculosis. Infect Immun. 2007;75:5127–5134. doi: 10.1128/IAI.00458-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Reiner NE. Altered cell signaling and mononuclear phagocyte deactivation during intracellular infection. Immunol Today. 1994;15:374–381. doi: 10.1016/0167-5699(94)90176-7. [DOI] [PubMed] [Google Scholar]
  • 219.Bhatt K, Salgame P. Host innate immune response to Mycobacterium tuberculosis. J Clin Immunol. 2007;27:347–362. doi: 10.1007/s10875-007-9084-0. [DOI] [PubMed] [Google Scholar]
  • 220.Dietrich J, Doherty TM. Interaction of Mycobacterium tuberculosis with the host: consequences for vaccine development. APMIS. 2009;117:440–457. doi: 10.1111/j.1600-0463.2009.02458.x. [DOI] [PubMed] [Google Scholar]
  • 221.Nguyen L, Pieters J. The Trojan horse: survival tactics of pathogenic mycobacteria in macrophages. Trends Cell Biol. 2005;15:269–276. doi: 10.1016/j.tcb.2005.03.009. [DOI] [PubMed] [Google Scholar]
  • 222.Deretic V, Singh S, Master S, Harris J, Roberts E, et al. Mycobacterium tuberculosis inhibition of phagolysosome biogenesis and autophagy as a host defence mechanism. Cell Microbiol. 2006;8:719–727. doi: 10.1111/j.1462-5822.2006.00705.x. [DOI] [PubMed] [Google Scholar]
  • 223.Russell DG. Mycobacterium tuberculosis: here today, and here tomorrow. Nature Reviews. 2001;2:1–9. doi: 10.1038/35085034. [DOI] [PubMed] [Google Scholar]
  • 224.Sturgill-Koszycki S, Schaible UE, Russell DG. Mycobacterium-containing phagosomes are accessible to early endosomes and reflect a transitional state in normal phagosome biogenesis. EMBO J. 1996;15:6960–6968. [PMC free article] [PubMed] [Google Scholar]
  • 225.Clemens DL, Horwitz MA. The Mycobacterium tuberculosis phagosome interacts with early endosomes and is accessible to exogenously administered transferrin. J Exp Med. 1996;184:1349–1355. doi: 10.1084/jem.184.4.1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Russell DG, Dant J, Sturgill-Koszycki S. Mycobacterium avium- and Mycobacterium tuberculosis- containing vacuoles are dynamic, fusion-competent vesicles that are accessible to glycosphingolipids from the host cell plasmalemma. J Immunol. 1996;156:4764–4773. [PubMed] [Google Scholar]
  • 227.Fratti RA, Chua J, Vergne I, Deretic V. Mycobacterium tuberculosis glycosylated phosphatidylinositol causes phagosome maturation arrest. Proc Natl Acad Sci U S A. 2003;100:5437–5442. doi: 10.1073/pnas.0737613100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Chua J, Vergne I, Master S, Deretic V. A tale of two lipids: Mycobacterium tuberculosis phagosome maturation arrest. Curr Opin Microbiol. 2004;7:71–77. doi: 10.1016/j.mib.2003.12.011. [DOI] [PubMed] [Google Scholar]
  • 229.Vergne I, Chua J, Deretic V. Tuberculosis toxin blocking phagosome maturation inhibits a novel Ca2+/calmodulin-PI3K hVPS34 cascade. J Exp Med. 2003;198:653–659. doi: 10.1084/jem.20030527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Handbook of Tuberculosis: Immunology and Cell Biology. 1. Wiley-Blackwell; 2008. [Google Scholar]
  • 231.Malik ZA, Thompson CR, Hashimi S, Porter B, Iyer SS, et al. Cutting Edge: Mycobacterium tuberculosis Blocks Ca(2+) Signaling and Phagosome Maturation in Human Macrophages Via Specific Inhibition of Sphingosine Kinase. J Immunol. 2003;170:2811–2815. doi: 10.4049/jimmunol.170.6.2811. [DOI] [PubMed] [Google Scholar]
  • 232.Fratti RA, Backer JM, Gruenberg J, Corvera S, Deretic V. Role of phosphatidylinositol 3-kinase and Rab5 effectors in phagosomal biogenesis and mycobacterial phagosome maturation arrest. J Cell Biol. 2001;154:631–644. doi: 10.1083/jcb.200106049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Kusner DJ. Mechanisms of mycobacterial persistence in tuberculosis. Clin Immunol. 2005;114:239–247. doi: 10.1016/j.clim.2004.07.016. [DOI] [PubMed] [Google Scholar]
  • 234.Connolly SF, Kusner DJ. The regulation of dendritic cell function by calcium-signaling and its inhibition by microbial pathogens. Immunol Res. 2007;39:115–127. doi: 10.1007/s12026-007-0076-1. [DOI] [PubMed] [Google Scholar]
  • 235.Deretic V, Vergne I, Chua J, Master S, Singh SB, et al. Endosomal membrane traffic: convergence point targeted by Mycobacterium tuberculosis and HIV. Cell Microbiol. 2004;6:999–1009. doi: 10.1111/j.1462-5822.2004.00449.x. [DOI] [PubMed] [Google Scholar]
  • 236.Clemens DL, Lee BY, Horwitz MA. Deviant expression of Rab5 on phagosomes containing the intracellular pathogens Mycobacterium tuberculosis and Legionella pneumophila is associated with altered phagosomal fate. Infect Immun. 2000;68:2671–2684. doi: 10.1128/iai.68.5.2671-2684.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Via LE, Deretic D, Ulmer RJ, Hibler NS, Huber LA, et al. Arrest of mycobacterial phagosome maturation is caused by a block in vesicle fusion between stages controlled by rab5 and rab7. J Biol Chem. 1997;272:13326–13331. doi: 10.1074/jbc.272.20.13326. [DOI] [PubMed] [Google Scholar]
