Remodelacion Pulmonar en Tuberculosis

download Remodelacion Pulmonar en Tuberculosis

of 10

Transcript of Remodelacion Pulmonar en Tuberculosis

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    1/10

    Remodeling in TB JID 2005:192 (1 October) 1201

    M A J O R A R T I C L E

    Lung Remodeling in Pulmonary Tuberculosis

    Keertan Dheda,1,2,3 Helen Booth,3 Jim F. Huggett,1 Margaret A. Johnson,2 Alimuddin Zumla,1 and Graham A. W. Rook1

    1Centre for Infectious Diseases and International Health, 2Department of Thoracic and HIV Medicine, Royal Free Hospital, and 3Department

    of Thoracic Medicine, Middlesex Hospital, Royal Free and University College Medical School, London, United Kingdom

    Tuberculosis is a global public health catastrophe responsible for 18 million cases of illness and 2 million

    deaths annually. Pulmonary cavitation with cough-generated aerosol is the principle means of spread, and

    lung remodeling (healed cavitation, fibrosis, and bronchiectasis) is a major cause of lung disability, surpassing

    all other diffuse parenchymal lung diseases combined. Efficient granuloma turnover is mycobactericidal, and

    extracellular matrix is disbanded without scarring. In many with progressive disease, however, there is dys-

    regulated granuloma turnover, liquefactive necrosis, and pathological scarring. The pathological mechanisms

    and the related immunological pathways underpinning these phenomena are reviewed in the present article.

    Further studies are needed to identify and develop specific immunotherapeutic interventions that target im-munopathology, since they have the potential to substantially reduce spread.

    The lung remodeling associated with pulmonary tuber-

    culosis (TB) (healed cavitation, fibrosis, and distorted

    architecture) has never been satisfactorily explained. This

    is despite TB being an international publichealth priority

    and despite cavitation, with associated tissue and lique-

    factive necrosis, being the mechanism by which disease

    transmission occurs. In addition to addressing the sig-

    nificant morbidity associated with lung remodeling, in-

    vestigations that can elucidate mechanisms and immu-

    nological pathways relevant to tissue necrosis might lead

    to strategies for interrupting aerosol-mediated transmis-

    sion. Such knowledge can be incorporated into immu-

    notherapeutic strategies aimed at halting person-to-per-

    son spread of Mycobacterium tuberculosis.

    Why do caseous and liquefactive necrosis occur? More-

    over, why is fibrosis present in a disease characterized by

    a potent interferon (IFN)g response, when this cytokine

    generally opposes fibrosis [1]? In this review, we reeval-

    uate current knowledge of the immunopathogenesis of

    tissue necrosis and fibrosis. Peer-reviewed data for the

    Received 16 December 2004; accepted 9 May 2005; electronically published

    29 August 2005.

    Potential conflicts of interest: none reported.

    Financial support: British Lung Foundation (support to K.D.).

    Reprints or correspondence: Dr. Keertan Dheda, Centre for Infectious Diseases

    and International Health, Royal Free and University College Medical School, 46

    Cleveland St., London W1T 4JF, United Kingdom ([email protected]).

    The Journal of Infectious Diseases 2005;192:120110

    2005 by the Infectious Diseases Society of America. All rights reserved.

    0022-1899/2005/19207-0012$15.00

    present article were identified by searches of the Medline

    and PubMed databases, up to and including October

    2004, in all languages, by use of the search terms tu-

    berculosis and remodeling, cavitation, fibrosis,

    immunopathology, extracellular matrix, protease,

    or necrosis. Other sources were the references cited in

    retrieved articles and referenced textbooks.

    DEFINITIONS

    In asthma and chronic obstructive pulmonary dis-

    ease, remodeling refers to anatomical and structural

    changes that are not easily reversible (laying down of

    extracellular matrix [ECM]), in contrast to reversible

    changes, such as edema and cellular infiltration) [2, 3].

    In this review, we have extended the term remodeling

    to include residual cavitation, lung fibrosis or scarring,

    distortion of lung architecture leading to volume loss,

    and tuberculous bronchiectasis, all of which represent

    an inappropriate response to lung injury. An appro-priate response occurs, for example, when granuloma

    formation occurs in a coordinated fashion, followed by

    disbanding of the granuloma, dissolution of ECM, and

    return to normal tissue architecture. Fibrosis implies

    a structural alteration, with laying down of collagenous

    ECM by fibroblasts and other cell types. The fibrosis

    may be interstitial; occur as a capsule around cavities;

    be bandlike, with distortion of the lung architecture;

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    2/10

    1202 JID 2005:192 (1 October) Dheda et al.

    Figure 1. Principle cell types and cytokines involved in competent granuloma formation. NK T (NKT) cells, CD4+ T cells, CD8+ T cells, and gd

    T cells produce type 1 cytokines that activate macrophages, which are mycobactericidal. Granulocytes (not shown) may be important in early granuloma

    formation. Mycobacteria (rods) may be present within cells or extracellularly. Mycobacterial killing can occur by macrophage apoptosis, cytotoxic T

    cell lysis (jagged arrow), or directly through granulysin (star)mediated destruction. Tumor necrosis factor (TNF)a is a likely requisite for macrophage

    mycobactericidal activity, necrosis, and formation of the fibrous capsule. Cellular recruitment into the granuloma is facilitated by a chemokine gradient.

    IFN-g, interferon-g; TGF-b, transforming growth factorb.

    or be a combination of these. The term tissue necrosis en-

    compasses both caseous and liquefactive necrosis.

    CLINICAL ASPECTS

    Cavitation into airways, with cough-induced aerosol genera-

    tion, is the principle method by which TB is spread. Untreated,

    there is a fatal outcome in 50% of individuals [4]. Despite che-

    motherapy, however, there may be widespread lung destruction

    with significant associated mortality [5]. Alternatively, subse-

    quent healing can result in extensive fibrosis, traction bronchi-

    ectasis [6, 7], and bronchostenosis [8], all of which may result

    in volume loss, a restrictive defect during pulmonary function

    testing [9, 10], and increased morbidity. Cavitation may erode

    blood vessels, and bronchiectasis may cause significanthemopty-

    sis [8, 11]. Poor drug penetration into cavities and fibrocaseousfoci, which are immunologically sealed off, may facilitate la-

    tency and selection of drug resistance. Residual distortion of the

    lung architecture depends on the degree to which the connective-

    tissue matrix of a granuloma is degraded and removed. The key

    question is what determines whether a granuloma resolves com-

    pletely without scarring or whether liquefactive necrosis and/or

    extensive scarring occurs.

    EFFICIENT GRANULOMA FORMATION

    AND DISSOLUTION

    The first cell types encountered by inhaled mycobacteria are

    the alveolar macrophages; phagocytosis is mediated by varioushost receptors [12, 13]. Lung gd T cells, NK T cells, and gran-

    ulocytes are important early arrivals that precede the second

    wave of expanded, IFN-g and tumor necrosis factor (TNF)

    aproducing effector T cell populations [1416]. Collectively,

    these cells initiate a chemokine and cytokine cascade that at-

    tracts other macrophages and, later, T cells to the site of in-

    fection [17] (figure 1). There is plasma exudation, and a fibrin

    clot is formed [18, 19]. Macrophage aggregation to form an

    early granuloma core is mediated by hyaluronic acid [20],

    which binds to macrophages via CD44 [21]. Detailed histo-

    logical analyses of developing granulomata in the rabbit model

    [22] led to the view that macrophage activation driven by cell-

    mediated immunity is followed by destruction of the same

    macrophages, which then accumulate as caseous necrosis, and

    is often associated with death of the contained bacilli. An al-

    ternative view is that granulocytes are early arrivals that medi-

    ate the cell lysis that forms the core of developing caseous

    necrosis [16, 23, 24]. Recent pathological studies of granulomas

    in tuberculous human lungs indicate that the peripheral lym-

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    3/10

    Remodeling in TB JID 2005:192 (1 October) 1203

    phocyte zones of the granuloma have secondary lymphoid fol-

    licles analogous to those found in lymph nodes [16].

