Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Activation of an NLRP3 Inflammasome Restricts Mycobacterium kansasii Infection

  • Chang-Chieh Chen ,

    Contributed equally to this work with: Chang-Chieh Chen, Sheng-Hui Tsai

    Affiliation Green Energy and Environment Research Laboratories, Industrial Technology Research Institute, Chutung, Hsinchu, Taiwan, Republic of China

  • Sheng-Hui Tsai ,

    Contributed equally to this work with: Chang-Chieh Chen, Sheng-Hui Tsai

    Affiliation Institute of Microbiology and Immunology, School of Life Science, National Yang-Ming University, Taipei, Taiwan, Republic of China

  • Chia-Chen Lu,

    Affiliation Department of Respiratory Therapy, Fu Jen Catholic University, Taipei, Taiwan, Republic of China

  • Shiau-Ting Hu,

    Affiliations Institute of Microbiology and Immunology, School of Life Science, National Yang-Ming University, Taipei, Taiwan, Republic of China, Department of Microbiology and Immunology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China

  • Ting-Shu Wu,

    Affiliation Department of Internal Medicine, Chang Gung Memorial Hospital and Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China

  • Tsung-Teng Huang,

    Affiliations Center for Molecular and Clinical Immunology, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Department of Medical Biotechnology and Laboratory Sciences, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Laboratory of Nanomaterials, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Research Center of Bacterial Pathogenesis, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China

  • Najwane Saïd-Sadier,

    Affiliation Health Sciences Research Institute and School of Natural Sciences, University of California Merced, Merced, California, United States of America

  • David M. Ojcius,

    Affiliations Center for Molecular and Clinical Immunology, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Health Sciences Research Institute and School of Natural Sciences, University of California Merced, Merced, California, United States of America

  • Hsin-Chih Lai

    hclai@mail.cgu.edu.tw

    Affiliations Center for Molecular and Clinical Immunology, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Department of Medical Biotechnology and Laboratory Sciences, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China, Research Center of Bacterial Pathogenesis, Chang Gung University, Kweishan, Taoyuan, Taiwan, Republic of China

Abstract

Mycobacterium kansasii has emerged as an important nontuberculous mycobacterium pathogen, whose incidence and prevalence have been increasing in the last decade. M. kansasii can cause pulmonary tuberculosis clinically and radiographically indistinguishable from that caused by Mycobacterium tuberculosis infection. Unlike the widely-studied M. tuberculosis, little is known about the innate immune response against M. kansasii infection. Although inflammasome activation plays an important role in host defense against bacterial infection, its role against atypical mycobacteria remains poorly understood. In this report, the role of inflammasome activity in THP-1 macrophages against M. kansasii infection was studied. Results indicated that viable, but not heat-killed, M. kansasii induced caspase-1-dependent IL-1β secretion in macrophages. The underlying mechanism was found to be through activation of an inflammasome containing the NLR (Nod-like receptor) family member NLRP3 and the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD). Further, potassium efflux, lysosomal acidification, ROS production and cathepsin B release played a role in M. kansasii-induced inflammasome activation. Finally, the secreted IL-1β derived from caspase-1 activation was shown to restrict intracellular M. kansasii. These findings demonstrate a biological role for the NLRP3 inflammasome in host defense against M. kansasii.

Introduction

Mycobacterium kansasii is an acid-fast bacillus that has emerged as an important pathogen of the group of nontuberculous mycobacteria (NTM). It is the second most-common nontuberculous opportunistic mycobacterial infection linked with AIDS, surpassed only by Mycobacterium avium complex (MAC) [1][3]. Furthermore, M. kansasii infects both immunocompetent and immunocompromised patients [4][9].

Although geographical variability of infection exists, M. kansasii is the most common cause of NTM-induced lung diseases in the United Kingdom and Western Europe [10][14]. M. kansasii causes pulmonary infection that resembles tuberculosis clinically and radiographically and is indistinguishable from Mycobacterium tuberculosis infection [12], [13], [15]. Comorbidity diseases are frequently closely related with M. kansasii pulmonary infection, including chronic obstructive pulmonary disease, bronchiectasis, pneumonoconiosis, previous tuberculosis, or carcinoma [16], [17]. In addition, extrapulmonary infection of M. kansasii can cause gastroenteritis, lymphadenitis, osteomyelitis, synovitis, cellulitis, empyema or pericarditis [14], [18], [19]. Furthermore, disseminated M. kansasii infections also commonly occur, especially in immunocompromised patients with advanced AIDS [20], [21]. Comparatively, unlike the widely-studied M. tuberculosis, most reports on M. kansasii focus on epidemiological and clinical features of infection [22][24]. Little is known about the innate immune response against M. kansasii infection.

Macrophages represent the first line of host defense against most bacterial pathogens. Following interaction with the bacteria, macrophages initiate inflammatory responses by secreting cytokines and chemokines [25], [26]. Among these, one of the key proinflammatory cytokines for antimicrobial responses is interleukin-1β (IL-1β) [27]. Two signaling systems control the synthesis, processing and secretion of IL-1β. Pathogen-recognition receptors such as Toll-like receptors (TLRs) control synthesis of pro-IL-1β, and the nucleotide binding and oligomerization domain (NOD)-like receptors (NLRs) lead to inflammasome activation and IL-1β maturation and secretion [28].

During infection with pathogenic bacteria, assembly and activation of the inflammasome result in caspase-1 activation and IL-1β secretion, which are critical for an effective immune response [29]. To date, among the four major inflammasomes described [30], the most thoroughly characterized is the NLRP3 inflammasome, which is activated by a number of diverse stimuli, including whole pathogens, microbial components and danger signals [31]. Upon activation, NLRP3 oligomerizes and recruits the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) through pyrin domain interactions. In turn, procaspase-1 is recruited by ASC via CARD-CARD interactions, thus forming the NLRP3 inflammasome and leading to caspase-1 activation. Caspase-1 is synthesized as a 45 kDa precursor (p45) before being cleaved into 20 kDa (p20) and 10 kDa (p10) mature proteins that form a hetero-tetrameric complex that express the enzymatic activity [32]. Thus, the appearance of p20 and p10 in culture supernatants reflects caspase-1 activation [33]. Similarly, upon stimulation of a pathogen recognition receptor such as the TLRs, proinflammatory cytokine IL-1β is generated as a 31 kDa proform which is proteolytically processed to the biologically active 17 kDa form by caspase-1 [34], and then released into the extracellular space through mechanisms that remain poorly characterized [35], [36].