  • 238.Hinchey J, Lee S, Jeon BY, Basaraba RJ, Venkataswamy MM, et al. Enhanced priming of adaptive immunity by a proapoptotic mutant of Mycobacterium tuberculosis. J Clin Invest. 2007;117:2279–2288. doi: 10.1172/JCI31947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Hou JM, D’Lima NG, Rigel NW, Gibbons HS, McCann JR, et al. ATPase activity of Mycobacterium tuberculosis SecA1 and SecA2 proteins and its importance for SecA2 function in macrophages. J Bacteriol. 2008;190:4880–4887. doi: 10.1128/JB.00412-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Tan T, Lee WL, Alexander DC, Grinstein S, Liu J. The ESAT-6/CFP-10 secretion system of Mycobacterium marinum modulates phagosome maturation. Cell Microbiol. 2006;8:1417–1429. doi: 10.1111/j.1462-5822.2006.00721.x. [DOI] [PubMed] [Google Scholar]
  • 241.Walburger A, Koul A, Ferrari G, Nguyen L, Prescianotto-Baschong C, et al. Protein kinase G from pathogenic mycobacteria promotes survival within macrophages. Science. 2004;304:1800–1804. doi: 10.1126/science.1099384. [DOI] [PubMed] [Google Scholar]
  • 242.Chan J, Fan X, Hunter SW, Brennan PJ, Bloom BR. Lipoarabinomannan, a possible virulence factor involved in persistence of Mycobacterium tuberculosis within macrophages. Infect Immun. 1991;59:1755–1761. doi: 10.1128/iai.59.5.1755-1761.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Knutson KL, Hmama Z, Herrera-Velit P, Rochford R, Reiner NE. Lipoarabinomannan of Mycobacterium tuberculosis promotes portein tyrosine dephosphorylation and inhibition of mitogen-activated protein kinase in human mononuclear phagocytes. J Biol Chem. 1997;273:645–652. doi: 10.1074/jbc.273.1.645. [DOI] [PubMed] [Google Scholar]
  • 244.Kan-Sutton C, Jagannath C, Hunter RL., Jr Trehalose 6,6′-dimycolate on the surface of Mycobacterium tuberculosis modulates surface marker expression for antigen presentation and costimulation in murine macrophages. Microbes Infect. 2009;11:40–48. doi: 10.1016/j.micinf.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Lee W, VanderVen BC, Fahey RJ, Russell DG. Intracellular Mycobacterium tuberculosis exploits host-derived fatty acids to limit metabolic stress. J Biol Chem. 2013 doi: 10.1074/jbc.M112.445056. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Daniel J, Maamar H, Deb C, Sirakova TD, Kolattukudy PE. Mycobacterium tuberculosis uses host triacylglycerol to accumulate lipid droplets and acquires a dormancy-like phenotype in lipid-loaded macrophages. PLoS Pathog. 2011;7:e1002093. doi: 10.1371/journal.ppat.1002093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Peyron P, Vaubourgeix J, Poquet Y, Levillain F, Botanch C, et al. Foamy macrophages from tuberculous patients’ granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence. PLoS Pathog. 2008;4:e1000204. doi: 10.1371/journal.ppat.1000204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Welin A, Lerm M. Inside or outside the phagosome? The controversy of the intracellular localization of Mycobacterium tuberculosis. Tuberculosis (Edinb ) 2012;92:113–120. doi: 10.1016/j.tube.2011.09.009. [DOI] [PubMed] [Google Scholar]
  • 249.Leake ES, Myrvik QN, Wright MJ. Phagosomal membranes of Mycobacterium bovis BCG-immune alveolar macrophages are resistant to disruption by Mycobacterium tuberculosis H37Rv. Infect Immun. 1984;45:443–446. doi: 10.1128/iai.45.2.443-446.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.McDonough KA, Kress Y, Bloom BR. Pathogenesis of tuberculosis: Interaction of Mycobacterium tuberculosis with macrophages. Infect Immun. 1993;61:2763–2773. doi: 10.1128/iai.61.7.2763-2773.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Myrvik QN, Leake ES, Wright MJ. Disruption of phagosomal membranes of normal alveolar macrophages by the H37Rv strain of Mycobacterium tuberculosis. A correlate of virulence. Am Rev Respir Dis. 1984;129:322–328. [PubMed] [Google Scholar]
  • 252.Stamm LM, Morisaki JH, Gao LY, Jeng RL, McDonald KL, et al. Mycobacterium marinum escapes from phagosomes and is propelled by actin-based motility. J Exp Med. 2003;198:1361–1368. doi: 10.1084/jem.20031072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Smith J, Manoranjan J, Pan M, Bohsali A, Xu J, et al. Evidence for pore formation in host cell membranes by ESX-1-secreted ESAT-6 and its role in Mycobacterium marinum escape from the vacuole. Infect Immun. 2008;76:5478–5487. doi: 10.1128/IAI.00614-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Hagedorn M, Rohde KH, Russell DG, Soldati T. Infection by tubercular mycobacteria is spread by nonlytic ejection from their amoeba hosts. Science. 2009;323:1729–1733. doi: 10.1126/science.1169381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.van der Wel N, Hava D, Houben D, Fluitsma D, van ZM, et al. M. tuberculosis and M. leprae translocate from the phagolysosome to the cytosol in myeloid cells. Cell. 2007;129:1287–1298. doi: 10.1016/j.cell.2007.05.059. [DOI] [PubMed] [Google Scholar]
  • 256.Houben D, Demangel C, van IJ, Perez J, Baldeon L, et al. ESX-1-mediated translocation to the cytosol controls virulence of mycobacteria. Cell Microbiol. 2012;14:1287–1298. doi: 10.1111/j.1462-5822.2012.01799.x. [DOI] [PubMed] [Google Scholar]