    Macrophages and other cell types, such as fibroblasts, endo-

    thelial cells, and neutrophils, also produce proteases (metal-

    loproteinases [collagenase, gelatinase, and stromelysin]; the ly-

    sosomal proteinases [cathepsins]; and the plasminogen/plas-

    min system and its activator, urokinase]). Such enzymes may

    facilitate granuloma formation by mediating antigen process-ing, removal of ECM and cellular debris, and processing of

    cytokines and hormones [25]. They are tightly regulated at

    multiple levels, including transcription, proenzyme formation,

    and signaling control, as well as by tissue inhibitors of metal-

    loproteinases (TIMPs) [26, 27].

    Within the granuloma, both T cells and macrophages secrete

    TNF-a and lymphotoxin a3. Not only is TNF-a crucial for

    host defense [2832], but it also facilitates the structural in-

    tegrity of the granuloma by mediating the formation of the en-

    capsulating fibrous wall [33], together with transforming growth

    factor (TGF)b [34, 35]. Macrophages also secrete insulin-like

    growth factor (GF)1, fibroblast GF, fibronectin, and platelet-derived GF [36, 37]. These GFs are chemotactic and support

    macrophage proliferation and, hence, the laying down of ECM

    comprising collagen, fibronectins, and glycosaminoglycans [38

    40]. Caseous necrosiswhich occurs at 23 weeks in the

    rabbit model [22]and its associated low oxygen content cre-

    ate unfavorable conditions for mycobacterial multiplication

    [41]. This, together with macrophage activation and CD3+ ef-

    fector cell mechanisms, culminates in mycobacterial steriliza-

    tion. It was shown in 1927 that healed primary lesions are usu-

    ally sterile within 5 years, although latent bacilli may persist in

    other, superficially normal, parts of the lung [42, 43].

    Controlled dissolution of the granuloma follows mycobac-

    terial containment, although this may be incomplete if there

    has been massive production of caseum. Proteases cleave com-

    ponents of the ECM [26]. Macrophages phagocytose the part-

    ly degraded ECM components, which are terminally degraded

    within the lysosome or transported out of the lung [4446].

    Minor residual scarring may remain.

    DYSREGULATED GRANULOMA TURNOVER

    AND LIQUEFACTIVE NECROSIS

    In a significant minority of infected individualsthose with pro-

    gressive primary and reactivation diseasethere is progressive

    disease and liquefactive necrosis. This forms an ideal culture

    medium for mycobacteria; they multiply extracellularly to large

    numbers [47], since macrophages are unable to survive in ne-

    crotic tissue, partly because of its toxic fatty acid content [41].

    The proteolytic enzymes [48] compromise the integrity of the

    fibrous granuloma capsule. Caseous material may discharge in-

    to surrounding blood vessels and airways, thereby facilitating

    systemic dissemination, and to the outside environment via

    cough-induced respiratory droplets. The cavity often has an

    external zone of collagen, internal to which is a zone of gran-

    ulation tissue rich in fibroblasts, inflammatory cells, and cap-

    illaries [47]. The role of vasculopathy as a primary event in the

    facilitation of cavitation is unclear. With antimicrobial therapy,

    the cavities may persist as fibrotic walled structures lined by

    metaplastic squamous epithelium or as emphysematous bullae,or they may become distorted from without by traction fibrosis

    [47].

    Although the exact mechanisms of liquefaction are unknown,

    the available data suggest that dysregulated proteolysis, direct

    mycobacterial toxicity, the Koch phenomenon [49] and Shwartz-

    man reaction [50], and host effector cells and cytokines are key

    players (figure 2). These concepts are further discussed below.

    Dysregulated Proteolysis and Direct Mycobacterial Toxicity

    A variety of host proteases are up-regulated in mycobacteri-al infection [48, 5153], and dysregulation of protease control

    mechanisms is likely to mediate proteolysis of structural com-

    ponents of the lung [54]. Cytokines such as TNF-a and in-

    terleukin (IL)1b can up-regulate proteases [5557] in mouse

    and human monocytes in response to mycobacterial proteins

    [58], and lipoarabinomannan induces collagenases in myeloid

    cells [54]. Macrophage destruction by cytotoxic T cells and the

    release of lysosomal contents are likely to play a significant role

    in tissue destruction and may explain some of the necrosis in

    the central part of the granuloma where macrophages pre-

    dominate. Granulocytes may be important facilitators of early

    necrosis [16, 23]. As discussed below, local tissue damage willbe more likely if the macrophages undergo necrosis rather than

    apoptosis. Although thought to be less important, mycobacte-

    ria themselves may produce endopeptidases [25] or other pro-

    necrotic virulence factors. Recently, however, a polyketide tox-

    ic factor, isolated from M. ulcerans, has been shown to have

    cytopathic properties and produced necrotic cutaneous le-

    sions in guinea pigs [59]. Since the M. tuberculosis genome

    contains many polyketide synthesis genes [60], it is reasonable

    to speculate that this toxin may represent one of a family of

    virulence factors associated with immunopathology in myco-

    bacterial diseases. Similarly, the secreted protein early secreted

    antigenic target 6 kDa (ESAT-6) is a virulence factor for M.

    tuberculosis and appears to cause lysis of lung epithelial cells

    and to facilitate local spread [61]. Its contribution to caseation

    is not known. Certain deletional mutants of M. tuberculosis

    have an unchanged ability to proliferate within the host but

    induce much less immunopathology, suggesting that myco-

    bacterial virulence factors actively induce tissue-damaging cell-

    mediated immune responses [62, 63].

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    4/10

    1204 JID 2005:192 (1 October) Dheda et al.

    Figure 2. Factors that might contribute to dysregulated granuloma formation. Mycobacterial antigens mediate uncoupled protease metabolism ac-

    tivity, apoptosis, the Koch phenomenon, and, perhaps, cell lysis mediated by early secreted antigenic target 6 kDa (ESAT-6) and induce the release

    of cytokines (tumor necrosis factor [TNF]a, mixed Th1/Th2, and transforming growth factor [TGF]b), which collectively facilitate liquefactive necrosis

    and deranged extracellular matrix (ECM) turnover. Mycobacteria (solid capsules) multiply extracellularly, compromise the fibrous granuloma capsule,

    and may discharge into a bronchial lumen or blood vessel.

    The Koch Phenomenon and Shwartzman Reaction

    Koch [49] showed that tuberculous guinea pigs developed ne-

    crosis locally and at distant sites of infection after rechallenge

    with tuberculin. High tuberculin reactivity occurs in experi-

    mental animals with liquefaction [64]. Similarly, the tubercu-

    lin test is frequently necrotic in humans with TB but not in

    healthy bacille Calmette-Guerin (BCG)vaccinated individuals

    [47]. More recently, an elegant study showed that M. tuber-

    culosisinfected mice developed increased lung inflammation

    and elevated TNF-a levels when rechallenged with mycobac-

    terial antigens [65]. Moreover, when prophylactic-vaccine can-

    didates are tested for therapeutic effects in mice with TB, they

    develop increased immunopathology [66] and classical Koch

    reactions characterized by cellular necrosis within granuloma-

    ta [67]. One explanation may be the systemic Shwartzman re-

    action [50], which referred originally to cutaneous necrosis at a

    site of a previous endotoxin injection after intravenous injec-

    tion of lipopolysaccharide (LPS). The concept is that the initial

    skin inflammation causes endothelial activation and accumula-

    tion of inflammatory cells. Then, a subsequent systemic cyto-

    kine-inducing signal administered 24 h later preferentially

    triggers necrosis in the prepared skin site. Indeed, M. tuber-

    culosisinfected C57BL/6 mice, which do not conventionally

    develop necrosis in this model, develop caseous necrosis after

    inoculation with LPS [68]. Moreover, LPS shares many physical

    properties and biological functions with a major mycobacterial

    antigen, lipoarabinomannan [69].

    Host Cytokines and Effector Mechanisms

    Necrosis and cavitation can occur in response to nonviable,

    nontoxic components of mycobacteria [48, 64, 70]. This sug-

    gests that host cytokines and enzymes are responsible for the

    necrosis. Although the precise mechanisms inducing progres-

    sion from controlled protease release and granuloma forma-

    tion to dysregulated protease production with liquefactive ne-

    crosis are unknown, a likely facilitator is TNF-a.

