Literature
Home医源资料库在线期刊传染病学杂志2005年第191卷第19期

Lung Remodeling in Pulmonary Tuberculosis

来源:传染病学杂志
摘要:CentreforInfectiousDiseasesandInternationalHealth,DepartmentofThoracicandHIVMedicine,RoyalFreeHospitalDepartmentofThoracicMedicine,MiddlesexHospital,RoyalFreeandUniversityCollegeMedicalSchool,London,UnitedKingdomTuberculosisisaglobalpublichealthcatastrophere......

点击显示 收起

    Centre for Infectious Diseases and International Health, Department of Thoracic and HIV Medicine, Royal Free Hospital
    Department of Thoracic Medicine, Middlesex Hospital, Royal Free and University College Medical School, London, United Kingdom

    Tuberculosis is a global public health catastrophe responsible for >8 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 dysregulated 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 immunopathology, since they have the potential to substantially reduce spread.

    The lung remodeling associated with pulmonary tuberculosis (TB) (healed cavitation, fibrosis, and distorted architecture) has never been satisfactorily explained. This is despite TB being an international public health priority and despite cavitation, with associated tissue and liquefactive necrosis, being the mechanism by which disease transmission occurs. In addition to addressing the significant morbidity associated with lung remodeling, investigations that can elucidate mechanisms and immunological pathways relevant to tissue necrosis might lead to strategies for interrupting aerosol-mediated transmission. Such knowledge can be incorporated into immunotherapeutic strategies aimed at halting person-to-person spread of Mycobacterium tuberculosis.

    Why do caseous and liquefactive necrosis occur Moreover, why is fibrosis present in a disease characterized by a potent interferon (IFN) response, when this cytokine generally opposes fibrosis [1] In this review, we reevaluate current knowledge of the immunopathogenesis of tissue necrosis and fibrosis. Peer-reviewed data for the 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 "tuberculosis" 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 disease, "remodeling" refers to anatomical and structural changes that are not easily reversible (laying down of extracellular matrix ), 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 appropriate 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; or be a combination of these. The term "tissue necrosis" encompasses both caseous and liquefactive necrosis.

    CLINICAL ASPECTS

    Cavitation into airways, with cough-induced aerosol generation, is the principle method by which TB is spread. Untreated, there is a fatal outcome in 50% of individuals [4]. Despite chemotherapy, however, there may be widespread lung destruction with significant associated mortality [5]. Alternatively, subsequent healing can result in extensive fibrosis, traction bronchiectasis [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 significant hemoptysis [8, 11]. Poor drug penetration into cavities and fibrocaseous foci, which are immunologically "sealed off," may facilitate latency 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 completely 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 various host receptors [12, 13]. Lung  T cells, NK T cells, and granulocytes are important early arrivals that precede the second wave of expanded, IFN- and tumor necrosis factor (TNF)producing effector T cell populations [1416]. Collectively, these cells initiate a chemokine and cytokine cascade that attracts other macrophages and, later, T cells to the site of infection [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 histological 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 alternative view is that granulocytes are early arrivals that mediate 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 lymphocyte zones of the granuloma have secondary lymphoid follicles analogous to those found in lymph nodes [16].

    Macrophages and other cell types, such as fibroblasts, endothelial cells, and neutrophils, also produce proteases (metalloproteinases [collagenase, gelatinase, and stromelysin]; the lysosomal proteinases ; and the plasminogen/plasmin system and its activator, urokinase]). Such enzymes may facilitate granuloma formation by mediating antigen processing, 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 metalloproteinases (TIMPs) [26, 27].

    Within the granuloma, both T cells and macrophages secrete TNF- and lymphotoxin 3. Not only is TNF- crucial for host defense [2832], but it also facilitates the structural integrity of the granuloma by mediating the formation of the encapsulating fibrous wall [33], together with transforming growth factor (TGF) [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 [3840]. Caseous necrosiswhich occurs at 23 weeks in the rabbit model [22]and its associated low oxygen content create unfavorable conditions for mycobacterial multiplication [41]. This, together with macrophage activation and CD3+ effector cell mechanisms, culminates in mycobacterial sterilization. It was shown in 1927 that healed primary lesions are usually 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 mycobacterial containment, although this may be incomplete if there has been massive production of caseum. Proteases cleave components of the ECM [26]. Macrophages phagocytose the partly 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 progressive 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 necrotic 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 into 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 granulation tissue rich in fibroblasts, inflammatory cells, and capillaries [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 Shwartzman 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 mycobacterial infection [48, 5153], and dysregulation of protease control mechanisms is likely to mediate proteolysis of structural components of the lung [54]. Cytokines such as TNF- and interleukin (IL)1 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 predominate. Granulocytes may be important facilitators of early necrosis [16, 23]. As discussed below, local tissue damage will be more likely if the macrophages undergo necrosis rather than apoptosis. Although thought to be less important, mycobacteria themselves may produce endopeptidases [25] or other pronecrotic virulence factors. Recently, however, a polyketide toxic factor, isolated from M. ulcerans, has been shown to have cytopathic properties and produced necrotic cutaneous lesions 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 mycobacterial 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 mycobacterial virulence factors actively induce tissue-damaging cell-mediated immune responses [62, 63].