Previous studies have indicated that M. marinum and M. tuberculosis and their components can activate an inflammasome consisting of NLRP3 and ASC [37], [38]. In addition to live M. tuberculosis, the ESAT-6 protein from the mycobacteria has been shown to induce activation of NLRP3/ASC inflammasome maturation and release of IL-1β from THP-1 macrophages [39]. Furthermore, studies using primary macrophages demonstrated that M. marinum activates the NLRP3/ASC inflammasome in an ESX-1-dependent manner [40]. Recently, a rapidly growing NTM, M. abscessus, has been reported to activate the NLRP3/ASC inflammasome via dectin-1/Syk-dependent signaling and the cytoplasmic scaffold protein p62/SQSTM1 in human macrophages [41]. However, whether M. kansasii, a slowly growing NTM, infection could induce caspase-1 activation and IL-1β secretion via the inflammasome activation has not been reported yet. Therefore, the role of inflammasome activation and secretion of IL-1β in prevention of M. kansasii infection was addressed in this study. Using the human macrophage cell line THP-1, we demonstrated that live intracellular M. kansasii triggers the activation of the NLRP3/ASC complex, caspase-1 activation, and IL-1β secretion. We further showed that potassium efflux, lysosomal acidification, cathepsin B release and production of reactive oxygen species (ROS) are required for the activation of the inflammasome. Finally, we demonstrate a major role for the secreted IL-1β in controlling M. kansasii infection. These results demonstrate an important biological function for the NLRP3 inflammasome in host defense against M. kansasii infection.

Results

Live intracellular M. kansasii triggers caspase-1 activation and IL-1β secretion in macrophages

To determine whether M. kansasii infection could induce caspase-1 activation and IL-1β secretion, THP-1 macrophages were challenged with M. kansasii at various multiplicities of infection (MOI) for 16 h. Caspase-1 cleavage and IL-1β processing were analyzed by ELISA and immunoblotting. Compared to untreated cells, bacterial challenge resulted in a dose-dependent caspase-1 activation and IL-1β secretion (Figure 1A and B), indicating that M. kansasii infection activates caspase-1 and promotes IL-1β release. By contrast, heat-killed M. kansasii failed to induce caspase-1 activation nor IL-1β secretion (Figure 1C). As macrophages can engulf mycobacteria, whether internalization of M. kansasii is required for the processing of IL-1β and caspase-1 was examined. Cytochalasin D, an inhibitor of actin polymerization, was used to block phagocytosis before M. kansasii infection. As shown in Figure 1D, caspase-1 activation and IL-1β maturation were abolished when internalization of M. kansasii by macrophages was inhibited. To determine whether activated caspase-1 is responsible for M. kansasii induced maturation and secretion of IL-1β, macrophages were pretreated with Z-YVAD-FMK, a cell-permeable and irreversible caspase-1 inhibitor. When caspase-1 activity was inhibited, the release of mature IL-1β into supernatants was reduced in a dose dependent manner, and both IL-1β processing and caspase-1 activation were reduced (Figure 2). Thus, viable intracellular M. kansasii induce caspase-1-dependent IL-1β secretion from macrophages.

thumbnail
Figure 1. Live intracellular M. kansasii activates caspase-1 and induces IL-1β secretion in THP-1 derived macrophages.

Macrophages were infected by M. kansasii, followed by analysis of caspase-1 activation and IL-1βsecretion in culture media. (A) The amount of IL-1β secreted from infected macrophages at an MOI of 0.1–10 for 16 h. (B) The secreted mature caspase-1 and IL-1β from infected macrophages at an MOI of 0.1–25 for 16 h. (C) Macrophages were challenged with live or heat-killed M. kansasii at an MOI of 10 for 16 h. Secreted IL-1β was measured by ELISA (left panel). Caspase-1 p20 and 17-kDa IL-1β were analyzed by immunoblotting (right panel). (D) Macrophages were treated with 1 µg/ml CytoD for 60 min to block phagocytosis and then incubated with M. kansasii at an MOI of 10 in the presence or absence of CytoD. Secreted IL-1β was measured by ELISA (left panel); activated caspase-1 and mature IL-1β were detected by Western blot analysis (right panel). Cells treated with 1 µg/ml LPS for 3 h and 1 µM nigericin for 1.5 h were used as control (LPS+Nig). Values represent the mean ± standard deviations of at least three independent experiments.

https://doi.org/10.1371/journal.pone.0036292.g001

thumbnail
Figure 2. Release of mature IL-1β by M. kansasii-infected cells requires caspase-1 activation.

Macrophages were infected with M. kansasii in the presence or absence of the caspase-1 inhibitor, Z-YVAD-FMK. At 16 h post-infection, secreted IL-1βwas quantified by ELISA (A), and secreted caspase-1 p20 and mature IL-1β were analyzed by Western blot analysis (B). Error bars represent standard deviations from at least three independent experiments. ** denote a p value of <0.001 compared to untreated infected cells.

https://doi.org/10.1371/journal.pone.0036292.g002

The NLRP3/ASC inflammasome contributes to caspase-1 activation and IL-1β secretion during M. kansasii infection

To define the role of NLRP3 inflammasome components in activation of caspase-1 by M. kansasii infection, NLRP3 or ASC in THP-1 cells were depleted by shRNA. Compared to non-target shRNA control, mRNA and protein levels of NLRP3 or ASC in the respective knockdown cells were significantly reduced (Figure 3A). NLRP3, ASC, or nontarget control knockdown THP-1 cells were then challenged with M. kansasii at an MOI of 10 for 16 h, and caspase-1 activation was revealed by the appearance of caspase-1 p20 and IL-1β p17 in Western blots. When compared to non-target control cells, depletion of either NLRP3 or ASC caused a significant reduction in the amount of secreted IL-1β(Figure 3B), while IL-6 production was unimpaired (Figure S1). Concomitantly, significant reduction of caspase-1 p20 levels was also observed (Figure 3C). Taken together, the results show that NLRP3 and ASC are required for M. kansasii induced caspase-1 activation and IL-1β processing in macrophages.

thumbnail
Figure 3. M. kansasii induces caspase-1 activation and IL-1β secretion through the NLRP3/ASC-dependent pathway.