  • 257.Wong KW, Jacobs WR., Jr Critical role for NLRP3 in necrotic death triggered by Mycobacterium tuberculosis. Cell Microbiol. 2011;13:1371–1384. doi: 10.1111/j.1462-5822.2011.01625.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Welin A, Eklund D, Stendahl O, Lerm M. Human macrophages infected with a high burden of ESAT-6-expressing M. tuberculosis undergo caspase-1- and cathepsin B-independent necrosis. PLoS ONE. 2011;6:e20302. doi: 10.1371/journal.pone.0020302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Simeone R, Bobard A, Lippmann J, Bitter W, Majlessi L, et al. Phagosomal rupture by Mycobacterium tuberculosis results in toxicity and host cell death. PLoS Pathog. 2012;8:e1002507. doi: 10.1371/journal.ppat.1002507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Manzanillo PS, Shiloh MU, Portnoy DA, Cox JS. Mycobacterium tuberculosis activates the DNA-dependent cytosolic surveillance pathway within macrophages. Cell Host Microbe. 2012;11:469–480. doi: 10.1016/j.chom.2012.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Watson RO, Manzanillo PS, Cox JS. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell. 2012;150:803–815. doi: 10.1016/j.cell.2012.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Harriff MJ, Purdy GE, Lewinsohn DM. Escape from the Phagosome: The Explanation for MHC-I Processing of Mycobacterial Antigens? Front Immunol. 2012;3:40. doi: 10.3389/fimmu.2012.00040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Weerdenburg EM, Peters PJ, van der Wel NN. How do mycobacteria activate CD8+ T cells? Trends Microbiol. 2010;18:1–10. doi: 10.1016/j.tim.2009.10.004. [DOI] [PubMed] [Google Scholar]
  • 264.Guermonprez P, Saveanu L, Kleijmeer M, Davoust J, van EP, et al. ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature. 2003;425:397–402. doi: 10.1038/nature01911. [DOI] [PubMed] [Google Scholar]
  • 265.Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol. 2001;19:93–129. doi: 10.1146/annurev.immunol.19.1.93. [DOI] [PubMed] [Google Scholar]
  • 266.Cooper AM. Cell-mediated immune responses in tuberculosis. Annu Rev Immunol. 2009;27:393–422. doi: 10.1146/annurev.immunol.021908.132703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Fenhalls G, Stevens L, Bezuidenhout J, Amphlett GE, Duncan K, et al. Distribution of IFN-gamma, IL-4 and TNF-alpha protein and CD8 T cells producing IL-12p40 mRNA in human lung tuberculous granulomas. Immunology. 2002;105:325–335. doi: 10.1046/j.1365-2567.2002.01378.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Herrera MT, Torres M, Nevels D, Perez-Redondo CN, Ellner JJ, et al. Compartmentalized bronchoalveolar IFN-gamma and IL-12 response in human pulmonary tuberculosis. Tuberculosis (Edinb ) 2009;89:38–47. doi: 10.1016/j.tube.2008.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Kellar KL, Gehrke J, Weis SE, Mahmutovic-Mayhew A, Davila B, et al. Multiple cytokines are released when blood from patients with tuberculosis is stimulated with Mycobacterium tuberculosis antigens. PLoS ONE. 2011;6:e26545. doi: 10.1371/journal.pone.0026545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Unsal E, Aksaray S, Koksal D, Sipit T. Potential role of interleukin 6 in reactive thrombocytosis and acute phase response in pulmonary tuberculosis. Postgrad Med J. 2005;81:604–607. doi: 10.1136/pgmj.2004.030544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Guler R, Parihar SP, Spohn G, Johansen P, Brombacher F, et al. Blocking IL-1alpha but not IL-1beta increases susceptibility to chronic Mycobacterium tuberculosis infection in mice. Vaccine. 2011;29:1339–1346. doi: 10.1016/j.vaccine.2010.10.045. [DOI] [PubMed] [Google Scholar]
  • 272.Zhang Y, Rom WN. Regulation of the interleukin-1b (IL-1b) gene by mycobacterial components and lipopolysaccharide is mediated by two nuclear factor-IL6 motifs. Mol Cell Biol. 1993;13:3831–3837. doi: 10.1128/mcb.13.6.3831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Kaufmann SH. How can immunology contribute to the control of tuberculosis? Nature Reviews Immunology. 2001;1:20–30. doi: 10.1038/35095558. [DOI] [PubMed] [Google Scholar]
  • 274.Roach DR, Bean AG, Demangel C, France MP, Briscoe H, et al. TNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infection. J Immunol. 2002;168:4620–4627. doi: 10.4049/jimmunol.168.9.4620. [DOI] [PubMed] [Google Scholar]
  • 275.Ray JC, Flynn JL, Kirschner DE. Synergy between individual TNF-dependent functions determines granuloma performance for controlling Mycobacterium tuberculosis infection. J Immunol. 2009;182:3706–3717. doi: 10.4049/jimmunol.0802297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Pompei L, Jang S, Zamlynny B, Ravikumar S, McBride A, et al. Disparity in IL-12 release in dendritic cells and macrophages in response to Mycobacterium tuberculosis is due to use of distinct TLRs. J Immunol. 2007;178:5192–5199. doi: 10.4049/jimmunol.178.8.5192. [DOI] [PubMed] [Google Scholar]
  • 277.Giacomini E, Iona E, Ferroni L, Miettinen M, Fattorini L, et al. Infection of human macrophages and dendritic cells with Mycobacterium tuberculosis induces a differential cytokine gene expression that modulates T cell response. J Immunol. 2001;166:7033–7041. doi: 10.4049/jimmunol.166.12.7033. [DOI] [PubMed] [Google Scholar]
  • 278.Hickman SP, Chan J, Salgame P. Mycobacterium tuberculosis induces differential cytokine production from dendritic cells and macrophages with divergent effects on naive T cell polarization. J Immunol. 2002;168:4636–4642. doi: 10.4049/jimmunol.168.9.4636. [DOI] [PubMed] [Google Scholar]