    The Koch Phenomenon and Shwartzman Reaction

    Koch [49] showed that tuberculous guinea pigs developed necrosis locally and at distant sites of infection after rechallenge with tuberculin. High tuberculin reactivity occurs in experimental animals with liquefaction [64]. Similarly, the tuberculin test is frequently necrotic in humans with TB but not in healthy bacille Calmette-Guérin (BCG)vaccinated individuals [47]. More recently, an elegant study showed that M. tuberculosisinfected mice developed increased lung inflammation and elevated TNF- levels when rechallenged with mycobacterial antigens [65]. Moreover, when prophylactic-vaccine candidates are tested for therapeutic effects in mice with TB, they develop increased immunopathology [66] and classical Koch reactions characterized by cellular necrosis within granulomata [67]. One explanation may be the systemic Shwartzman reaction [50], which referred originally to cutaneous necrosis at a site of a previous endotoxin injection after intravenous injection of lipopolysaccharide (LPS). The concept is that the initial skin inflammation causes endothelial activation and accumulation of inflammatory cells. Then, a subsequent systemic cytokine-inducing signal administered 24 h later preferentially triggers necrosis in the "prepared" skin site. Indeed, M. tuberculosisinfected 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 suggests that host cytokines and enzymes are responsible for the necrosis. Although the precise mechanisms inducing progression from controlled protease release and granuloma formation to dysregulated protease production with liquefactive necrosis are unknown, a likely facilitator is TNF-.

    TNF-.

    Although TNF- is essential for immunity to TB, in progressive disease TNF- is associated with fever and wasting [7173] and correlates with disease activity and immunopathology [29, 7477]. Cells containing M. tuberculosis are rendered exquisitely sensitive to killing by TNF- [75, 78]. As outlined above, TNF- can up-regulate metalloproteinases and urokinase and thereby facilitate proteolysis of structural lung elements. However, under what conditions is the protective TNF- 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 dependent [80]. In M. tuberculosisinfected mice, susceptibility to the toxic effects of TNF- injected into the footpads coincided temporally with the emergence of Th2 cytokines in the lungs [81]. IL-4 may also regulate TNF-mediated enteropathy in Trichinella spiralis infection [82], and necrosis in a schistosomiasis model coincides precisely with the superimposition of Th2 response on an existing Th1 pattern [83]. Mouse (BALB/c) models of TB showed high IFN- levels early during infection, 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 damage by enhancing the pathological effect of TNF- [81]. More recent evidence, from IL-4 gene knockout experiments in BALB/c mice, showed an absence of TNF-mediated toxicity after TNF- challenge in the absence of IL-4 [86]. Collectively, these murine data suggest that, under the influence of superimposed Th2 cytokines, TNF- is toxic in dominantly Th1-mediated lesions. 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 subsets in those with cavitary TB [87] and the presence of a Th1 cytokine profile in those with noncavitary disease [74]. Similarly, expression of IL-4 in peripheral blood lymphocytes correlates with cavitary disease [88].

    In other murine knockout models of immunopathology induced by mycobacteria, / T cells [89], /-positive T cells, IL-12, IFN-, and TNF- [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 logarithmic 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. tuberculosis has evolved components that exploit the host's tissue-damaging protective responses by driving a Th2 response and necrosis. It is interesting that particularly virulent Beijing strains of M. tuberculosis cause human monocytes to express IL-4 and IL-13 [94].

    Apoptosis.

    Caseation is a mixture of necrosis and apoptosis. Large numbers of apoptotic T cells and macrophages are seen in caseous foci [98]. Immunohistochemical analysis and in situ hybridization have shown that macrophages surrounding caseous foci are negative for the antiapoptotic protein bcl2 but positive for the proapoptotic protein bax, whereas the associated T cells express IFN- 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 granule-mediated pathway to induce apoptosis (as opposed to the FasFas ligand pathway) lead to killing of the contained mycobacteria too. The role of the granule contents, particularly granulysin, has been reviewed [101]. Apoptosis induced by ATP [102, 103], TNF- independent of IFN- [104], Fas ligand [105], and hydrogen peroxide [106] all promote killing of virulent M. tuberculosis within macrophages, whereas the necrotic mode of death does not [105, 107]. Moreover, M. tuberculosis has mechanisms that tend to inhibit apoptosis. Mannose-capped lipoarabinomannan (Man-LAM) stimulates phosphorylation of Bad, a proapoptotic protein, and so inhibits apoptosis [108]. Another group has shown that Man-LAM inhibits apoptosis by a mechanism involving calcium-dependent signaling [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, possible that the material that accumulates as caseum is, in fact, a product of inappropriate necrosis. Apoptotic cells are usually removed by surrounding cells with minimal inflammatory consequences, whereas necrotic cells accumulate. However, it is 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-, TGF-, and the pathogenesis of fibrosis.