(A) THP-1 cells were stably transfected with shRNAs that target NLRP3 or ASC, and mRNA expression of NLRP3 and ASC was determined by RT-PCR and compared with nontarget control (sh Ctrl) (left panel). Protein levels of NLRP3 or ASC in the respective knockdown cells were analyzed by Western blot analysis (right panel). (B and C) NLRP3, ASC, or nontarget control (sh Ctrl) knockdown cells were infected with M. kansasii at an MOI of 10 for 16 h. Secreted IL-1β(B) and activated caspase-1 (C) were examined by Western blot analysis.

https://doi.org/10.1371/journal.pone.0036292.g003

Potassium efflux, lysosomal acidification, ROS production and cathepsin B release are involved in M. kansasii-induced inflammasome activation

Previous studies have reported a number of signaling mechanisms playing important roles in activating the NLRP3/ASC inflammasome, including potassium efflux, lysosomal acidification, generation of reactive oxygen species (ROS) and cathepsin B release from lysosomes. To explore whether any of these factors are involved in M. kansasii-induced inflammasome activation, a series of defined inhibitors were used to treat M. kansasii-challenged THP-1 macrophages. The amount of secreted IL-1β in culture supernatants was used as an indicator of inflammasome activation. A high extracellular potassium concentration (130 mM) was first used to test if inhibiting K+ efflux can influence M. kansasii-mediated activation of the NLRP3/ASC inflammasome. Following infection of THP-1 macrophages in high extracellular [K+], M. kansasii-induced IL-1β release was significantly inhibited (Figure 4A). Moreover, glibenclamide, a selective inhibitor for ATP-dependent potassium channels, was used to block potassium efflux. Addition of 50 µM glibenclamide to macrophages prior to bacterial challenge significantly reduced IL-1β secretion (Figure 4A). Since ROS generation was reported to activate the NLRP3/ASC inflammasome [42], the anti-oxidant, N-acetyl cysteine (NAC), was used to determine whether ROS are involved in M. kansasii-mediated NLRP3/ASC inflammasome activation. IL-1β secretion was significantly reduced upon treatment with 25 µM NAC (Figure 4B), indicating that ROS also contributes to NLRP3/ASC inflammasome activation during M. kansasii infection. As the NLRP3 inflammasome can be activated due to lysosomal damage and release of cathepsin B, ammonium chloride (NH4Cl) and chloroquine diphosphate (CQ), which inhibit endosomal-lysosomal system acidification, and CA-074-Me, which acts as a cell-permeable inhibitor of thiol proteases, were used to block lysosomal acidification and cathepsin B activity, respectively. The three inhibitors resulted in a significant reduction in IL-1β release after M. kansasii challenge (Figure 4C), suggesting a role for lysosomes in NLRP3/ASC inflammasome activation in response to M. kansasii infection. However, these marked inhibitory effects were not due to cytotoxicity from these treatments (Fig S2). Thus, the results suggest that NLRP3/ASC activation during M. kansasii infection involves most of the pathways known to activate the NLRP3 inflammasome: potassium efflux, lysosomal acidification, cathepsin B release, and ROS production.

thumbnail
Figure 4. Potassium efflux, lysosomal acidification, cathepsin B release and ROS production are involved in inflammasome activation by M. kansasii.

(A) Macrophages were treated with KCl and Glibenclamide (Gliben) at the indicated concentration for 30 min, and subsequently infected with M. kansasii at an MOI of 10 for 16 h. Supernatants from infected cells were harvested and assayed for IL-1β by ELISA. (B and C) THP-1 derived macrophages were infected with M. kansasii at an MOI of 10. N-acetyl cysteine (NAC), NH4Cl, chloroquine (CQ) and CA-074-Me (CA-074) were added at the indicated concentration as described in Materials and Methods. Supernatants harvested at 16 h post-infection were assayed for IL-1βsecretion by ELISA. Error bars represent standard deviations of at least three independent experiments, and significance was calculated using a two-tailed t test. ** denote a p value of <0.001 compared to untreated infected cells.

https://doi.org/10.1371/journal.pone.0036292.g004

IL-1β derived from caspase-1 activation restricts M. kansasii infection

Caspase-1 activation has been reported to restrict bacterial infection either directly, by affecting bacterial growth, or indirectly, through IL-1β-mediated inhibition of infection [38], [39], [43][46]. To address the contribution of caspase-1 in the control of M. kansasii infection, macrophages were treated with caspase-1 inhibitor, Z-YVAD-FMK, before M. kansasii challenge. Intracellular bacterial growth was evaluated by quantifying the colony forming units (CFUs) for 96 h following bacterial infection. The number of CFUs recovered from macrophages treated with caspase-1 inhibitor was greater than those from untreated cells, indicating that caspase-1 activation contributes to control of M. kansasii infection in macrophages (Figure 5A).

thumbnail
Figure 5. Caspase-1 activation and IL-1β secretion restrict M. kansasii growth.

(A) THP-1 macrophages were infected with M. kansasii at an MOI of 1 for 1 h, before treatment with 50 µM caspase-1 inhibitor (Z-YVAD-FMK) or dimethylsulfoxide (DMSO) alone as control. Cells were then lysed and the intracellular bacterial load was quantified at indicated time points. (B) The M. kansasii-infected macrophages (MOI 1, 1 h) were treated with neutralizing antibodies specific for IL-1βor with exogenous IL-1β for 48 h. The intracellular bacterial CFU was then determined. Results represent the mean ± standard deviations of three independent experiments. Data were analyzed by Student's t test. *p<0.05 compared to untreated infected cells.

https://doi.org/10.1371/journal.pone.0036292.g005

Studies from knockout mice with deficiency in either IL-1β or its receptor, IL-1R, suggested that IL-1β plays an important role in the immune response to M. tuberculosis or M. kansasii infection in mice [47][49]. Whether IL-1β also affects M. kansasii infection of human macrophages in vitro was next investigated. THP-1 macrophages were treated with IL-1β neutralizing antibody during bacterial infection. The intracellular bacterial number was evaluated by quantifying the CFUs at 48 h post-infection. Blocking IL-1β signaling with IL-1β specific neutralizing antibody resulted in a nearly 5-fold increase in the number of intracellular M. kansasii (Figure 5B). Conversely, exogenously-added IL-1βsignificantly reduced the bacterial counts in macrophages. These results suggest that IL-1β secreted due to NLRP3 inflammasome activation is critical for the control of intracellular M. kansasii, and that caspase-1 most likely affects infection indirectly, through processing of IL-1β.