    TNF-a. Although TNF-a is essential for immunity to TB,

    in progressive disease TNF-a is associated with fever and wast-

    ing [7173] and correlates with disease activity and immuno-

    pathology [29, 7477]. Cells containing M. tuberculosisare ren-

    dered exquisitely sensitive to killing by TNF-a [75, 78]. As out-

    lined above, TNF-a can up-regulate metalloproteinases and

    urokinase and thereby facilitate proteolysis of structural lung

    elements. However, under what conditions is the protective

    TNF-a molecule toxic? Data, as outlined below, support a

    possible role for Th2 cytokines in such toxicity.

    Th1 and Th2 cytokines. Recent data have convincingly

    demonstrated the presence of Th2 cytokines in human TB [79].

    Th2 cells may mediate local tissue damage that is IL-4 depen-

    dent [80]. In M. tuberculosisinfected mice, susceptibility to the

    toxic effects of TNF-a injected into the footpads coincided

    temporally with the emergence of Th2 cytokines in the lungs

    [81]. IL-4 may also regulate TNF-amediated enteropathy inTrichinella spiralis infection [82], and necrosis in a schistoso-

    miasis model coincides precisely with the superimposition of

    Th2 response on an existing Th1 pattern [83]. Mouse (BALB/

    c) models of TB showed high IFN-g levels early during infec-

    tion, with increased levels of IL-4 during the chronic phase of

    infection, which was characterized by progressive fibrosis and

    necrosis [84, 85]. The Th2 response may exacerbate tissue dam-

    age by enhancing the pathological effect of TNF-a [81]. More

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    5/10

    Remodeling in TB JID 2005:192 (1 October) 1205

    Figure 3. The roles of the subversive Th2-like component of the immune response in progressive human tuberculosis. This Th2-like interleukin

    (IL)4 response, modulated by IL-4d2, may be primed by environmental mycobacteria and helminths and is most striking in developing countries [92,

    93]. After exposure to Mycobacterium tuberculosis, certain cell-wall components and protein antigens [9497] are then able to drive and enhance

    this IL-4 response, which may contribute to deactivation of macrophages, as well as to necrosis and fibrosis. TGF-b, transforming growth factor b;

    TNF-a, tumor necrosis factor a.

    recent evidence, from IL-4 gene knockout experiments in BALB/

    c mice, showed an absence of TNF-amediated toxicity after

    TNF-a challenge in the absence of IL-4 [86]. Collectively, these

    murine data suggest that, under the influence of superimposed

    Th2 cytokines, TNF-a is toxic in dominantly Th1-mediated le-

    sions. It is not unreasonable to suggest that a similar mechanism

    operates in humans. Studies of lavage fluid from patients with

    TB show the presence of IL-4producing Th2 lymphocyte sub-sets in those with cavitary TB [87] and the presence of a Th1

    cytokine profile in those with noncavitary disease [74]. Simi-

    larly, expression of IL-4 in peripheral blood lymphocytes cor-

    relates with cavitary disease [88].

    In other murine knockout models of immunopathology in-

    duced by mycobacteria, g/d T cells [89], a/b-positive T cells,

    IL-12, IFN-g, and TNF-a [90, 91] also seem to be essential for

    necrosis of granulomata (figure 3). It has been suggested that

    caseous necrosis is a protective mechanism to reduce the log-

    arithmic growth of the organisms [47]. However, it is apoptosis

    rather than necrosis that destroys M. tuberculosis (see below),

    so it is intriguing to speculate that M. tuberculosishas evolvedcomponents that exploit the hosts tissue-damaging protective

    responses by driving a Th2 response and necrosis. It is inter-

    esting that particularly virulent Beijing strains ofM. tuberculosis

    cause human monocytes to express IL-4 and IL-13 [94].

    Apoptosis. Caseation is a mixture of necrosis and apopto-

    sis. Large numbers of apoptotic T cells and macrophages are

    seen in caseous foci [98]. Immunohistochemical analysis and

    in situ hybridization have shown that macrophages surround-

    ing caseous foci are negative for the antiapoptotic protein bcl2

    but positive for the proapoptotic protein bax, whereas the as-

    sociated T cells express IFN-g and FasL [98]. Apoptosis of

    macrophages may be beneficial to the host. Both in mice [99]

    and in humans [100], CD8+ cytotoxic T cells that use the gran-

    ule-mediated pathway to induce apoptosis (as opposed to the

    FasFas ligand pathway) lead to killing of the contained my-

    cobacteria too. The role of the granule contents, particularly

    granulysin, has been reviewed [101]. Apoptosis induced by ATP

    [102, 103], TNF-a independent of IFN-g [104], Fas ligand

    [105], and hydrogen peroxide [106] all promote killing of vir-

    ulent M. tuberculosiswithin macrophages, whereas the necrotic

    mode of death does not [105, 107]. Moreover, M. tuberculosishas mechanisms that tend to inhibit apoptosis. Mannose-cap-

    ped lipoarabinomannan (Man-LAM) stimulates phosphoryla-

    tion of Bad, a proapoptotic protein, and so inhibits apoptosis

    [108]. Another group has shown that Man-LAM inhibits ap-

    optosis by a mechanism involving calcium-dependent signal-

    ing [109]. Recently, cathepsins have been shown to modulate

    apoptosis in human pulmonary epithelial cells [110], although

    their role in TB has not been investigated. It is, therefore, pos-

    sible that the material that accumulates as caseum is, in fact,

    a product of inappropriate necrosis. Apoptotic cells are usu-

    ally removed by surrounding cells with minimal inflammatory

    consequences, whereas necrotic cells accumulate. However, itis also possible that, when a sufficient percentage of the cells

    are undergoing apoptosis, the normal clearance mechanism

    fails, and apoptotic cells can themselves contribute to caseum.

    FIBROSIS IN TB

    TNF-a, TGF-b, and the pathogenesis of fibrosis. Extensive

    deposition of ECM occurs in the guinea pig model of TB [111].

    Available data suggest that TGF-b, together with TNF-a, plays

    a key role in the formation of the fibrous wall that encapsu-

    lates the tuberculous granuloma [3335]. The importance of

    TGF-b with respect to pulmonary fibrosis has been established

    in human [38, 112] and animal [113, 114] models. Although

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    6/10

    1206 JID 2005:192 (1 October) Dheda et al.

    the exact signaling pathways that drive fibroblasts to lay down

    ECM and macrophages to dissolve ECM are poorly under-

    stood, it is reasonable to speculate that TGF-b may contribute

    to the dysregulation of ECM turnover in TB. Indeed, TGF-b

    may also perpetuate fibrogenesis by inhibiting apoptosis of

    fibroblasts [115] and mediating localized production of inhib-

    itors of TIMP [116]. However, it seems that TGF-b, although

    necessary, is not sufficient for fibrosis. The importance of othermolecules, such as TNF-a, in lung fibrosis is illustrated by

    models of bleomycin and hypersensitivity pneumonitis, in

    which antiTNF-a antibodies ameliorate lung fibrosis [117,

    118]. The role of other cell typessuch as alveolar epithelial

    cells, which can harbor M. tuberculosis [42] and can be an im-

    portant source of cytokines and GFs in idiopathic pulmonary

    fibrosis [119]merits further investigation.

    Two paradoxes immediately arise. The first is the presence

    of increased ECM at the periphery of the granuloma but dis-

    solution of ECM in the central part of the granuloma. Possible

    explanations include a cytokine gradient within the granuloma

    [120], including low IFN-g levels in the central macrophage-

    dominant part of the granuloma or selective binding of cyto-

    kines to glycosaminoglycans in different parts of the granuloma

    [121]. Therefore, in the center of the granuloma, high TNF-a

    concentrations, protease activity, and lysis of infected macro-

    phages predominate, whereas, in the periphery, TGF-b activity

    and fibroblast activation predominate.

    The second paradox is the presence of fibrosis where there

    are potent IFN-g responses, which down-regulate TGF-b and

    production of collagen by fibroblasts [122, 123]. The answer

    may lie in the significant Th2 response that develops parallel

    to and within the framework of a Th1 response.Role of Th2 cytokines in fibrosis. IL-4 and IL-13 are pro-

    fibrotic and enhance collagen production by fibroblasts [123].