  • 279.Cooper AM, Solache A, Khader SA. Interleukin-12 and tuberculosis: an old story revisited. Curr Opin Immunol. 2007;19:441–447. doi: 10.1016/j.coi.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Chen Q, Ghilardi N, Wang H, Baker T, Xie MH, et al. Development of Th1-type immune responses requires the type I cytokine receptor TCCR. Nature. 2000;407:916–920. doi: 10.1038/35038103. [DOI] [PubMed] [Google Scholar]
  • 281.Trinchieri G, Pflanz S, Kastelein RA. The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses. Immunity. 2003;19:641–644. doi: 10.1016/s1074-7613(03)00296-6. [DOI] [PubMed] [Google Scholar]
  • 282.Pearl JE, Khader SA, Solache A, Gilmartin L, Ghilardi N, et al. IL-27 signaling compromises control of bacterial growth in mycobacteria-infected mice. J Immunol. 2004;173:7490–7496. doi: 10.4049/jimmunol.173.12.7490. [DOI] [PubMed] [Google Scholar]
  • 283.Holscher C, Holscher A, Ruckerl D, Yoshimoto T, Yoshida H, et al. The IL-27 receptor chain WSX-1 differentially regulates antibacterial immunity and survival during experimental tuberculosis. J Immunol. 2005;174:3534–3544. doi: 10.4049/jimmunol.174.6.3534. [DOI] [PubMed] [Google Scholar]
  • 284.Robinson CM, Jung JY, Nau GJ. Interferon-gamma, tumor necrosis factor, and interleukin-18 cooperate to control growth of Mycobacterium tuberculosis in human macrophages. Cytokine. 2012;60:233–241. doi: 10.1016/j.cyto.2012.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, et al. IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat Immunol. 2007;8:369–377. doi: 10.1038/ni1449. [DOI] [PubMed] [Google Scholar]
  • 286.Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol. 2009;2:403–411. doi: 10.1038/mi.2009.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Lockhart E, Green AM, Flynn JL. IL-17 production is dominated by gammadelta T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J Immunol. 2006;177:4662–4669. doi: 10.4049/jimmunol.177.7.4662. [DOI] [PubMed] [Google Scholar]
  • 288.Sergejeva S, Ivanov S, Lotvall J, Linden A. Interleukin-17 as a recruitment and survival factor for airway macrophages in allergic airway inflammation. Am J Respir Cell Mol Biol. 2005;33:248–253. doi: 10.1165/rcmb.2004-0213OC. [DOI] [PubMed] [Google Scholar]
  • 289.Kolls JK, Linden A. Interleukin-17 family members and inflammation. Immunity. 2004;21:467–476. doi: 10.1016/j.immuni.2004.08.018. [DOI] [PubMed] [Google Scholar]
  • 290.Khader SA, Cooper AM. IL-23 and IL-17 in tuberculosis. Cytokine. 2008;41:79–83. doi: 10.1016/j.cyto.2007.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Khader SA, Guglani L, Rangel-Moreno J, Gopal R, Junecko BA, et al. IL-23 is required for long-term control of Mycobacterium tuberculosis and B cell follicle formation in the infected lung. J Immunol. 2011;187:5402–5407. doi: 10.4049/jimmunol.1101377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Akira S, Taga T, Kishimoto T. Interleukin-6 in biology and medicine. Adv Immunol. 1993;54:1–78. doi: 10.1016/s0065-2776(08)60532-5. [DOI] [PubMed] [Google Scholar]
  • 293.Kishimoto T, Akira S, Taga T. Interleukin-6 and its receptor: a paradigm for cytokines. Science. 1992;258:593–597. doi: 10.1126/science.1411569. [DOI] [PubMed] [Google Scholar]
  • 294.Chan ED, Morris KR, Belisle JT, Hill P, Remigio LK, et al. Induction of inducible nitric oxide synthase-NO• by lipoarabinomannan of Mycobacterium tuberculosis is mediated by MEK1-ERK, MKK7-JNK, and NF-kB signaling pathways. Infect Immun. 2001;69:2001–2010. doi: 10.1128/IAI.69.4.2001-2010.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–550. doi: 10.1146/annurev.immunol.021908.132612. [DOI] [PubMed] [Google Scholar]
  • 296.Hernandez-Pando R, Orozco-Esteves H, Maldonado HA, guilar-Leon D, Vilchis-Landeros MM, et al. A combination of a transforming growth factor-beta antagonist and an inhibitor of cyclooxygenase is an effective treatment for murine pulmonary tuberculosis. Clin Exp Immunol. 2006;144:264–272. doi: 10.1111/j.1365-2249.2006.03049.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Yang CS, Yuk JM, Shin DM, Kang J, Lee SJ, et al. Secretory phospholipase A2 plays an essential role in microglial inflammatory responses to Mycobacterium tuberculosis. Glia. 2009;57:1091–1103. doi: 10.1002/glia.20832. [DOI] [PubMed] [Google Scholar]
  • 298.Sabat R. IL-10 family of cytokines. Cytokine Growth Factor Rev. 2010;21:315–324. doi: 10.1016/j.cytogfr.2010.11.001. [DOI] [PubMed] [Google Scholar]
  • 299.Sabat R, Grutz G, Warszawska K, Kirsch S, Witte E, et al. Biology of interleukin-10. Cytokine Growth Factor Rev. 2010 doi: 10.1016/j.cytogfr.2010.09.002. [DOI] [PubMed] [Google Scholar]
  • 300.Bogdan C, Vodovotz Y, Nathan C. Macrophage deactivation by interleukin 10. J Exp Med. 1991;174:1549–1555. doi: 10.1084/jem.174.6.1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.de Waal Malefyt R, Abrams J, Bennett B, Figdor CG, De Vries JE. Interleukin 10(IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes. J Exp Med. 1991;174:1209–1220. doi: 10.1084/jem.174.5.1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. IL-10 inhibits cytokine production by activated macrophages. J Immunol. 1991;147:3815–3822. [PubMed] [Google Scholar]