Discussion

In this report, we demonstrated that live and intracellular M. kansasii can trigger caspase-1 activation and IL-1β secretion from human macrophages by activating the NLRP3/ASC inflammasome. Heat-killed and extracellular M. kansasii failed to activate the NLRP3/ASC inflammasome, suggesting that simple surface contact of M. kansasii with macrophages is not sufficient for activation of the inflammasome. Although the precise mechanisms behind responsible for NLRP3/ASC inflammasome activation are still under investigation, our results suggest that M. kansasii infection of macrophages can induce potassium efflux, cathepsin B release and ROS production, all of which are involved in NLRP3/ASC inflammasome activation. Additionally, the results from the intracellular survival assay revealed that M. kansasii-induced caspase-1 activation and subsequent IL-1β secretion restrict growth of intracellular M. kansasii. This is the first report showing inflammasome activation induced by M. kansasii and demonstrating a biological function for the NLRP3/ASC inflammasome in host defense against this slow-growing NTM.

M. kansasii can cause a pneumonia that resembles classical lung tuberculosis in many features, reflecting similarities in the pathogenesis between M. kansasii and M. tuberculosis infection; however, several reports have also referred to differences in host and cellular responses to M. kansasii and M. tuberculosis. During infection by M. tuberculosis or M. avium complex which is also a pathogenic NTM, CD4+ T cells, interferon (IFN)-γ, or IL-12p40 are crucial for the development of protective immunity in mice [50][57]. However, CD4+ and IFN-γ deficient mice display normal resistance against pulmonary infection with M. kansasii, indicating that a T helper cell type 1 response is not sufficient for control of M. kansasii infection [58]. Since macrophages are the first line of defense against pathogens such as mycobacteria, and following phagocytosis, Mycobacterium species reside and multiply in macrophages, we characterized the ability of macrophages to control M. kansasii infection in vitro. Recent studies have demonstrated that the pathogenic mycobacteria, M. tuberculosis, M. marinum and M. abscessus, induce activation of the NLRP3/ASC inflammasome and subsequent release of IL-1βin macrophages [37], [39][41], [59], [60]. Here, we found that macrophages infected by M. kansasii also secrete IL-1β via activation of the NLRP3 inflammasome, suggesting that all pathogenic mycobacteria may stimulate inflammasome activation and IL-1β secretion.

The mechanisms leading to NLRP3 inflammasome activation comprise mainly three signaling pathways, including potassium efflux, cathepsin B release, and generation of ROS [30]. The current results showed that ROS production, potassium efflux, lysosomal acidification, and cathepsin B release are all required for M. kansasii induced NLRP3-dependent caspase-1 activation and IL-1β secretion. In this study, a potent cathepsin B inhibitor, CA-074-Me, was used to determine the involvement of cathepsin B in NLRP3 inflammasome activation during M. kansasii infection. However, CA-074-Me has been proposed to act on other cellular proteases [61], suggesting the possibility that CA-074-Me may inhibit M. kansasii-induced IL-1β secretion through a cathepsin B-independent manner. Even so, a recent study with cathepsin B knockdown cells confirmed that cathepsin B is involved in inflammasome activation upon mycobacteria infection [62].

It has been shown that infection by M. tuberculosis and M. marinum may induce ESX-1-dependent NLRP3 inflammasome activation [37], [39], [40], [60]. ESX-1 has been identified as a critical virulence factor in pathogenic mycobacteria and is involved in immune signaling, cytolysis, phagosome escape and membrane pore formation [63][67]. As M. kansasii also expresses ESX-1 [68], it seems reasonable to assume that pore formation of cell membranes by M. kansasii ESX-1 could cause potassium efflux and subsequent activation of the NLRP3 inflammasome. In addition, ESX-1-induced vacuole escape could lead to lysosomal damage and cathepsin B release, which have been implicated as potential activators of the NLRP3 inflammasome [63], [66], [67]. Recently, ESAT-6, one of the secreted effectors of ESX-1, has been shown to be a potent activator of the NLRP3/ASC inflammasome, possibly due to its membrane-lysing activity [39]. Genetic analysis and the nucleotide sequences revealed that the virulence genes, esx-1 and esat-6 of M. tuberculosis, are lacking in most environmental mycobacteria except for M. kansasii and M. marinum [69][71], both of which can cause disease in apparently immunocompetent persons. Moreover, analysis of immunoblotting with specific antibodies indicated that the ESAT-6 was expressed by M. kansasii [68], [71]. Thus, we propose that the activation of the NLRP3 inflammasome by live intracellular M. kansasii might be through ESX-1 or ESAT-6.

Although the role of ROS in the activation of NLRP3 inflammasome is controversial [72][78], several studies have demonstrated that ROS are required for inflammasome activation during bacterial infections [42], [79][82]. In this study, NAC significantly diminished IL-1β secretion triggered by mycobacterial infection, suggesting that ROS are involved in M. kansasii-induced NLRP3 inflammasome activation; however the source of ROS is currently unknown. The intracellular ROS are mainly generated from two sources: the mitochondrial electron transport chain complex, and NADPH oxidase at the plasma membrane or phagosomal membrane of phagocytes [45], [83][85]. It has been shown that M. tuberculosis infection leads to intracellular ROS production via NADPH oxidase at phagosomal membranes, and ESAT-6 treatment induces a robust burst of intracellular ROS production in human alveolar epithelial cells [86], [87]. Moreover, a low intracellular K+ concentration has been proposed to trigger ROS generation [73], indicating the possibility that ESAT-6, a pore-forming protein, could initiate potassium efflux and subsequently ROS production. Recent studies indicated that besides NADPH oxidase, NLRX1, a member of the Nod-like receptor (NLR) family that is localized in mitochondria, can enhance ROS production following infections by Shigella flexneri and Chlamydia trachomatis infection [80], [88]. Moreover, mitochondrial dysfunction-derived ROS has been shown to activate NLRP3 inflammasome [89], [90]. Thus, whether ESAT-6, NADPH oxidase, NLRX1 or mitochondrial dysfunction are involved in M. kansasii-induced ROS production remains to be determined.