    Extensive deposition of ECM occurs in the guinea pig model of TB [111]. Available data suggest that TGF-, together with TNF-, plays a key role in the formation of the fibrous wall that encapsulates the tuberculous granuloma [3335]. The importance of TGF- with respect to pulmonary fibrosis has been established in human [38, 112] and animal [113, 114] models. Although the exact signaling pathways that drive fibroblasts to lay down ECM and macrophages to dissolve ECM are poorly understood, it is reasonable to speculate that TGF- may contribute to the dysregulation of ECM turnover in TB. Indeed, TGF- may also perpetuate fibrogenesis by inhibiting apoptosis of fibroblasts [115] and mediating localized production of inhibitors of TIMP [116]. However, it seems that TGF-, although necessary, is not sufficient for fibrosis. The importance of other molecules, such as TNF-, in lung fibrosis is illustrated by models of bleomycin and hypersensitivity pneumonitis, in which antiTNF- 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 important 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 dissolution of ECM in the central part of the granuloma. Possible explanations include a cytokine gradient within the granuloma [120], including low IFN- levels in the central macrophage-dominant part of the granuloma or selective binding of cytokines to glycosaminoglycans in different parts of the granuloma [121]. Therefore, in the center of the granuloma, high TNF- concentrations, protease activity, and lysis of infected macrophages predominate, whereas, in the periphery, TGF- activity and fibroblast activation predominate.

    The second paradox is the presence of fibrosis where there are potent IFN- responses, which down-regulate TGF- 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 profibrotic and enhance collagen production by fibroblasts [123]. Mice, when infected with saprophytic mycobacteria, only develop 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, radiation-induced pulmonary fibrosis, and chronic lung allograft rejection; reviewed in [125]), there is expression of type 2 cytokines. The same is true in the bleomycin murine model [125] and in schistosomiasis, in which fibrosis and tissue remodeling do not occur if induction of type 2 cytokines by the ova is blocked by preimmunization with ova plus IL-12 [126]. Notably, 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, followed by a Th2 response that seeks to "wall off," or isolate, a persistent antigen from the host [128130]. M. tuberculosis may 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 cytokines (IL-4, IL-13, and IL-42) and reciprocally inhibited by Th1 cytokines such as IFN- and IL-12 [1, 126]. Moreover, granulomatous inflammation and fibrosis are significantly reduced 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-42 [135], activates fibroblasts and promotes collagen formation. However, IL-13 also drives expression and activation of TGF- [125]. Significantly, the increase in expression of type 2 cytokines in human TB extends to IL-13, which, like IL-4 and IL-42, is increased 80100-fold [136, 137].

    Smoking can reactivate TB, probably because nicotine switches off secretion of TNF- by macrophages, leaving IL-10 secretion unchanged [138]. Similarly, TNF-mediated fibrotic lung diseases 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 fibrosis [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-.

    IL-4 or IL-42 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-42, raising the likelihood that the CD8+ T cells making IL-4 in TB reported by van Crevel et al. might be really making IL-42 (van Crevel et al. used reagents that do not distinguish between the 2 cytokines [88]). IL-42 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 mechanism [94]. Similarly, dendritic cells derived from BCG-infected precursors can drive a Th2-like response [145]. Nevertheless, 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 protein do so [95]. Identification of the antigens or epitopes that drive IL-4 production will allow design of vaccines that suppress the unwanted Th2 component [79]. Agents like M. vaccae, which attenuate Th2 responses [147, 148], have demonstrated 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 and fibrosis are required. This could yield clinical benefit and has the potential to interrupt transmission. For instance, vaccines that target immunopathology (either host responses or bacterial virulence factors), even if they are not completely protective, may have the ability to reduce global disease burden by minimizing cavitation and interrupting transmission. Like the antiTNF- agents [152154], other immunotherapeutic modalities could target pathways relating to cytokine signaling, apoptosis, protease activity, and ECM turnover. However, such goals are tenable only if adequate funding is committed by government agencies, the pharmaceutical industry, and international charities. Moreover, there will have to be a global organizational structure committed 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. First citation in article

    2.  Caughey GH. Chairman's summary: mechanisms of airway remodeling. Am J Respir Crit Care Med 2001; 164:S267. First citation in article

    3.  Jeffery PK. Remodeling in asthma and chronic obstructive lung disease. Am J Respir Crit Care Med 2001; 164:S2838. First citation in article

    4.  Smith PG, Moss AR. Epidemiology of tuberculosis. In: Bloom BR, ed. Tuberculosis: pathogenesis, protection, and control. 1st ed. Washington DC, American Society for Microbiology, 1994:4759. First citation in article