  • 303.D’Andrea A, Aste-Amezaga M, Valiante NM, Ma X, Kubin M, et al. Interleukin 10 (IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells. J Exp Med. 1993;178:1041–1048. doi: 10.1084/jem.178.3.1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Gruber MF, Williams CC, Gerrard TL. Macrophage-colony-stimulating factor expression by anti- CD45 stimulated human monocytes is transcriptionally up- regulated by IL-1b and inhibited by IL-4 and IL-10. J Immunol. 1994;152:1354–1361. [PubMed] [Google Scholar]
  • 305.Aste-Amezaga M, Ma X, Sartori A, Trinchieri G. Molecular mechanisms of the induction of IL-12 and its inhibition by IL-10. J Immunol. 1998;160:5936–5944. [PubMed] [Google Scholar]
  • 306.Cunha FQ, Moncada S, Liew FY. Interleukin-10 (IL-10) inhibits the induction of nitric oxide synthase by interferon-gamma in murine macrophages. Biochem Biophys Res Commun. 1992;182:1155–1159. doi: 10.1016/0006-291x(92)91852-h. [DOI] [PubMed] [Google Scholar]
  • 307.Kuga S, Otsuka T, Niiro H, Nunoi H, Nemoto Y, et al. Suppression of superoxide anion production by interleukin-10 is accompanied by a downregulation of the genes for subunit proteins of NADPH oxidase. Exp Hematol. 1996;24:151–157. [PubMed] [Google Scholar]
  • 308.Moore KW, de Waal MR, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683–765. doi: 10.1146/annurev.immunol.19.1.683. [DOI] [PubMed] [Google Scholar]
  • 309.O’Leary S, O’Sullivan MP, Keane J. IL-10 Blocks Phagosome Maturation in Mycobacterium tuberculosis-infected Human Macrophages. Am J Respir Cell Mol Biol. 2010 doi: 10.1165/rcmb.2010-0319OC. [DOI] [PubMed] [Google Scholar]
  • 310.Flynn JL, Chan J. What’s good for the host is good for the bug. Trends Microbiol. 2005;13:98–102. doi: 10.1016/j.tim.2005.01.005. [DOI] [PubMed] [Google Scholar]
  • 311.Mendez-Samperio P. Expression and regulation of chemokines in mycobacterial infection. J Infect. 2008;57:374–384. doi: 10.1016/j.jinf.2008.08.010. [DOI] [PubMed] [Google Scholar]
  • 312.Saunders BM, Britton WJ. Life and death in the granuloma: immunopathology of tuberculosis. Immunol Cell Biol. 2007;85:103–111. doi: 10.1038/sj.icb.7100027. [DOI] [PubMed] [Google Scholar]
  • 313.Algood HM, Chan J, Flynn JL. Chemokines and tuberculosis. Cytokine Growth Factor Rev. 2003;14:467–477. doi: 10.1016/s1359-6101(03)00054-6. [DOI] [PubMed] [Google Scholar]
  • 314.Sadek MI, Sada E, Toossi Z, Schwander SK, Rich EA. Chemokines induced by infection of mononuclear phagocytes with mycobacteria and present in lung alveoli during active pulmonary tuberculosis. Am J Respir Cell Mol Biol. 1998;19:513–521. doi: 10.1165/ajrcmb.19.3.2815. [DOI] [PubMed] [Google Scholar]
  • 315.Saukkonen JJ, Bazydlo B, Thomas M, Strieter RM, Keane J, et al. Beta-chemokines are induced by Mycobacterium tuberculosis and inhibit its growth. Infect Immun. 2002;70:1684–1693. doi: 10.1128/IAI.70.4.1684-1693.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Taub DD, Turcovski-Corrales SM, Key ML, Longo DL, Murphy WJ. Chemokines and T lymphocyte activation: I. Beta chemokines costimulate human T lymphocyte activation in vitro. J Immunol. 1996;156:2095–2103. [PubMed] [Google Scholar]
  • 317.Fahey TJ, III, Tracey KJ, Tekamp-Olson P, Cousens LS, Jones WG, et al. Macrophage inflammatory protein 1 modulates macrophage function. J Immunol. 1992;148:2764–2769. [PubMed] [Google Scholar]
  • 318.Karpus WJ, Kennedy KJ. MIP-1alpha and MCP-1 differentially regulate acute and relapsing autoimmune encephalomyelitis as well as Th1/Th2 lymphocyte differentiation. J Leukoc Biol. 1997;62:681–687. [PubMed] [Google Scholar]
  • 319.Kurashima K, Mukaida N, Fujimura M, Yasui M, Nakazumi Y, et al. Elevated chemokine levels in bronchoalveolar lavage fluid of tuberculosis patients. Am J Respir Crit Care Med. 1997;155:1474–1477. doi: 10.1164/ajrccm.155.4.9105097. [DOI] [PubMed] [Google Scholar]
  • 320.Chensue SW, Warmington KS, Allenspach EJ, Lu B, Gerard C, et al. Differential expression and cross-regulatory function of RANTES during mycobacterial (type 1) and schistosomal (type 2) antigen-elicited granulomatous inflammation. J Immunol. 1999;163:165–173. [PubMed] [Google Scholar]
  • 321.Legler DF, Loetscher M, Roos RS, Clark-Lewis I, Baggiolini M, et al. B cell-attracting chemokine 1, a human CXC chemokine expressed in lymphoid tissues, selectively attracts B lymphocytes via BLR1/CXCR5. J Exp Med. 1998;187:655–660. doi: 10.1084/jem.187.4.655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Jones BW, Heldwein KA, Means TK, Saukkonen JJ, Fenton MJ. Differential roles of Toll-like receptors in the elicitation of proinflammatory responses by macrophages. Ann Rheum Dis. 2001;60(Suppl 3):iii6–12. doi: 10.1136/ard.60.90003.iii6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Martin CJ, Booty MG, Rosebrock TR, Nunes-Alves C, Desjardins DM, et al. Efferocytosis is an innate antibacterial mechanism. Cell Host Microbe. 2012;12:289–300. doi: 10.1016/j.chom.2012.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Harris J, Keane J. How tumour necrosis factor blockers interfere with tuberculosis immunity. Clin Exp Immunol. 2010;161:1–9. doi: 10.1111/j.1365-2249.2010.04146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Persson YA, Blomgran-Julinder R, Rahman S, Zheng L, Stendahl O. Mycobacterium tuberculosis-induced apoptotic neutrophils trigger a pro-inflammatory response in macrophages through release of heat shock protein 72, acting in synergy with the bacteria. Microbes Infect. 2008;10:233–240. doi: 10.1016/j.micinf.2007.11.007. [DOI] [PubMed] [Google Scholar]