Caspase-1, known as an inflammatory caspase, plays a key role in the innate immune response of macrophages to various infections [91][93]. Activation of caspase-1 is responsible for the processing and secretion of the proinflammatory cytokines, IL-1β and IL-18 [34], [94], [95]. In addition, active caspase-1 mediates either cell death or survival, and regulates unconventional secretion of leaderless proteins [33]. Even so, much remains to be learned regarding the role of caspase-1 activation in the control of bacterial infection. In cervical epithelial cells infected by C. trachomatis, caspase-1activation contributes to the development of chlamydial infection [81], but caspase-1-dependent caspase-7 activation restricts Legionella pneumophila replication in macrophages and in mice [96]. In mycobacterial infection, overexpression of caspase-1 represses M. tuberculosis growth in THP-1 macrophages [39]. In this study, inhibition of caspase-1 activity by Z-YVAD-FMK resulted in higher bacterial growth, suggesting that caspase-1 activity is required for restriction of M. kansasii growth in macrophages. Secretion of IL-1β is downstream from caspase-1 activation, and IL-1 has been implicated in controlling a variety of intracellular pathogens such as Listeria, Leishmania, and M. tuberculosis [50], [51], [97][100]. Our result that blocking IL-1β with IL-1β specific neutralizing antibody led to significantly higher levels of intracellular M. kansasii, and that treatment of infected cells with IL-1β reduced the bacterial load, suggested that caspase-1-dependent IL-1β secretion is critical for control of M. kansasii infection. The secreted IL-1β might be sensed by IL-1R. Possibly, IL-1R signaling promotes phagolysosomal maturation, which enhances bacterial degradation and clearance, as reported by Master et al. [38]. Concordantly, a previous study with IL-1R1 knockout mice demonstrated that blocking of IL-1-mediated signaling reduced the ability to clear M. kansasii from the lungs of IL-1R1 deficient mice [48]. These results highlighted the important role of IL-1βand IL-1R signaling pathway in defence against M. kansasii infection, and provided another evidence for the protective role of IL-1β in mycobacterial infection.

IL-1β signaling has been reported to play an important role in the control of mycobacterial infection and granuloma formation [38], [39], [47][49], [99][103]. In humans, IL-1β is upregulated at the site of mycobacterial infection, and genetic studies demonstrated an association of polymorphisms in the IL-1 or IL-1R genes with tuberculosis susceptibility and disease expression [104][107]. Notably, higher levels of IL-1β and NLRP3 mRNA were observed in monocyte-derived macrophage from active tuberculosis patients as compared with healthy subjects [39], suggesting the involvement of NLRP3 inflammasome in human response to mycobacterial infection. Consistent with our results, the involvement of NLRP3/ASC in controlling mycobacterial infection in vitro has been reported [41]. However, recent studies using NLRP3-, ASC-, or caspase-1-deficient mice demonstrated that NLRP3/ASC inflammasome is not essential for the control of M. tuberculosis infection in vivo [108][110], although it cannot be excluded that potential compensatory mechanisms can overcome NLRP3/ASC inflammasome dependence in these deficient mice [110]. Thus, the role of NLRP3 inflammasome for antimycobacterial response seems to be controversial. A recent study demonstrated that NLRP3 inflammasome activation is disparate between human and mouse during Francisella infection [111], [112], indicating that the human innate response to intracellular pathogens may be distinct from the murine response. Accordingly, the exact role of NLRP3 inflammasome for the control of mycobacterial infection should be carefully evaluated. Furthermore, the protective role of NLRP3 inflammasome against M. kansasii infection in vivo will be clarified in future studies.

In conclusion, this report demonstrates that the NLRP3 inflammasome was activated by live intracellular M. kansasii through a process involving low intracellular potassium concentration, higher ROS, and active cathepsin B. As a consequence, activated caspase-1 by inflammasome activation triggers the processing and release of IL-1β,which is required for macrophage immunity against M. kansasii infection (Figure 6).

thumbnail
Figure 6. NLRP3/ASC inflammasome activation restricts Mycobacterium kansasii infection.

During M. kansasii infection, live and intracellular bacteria trigger potassium efflux, ROS production and lysosomal damage with cathepsin B release (1), and then lead to NLRP3/ASC inflammasome activation, resulting caspase-1 activation and IL-1βsecretion (2). IL-1β derived from inflammasome activation is released and followed by the engagement of its receptor (IL-1R) (3). IL-1R signaling promotes phagosome maturation that ultimately leads to phagolysosome fusion and bacterial degradation (4).

https://doi.org/10.1371/journal.pone.0036292.g006

Materials and Methods

Cells, Bacteria, and Chemical Reagents

THP-1 cells, a human acute monocytic leukemia cell line, were obtained from American Type Culture Collection (ATCC) and were cultured in RPMI 1640 complete medium (Invitrogen) with 10% heat-inactivated fetal bovine serum (HyClone) and 1× antibiotic-antimycotic (Invitrogen) at 37°C with 5% CO2 in a humidified incubator. THP-1 stably expressing shRNA against NLRP3, ASC, and nontarget control were obtained as previously described [113], [114]. The M. kansasii strain (ATCC12478) was obtained from ATCC and grown at 35°C on Middlebrook 7H11 agar medium (Difco Laboratories) supplemented with 10% OADC (oleic acid, albumin, dextrose, catalase; Becton Dickinson). Heat-killed bacteria were prepared by incubation for 30 min at 80°C, and loss of viability was confirmed by plating on 7H11 plates [115]. Phorbol 12-myristate 13-acetate (PMA), glibenclamide, bafilomycin A1, and Z-YVAD-FMK were purchased from Enzo Life Sciences. LPS (Escherichia coli serotype O111:B4), nigericin, N-acetyl-L-cysteine (NAC), cytochalasin D, chloroquine diphosphate, potassium chloride (KCl), ammonium chloride (NH4Cl) and dimethyl sulfoxide (DMSO) were from Sigma-Aldrich. The cathepsin B inhibitor, CA-074-Me, was from Calbiochem. The cytotoxicity of reagents used for inhibition studies were evaluated by using the CytoTox 96 Non-Radioactive Cytotoxicity Assay according to the manufacturer's instructions (Promega) (Figure S2).

Bacterial infection of macrophages

THP-1 cells were differentiated into adherent macrophages by overnight culture in complete medium supplemented with 500 ng/ml PMA, and allowed to rest for 2 days prior to infection. THP-1 derived macrophages were challenged with bacterial suspensions prepared in supplemented medium without antibiotics at the indicated multiplicity of infection (MOI). When using inhibitors or other reagents, cells were preincubated 60 min with inhibitors or other reagents at the indicated concentrations before bacterial infection.

Cytokine measurement by enzyme-linked immunosorbent assay (ELISA)

To determine IL-1β levels in supernatants from M. kansasii-infected cells, the DuoSet ELISA development system kit (R&D Systems) for human IL-1β was used according to the manufacturer's directions. ELISA plates were analyzed using an Emax Microplate Reader (Molecular Devices) at 450 nm.