    Mice, when infected with saprophytic mycobacteria, only de-

    velop peribronchial and interstitial fibrosis when primed for IL-

    4 [84]. Even a single epitope (16 aa) inducing a Th2 response

    can drive fibrosis in a murine model of TB [124].

    In all human diseases characterized by marked pulmonary

    fibrosis (systemic sclerosis, idiopathic pulmonary fibrosis, ra-

    diation-induced pulmonary fibrosis, and chronic lung allograft

    rejection; reviewed in [125]), there is expression of type 2 cy-

    tokines. The same is true in the bleomycin murine model [125]

    and in schistosomiasis, in which fibrosis and tissue remodel-ing do not occur if induction of type 2 cytokines by the ova

    is blocked by preimmunization with ova plus IL-12 [126]. No-

    tably, there are no data on cytokine profiles in the late stages

    of fibrotic sarcoidosis [127]. These observations have led to the

    type 2 cytokine hypothesis of fibrosis [125]. This paradigm

    implies an initial Th1-type response to antigenic challenge, fol-

    lowed by a Th2 response that seeks to wall off, or isolate, a

    persistent antigen from the host [128130]. M. tuberculosismay

    be exploiting this host response to remain immunologically

    sealed off in fibrocaseous foci.

    The Th2 phenotype causes fibroblast activation and collagen

    deposition in human [131, 132] and animal [114, 133] models.

    Collagen synthesis in granulomata is stimulated by Th2 cyto-

    kines (IL-4, IL-13, and IL-4d2) and reciprocally inhibited by

    Th1 cytokines such as IFN-g and IL-12 [1, 126]. Moreover,

    granulomatous inflammation and fibrosis are significantly re-duced in Stat6/ mice [134]; IL-4 and IL-13 are the major

    activators of Stat6. Fibrosis is particularly pronounced in mice

    overexpressing IL-13 [125]. This cytokine, like IL-4 and IL-4d2

    [135], activates fibroblasts and promotes collagen formation.

    However, IL-13 also drives expression and activation of TGF-

    b [125]. Significantly, the increase in expression of type 2 cy-

    tokines in human TB extends to IL-13, which, like IL-4 and

    IL-4d2, is increased 80100-fold [136, 137].

    Smoking can reactivate TB, probably because nicotine switches

    off secretion of TNF-a by macrophages, leaving IL-10 secretion

    unchanged [138]. Similarly, TNF-amediated fibrotic lung dis-

    eases like sarcoidosis and extrinsic allergic alveolitis [139, 140]are less common in smokers, and smoking improves survival in

    a Th2-type fibrotic lung condition, idiopathic pulmonary fibro-

    sis [141, 142]. The common denominator facilitating fibrosis in

    all of these disorders might be the presence of a Th2 cytokine

    component acting synergistically with TNF-a.

    IL-4 or IL-4d2 from CD8+ T cells and their role in fibrosis.

    In systemic sclerosis, Th2 cytokine expression is associated with

    pulmonary fibrosis [135]. Interestingly, in several patients, the

    IL-4, made mostly by the CD8+ T cells, was in fact not IL-

    4 at all but rather IL-4d2, raising the likelihood that the CD8+

    T cells making IL-4 in TB reported by van Crevel et al. might

    be really making IL-4d2 (van Crevel et al. used reagents that

    do not distinguish between the 2 cytokines [88]). IL-4d2 is

    antagonistic to IL-4 with regard to lymphocyte function [143,

    144], but it is an agonist on fibroblasts [135].

    ARE THERE ANTIGENS THAT PREFERENTIALLY

    DRIVE NECROSIS AND FIBROSIS?

    IMPLICATIONS FOR IMMUNOTHERAPY

    It is not yet clear whether there are antigens that preferentially

    drive Th2, or whether M. tuberculosis merely contains Th2

    adjuvant activity. The recent discovery that virulent Beijing

    strains contain lipids that cause human monocytes to make

    IL-4 and IL-13 suggests that adjuvanticity is part of the mech-

    anism [94]. Similarly, dendritic cells derived from BCG-in-

    fected precursors can drive a Th2-like response [145]. Nev-

    ertheless, there does seem to be some antigenic specificity to

    the Th2 response. ESAT-6 fails to drive IL-4 production [146],

    but certain epitopes from within the 16-kDa heat-shock pro-

    tein do so [95]. Identification of the antigens or epitopes that

    drive IL-4 production will allow design of vaccines that sup-

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    7/10

    Remodeling in TB JID 2005:192 (1 October) 1207

    press the unwanted Th2 component [79]. Agents like M. vac-

    cae, which attenuate Th2 responses [147, 148], have demon-

    strated the ability to modulate lung remodeling and radiographic

    absorbance in both drug-sensitive and resistant TB [149, 150];

    clinical trials using multiple dose regimes are currently under

    way [151].

    In conclusion, further research into the genetic, molecular,

    immunological, and cellular pathways that drive cavitation andfibrosis are required. This could yield clinical benefit and has

    the potential to interrupt transmission. For instance, vaccines

    that target immunopathology (either host responses or bacte-

    rial virulence factors), even if they are not completely protec-

    tive, may have the ability to reduce global disease burden by

    minimizing cavitation and interrupting transmission. Like the

    antiTNF-a agents [152154], other immunotherapeutic mo-

    dalities could target pathways relating to cytokine signaling, ap-

    optosis, protease activity,and ECM turnover. However, such goals

    are tenable only if adequate funding is committed by government

    agencies, the pharmaceutical industry,and internationalcharities.

    Moreover, there will have to be a global organizational structurecommitted to achieving such goals.

    References

    1. Sempowski GD, Derdak S, Phipps RP. Interleukin-4 and interferon-

    gamma discordantly regulate collagen biosynthesis by functionally

    distinct lung fibroblast subsets. J Cell Physiol 1996; 167:2906.

    2. Caughey GH. Chairmans summary: mechanisms of airway remod-

    eling. Am J Respir Crit Care Med 2001; 164:S267.

    3. Jeffery PK. Remodeling in asthma and chronic obstructive lung dis-

    ease. Am J Respir Crit Care Med 2001; 164:S2838.

    4. Smith PG, Moss AR. Epidemiology of tuberculosis. In: Bloom BR,

    ed. Tuberculosis: pathogenesis, protection, and control. 1st ed. Wash-ington DC, American Society for Microbiology, 1994:4759.

    5. Bobrowitz ID. The destroyed tuberculous lung. Scand J Respir Dis

    1974; 55:828.

    6. Curtis JK. The significance of bronchiectasis associated with pulmonary

    tuberculosis. Am J Med 1957; 22:894903.

    7. Rosenzweig DY. The role of tuberculosis and other forms of bron-

    chopulmonary necrosis in the pathogenesis of bronchiectasis. Am Rev

    Respir Dis 1966; 93:76985.

    8. Kim YH, Kim HT, Lee KS, Uh ST, Cung YT, Park CS. Serial fiberoptic

    bronchoscopic observations of endobronchial tuberculosis before and

    early after antituberulosis chemotherapy. Chest 1993; 103:6737.

    9. Lopez-Majano V. Ventilation and transfer of gases in pulmonary tu-

    berculosis. Respiration 1973; 30:4863.

    10. Skoogh BE. Lung mechanics in pulmonary tuberculosis. I. Static lung

    volumes. Scand J Respir Dis 1973; 54:14856.

    11. Thompson JR. Mechanisms of fatal pulmonary hemorrhage in tu-

    berculosis. Am J Surg 1955; 89:63744.

    12. Ernst JD. Macrophage receptors for Mycobacterium tuberculosis. Infect

    Immun 1998; 66:127781.

    13. Heldwein KA, Fenton MJ. The role of Toll-like receptors in immunity

    against mycobacterial infection. Microbes Infect 2002; 4:93744.

    14. Apostolou I, Takahama Y, Belmant C, et al. Murine natural killer T

    (NKT) cells [correction of natural killer cells] contribute to the gran-

    ulomatous reaction caused by mycobacterial cell walls. Proc Natl Acad

    Sci USA 1999; 96:51416.

    15. Kaufmann SH. g/d and other unconventional T lymphocytes: what

    do they see and what do they do? Proc Natl Acad Sci USA 1996;93:

    22729.