  • 326.Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition. Trends Biochem Sci. 2007;32:37–43. doi: 10.1016/j.tibs.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • 327.Repasy T, Lee J, Marino S, Martinez N, Kirschner DE, et al. Intracellular Bacillary Burden Reflects a Burst Size for Mycobacterium tuberculosis In Vivo. PLoS Pathog. 2013;9:e1003190. doi: 10.1371/journal.ppat.1003190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Koo IC, Wang C, Raghavan S, Morisaki JH, Cox JS, et al. ESX-1-dependent cytolysis in lysosome secretion and inflammasome activation during mycobacterial infection. Cell Microbiol. 2008;10:1866–1878. doi: 10.1111/j.1462-5822.2008.01177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Carlsson F, Kim J, Dumitru C, Barck KH, Carano RA, et al. Host-detrimental role of Esx-1-mediated inflammasome activation in mycobacterial infection. PLoS Pathog. 2010;6:e1000895. doi: 10.1371/journal.ppat.1000895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Mishra BB, Moura-Alves P, Sonawane A, Hacohen N, Griffiths G, et al. Mycobacterium tuberculosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome. Cell Microbiol. 2010;12:1046–1063. doi: 10.1111/j.1462-5822.2010.01450.x. [DOI] [PubMed] [Google Scholar]
  • 331.Elkington PT, Friedland JS. Matrix metalloproteinases in destructive pulmonary pathology. Thorax. 2006;61:259–266. doi: 10.1136/thx.2005.051979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Elkington PT, Ugarte-Gil CA, Friedland JS. Matrix metalloproteinases in tuberculosis. Eur Respir J. 2011;38:456–464. doi: 10.1183/09031936.00015411. [DOI] [PubMed] [Google Scholar]
  • 333.Elkington PT, D’Armiento JM, Friedland JS. Tuberculosis immunopathology: the neglected role of extracellular matrix destruction. Sci Transl Med. 2011;3:71ps6. doi: 10.1126/scitranslmed.3001847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Davidson JM. Biochemistry and turnover of lung interstitium. Eur Respir J. 1990;3:1048–1063. [PubMed] [Google Scholar]
  • 335.Chang JC, Wysocki A, Tchou-Wong KM, Moskowitz N, Zhang YH, et al. Effect of Mycobacterium tuberculosis and its components on macrophages and the release of matrix metalloproteinases. Thorax. 1996;51:306–311. doi: 10.1136/thx.51.3.306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Rivera-Marrero CA, Schuyler W, Roser S, Ritzenthaler JD, Newburn SA, et al. M. tuberculosis induction of matrix metalloproteinase-9: the role of mannose and receptor-mediated mechanisms. Am J Physiol Lung Cell Mol Physiol. 2002;282:L546–L555. doi: 10.1152/ajplung.00175.2001. [DOI] [PubMed] [Google Scholar]
  • 337.Elass E, Aubry L, Masson M, Denys A, Guerardel Y, et al. Mycobacterial lipomannan induces matrix metalloproteinase-9 expression in human macrophagic cells through a Toll-like receptor 1 (TLR1)/ Infect Immun. 2005;73:7064–7068. doi: 10.1128/IAI.73.10.7064-7068.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Elkington PT, Nuttall RK, Boyle JJ, O’Kane CM, Horncastle DE, et al. Mycobacterium tuberculosis, but not vaccine BCG, specifically upregulates matrix metalloproteinase-1. Am J Respir Crit Care Med. 2005;172:1596–1604. doi: 10.1164/rccm.200505-753OC. [DOI] [PubMed] [Google Scholar]
  • 339.Subbian S, Tsenova L, O’Brien P, Yang G, Koo MS, et al. Phosphodiesterase-4 inhibition combined with isoniazid treatment of rabbits with pulmonary tuberculosis reduces macrophage activation and lung pathology. Am J Pathol. 2011;179:289–301. doi: 10.1016/j.ajpath.2011.03.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Elkington P, Shiomi T, Breen R, Nuttall RK, Ugarte-Gil CA, et al. MMP-1 drives immunopathology in human tuberculosis and transgenic mice. J Clin Invest. 2011;121:1827–1833. doi: 10.1172/JCI45666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Sundararajan S, Babu S, Das SD. Comparison of localized versus systemic levels of Matrix metalloproteinases (MMPs), its tissue inhibitors (TIMPs) and cytokines in tuberculous and non-tuberculous pleuritis patients. Hum Immunol. 2012;73:985–991. doi: 10.1016/j.humimm.2012.07.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Elkington PT, Green JA, Emerson JE, Lopez-Pascua LD, Boyle JJ, et al. Synergistic up-regulation of epithelial cell matrix metalloproteinase-9 secretion in tuberculosis. Am J Respir Cell Mol Biol. 2007;37:431–437. doi: 10.1165/rcmb.2007-0011OC. [DOI] [PubMed] [Google Scholar]