Western blotting

Cell culture supernatants from infected macrophages were resolved on 12% SDS-polyacrylamide gels and electrotransferred onto the polyvinylidene difluoride membranes (Millipore). For detection of the active caspase-1 subunit (p20), the membranes were probed with 1∶1000 diluted rabbit anti-human caspase-1 antibody (Millipore) and 1∶10000 diluted horseradish peroxidase-conjugated anti-rabbit IgG antibodies (Santa Cruz Biotechnology). For detection of pro-IL1β and mature IL-1β (p17), the blot was probed with 1∶1000 rabbit anti-human IL-1β antibody (Santa Cruz Biotechnology) and cleaved IL-1β antibody (Cell Signaling), respectively. To detect NLRP3 and ASC, the blots were probed with 1∶1000 rabbit anti-human NLRP3 antibody (Sigma) and mouse anti-human ASC antibody (Santa Cruz Biotechnology), respectively. The signals on the blots were visualized using the enhanced chemiluminescence system (Millipore).

RNA isolation and PCR

Total RNA was isolated using the Total RNA Mini Kit (Geneaid), reverse transcribed into cDNA (Superscript III, Invitrogen) and analyzed for NLRP3 and ASC mRNA expression by RT-PCR using the following primer pairs. The primers for human GAPDH were 5′-AACGGATTTGGTCGTATTGGGC-3′ forward and 5′-CTTGACGGTGCCATGGAATTTG-3′ reverse. Primers for human NLRP3 were 5′-CTTCTCTGATGAGGCCCAAG-3′ forward and 5′-GCAGCAAACTGGAAAGGAAG-3′ reverse. Primers for human ASC were 5′-ATCCAGGCCCCTCCTCAGT-3′ forward and 5′-GTTTGTGACCCTCGCGATAAG-3′ reverse.

Evaluation of intracellular bacterial viability by the Colony Forming Unit assay

THP-1 cells (5×105 cells/well) were added to 24-well plates and differentiated into macrophages with PMA. Monolayers of macrophages were infected by M. kansasii at an MOI of 1. After 1 h, the medium was removed and the wells were washed with serum-free medium to remove extracellular bacteria and then fresh medium was added. The infected cells were further incubated at 37°C for the indicated time. Some of the infected cells were treated with caspase-1 inhibitor, IL-1β neutralizing antibody (R&D Systems) or exogenous IL-1β (R&D Systems) for different time intervals as described. To evaluate the intracellular bacterial load, cells were lysed with 500 µl of sterile water with 0.1% triton X-100, and the number of viable intracellular bacteria was counted by plating serial dilutions of the lysis solution onto Middlebrook 7H11 agar plates.

Statistical analysis

All experiments were performed at least three times, and the results are presented as the mean ± standard deviation (SD). Statistical comparisons were performed using Student's t test.

Supporting Information

Figure S1.

IL-6 production is unimpaired in NLRP3 or ASC knockdown cells. To determine whether the ability to generate pro-IL-1β in response to LPS is diminished in NLRP3 or ASC knockdown cells. ASC, NLRP3, or nontarget control (sh Ctrl) knockdown cells were treated with 1 µg/ml LPS or M. kansasii at an MOI of 10. IL-6 in supernatant was measured by ELISA (R&D Systems). Values represent the mean ± standard deviations of at least three independent experiments. These results indicated that ASC and NLRP3 knockdown cells can produce IL-6 normally in response to LPS or M. kansasii.

https://doi.org/10.1371/journal.pone.0036292.s001

(TIF)

Figure S2.

No apparent cytotoxic effects of inhibitors on THP-1 cells in the experimental conditions. To evaluate cytotoxic effects of inhibitors used in this study, THP-1 derived macrophages were treated with the indicated pharmacological inhibitors. Cytotoxicity was quantitated by measurement of lactate dehydrogenase (LDH) activity in the culture supernatants using a CytoTox 96 assay kit (Promega) according to the manufacturer's protocol. Error bars represent standard deviation of at least three independent experiments. These results indicated that the experimental treatments have no apparent cytotoxic effects.

https://doi.org/10.1371/journal.pone.0036292.s002

(TIF)

Author Contributions

Conceived and designed the experiments: CCC DMO HCL. Performed the experiments: CCC SHT. Analyzed the data: CCC SHT DMO HCL. Contributed reagents/materials/analysis tools: CCL STH TSW TTH NSS. Wrote the paper: CCC SHT DMO HCL.