    16. Ulrichs T, Kosmiadi GA, Trusov V, et al. Human tuberculous gran-

    ulomas induce peripheral lymphoid follicle-like structures to orches-

    trate local host defence in the lung. J Pathol 2004; 204:21728.

    17. Ulrichs T, Kaufmann SHE. Cell-mediated immune response. In: Rom

    WN, Garay SM, eds. Tuberculosis. 2nd ed. Philadelphia: Lippincott

    Williams & Wilkins, 2004:25162.

    18. Behling CA, Perez RL, Kidd MR, Staton GW Jr, Hunter RL. Induction

    of pulmonary granulomas, macrophage procoagulant activity, and

    tumor necrosis factor-alpha by trehalose glycolipids. Ann Clin Lab

    Sci 1993; 23:25666.

    19. Perez RL, Roman J, Staton GW Jr, Hunter RL. Extravascular coag-

    ulation and fibrinolysis in murine lung inflammation induced by the

    mycobacterial cord factor trehalose-6,6-dimycolate. Am J Respir Crit

    Care Med 1994; 149:5108.

    20. Love SH, Shannon BT, Myrvik QN, Lynn WS. Characterization of

    macrophage agglutinating factor as a hyaluronic acid-protein com-

    plex. J Reticuloendothel Soc 1979; 25:26982.

    21. Green SJ, Tarone G, Underhill CB. Aggregation of macrophages and

    fibroblasts is inhibited by a monoclonal antibody to the hyaluronate

    receptor. Exp Cell Res 1988; 178:22432.

    22. Lurie MB, Zappasodi P, Tickner C. On the nature of genetic resistance

    to tuberculosis in the light of the host-parasite relationshipsin natively

    resistant and susceptible rabbits. Am Rev Tuberc 1955; 72:297329.

    23. Seiler P, Aichele P, Bandermann S, et al. Early granuloma formationafter aerosol Mycobacterium tuberculosisinfection is regulated by neu-

    trophils via CXCR3-signaling chemokines. Eur J Immunol 2003;33:

    267686.

    24. Turner OC, Basaraba RJ, Orme IM. Immunopathogenesis of pulmonary

    granulomas in the guinea pig after infection with Mycobacterium tu-

    berculosis. Infect Immun 2003; 71:86471.

    25. Munger, JS, Chapman HA Jr. Tissue destruction by proteases. In: Rom

    WN, Garay SM, eds. Tuberculosis. 2nd ed. Philadelphia: Lippincott

    Williams & Wilkins, 2004:35362.

    26. Chapman HA Jr. Role of enzyme receptors and inhibitors in regulating

    proteolytic activities of macrophages. Ann N Y Acad Sci 1991;624:

    8796.

    27. Mauviel A. Cytokine regulation of metalloproteinase gene expression.

    J Cell Biochem 1993; 53:28895.

    28. Appelberg R. Protective role of interferon gamma, tumor necrosisfactor alpha and interleukin-6 in Mycobacterium tuberculosis and M.

    avium infections. Immunobiology 1994; 191:5205.

    29. Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with in-

    fliximab, a tumor necrosis factor a-neutralizing agent. N Engl J Med

    2001; 345:1098104.

    30. Mohan VP, Scanga CA, Yu K, et al. Effects of tumor necrosis factor

    alpha on host immune response in chronic persistent tuberculosis:

    possible role for limiting pathology. Infect Immun 2001; 69:184755.

    31. Ogawa T, Uchida H, Kusumoto Y, Mori Y, Yamamura Y, Hamada S.

    Increase in tumor necrosis factor alpha- and interleukin-6-secreting

    cells in peripheral blood mononuclear cells from subjects infected

    with Mycobacterium tuberculosis. Infect Immun 1991; 59:30215.

    32. Roach DR, Briscoe H, Saunders B, France MP, Riminton S, Britton

    WJ. Secreted lymphotoxin-alpha is essential for the control of an

    intracellular bacterial infection. J Exp Med 2001; 193:23946.

    33. Lukacs NW, Chensue SW, Strieter RM, Warmington K, Kunkel SL.

    Inflammatory granuloma formation is mediated by TNF-a-inducible

    intercellular adhesion molecule-1. J Immunol 1994; 152:58839.

    34. Aung H, Toossi Z, McKenna SM, et al. Expression of transforming

    growth factor-b but not tumor necrosis factor-a, interferon-g, and

    interleukin-4 in granulomatous lung lesions in tuberculosis. Tuber

    Lung Dis 2000; 80:617.

    35. Marshall BG, Wangoo A, Cook HT, Shaw RJ. Increased inflammatory

    cytokines and new collagen formation in cutaneous tuberculosis and

    sarcoidosis. Thorax 1996; 51:125361.

    36. Wangoo A, Taylor IK, Haynes AR, Shaw RJ. Up-regulation of alveolar

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    8/10

    1208 JID 2005:192 (1 October) Dheda et al.

    macrophage platelet-derived growth factor-B (PDGF-B) mRNA by

    interferon-gamma from Mycobacterium tuberculosis antigen (PPD)-

    stimulated lymphocytes. Clin Exp Immunol 1993; 94:4350.

    37. Yeager H Jr, Azumi N, Underhill CB. Fibrosis: the formation of the

    granuloma matrix. In: Rom WN, Garay SM, eds. Tuberculosis. 2nd

    ed. Philadelphia: Lippincott Williams & Wilkins, 2004:36372.

    38. Limper AH, Colby TV, Sanders MS, Asakura S, Roche PC, DeRemee

    RA. Immunohistochemical localization of transforming growth fac-

    tor-beta 1 in the nonnecrotizing granulomas of pulmonary sarcoid-

    osis. Am J Respir Crit Care Med 1994; 149:197204.

    39. Mornex JF, Leroux C, Greenland T, Ecochard D. From granuloma to

    fibrosis in interstitial lung diseases: molecular and cellular interactions.

    Eur Respir J 1994; 7:77985.

    40. Peyrol S, Takiya C, Cordier JF, Grimaud JA. Organization of the con-

    nective matrix of the sarcoid granuloma: evolution and cell-matrix in-

    teractions. Ann N Y Acad Sci 1986; 465:26885.

    41. Hemsworth GR, Kochan I. Secretion of antimycobacterial fatty acids

    by normal and activated macrophages. Infect Immun 1978; 19:1707.

    42. Hernandez-Pando R, Jeyanathan M, Mengistu G, et al. Persistence of

    DNA from Mycobacterium tuberculosis in superficially normal lung

    tissue during latent infection. Lancet 2000; 356:21338.

    43. Opie EL, Aronson JD. Tubercle bacilli in latent tuberculosis lesions

    and in lung tissue without tuberculous lesions. Arch Pathol 1927; 4:

    121.

    44. Culty M, OMara TE, Underhill CB, Yeager H Jr, Swartz RP. Hyalu-

    ronan receptor (CD44) expression and function in human peripheralblood monocytes and alveolar macrophages. J Leukoc Biol 1994;56:

    60511.

    45. Parakkal PF. Involvement of macrophages in collagen resorption. J

    Cell Biol 1969; 41:34554.

    46. Werb Z, Bainton DF, Jones PA. Degradation of connective tissue ma-

    trices by macrophages. III. Morphological and biochemical studies on

    extracellular, pericellular, and intracellular events in matrix proteolysis

    by macrophages in culture. J Exp Med 1980; 152:153753.

    47. Dannenburg AM, Rook GAW. Pathogenesis of pulmonary tubercu-

    losis: an interplay of tissue-damaging and macrophage-activatingim-

    mune responsesdual mechanisms that control bacillary multipli-

    cation. In: Bloom BR, ed. Tuberculosis: pathogenesis, protection, and

    control. 1st ed. Washington DC, American Society for Microbiology,

    1994:45983.

    48. Dannenberg AM Jr, Sugimoto M. Liquefaction of caseous foci in tu-berculosis. Am Rev Respir Dis 1976; 113:2579.

    49. Koch R. Fortsetzung uber ein Heilmittel gegen Tuberculose. Dtsch

    Med Wochenschr 1989; 17:1012.

    50. Rook GA, Al Attiyah R. Cytokines and the Koch phenomenon. Tu-

    bercle 1991; 72:1320.