  • 343.Lopez B, Aguilar D, Orozco H, Burger M, Espitia C, et al. A marked difference in pathogenesis and immune response induced by different Mycobacterium tuberculosis genotypes. Clin Exp Immunol. 2003;133:30–37. doi: 10.1046/j.1365-2249.2003.02171.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Manca C, Tsenova L, Bergtold A, Freeman S, Tovey M, et al. Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-a/B. Proc Natl Acad Sci USA. 2001;98:5752–5757. doi: 10.1073/pnas.091096998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Manabe YC, Dannenberg AM, Jr, Tyagi SK, Hatem CL, Yoder M, et al. Different strains of Mycobacterium tuberculosis cause various spectrums of disease in the rabbit model of tuberculosis. Infect Immun. 2003;71:6004–6011. doi: 10.1128/IAI.71.10.6004-6011.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Manca C, Reed MB, Freeman S, Mathema B, Kreiswirth B, et al. Differential monocyte activation underlies strain-specific Mycobacterium tuberculosis pathogenesis. Infect Immun. 2004;72:5511–5514. doi: 10.1128/IAI.72.9.5511-5514.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Gagneux S, Small PM. Global phylogeography of Mycobacterium tuberculosis and implications for tuberculosis product development. Lancet Infect Dis. 2007;7:328–337. doi: 10.1016/S1473-3099(07)70108-1. [DOI] [PubMed] [Google Scholar]
  • 348.Mathema B, Kurepina NE, Bifani PJ, Kreiswirth BN. Molecular epidemiology of tuberculosis: current insights. Clin Microbiol Rev. 2006;19:658–685. doi: 10.1128/CMR.00061-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Comas I, Chakravartti J, Small PM, Galagan J, Niemann S, et al. Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconserved. Nat Genet. 2010;42:498–503. doi: 10.1038/ng.590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Niemann S, Koser CU, Gagneux S, Plinke C, Homolka S, et al. Genomic diversity among drug sensitive and multidrug resistant isolates of Mycobacterium tuberculosis with identical DNA fingerprints. PLoS ONE. 2009;4:e7407. doi: 10.1371/journal.pone.0007407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Homolka S, Niemann S, Russell DG, Rohde KH. Functional genetic diversity among Mycobacterium tuberculosis complex clinical isolates: delineation of conserved core and lineage-specific transcriptomes during intracellular survival. PLoS Pathog. 2010;6:e1000988. doi: 10.1371/journal.ppat.1000988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Achtman M, Zurth K, Morelli G, Torrea G, Guiyoule A, et al. Yersinia pestis, the cause of plague, is a recently emerged clone of Yersinia pseudotuberculosis. Proc Natl Acad Sci U S A. 1999;96:14043–14048. doi: 10.1073/pnas.96.24.14043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Hirsh AE, Tsolaki AG, Deriemer K, Feldman MW, Small PM. Stable association between strains of Mycobacterium tuberculosis and their human host populations. Proc Natl Acad Sci U S A. 2004;101:4871–4876. doi: 10.1073/pnas.0305627101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Levin BR, Lipsitch M, Bonhoeffer S. Population biology, evolution, and infectious disease: convergence and synthesis. Science. 1999;283:806–809. doi: 10.1126/science.283.5403.806. [DOI] [PubMed] [Google Scholar]
  • 355.Musser JM. Molecular population genetic analysis of emerged bacterial pathogens: selected insights. Emerg Infect Dis. 1996;2:1–17. doi: 10.3201/eid0201.960101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Friedman CR, Quinn GC, Kreiswirth BN, Perlman DC, Salomon N, et al. Widespread dissemination of a drug-susceptible strain of Mycobacterium tuberculosis. J Infect Dis. 1997;176:478–484. doi: 10.1086/514067. [DOI] [PubMed] [Google Scholar]
  • 357.Soto CY, Menendez MC, Perez E, Samper S, Gomez AB, et al. IS6110 mediates increased transcription of the phoP virulence gene in a multidrug-resistant clinical isolate responsible for tuberculosis outbreaks. J Clin Microbiol. 2004;42:212–219. doi: 10.1128/JCM.42.1.212-219.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Valway SE, Sanchez MPC, Shinnick TF, Orme I, Agerton T, et al. An outbreak involving extensive transmission of a virulent strain of Mycobacerium tuberculosis. N Engl J Med. 1998;338:633–639. doi: 10.1056/NEJM199803053381001. [DOI] [PubMed] [Google Scholar]
  • 359.Zhang M, Gong J, Yang Z, Samten B, Cave MD, et al. Enhanced capacity of a widespread strain of Mycobacterium tuberculosis to grow in human macrophages. J Infect Dis. 1999;179:1213–1217. doi: 10.1086/314738. [DOI] [PubMed] [Google Scholar]
  • 360.Reed MB, Domenech P, Manca C, Su H, Barczak AK, et al. A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature. 2004;431:84–87. doi: 10.1038/nature02837. [DOI] [PubMed] [Google Scholar]
  • 361.Manca C, Tsenova L, Barry CE, III, Bergtold A, Freeman S, et al. Mycobacterium tuberculosis CDC1551 induces a more vigorous host response in vivo and in vitro, but is not more virulent than other clinical isolates. J Immunol. 1999;162:6740–6746. [PubMed] [Google Scholar]