    5.  Bobrowitz ID. The destroyed tuberculous lung. Scand J Respir Dis 1974; 55:828. First citation in article

    6.  Curtis JK. The significance of bronchiectasis associated with pulmonary tuberculosis. Am J Med 1957; 22:894903. First citation in article

    7.  Rosenzweig DY. The role of tuberculosis and other forms of bronchopulmonary necrosis in the pathogenesis of bronchiectasis. Am Rev Respir Dis 1966; 93:76985. First citation in article

    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. First citation in article

    9.  Lopez-Majano V. Ventilation and transfer of gases in pulmonary tuberculosis. Respiration 1973; 30:4863. First citation in article

    10.  Skoogh BE. Lung mechanics in pulmonary tuberculosis. I. Static lung volumes. Scand J Respir Dis 1973; 54:14856. First citation in article

    11.  Thompson JR. Mechanisms of fatal pulmonary hemorrhage in tuberculosis. Am J Surg 1955; 89:63744. First citation in article

    12.  Ernst JD. Macrophage receptors for Mycobacterium tuberculosis. Infect Immun 1998; 66:127781. First citation in article

    13.  Heldwein KA, Fenton MJ. The role of Toll-like receptors in immunity against mycobacterial infection. Microbes Infect 2002; 4:93744. First citation in article

    14.  Apostolou I, Takahama Y, Belmant C, et al. Murine natural killer T (NKT) cells [correction of natural killer cells] contribute to the granulomatous reaction caused by mycobacterial cell walls. Proc Natl Acad Sci USA 1999; 96:51416. First citation in article

    15.  Kaufmann SH. / and other unconventional T lymphocytes: what do they see and what do they do Proc Natl Acad Sci USA 1996; 93:22729. First citation in article

    16.  Ulrichs T, Kosmiadi GA, Trusov V, et al. Human tuberculous granulomas induce peripheral lymphoid follicle-like structures to orchestrate local host defence in the lung. J Pathol 2004; 204:21728. First citation in article

    17.  Ulrichs T, Kaufmann SHE. Cell-mediated immune response. In: Rom WN, Garay SM, eds. Tuberculosis. 2nd ed. Philadelphia: Lippincott Williams & Wilkins, 2004:25162. First citation in article

    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. First citation in article

    19.  Perez RL, Roman J, Staton GW Jr, Hunter RL. Extravascular coagulation 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. First citation in article

    20.  Love SH, Shannon BT, Myrvik QN, Lynn WS. Characterization of macrophage agglutinating factor as a hyaluronic acid-protein complex. J Reticuloendothel Soc 1979; 25:26982. First citation in article

    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. First citation in article

    22.  Lurie MB, Zappasodi P, Tickner C. On the nature of genetic resistance to tuberculosis in the light of the host-parasite relationships in natively resistant and susceptible rabbits. Am Rev Tuberc 1955; 72:297329. First citation in article

    23.  Seiler P, Aichele P, Bandermann S, et al. Early granuloma formation after aerosol Mycobacterium tuberculosis infection is regulated by neutrophils via CXCR3-signaling chemokines. Eur J Immunol 2003; 33:267686. First citation in article

    24.  Turner OC, Basaraba RJ, Orme IM. Immunopathogenesis of pulmonary granulomas in the guinea pig after infection with Mycobacterium tuberculosis. Infect Immun 2003; 71:86471. First citation in article

    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. First citation in article

    26.  Chapman HA Jr. Role of enzyme receptors and inhibitors in regulating proteolytic activities of macrophages. Ann N Y Acad Sci 1991; 624:8796. First citation in article

    27.  Mauviel A. Cytokine regulation of metalloproteinase gene expression. J Cell Biochem 1993; 53:28895. First citation in article

    28.  Appelberg R. Protective role of interferon gamma, tumor necrosis factor alpha and interleukin-6 in Mycobacterium tuberculosis and M. avium infections. Immunobiology 1994; 191:5205. First citation in article

    29.  Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with infliximab, a tumor necrosis factor -neutralizing agent. N Engl J Med 2001; 345:1098104. First citation in article

    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. First citation in article

    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. First citation in article

    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. First citation in article

    33.  Lukacs NW, Chensue SW, Strieter RM, Warmington K, Kunkel SL. Inflammatory granuloma formation is mediated by TNF--inducible intercellular adhesion molecule-1. J Immunol 1994; 152:58839. First citation in article

    34.  Aung H, Toossi Z, McKenna SM, et al. Expression of transforming growth factor- but not tumor necrosis factor-, interferon-, and interleukin-4 in granulomatous lung lesions in tuberculosis. Tuber Lung Dis 2000; 80:617. First citation in article