References

  1. 1. Tsukamura M, Kita N, Shimoide H, Arakawa H, Kuze A (1988) Studies on the epidemiology of nontuberculous mycobacteriosis in Japan. Am Rev Respir Dis 137: 1280–1284.
  2. 2. Lillo M, Orengo S, Cernoch P, Harris RL (1990) Pulmonary and disseminated infection due to Mycobacterium kansasii: a decade of experience. Rev Infect Dis 12: 760–767.
  3. 3. Wallace RJ Jr, Zhang Y, Brown BA, Dawson D, Murphy DT, et al. (1998) Polyclonal Mycobacterium avium complex infections in patients with nodular bronchiectasis. Am J Respir Crit Care Med 158: 1235–1244.
  4. 4. Shafer RW, Sierra MF (1992) Mycobacterium xenopi, Mycobacterium fortuitum, Mycobacterium kansasii, and other nontuberculous mycobacteria in an area of endemicity for AIDS. Clin Infect Dis 15: 161–162.
  5. 5. Witzig RS, Fazal BA, Mera RM, Mushatt DM, Dejace PM, et al. (1995) Clinical manifestations and implications of coinfection with Mycobacterium kansasii and human immunodeficiency virus type 1. Clin Infect Dis 21: 77–85.
  6. 6. Bittner MJ, Horowitz EA, Safranek TJ, Preheim LC (1996) Emergence of Mycobacterium kansasii as the leading mycobacterial pathogen isolated over a 20-year period at a midwestern Veterans Affairs hospital. Clin Infect Dis 22: 1109–1110.
  7. 7. Corbett EL, Blumberg L, Churchyard GJ, Moloi N, Mallory K, et al. (1999) Nontuberculous mycobacteria: defining disease in a prospective cohort of South African miners. Am J Respir Crit Care Med 160: 15–21.
  8. 8. Bartralot R, Pujol RM, Garcia-Patos V, Sitjas D, Martin-Casabona N, et al. (2000) Cutaneous infections due to nontuberculous mycobacteria: histopathological review of 28 cases. Comparative study between lesions observed in immunosuppressed patients and normal hosts. J Cutan Pathol 27: 124–129.
  9. 9. Arend SM, Cerda de Palou E, de Haas P, Janssen R, Hoeve MA, et al. (2004) Pneumonia caused by Mycobacterium kansasii in a series of patients without recognised immune defect. Clin Microbiol Infect 10: 738–748.
  10. 10. Wolinsky E (1979) Nontuberculous mycobacteria and associated diseases. Am Rev Respir Dis 119: 107–159.
  11. 11. Buckner CB, Leithiser RE, Walker CW, Allison JW (1991) The changing epidemiology of tuberculosis and other mycobacterial infections in the United States: implications for the radiologist. AJR Am J Roentgenol 156: 255–264.
  12. 12. Evans SA, Colville A, Evans AJ, Crisp AJ, Johnston ID (1996) Pulmonary Mycobacterium kansasii infection: comparison of the clinical features, treatment and outcome with pulmonary tuberculosis. Thorax 51: 1248–1252.
  13. 13. Evans AJ, Crisp AJ, Hubbard RB, Colville A, Evans SA, et al. (1996) Pulmonary Mycobacterium kansasii infection: comparison of radiological appearances with pulmonary tuberculosis. Thorax 51: 1243–1247.
  14. 14. Bloch KC, Zwerling L, Pletcher MJ, Hahn JA, Gerberding JL, et al. (1998) Incidence and clinical implications of isolation of Mycobacterium kansasii: results of a 5-year, population-based study. Ann Intern Med 129: 698–704.
  15. 15. Gadkowski LB, Stout JE (2008) Cavitary pulmonary disease. Clin Microbiol Rev 21: 305–333. table of contents.
  16. 16. Shitrit D, Priess R, Peled N, Bishara G, Shlomi D, et al. (2007) Differentiation of Mycobacterium kansasii infection from Mycobacterium tuberculosis infection: comparison of clinical features, radiological appearance, and outcome. Eur J Clin Microbiol Infect Dis 26: 679–684.
  17. 17. Wu TS, Leu HS, Chiu CH, Lee MH, Chiang PC, et al. (2009) Clinical manifestations, antibiotic susceptibility and molecular analysis of Mycobacterium kansasii isolates from a university hospital in Taiwan. J Antimicrob Chemother 64: 511–514.
  18. 18. Pintado V, Fortun J, Casado JL, Gomez-Mampaso E (2001) Mycobacterium kansasii pericarditis as a presentation of AIDS. Infection 29: 48–50.
  19. 19. Paull DE, Decker GR, Brown RL (2003) Mycobacterium kansasii empyema in a renal transplant recipient case report and review of the literature. Transplantation 76: 270–271.
  20. 20. Sherer R, Sable R, Sonnenberg M, Cooper S, Spencer P, et al. (1986) Disseminated infection with Mycobacterium kansasii in the acquired immunodeficiency syndrome. Ann Intern Med 105: 710–712.
  21. 21. Valainis GT, Cardona LM, Greer DL (1991) The spectrum of Mycobacterium kansasii disease associated with HIV-1 infected patients. J Acquir Immune Defic Syndr 4: 516–520.
  22. 22. Taillard C, Greub G, Weber R, Pfyffer GE, Bodmer T, et al. (2003) Clinical implications of Mycobacterium kansasii species heterogeneity: Swiss National Survey. J Clin Microbiol 41: 1240–1244.
  23. 23. Field SK, Cowie RL (2006) Lung disease due to the more common nontuberculous mycobacteria. Chest 129: 1653–1672.
  24. 24. Koh WJ, Yu CM, Suh GY, Chung MP, Kim H, et al. (2006) Pulmonary TB and NTM lung disease: comparison of characteristics in patients with AFB smear-positive sputum. Int J Tuberc Lung Dis 10: 1001–1007.
  25. 25. Delbridge LM, O'Riordan MX (2007) Innate recognition of intracellular bacteria. Curr Opin Immunol 19: 10–16.
  26. 26. Medzhitov R (2010) Inflammation 2010: new adventures of an old flame. Cell 140: 771–776.
  27. 27. van de Veerdonk FL, Netea MG, Dinarello CA, Joosten LA (2011) Inflammasome activation and IL-1beta and IL-18 processing during infection. Trends Immunol 32: 110–116.
  28. 28. Meylan E, Tschopp J, Karin M (2006) Intracellular pattern recognition receptors in the host response. Nature 442: 39–44.
  29. 29. Sutterwala FS, Mijares LA, Li L, Ogura Y, Kazmierczak BI, et al. (2007) Immune recognition of Pseudomonas aeruginosa mediated by the IPAF/NLRC4 inflammasome. J Exp Med 204: 3235–3245.
  30. 30. Schroder K, Tschopp J (2010) The inflammasomes. Cell 140: 821–832.
  31. 31. Jin C, Flavell RA (2010) Molecular mechanism of NLRP3 inflammasome activation. J Clin Immunol 30: 628–631.
  32. 32. Wilson KP, Black JA, Thomson JA, Kim EE, Griffith JP, et al. (1994) Structure and mechanism of interleukin-1 beta converting enzyme. Nature 370: 270–275.
  33. 33. Keller M, Ruegg A, Werner S, Beer HD (2008) Active caspase-1 is a regulator of unconventional protein secretion. Cell 132: 818–831.
  34. 34. Fantuzzi G, Dinarello CA (1999) Interleukin-18 and interleukin-1 beta: two cytokine substrates for ICE (caspase-1). J Clin Immunol 19: 1–11.