  • 362.Newton SM, Smith RJ, Wilkinson KA, Nicol MP, Garton NJ, et al. A deletion defining a common Asian lineage of Mycobacterium tuberculosis associates with immune subversion. Proc Natl Acad Sci U S A. 2006;103:15594–15598. doi: 10.1073/pnas.0604283103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Portevin D, Gagneux S, Comas I, Young D. Human macrophage responses to clinical isolates from the Mycobacterium tuberculosis complex discriminate between ancient and modern lineages. PLoS Pathog. 2011;7:e1001307. doi: 10.1371/journal.ppat.1001307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Chacon-Salinas R, Serafin-Lopez J, Ramos-Payan R, Mendez-Aragon P, Hernandez-Pando R, et al. Differential pattern of cytokine expression by macrophages infected in vitro with different Mycobacterium tuberculosis genotypes. Clin Exp Immunol. 2005;140:443–449. doi: 10.1111/j.1365-2249.2005.02797.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Dormans J, Burger M, Aguilar D, Hernandez-Pando R, Kremer K, et al. Correlation of virulence, lung pathology, bacterial load and delayed type hypersensitivity responses after infection with different Mycobacterium tuberculosis genotypes in a BALB/c mouse model. Clin Exp Immunol. 2004;137:460–468. doi: 10.1111/j.1365-2249.2004.02551.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Shimono N, Morici L, Casali N, Cantrell S, Sidders B, et al. Hypervirulent mutant of Mycobacterium tuberculosis resulting from disruption of the mce1 operon. Proc Natl Acad Sci U S A. 2003;100:15918–15923. doi: 10.1073/pnas.2433882100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Subbian S, Tsenova L, Yang G, O’Brien P, Parsons S, et al. Chronic pulmonary cavitary tuberculosis in rabbits: a failed host immune response. Open Biol. 2011;1:110016. doi: 10.1098/rsob.110016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Subbian S, Tsenova L, O’Brien P, Yang G, Kushner NL, et al. Spontaneous latency in a rabbit model of pulmonary tuberculosis. Am J Pathol. 2012;181:1711–1724. doi: 10.1016/j.ajpath.2012.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Subbian S, O’Brien P, Kushner NL, Yang G, Tsenova L, et al. Molecular immunologic correlates of spontaneous latency in a rabbit model of pulmonary tuberculosis. Cell Commun Signal. 2013;11:16. doi: 10.1186/1478-811X-11-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Briken V, Porcelli SA, Besra GS, Kremer L. Mycobacterial lipoarabinomannan and related lipoglycans: from biogenesis to modulation of the immune response. Mol Microbiol. 2004;53:391–403. doi: 10.1111/j.1365-2958.2004.04183.x. [DOI] [PubMed] [Google Scholar]
  • 371.Torrelles JB, Schlesinger LS. Diversity in Mycobacterium tuberculosis mannosylated cell wall determinants impacts adaptation to the host. Tuberculosis. 2010;90:84–93. doi: 10.1016/j.tube.2010.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Crick DC, Brennan PJ, McNeil MR. The cell wall of Mycobacterium tuberculosis. In: Rom WM, Garay SM, editors. Tuberculosis. 2. Lippincott Williams and Wilkins; Philadelphia: 2003. [Google Scholar]
  • 373.Rajaram MV, Ni B, Morris JD, Brooks MN, Carlson TK, et al. Mycobacterium tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-activated protein kinase 2 (MK2) and microRNA miR-125b. Proc Natl Acad Sci U S A. 2011;108:17408–17413. doi: 10.1073/pnas.1112660108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Torrelles JB, Sieling PA, Arcos J, Knaup R, Bartling C, et al. Structural differences in lipomannans from pathogenic and nonpathogenic mycobacteria that impact CD1b-restricted T cell responses. J Biol Chem. 2011;286:35438–35446. doi: 10.1074/jbc.M111.232587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Yadav M, Roach SK, Schorey JS. Increased mitogen-activated protein kinase activity and TNF-alpha production associated with Mycobacterium smegmatis- but not Mycobacterium avium-infected macrophages requires prolonged stimulation of the calmodulin/calmodulin kinase and cyclic AMP/protein kinase A pathways. J Immunol. 2004;172:5588–5597. doi: 10.4049/jimmunol.172.9.5588. [DOI] [PubMed] [Google Scholar]
  • 376.Doz E, Rose S, Nigou J, Gilleron M, Puzo G, et al. Acylation determines the toll-like receptor (TLR)-dependent positive versus TLR2-, mannose receptor-, and SIGNR1-independent negative regulation of pro-inflammatory cytokines by mycobacterial lipomannan. J Biol Chem. 2007;282:26014–26025. doi: 10.1074/jbc.M702690200. [DOI] [PubMed] [Google Scholar]
  • 377.Guerardel Y, Maes E, Elass E, Leroy Y, Timmerman P, et al. Structural study of lipomannan and lipoarabinomannan from Mycobacterium chelonae. Presence of unusual components with alpha 1,3-mannopyranose side chains. J Biol Chem. 2002;277:30635–30648. doi: 10.1074/jbc.M204398200. [DOI] [PubMed] [Google Scholar]
  • 378.Hance AJ, Saltini C, Crystal RG. Does de novo immunoglobin synthesis occur on the epithelial surface of the human lower respiratory tract? Am Rev Resp Dis. 1988;137:17–24. doi: 10.1164/ajrccm/137.1.17. [DOI] [PubMed] [Google Scholar]
  • 379.Khoo K-H, Dell A, Morris HR, Brennan PJ, Chatterjee D. Inositol phosphate capping of the nonreducing termini of lipoarabinomannan from rapidly growing strains of Mycobacterium. J Biol Chem. 1995;270:12380–12389. doi: 10.1074/jbc.270.21.12380. [DOI] [PubMed] [Google Scholar]
  • 380.Afonso-Barroso A, Clark SO, Williams A, Rosa GT, Nobrega C, et al. Lipoarabinomannan mannose caps do not affect mycobacterial virulence or the induction of protective immunity in experimental animal models of infection and have minimal impact on in vitro inflammatory responses. Cell Microbiol. 2012 doi: 10.1111/cmi.12065. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]

RESOURCES