    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. First citation in article

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

    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. First citation in article

    38.  Limper AH, Colby TV, Sanders MS, Asakura S, Roche PC, DeRemee RA. Immunohistochemical localization of transforming growth factor-beta 1 in the nonnecrotizing granulomas of pulmonary sarcoidosis. Am J Respir Crit Care Med 1994; 149:197204. First citation in article

    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. First citation in article

    40.  Peyrol S, Takiya C, Cordier JF, Grimaud JA. Organization of the connective matrix of the sarcoid granuloma: evolution and cell-matrix interactions. Ann N Y Acad Sci 1986; 465:26885. First citation in article

    41.  Hemsworth GR, Kochan I. Secretion of antimycobacterial fatty acids by normal and activated macrophages. Infect Immun 1978; 19:1707. First citation in article

    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. First citation in article

    43.  Opie EL, Aronson JD. Tubercle bacilli in latent tuberculosis lesions and in lung tissue without tuberculous lesions. Arch Pathol 1927; 4:121. First citation in article

    44.  Culty M, O'Mara TE, Underhill CB, Yeager H Jr, Swartz RP. Hyaluronan receptor (CD44) expression and function in human peripheral blood monocytes and alveolar macrophages. J Leukoc Biol 1994; 56:60511. First citation in article

    45.  Parakkal PF. Involvement of macrophages in collagen resorption. J Cell Biol 1969; 41:34554. First citation in article

    46.  Werb Z, Bainton DF, Jones PA. Degradation of connective tissue matrices 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. First citation in article

    47.  Dannenburg AM, Rook GAW. Pathogenesis of pulmonary tuberculosis: an interplay of tissue-damaging and macrophage-activating immune responsesdual mechanisms that control bacillary multiplication. In: Bloom BR, ed. Tuberculosis: pathogenesis, protection, and control. 1st ed. Washington DC, American Society for Microbiology, 1994:45983. First citation in article

    48.  Dannenberg AM Jr, Sugimoto M. Liquefaction of caseous foci in tuberculosis. Am Rev Respir Dis 1976; 113:2579. First citation in article

    49.  Koch R. Fortsetzung uber ein Heilmittel gegen Tuberculose. Dtsch Med Wochenschr 1989; 17:1012. First citation in article

    50.  Rook GA, Al Attiyah R. Cytokines and the Koch phenomenon. Tubercle 1991; 72:1320. First citation in article

    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-antiperoxidase complex. Am J Pathol 1974; 74:117. First citation in article

    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 developing 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. First citation in article

    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. First citation in article

    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. Thorax 1996; 51:30611. First citation in article

    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. First citation in article

    56.  Gyetko MR, Shollenberger SB, Sitrin RG. Urokinase expression in mononuclear phagocytes: cytokine-specific modulation by interferon-gamma and tumor necrosis factor-alpha. J Leukoc Biol 1992; 51:25663. First citation in article

    57.  Gyetko MR, Wilkinson CC, Sitrin RG. Monocyte urokinase expression: modulation by interleukins. J Leukoc Biol 1993; 53:598601. First citation in article

    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. First citation in article

    59.  George KM, Pascopella L, Welty DM, Small PL. A Mycobacterium ulcerans toxin, mycolactone, causes apoptosis in guinea pig ulcers and tissue culture cells. Infect Immun 2000; 68:87783. First citation in article

    60.  Cole ST, Brosch R, Parkhill J, et al. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature 1998; 393:53744. First citation in article

    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. First citation in article

    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. First citation in article

    63.  Kaushal D, Schroeder BG, Tyagi S, et al. Reduced immunopathology and mortality despite tissue persistence in a Mycobacterium tuberculosis mutant lacking alternative sigma factor, SigH. Proc Natl Acad Sci USA 2002; 99:83305. First citation in article

    64.  Yamamura Y, Ogawa Y, Maeda H, Yamamura Y. Prevention of tuberculous cavity formation by desensitization with tuberculin-active peptide. Am Rev Respir Dis 1974; 109:594601. First citation in article

    65.  Moreira AL, Tsenova L, Aman MH, et al. Mycobacterial antigens exacerbate disease manifestations in Mycobacterium tuberculosis-infected mice. Infect Immun 2002; 70:21007. First citation in article

    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. First citation in article

    67.  Taylor JL, Turner OC, Basaraba RJ, Belisle JT, Huygen K, Orme IM. Pulmonary necrosis resulting from DNA vaccination against tuberculosis. Infect Immun 2003; 71:21928. First citation in article

    68.  Cardona PJ, Llatjos R, Gordillo S, et al. Towards a "human-like" model of tuberculosis: intranasal inoculation of LPS induces intragranulomatous lung necrosis in mice infected aerogenically with Mycobacterium tuberculosis. Scand J Immunol 2001; 53:6571. First citation in article