    51. Rojas-Espinosa O, Dannenberg AM Jr, Sternberger LA, Tsuda T. The

    role of cathepsin D in the pathogenesis of tuberculosis:a histochemical

    study employing unlabeled antibodies and the peroxidase-antiperox-

    idase complex. Am J Pathol 1974; 74:117.

    52. Tsuda T, Dannenberg AM Jr, Ando M, Rojas-Espinosa O, Shima K.

    Enzymes in tuberculous lesions hydrolyzing protein, hyaluronic acid

    and chondroitin sulfate: a study of isolated macrophages and devel-

    oping and healing rabbit BCG lesions with substrate film techniques;

    the shift of enzyme pH optima towards neutrality in intact cells

    and tissues. J Reticuloendothel Soc 1974; 16:22031.

    53. Yagel S, Gallily R, Weiss DW. Effect of treatment with the MER fraction

    of tubercle bacilli on hydrolytic lysosomal enzyme activity of mouse

    peritoneal macrophages. Cell Immunol 1975; 19:3816.

    54. Chang JC, Wysocki A, Tchou-Wong KM, Moskowitz N, Zhang Y,

    Rom WN. Effect of Mycobacterium tuberculosis and its components

    on macrophages and the release of matrix metalloproteinases. Tho-

    rax 1996; 51:30611.

    55. Schonthal A, Herrlich P, Rahmsdorf HJ, Ponta H. Requirement for

    fos gene expression in the transcriptional activation of collagenase by

    other oncogenes and phorbol esters. Cell 1988; 54:32534.

    56. Gyetko MR, Shollenberger SB, Sitrin RG. Urokinase expression in mono-

    nuclear phagocytes: cytokine-specific modulation by interferon-gamma

    and tumor necrosis factor-alpha. J Leukoc Biol 1992; 51:25663.

    57. Gyetko MR, Wilkinson CC, Sitrin RG. Monocyte urokinase expres-

    sion: modulation by interleukins. J Leukoc Biol 1993; 53:598601.

    58. Gordon S, Cohn ZA. Bacille Calmette-Guerin infection in the mouse:

    regulation of macrophage plasminogen activator by T lymphocytes

    and specific antigen. J Exp Med 1978; 147:117588.

    59. George KM, Pascopella L, Welty DM, Small PL. A Mycobacterium ul-

    cerans toxin, mycolactone, causes apoptosis in guinea pig ulcers and

    tissue culture cells. Infect Immun 2000; 68:87783.60. Cole ST, Brosch R, Parkhill J, et al. Deciphering the biology ofMy-

    cobacterium tuberculosisfrom the complete genome sequence. Nature

    1998; 393:53744.

    61. Hsu T, Hingley-Wilson SM, Chen B, et al. The primary mechanism

    of attenuation of bacillus Calmette-Guerin is a loss of secreted lytic

    function required for invasion of lung interstitial tissue. Proc Natl

    Acad Sci USA 2003; 100:124205.

    62. Steyn AJ, Collins DM, Hondalus MK, Jacobs WR Jr, Kawakami RP,

    Bloom BR. Mycobacterium tuberculosis WhiB3 interacts with RpoV

    to affect host survival but is dispensable for in vivo growth. Proc Natl

    Acad Sci USA 2002; 99:314752.

    63. Kaushal D, Schroeder BG, Tyagi S, et al. Reduced immunopathology

    and mortality despite tissue persistence in a Mycobacterium tuber-

    culosis mutant lacking alternative sigma factor, SigH. Proc Natl Acad

    Sci USA 2002; 99:83305.

    64. Yamamura Y, Ogawa Y, Maeda H, Yamamura Y. Prevention of tu-

    berculous cavity formation by desensitization with tuberculin-active

    peptide. Am Rev Respir Dis 1974; 109:594601.

    65. Moreira AL, Tsenova L, Aman MH, et al. Mycobacterial antigens

    exacerbate disease manifestations in Mycobacterium tuberculosis-in-

    fected mice. Infect Immun 2002; 70:21007.

    66. Turner J, Rhoades ER, Keen M, Belisle JT, Frank AA, Orme IM.

    Effective preexposure tuberculosis vaccines fail to protect when they

    are given in an immunotherapeutic mode. Infect Immun 2000;68:

    17069.

    67. Taylor JL, Turner OC, Basaraba RJ, Belisle JT, Huygen K, Orme IM.

    Pulmonary necrosis resulting from DNA vaccination against tuber-

    culosis. Infect Immun 2003; 71:21928.

    68. Cardona PJ, Llatjos R, Gordillo S, et al. Towards a human-likemodel

    of tuberculosis: intranasal inoculation of LPS induces intragranulom-

    atous lung necrosis in mice infected aerogenically with Mycobacterium

    tuberculosis. Scand J Immunol 2001; 53:6571.

    69. Roach TI, Barton CH, Chatterjee D, Blackwell JM. Macrophage acti-

    vation: lipoarabinomannan from avirulent and virulent strains ofMy-

    cobacterium tuberculosis differentially induces the early genes c-fos, KC,

    JE, and tumor necrosis factor-a. J Immunol 1993; 150:188696.

    70. Yamamura Y. The pathogenesis of tuberculous cavities. Bibl Tuberc

    1958; 13:1337.

    71. Cadranel J, Philippe C, Perez J, et al. In vitro production of tumour

    necrosis factor and prostaglandin E2 by peripheral blood mononuclear

    cells from tuberculosis patients. Clin Exp Immunol 1990; 81:31924.

    72. Flynn JL, Goldstein MM, Chan J, et al. Tumor necrosis factor-alpha

    is required in the protective immune response against Mycobacterium

    tuberculosis in mice. Immunity 1995; 2:56172.

    73. Takashima T, Ueta C, Tsuyuguchi I, Kishimoto S. Productionof tumor

    necrosis factor alpha by monocytes from patients with pulmonary

    tuberculosis. Infect Immun 1990; 58:328692.

    74. Condos R, Rom WN, Liu YM, Schluger NW. Local immuneresponses

    correlate with presentation and outcome in tuberculosis. Am J Respir

    Crit Care Med 1998; 157:72935.

    75. Filley EA, Bull HA, Dowd PM, Rook GA. The effect ofMycobacterium

    tuberculosis on the susceptibility of human cells to the stimulatory and

    toxic effects of tumour necrosis factor. Immunology1992; 77:5059.

    76. Olobo JO, Geletu M, Demissie A, et al. Circulating TNF-a, TGF-b,

    and IL-10 in tuberculosis patients and healthy contacts. Scand J Im-

    munol 2001; 53:8591.

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    9/10

    Remodeling in TB JID 2005:192 (1 October) 1209

    77. Saltini C, Colizzi V. Soluble immunological markers of disease activity

    in tuberculosis. Eur Respir J 1999; 14:4856.

    78. Filley EA, Rook GA. Effect of mycobacteria on sensitivity to the cytotoxic

    effects of tumor necrosis factor. Infect Immun 1991; 59:256772.

    79. Rook GA, Hernandez-Pando R, Dheda K, Teng SG. IL-4 in tuberculosis:

    implications for vaccine design. Trends Immunol 2004; 25:4838.

    80. Muller KM, Jaunin F, Masouye I, Saurat JH, Hauser C. Th2 cells medi-

    ate IL-4-dependent local tissue inflammation. J Immunol 1993;150:

    557684.

    81. Hernandez-Pando R, Rook GA. The role of TNF-alpha in T-cell-me-

    diated inflammation depends on the Th1/Th2 cytokine balance. Im-

    munology 1994; 82:5915.

    82. Lawrence CE, Paterson JC, Higgins LM, MacDonald TT, Kennedy

    MW, Garside P. IL-4-regulated enteropathy in an intestinal nematode

    infection. Eur J Immunol 1998; 28:267284.

    83. Grzych JM, Pearce E, Cheever A, et al. Egg deposition is the major

    stimulus for the production of Th2 cytokines in murine schistoso-

    miasis mansoni. J Immunol 1991; 146:13227.

    84. Hernandez-Pando R, Orozcoe H, Sampieri A, et al. Correlation between

    the kinetics of Th1, Th2 cells and pathology in a murine model of ex-

    perimental pulmonary tuberculosis. Immunology 1996; 89:2633.

    85. Orme IM, Roberts AD, Griffin JP, Abrams JS. Cytokine secretion by

    CD4 T lymphocytes acquired in response to Mycobacterium tubercu-

    losis infection. J Immunol 1993; 151:51825.