    69.  Roach TI, Barton CH, Chatterjee D, Blackwell JM. Macrophage activation: lipoarabinomannan from avirulent and virulent strains of Mycobacterium tuberculosis differentially induces the early genes c-fos, KC, JE, and tumor necrosis factor-. J Immunol 1993; 150:188696. First citation in article

    70.  Yamamura Y. The pathogenesis of tuberculous cavities. Bibl Tuberc 1958; 13:1337. First citation in article

    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. First citation in article

    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. First citation in article

    73.  Takashima T, Ueta C, Tsuyuguchi I, Kishimoto S. Production of tumor necrosis factor alpha by monocytes from patients with pulmonary tuberculosis. Infect Immun 1990; 58:328692. First citation in article

    74.  Condos R, Rom WN, Liu YM, Schluger NW. Local immune responses correlate with presentation and outcome in tuberculosis. Am J Respir Crit Care Med 1998; 157:72935. First citation in article

    75.  Filley EA, Bull HA, Dowd PM, Rook GA. The effect of Mycobacterium tuberculosis on the susceptibility of human cells to the stimulatory and toxic effects of tumour necrosis factor. Immunology 1992; 77:5059. First citation in article

    76.  Olobo JO, Geletu M, Demissie A, et al. Circulating TNF-, TGF-, and IL-10 in tuberculosis patients and healthy contacts. Scand J Immunol 2001; 53:8591. First citation in article

    77.  Saltini C, Colizzi V. Soluble immunological markers of disease activity in tuberculosis. Eur Respir J 1999; 14:4856. First citation in article

    78.  Filley EA, Rook GA. Effect of mycobacteria on sensitivity to the cytotoxic effects of tumor necrosis factor. Infect Immun 1991; 59:256772. First citation in article

    79.  Rook GA, Hernandez-Pando R, Dheda K, Teng SG. IL-4 in tuberculosis: implications for vaccine design. Trends Immunol 2004; 25:4838. First citation in article

    80.  Muller KM, Jaunin F, Masouye I, Saurat JH, Hauser C. Th2 cells mediate IL-4-dependent local tissue inflammation. J Immunol 1993; 150:557684. First citation in article

    81.  Hernandez-Pando R, Rook GA. The role of TNF-alpha in T-cell-mediated inflammation depends on the Th1/Th2 cytokine balance. Immunology 1994; 82:5915. First citation in article

    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. First citation in article

    83.  Grzych JM, Pearce E, Cheever A, et al. Egg deposition is the major stimulus for the production of Th2 cytokines in murine schistosomiasis mansoni. J Immunol 1991; 146:13227. First citation in article

    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 experimental pulmonary tuberculosis. Immunology 1996; 89:2633. First citation in article

    85.  Orme IM, Roberts AD, Griffin JP, Abrams JS. Cytokine secretion by CD4 T lymphocytes acquired in response to Mycobacterium tuberculosis infection. J Immunol 1993; 151:51825. First citation in article

    86.  Hernandez-Pando R, Aguilar D, Hernandez ML, Orozco H, Rook G. Pulmonary tuberculosis in BALB/c mice with non-functional IL-4 genes: changes in the inflammatory effects of TNF- and in the regulation of fibrosis. Eur J Immunol 2004; 34:17483. First citation in article

    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. First citation in article

    88.  van Crevel R, Karyadi E, Preyers F, et al. Increased production of interleukin 4 by CD4+ and CD8+ T cells from patients with tuberculosis is related to the presence of pulmonary cavities. J Infect Dis 2000; 181:11947. First citation in article

    89.  Saunders BM, Frank AA, Cooper AM, Orme IM. Role of  T cells in immunopathology of pulmonary Mycobacterium avium infection in mice. Infect Immun 1998; 66:550814. First citation in article

    90.  Ehlers S, Benini J, Held HD, Roeck C, Alber G, Uhlig S.  T cell receptor-positive cells and interferon-, but not inducible nitric oxide synthase, are critical for granuloma necrosis in a mouse model of mycobacteria-induced pulmonary immunopathology. J Exp Med 2001; 194:184759. First citation in article

    91.  Florido M, Cooper AM, Appelberg R. Immunological basis of the development of necrotic lesions following Mycobacterium avium infection. Immunology 2002; 106:590601. First citation in article

    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. First citation in article

    93.  Rook GAW, Dheda K, Zumla A. Do successful tuberculosis vaccines need to be immunoregulatory rather than merely Th1-boosting Vaccine 2005; 23:211520. First citation in article

    94.  Manca C, Reed MB, Freeman S, et al. Differential monocyte activation underlies strain-specific Mycobacterium tuberculosis pathogenesis. Infect Immun 2004; 72:55114. First citation in article

    95.  Agrewala JN, Wilkinson RJ. Differential regulation of Th1 and Th2 cells by p91110 and p2140 peptides of the 16-kD -crystallin antigen of Mycobacterium tuberculosis. Clin Exp Immunol 1998; 114:3927. First citation in article

    96.  Seah GT, Rook GA. IL-4 influences apoptosis of mycobacterium-reactive lymphocytes in the presence of TNF-. J Immunol 2001; 167:12307. First citation in article

    97.  Dheda K, Chang JS, Breen RA, et al. In vivo and in vitro studies of a novel cytokine, interleukin 42, in pulmonary tuberculosis. Am J Respir Crit Care Med 2005; 172:5018. First citation in article

    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. First citation in article

    99.  Silva CL, Lowrie DB. Identification and characterization of murine cytotoxic T cells that kill Mycobacterium tuberculosis. Infect Immun 2000; 68:326974. First citation in article

    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. First citation in article

    101.  Krensky AM. Granulysin: a novel antimicrobial peptide of cytolytic T lymphocytes and natural killer cells. Biochem Pharmacol 2000; 59:31720. First citation in article

    102.  Kusner DJ, Barton JA. ATP stimulates human macrophages to kill intracellular virulent Mycobacterium tuberculosis via calcium-dependent phagosome-lysosome fusion. J Immunol 2001; 167:330815. First citation in article

    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. Immunity 1997; 7:43344. First citation in article

    104.  Keane J, Shurtleff B, Kornfeld H. TNF-dependent BALB/c murine macrophage apoptosis following Mycobacterium tuberculosis infection inhibits bacillary growth in an IFN-gamma independent manner. Tuberculosis (Edinb) 2002; 82:5561. First citation in article

    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 Immunol 1998; 160:544854. First citation in article

    106.  Laochumroonvorapong P, Paul S, Manca C, Freedman VH, Kaplan G. Mycobacterial growth and sensitivity to H2O2 killing in human monocytes in vitro. Infect Immun 1997; 65:48507. First citation in article

    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. First citation in article

    108.  Maiti D, Bhattacharyya A, Basu J. Lipoarabinomannan from Mycobacterium tuberculosis promotes macrophage survival by phosphorylating Bad through a phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem 2001; 276:32933. First citation in article

    109.  Rojas M, Garcia LF, Nigou J, Puzo G, Olivier M. Mannosylated lipoarabinomannan antagonizes Mycobacterium tuberculosisinduced macrophage apoptosis by altering Ca+2-dependent cell signaling. J Infect Dis 2000; 182:24051. First citation in article

    110.  Wille A, Gerber A, Heimburg A, et al. Cathepsin L is involved in cathepsin D processing and regulation of apoptosis in A549 human lung epithelial cells. Biol Chem 2004; 385:66570. First citation in article

    111.  Jayasankar K, Shakila H, Umapathy KC, Ramanathan VD. Biochemical and histochemical changes pertaining to active and healed cutaneous tuberculosis. Br J Dermatol 2002; 146:97782. First citation in article

    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. First citation in article

    113.  Liu JY, Sime PJ, Wu T, et al. Transforming growth factor-1 overexpression in tumor necrosis factor- receptor knockout mice induces fibroproliferative lung disease. Am J Respir Cell Mol Biol 2001; 25:37. First citation in article

    114.  Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-mediated gene transfer of active transforming growth factor-1 induces prolonged severe fibrosis in rat lung. J Clin Invest 1997; 100:76876. First citation in article

    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. First citation in article

    116.  Ramos C, Montano M, Garcia-Alvarez J, et al. Fibroblasts from idiopathic 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. First citation in article

    117.  Denis M, Ghadirian E. Transforming growth factor-beta is generated in the course of hypersensitivity pneumonitis: contribution to collagen synthesis. Am J Respir Cell Mol Biol 1992; 7:15660. First citation in article

    118.  Piguet PF, Collart MA, Grau GE, Sappino AP, Vassalli P. Requirement of tumour necrosis factor for development of silica-induced pulmonary fibrosis. Nature 1990; 344:2457. First citation in article

    119.  Selman M, Pardo A. The epithelial/fibroblastic pathway in the pathogenesis of idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol 2003; 29(Suppl):S937. First citation in article

    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. First citation in article

    121.  Rider C. Many cytokines and interleukins bind to glycosaminoglycans . Immunol Today 1993; 14:615. First citation in article

    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. First citation in article

    123.  Serpier H, Gillery P, Salmon-Ehr V, et al. Antagonistic effects of interferon-gamma and interleukin-4 on fibroblast cultures. J Invest Dermatol 1997; 109:15862. First citation in article

    124.  Wangoo A, Sparer T, Brown IN, et al. Contribution of Th1 and Th2 cells to protection and pathology in experimental models of granulomatous lung disease. J Immunol 2001; 166:34329. First citation in article

    125.  Lee CG, Homer RJ, Zhu Z, et al. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor 1. J Exp Med 2001; 194:80921. First citation in article

    126.  Wynn TA, Cheever AW, Jankovic D, et al. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature 1995; 376:5946. First citation in article