    86. Hernandez-Pando R, Aguilar D, Hernandez ML, Orozco H, Rook G.

    Pulmonary tuberculosis in BALB/c mice with non-functional IL-4genes: changes in the inflammatory effects of TNF-a and in the reg-

    ulation of fibrosis. Eur J Immunol 2004; 34:17483.

    87. Mazzarella G, Bianco A, Perna F, et al. T lymphocyte phenotypic profile

    in lung segments affected by cavitary and non-cavitary tuberculosis.

    Clin Exp Immunol 2003; 132:2838.

    88. van Crevel R, Karyadi E, Preyers F, et al. Increased production of

    interleukin 4 by CD4+ and CD8+ T cells from patients with tuber-

    culosis is related to the presence of pulmonary cavities. J Infect Dis

    2000; 181:11947.

    89. Saunders BM, Frank AA, Cooper AM, Orme IM. Role ofgd T cells

    in immunopathology of pulmonary Mycobacterium avium infection

    in mice. Infect Immun 1998; 66:550814.

    90. Ehlers S, Benini J, Held HD, Roeck C, Alber G, Uhlig S. ab T cell

    receptor-positive cells and interferon-g, but not inducible nitric oxide

    synthase, are critical for granuloma necrosis in a mouse model ofmycobacteria-induced pulmonary immunopathology. J Exp Med 2001;

    194:184759.

    91. Florido M, Cooper AM, Appelberg R. Immunological basis of the

    development of necrotic lesions following Mycobacterium avium in-

    fection. Immunology 2002; 106:590601.

    92. Malhotra I, Mungai P, Wamachi A, et al. Helminth- and bacillus

    Calmette-Guerin-induced immunity in children sensitized in utero

    to filariasis and schistosomiasis. J Immunol 1999; 162:68438.

    93. Rook GAW, Dheda K, Zumla A. Do successful tuberculosis vaccines

    need to be immunoregulatory rather than merely Th1-boosting? Vac-

    cine 2005; 23:211520.

    94. Manca C, Reed MB, Freeman S, et al. Differential monocyte activation

    underlies strain-specific Mycobacterium tuberculosispathogenesis. In-

    fect Immun 2004; 72:55114.

    95. Agrewala JN, Wilkinson RJ. Differential regulation of Th1 and Th2 cells

    by p91110 and p2140 peptides of the 16-kD a-crystallin antigen of

    Mycobacterium tuberculosis. Clin Exp Immunol 1998; 114:3927.

    96. Seah GT, Rook GA. IL-4 influences apoptosis of mycobacterium-re-

    active lymphocytes in the presence of TNF-a. J Immunol 2001; 167:

    12307.

    97. Dheda K, Chang JS, Breen RA, et al. In vivo and in vitro studies of

    a novel cytokine, interleukin 4d2, in pulmonary tuberculosis. Am J

    Respir Crit Care Med 2005; 172:5018.

    98. Fayyazi A, Eichmeyer B, Soruri A, et al. Apoptosis of macrophages

    and T cells in tuberculosis associated caseous necrosis. J Pathol 2000;

    191:41725.

    99. Silva CL, Lowrie DB. Identification and characterization of murine

    cytotoxic T cells that kill Mycobacterium tuberculosis. Infect Immun

    2000; 68:326974.

    100. Cho S, Mehra V, Thoma-Uszynski S, et al. Antimicrobial activity of

    MHC class I-restricted CD8+ T cells in human tuberculosis. Proc

    Natl Acad Sci USA 2000; 97:122105.

    101. Krensky AM. Granulysin: a novel antimicrobial peptide of cytolytic

    T lymphocytes and natural killer cells. Biochem Pharmacol 2000;59:

    31720.

    102. Kusner DJ, Barton JA. ATP stimulates human macrophages to kill

    intracellular virulent Mycobacterium tuberculosis via calcium-depen-

    dent phagosome-lysosome fusion. J Immunol 2001; 167:330815.

    103. Lammas DA, Stober C, Harvey CJ, Kendrick N, Panchalingam S,

    Kumararatne DS. ATP-induced killing of mycobacteria by human

    macrophages is mediated by purinergic P2Z(P2X7) receptors. Im-

    munity 1997; 7:43344.

    104. Keane J, Shurtleff B, Kornfeld H. TNF-dependent BALB/c murine

    macrophage apoptosis following Mycobacterium tuberculosisinfection

    inhibits bacillary growth in an IFN-gamma independent manner. Tu-

    berculosis (Edinb) 2002; 82:5561.

    105. Oddo M, Renno T, Attinger A, Bakker T, MacDonald HR, Meylan

    PR. Fas ligand-induced apoptosis of infected human macrophages

    reduces the viability of intracellular Mycobacterium tuberculosis. J Im-

    munol 1998; 160:544854.

    106. Laochumroonvorapong P, Paul S, Manca C, Freedman VH, Kaplan G.

    Mycobacterial growth and sensitivity to H2O2 killing in human mono-cytes in vitro. Infect Immun 1997; 65:48507.

    107. Molloy A, Laochumroonvorapong P, Kaplan G. Apoptosis, but not

    necrosis, of infected monocytes is coupled with killing of intracellular

    bacillus Calmette-Guerin. J Exp Med 1994; 180:1499509.

    108. Maiti D, Bhattacharyya A, Basu J. Lipoarabinomannan from Myco-

    bacterium tuberculosis promotes macrophage survival by phosphor-

    ylating Bad through a phosphatidylinositol 3-kinase/Akt pathway. J

    Biol Chem 2001; 276:32933.

    109. Rojas M, Garcia LF, Nigou J, Puzo G, Olivier M. Mannosylatedlipoara-

    binomannan antagonizes Mycobacterium tuberculosisinduced macro-

    phage apoptosis by altering Ca+2-dependent cell signaling. J Infect Dis

    2000; 182:24051.

    110. Wille A, Gerber A, Heimburg A, et al. Cathepsin L is involved in ca-

    thepsin D processing and regulation of apoptosis in A549 human lung

    epithelial cells. Biol Chem 2004; 385:66570.111. Jayasankar K, Shakila H, Umapathy KC, Ramanathan VD. Biochem-

    ical and histochemical changes pertaining to active and healed cu-

    taneous tuberculosis. Br J Dermatol 2002; 146:97782.

    112. Broekelmann TJ, Limper AH, Colby TV, McDonald JA. Transforming

    growth factor beta 1 is present at sites of extracellular matrix gene

    expression in human pulmonary fibrosis. Proc Natl Acad Sci USA

    1991; 88:66426.

    113. Liu JY, Sime PJ, Wu T, et al. Transforming growth factor-b1

    overex-

    pression in tumor necrosis factor-a receptor knockout mice induces

    fibroproliferative lung disease. Am J Respir Cell Mol Biol 2001; 25:37.

    114. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-me-

    diated gene transfer of active transforming growth factor-b1 induces

    prolonged severe fibrosis in rat lung. J Clin Invest 1997; 100:76876.

    115. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates

    the decrease in cellularity during the transition between granulation

    tissue and scar. Am J Pathol 1995; 146:5666.

    116. Ramos C, Montano M, Garcia-Alvarez J, et al. Fibroblasts from id-

    iopathic pulmonary fibrosis and normal lungs differ in growth rate,

    apoptosis, and tissue inhibitor of metalloproteinases expression. Am

    J Respir Cell Mol Biol 2001; 24:5918.

    117. Denis M, Ghadirian E. Transforming growth factor-beta is generated

    in the course of hypersensitivity pneumonitis: contributionto collagen

    synthesis. Am J Respir Cell Mol Biol 1992; 7:15660.

    118. Piguet PF, Collart MA, Grau GE, Sappino AP, Vassalli P. Requirement

    of tumour necrosis factor for development of silica-induced pulmo-

    nary fibrosis. Nature 1990; 344:2457.

  • 7/29/2019 Remodelacion Pulmonar en Tuberculosis

    10/10

    1210 JID 2005:192 (1 October) Dheda et al.

    119. Selman M, Pardo A. The epithelial/fibroblastic pathway in the path-

    ogenesis of idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol

    2003; 29(Suppl):S937.

    120. Elias JA, Freundlich B, Kern JA, Rosenbloom J. Cytokine networks in

    the regulation of inflammation and fibrosis in the lung. Chest 1990;

    97:143945.

    121. Rider C. Many cytokines and interleukins bind to glycosaminoglycans

    [letter]. Immunol Today 1993; 14:615.

    122. Giri SN, Hyde DM, Marafino BJ Jr. Ameliorating effect of murine

    interferon gamma on bleomycin-induced lung collagen fibrosis in

    mice. Biochem Med Metab Biol 1986; 36:1947.

    123. Serpier H, Gillery P, Salmon-Ehr V, et al. Antagonistic effects of in-

    terferon-gamma and interleukin-4 on fibroblast cultures. J Invest Der-

    matol 1997; 109:15862.

    124. Wangoo A, Sparer T, Brown IN, et al. Contribution of Th1 and Th2

    cells to protection and pathology in experimental models of granu-

    lomatous lung disease. J Immunol 2001; 166:34329.

    125. Lee CG, Homer RJ, Zhu Z, et al. Interleukin-13 induces tissue fibrosis

    by selectively stimulating and activating transforming growth factor

    b1. J Exp Med 2001; 194:80921.

    126. Wynn TA, Cheever AW, Jankovic D, et al. An IL-12-based vaccination

    method for preventing fibrosis induced by schistosome infection. Na-

    ture 1995; 376:5946.

    127. Moller DR. Pulmonary fibrosis of sarcoidosis: new approaches, old

    ideas. Am J Respir Cell Mol Biol 2003; 29(Suppl):S3741.

    128. Kunkel SL, Chensue SW, Lukacs N, Hogaboam C. Cytokine phe-notypes serve as a paradigms for experimental immune-mediatedlung

    diseases and remodeling. Am J Respir Cell Mol Biol 2003; 29(Suppl):

    S636.

    129. Chensue SW, Warmington K, Ruth J, Lincoln P, Kuo MC, Kunkel

    SL. Cytokine responses during mycobacterial and schistosomal an-

    tigen-induced pulmonary granuloma formation: production of Th1

    and Th2 cytokines and relative contribution of tumor necrosis fac-

    tor. Am J Pathol 1994; 145:110513.

    130. Wynn TA, Eltoum I, Cheever AW, Lewis FA, Gause WC, Sher A.

    Analysis of cytokine mRNA expression during primary granuloma

    formation induced by eggs of Schistosoma mansoni. J Immunol 1993;

    151:143040.

    131. Gillery P, Fertin C, Nicolas JF, et al. Interleukin-4 stimulates colla-

    gen gene expression in human fibroblast monolayer cultures: poten-

    tial role in fibrosis. FEBS Lett 1992; 302:2314.132. Oriente A, Fedarko NS, Pacocha SE, Huang SK, Lichtenstein LM, Es-

    sayan DM. Interleukin-13 modulates collagen homeostasis in human

    skin and keloid fibroblasts. J Pharmacol Exp Ther 2000; 292:98894.

    133. Zhu Z, Homer RJ, Wang Z, et al. Pulmonary expression of interleu-

    kin-13 causes inflammation, mucus hypersecretion, subepithelial fi-

    brosis, physiologic abnormalities, and eotaxin production. J Clin In-

    vest 1999; 103:77988.

    134. Kaplan MH, Whitfield JR, Boros DL, Grusby MJ. Th2 cells are re-

    quired for the Schistosoma mansoni egg-induced granulomatous re-

    sponse. J Immunol 1998; 160:18506.

    135. Atamas SP, Yurovsky VV, Wise R, et al. Production of type 2 cytokines

    by CD8+ lung cells is associated with greater decline in pulmonary

    function in patients with systemic sclerosis. Arthritis Rheum 1999;

    42:116878.

    136. Seah GT, Scott GM, Rook GA. Type 2 cytokine gene activation and

    its relationship to extent of disease in patients with tuberculosis. J In-

    fect Dis 2000; 181:3859.

    137. Seah GT, Rook GA. High levels of mRNA encoding IL-4 in unstim-

    ulated peripheral blood mononuclear cells from tuberculosis patients

    revealed by quantitative nested reverse transcriptase-polymerasechain

    reaction; correlations with serum IgE levels. Scand J Infect Dis 2001;

    33:1069.

    138. Dheda K, Johnson MA, Zumla A, Rook GA. Smoking is not beneficial

    for tuberculosis [letter]. Am J Respir Crit Care Med 2004; 170:821.

    139. Hance AJ, Basset F, Saumon G, et al. Smoking and interstitial lung

    disease: the effect of cigarette smoking on the incidence of pulmonary

    histiocytosis X and sarcoidosis. Ann N Y Acad Sci 1986; 465:64356.140. Warren CP. Extrinsic allergic alveolitis: a disease commoner in non-

    smokers. Thorax 1977; 32:5679.

    141. King TE Jr, Tooze JA, Schwarz MI, Brown KR, Cherniack RM. Pre-

    dicting survival in idiopathic pulmonary fibrosis: scoring system and

    survival model. Am J Respir Crit Care Med 2001; 164:117181.

    142. King TE Jr, Schwarz MI, Brown K, et al. Idiopathic pulmonary fibro-

    sis: relationship between histopathologic features and mortality. Am

    J Respir Crit Care Med 2001; 164:102532.

    143. Arinobu Y, Atamas SP, Otsuka T, et al. Antagonistic effects of an

    alternative splice variant of human IL-4, IL-4d2, on IL-4 activities in

    human monocytes and B cells. Cell Immunol 1999; 191:1617.

    144. Atamas SP, Choi J, Yurovsky VV, White B. An alternative splice var-

    iant of human IL-4, IL-4d2, inhibits IL-4-stimulated T cell prolifer-

    ation. J Immunol 1996; 156:43541.

    145. Martino A, Sacchi A, Sanarico N, et al. Dendritic cells derived from

    BCG-infected precursors induce Th2-like immune response. J Leukoc

    Biol 2004; 76:82734.

    146. Pathan AA, Wilkinson KA, Klenerman P, et al. Direct ex vivo analysis

    of antigen-specific IFN-g-secreting CD4 T cells in Mycobacterium tu-

    berculosis-infected individuals: associations with clinical disease state

    and effect of treatment. J Immunol 2001; 167:521725.

    147. Zuany-Amorim C, Sawicka E, Manlius C, et al. Suppression of airway

    eosinophilia by killed Mycobacterium vaccae-induced allergen-specific

    regulatory T-cells. Nat Med 2002; 8:6259.

    148. Zuany-Amorim C, Manlius C, Trifilieff A, et al. Long-term protective

    and antigen-specific effect of heat-killed Mycobacterium vaccae in a

    murine model of allergic pulmonary inflammation. J Immunol 2002;

    169:14929.

    149. Johnson JL, Kamya RM, Okwera A, et al. Randomized controlledtrial

    of Mycobacterium vaccae immunotherapy in nonhuman immuno-

    deficiency virusinfected Ugandan adults with newly diagnosed pul-

    monary tuberculosis. J Infect Dis 2000; 181:130412.

    150. Luo Y, Lu S, Guo S. Immunotherapeutic effect of Mycobacterium vaccae

    on multi-drug resistant pulmonary tuberculosis [in Chinese]. Zhonghua

    Jie He He Hu Xi Za Zhi 2000; 23:858.

    151. Stanford J, Stanford C, Grange J. Immunotherapywith Mycobacterium

    vaccae in the treatment of tuberculosis. Front Biosci 2004; 9:170119.

    152. Sampaio EP, Hernandez MO, Carvalho DS, Sarno EN. Management

    of erythema nodosum leprosum by thalidomide: thalidomide ana-

    logues inhibit M. leprae-induced TNFa production in vitro. Biomed

    Pharmacother 2002; 56:139.

    153. Tramontana JM, Utaipat U, Molloy A, et al. Thalidomide treatment

    reduces tumor necrosis factor alpha production and enhances weight

    gain in patients with pulmonary tuberculosis. Mol Med 1995; 1:38497.

    154. Weiner HL. Oral tolerance: immune mechanisms and the genera-

    tion of Th3-type TGF-beta-secreting regulatory cells. Microbes Infect

    2001; 3:94754.