    127.  Moller DR. Pulmonary fibrosis of sarcoidosis: new approaches, old ideas. Am J Respir Cell Mol Biol 2003; 29(Suppl):S3741. First citation in article

    128.  Kunkel SL, Chensue SW, Lukacs N, Hogaboam C. Cytokine phenotypes serve as a paradigms for experimental immune-mediated lung diseases and remodeling. Am J Respir Cell Mol Biol 2003; 29(Suppl):S636. First citation in article

    129.  Chensue SW, Warmington K, Ruth J, Lincoln P, Kuo MC, Kunkel SL. Cytokine responses during mycobacterial and schistosomal antigen-induced pulmonary granuloma formation: production of Th1 and Th2 cytokines and relative contribution of tumor necrosis factor. Am J Pathol 1994; 145:110513. First citation in article

    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. First citation in article

    131.  Gillery P, Fertin C, Nicolas JF, et al. Interleukin-4 stimulates collagen gene expression in human fibroblast monolayer cultures: potential role in fibrosis. FEBS Lett 1992; 302:2314. First citation in article

    132.  Oriente A, Fedarko NS, Pacocha SE, Huang SK, Lichtenstein LM, Essayan DM. Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther 2000; 292:98894. First citation in article

    133.  Zhu Z, Homer RJ, Wang Z, et al. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest 1999; 103:77988. First citation in article

    134.  Kaplan MH, Whitfield JR, Boros DL, Grusby MJ. Th2 cells are required for the Schistosoma mansoni egg-induced granulomatous response. J Immunol 1998; 160:18506. First citation in article

    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. First citation in article

    136.  Seah GT, Scott GM, Rook GA. Type 2 cytokine gene activation and its relationship to extent of disease in patients with tuberculosis. J Infect Dis 2000; 181:3859. First citation in article

    137.  Seah GT, Rook GA. High levels of mRNA encoding IL-4 in unstimulated peripheral blood mononuclear cells from tuberculosis patients revealed by quantitative nested reverse transcriptase-polymerase chain reaction; correlations with serum IgE levels. Scand J Infect Dis 2001; 33:1069. First citation in article

    138.  Dheda K, Johnson MA, Zumla A, Rook GA. Smoking is not beneficial for tuberculosis . Am J Respir Crit Care Med 2004; 170:821. First citation in article

    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. First citation in article

    140.  Warren CP. Extrinsic allergic alveolitis: a disease commoner in non-smokers. Thorax 1977; 32:5679. First citation in article

    141.  King TE Jr, Tooze JA, Schwarz MI, Brown KR, Cherniack RM. Predicting survival in idiopathic pulmonary fibrosis: scoring system and survival model. Am J Respir Crit Care Med 2001; 164:117181. First citation in article

    142.  King TE Jr, Schwarz MI, Brown K, et al. Idiopathic pulmonary fibrosis: relationship between histopathologic features and mortality. Am J Respir Crit Care Med 2001; 164:102532. First citation in article

    143.  Arinobu Y, Atamas SP, Otsuka T, et al. Antagonistic effects of an alternative splice variant of human IL-4, IL-42, on IL-4 activities in human monocytes and B cells. Cell Immunol 1999; 191:1617. First citation in article

    144.  Atamas SP, Choi J, Yurovsky VV, White B. An alternative splice variant of human IL-4, IL-42, inhibits IL-4-stimulated T cell proliferation. J Immunol 1996; 156:43541. First citation in article

    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. First citation in article

    146.  Pathan AA, Wilkinson KA, Klenerman P, et al. Direct ex vivo analysis of antigen-specific IFN--secreting CD4 T cells in Mycobacterium tuberculosis-infected individuals: associations with clinical disease state and effect of treatment. J Immunol 2001; 167:521725. First citation in article

    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. First citation in article

    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. First citation in article

    149.  Johnson JL, Kamya RM, Okwera A, et al. Randomized controlled trial of Mycobacterium vaccae immunotherapy in nonhuman immunodeficiency virusinfected Ugandan adults with newly diagnosed pulmonary tuberculosis. J Infect Dis 2000; 181:130412. First citation in article

    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. First citation in article

    151.  Stanford J, Stanford C, Grange J. Immunotherapy with Mycobacterium vaccae in the treatment of tuberculosis. Front Biosci 2004; 9:170119. First citation in article

    152.  Sampaio EP, Hernandez MO, Carvalho DS, Sarno EN. Management of erythema nodosum leprosum by thalidomide: thalidomide analogues inhibit M. leprae-induced TNF production in vitro. Biomed Pharmacother 2002; 56:139. First citation in article

    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. First citation in article

    154.  Weiner HL. Oral tolerance: immune mechanisms and the generation of Th3-type TGF-beta-secreting regulatory cells. Microbes Infect 2001; 3:94754. First citation in article

作者: Keertan Dheda, Helen Booth, Jim F. Huggett, Margar 2007-5-15
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具