  35. 35. Singer , Scott S, Chin J, Bayne EK, Limjuco G, et al. (1995) The interleukin-1 beta-converting enzyme (ICE) is localized on the external cell surface membranes and in the cytoplasmic ground substance of human monocytes by immuno-electron microscopy. J Exp Med 182: 1447–1459.
  36. 36. Netea MG, van de Veerdonk FL, Kullberg BJ, Van der Meer JW, Joosten LA (2008) The role of NLRs and TLRs in the activation of the inflammasome. Expert Opin Biol Ther 8: 1867–1872.
  37. 37. Koo IC, Wang C, Raghavan S, Morisaki JH, Cox JS, et al. (2008) ESX-1-dependent cytolysis in lysosome secretion and inflammasome activation during mycobacterial infection. Cell Microbiol 10: 1866–1878.
  38. 38. Master SS, Rampini SK, Davis AS, Keller C, Ehlers S, et al. (2008) Mycobacterium tuberculosis prevents inflammasome activation. Cell Host Microbe 3: 224–232.
  39. 39. Mishra BB, Moura-Alves P, Sonawane A, Hacohen N, Griffiths G, et al. (2010) Mycobacterium tuberculosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome. Cell Microbiol 12: 1046–1063.
  40. 40. Carlsson F, Kim J, Dumitru C, Barck KH, Carano RA, et al. (2010) Host-detrimental role of Esx-1-mediated inflammasome activation in mycobacterial infection. PLoS Pathog 6: e1000895.
  41. 41. Lee HM, Yuk JM, Kim KH, Jang J, Kang G, et al. (2011) Mycobacterium abscessus activates the NLRP3 inflammasome via Dectin-1-Syk and p62/SQSTM1. Immunol Cell Biol.
  42. 42. Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, et al. (2008) Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 320: 674–677.
  43. 43. Sansonetti PJ, Phalipon A, Arondel J, Thirumalai K, Banerjee S, et al. (2000) Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity 12: 581–590.
  44. 44. Mariathasan S, Weiss DS, Dixit VM, Monack DM (2005) Innate immunity against Francisella tularensis is dependent on the ASC/caspase-1 axis. J Exp Med 202: 1043–1049.
  45. 45. Raupach B, Peuschel SK, Monack DM, Zychlinsky A (2006) Caspase-1-mediated activation of interleukin-1beta (IL-1beta) and IL-18 contributes to innate immune defenses against Salmonella enterica serovar Typhimurium infection. Infect Immun 74: 4922–4926.
  46. 46. He X, Mekasha S, Mavrogiorgos N, Fitzgerald KA, Lien E, et al. (2010) Inflammation and fibrosis during Chlamydia pneumoniae infection is regulated by IL-1 and the NLRP3/ASC inflammasome. J Immunol 184: 5743–5754.
  47. 47. Juffermans NP, Florquin S, Camoglio L, Verbon A, Kolk AH, et al. (2000) Interleukin-1 signaling is essential for host defense during murine pulmonary tuberculosis. J Infect Dis 182: 902–908.
  48. 48. Sugawara I, Yamada H, Hua S, Mizuno S (2001) Role of interleukin (IL)-1 type 1 receptor in mycobacterial infection. Microbiol Immunol 45: 743–750.
  49. 49. Wieland CW, Florquin S, Pater JM, Weijer S, van der Poll T (2006) Interleukin-1 contributes to an effective clearance of Mycobacterium kansasii from the respiratory tract. Microbes Infect 8: 2409–2413.
  50. 50. Muller I, Cobbold SP, Waldmann H, Kaufmann SH (1987) Impaired resistance to Mycobacterium tuberculosis infection after selective in vivo depletion of L3T4+ and Lyt-2+ T cells. Infect Immun 55: 2037–2041.
  51. 51. Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, et al. (1999) Mice deficient in CD4 T cells have only transiently diminished levels of IFN-gamma, yet succumb to tuberculosis. J Immunol 162: 5407–5416.
  52. 52. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, et al. (1993) Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med 178: 2243–2247.
  53. 53. Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, et al. (1993) An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med 178: 2249–2254.
  54. 54. Cooper AM, Magram J, Ferrante J, Orme IM (1997) Interleukin 12 (IL-12) is crucial to the development of protective immunity in mice intravenously infected with Mycobacterium tuberculosis. J Exp Med 186: 39–45.
  55. 55. Silva RA, Florido M, Appelberg R (2001) Interleukin-12 primes CD4+ T cells for interferon-gamma production and protective immunity during Mycobacterium avium infection. Immunology 103: 368–374.
  56. 56. Florido M, Cooper AM, Appelberg R (2002) Immunological basis of the development of necrotic lesions following Mycobacterium avium infection. Immunology 106: 590–601.
  57. 57. Saunders BM, Frank AA, Orme IM, Cooper AM (2002) CD4 is required for the development of a protective granulomatous response to pulmonary tuberculosis. Cell Immunol 216: 65–72.
  58. 58. Wieland CW, Florquin S, Pater JM, Weijer S, van der Poll T (2006) CD4+ cells play a limited role in murine lung infection with Mycobacterium kansasii. Am J Respir Cell Mol Biol 34: 167–173.
  59. 59. Kleinnijenhuis J, Joosten LA, van de Veerdonk FL, Savage N, van Crevel R, et al. (2009) Transcriptional and inflammasome-mediated pathways for the induction of IL-1beta production by Mycobacterium tuberculosis. Eur J Immunol 39: 1914–1922.
  60. 60. Kurenuma T, Kawamura I, Hara H, Uchiyama R, Daim S, et al. (2009) The RD1 locus in the Mycobacterium tuberculosis genome contributes to activation of caspase-1 via induction of potassium ion efflux in infected macrophages. Infect Immun 77: 3992–4001.
  61. 61. Newman ZL, Leppla SH, Moayeri M (2009) CA-074Me protection against anthrax lethal toxin. Infect Immun 77: 4327–4336.
  62. 62. Abdallah AM, Bestebroer J, Savage ND, de Punder K, van Zon M, et al. (2011) Mycobacterial secretion systems ESX-1 and ESX-5 play distinct roles in host cell death and inflammasome activation. J Immunol 187: 4744–4753.
  63. 63. Gao LY, Guo S, McLaughlin B, Morisaki H, Engel JN, et al. (2004) A mycobacterial virulence gene cluster extending RD1 is required for cytolysis, bacterial spreading and ESAT-6 secretion. Mol Microbiol 53: 1677–1693.
  64. 64. McLaughlin B, Chon JS, MacGurn JA, Carlsson F, Cheng TL, et al. (2007) A mycobacterium ESX-1-secreted virulence factor with unique requirements for export. PLoS Pathog 3: e105.
  65. 65. Stanley SA, Johndrow JE, Manzanillo P, Cox JS (2007) The Type I IFN response to infection with Mycobacterium tuberculosis requires ESX-1-mediated secretion and contributes to pathogenesis. J Immunol 178: 3143–3152.
  66. 66. van der Wel N, Hava D, Houben D, Fluitsma D, van Zon M, et al. (2007) M. tuberculosis and M. leprae translocate from the phagolysosome to the cytosol in myeloid cells. Cell 129: 1287–1298.
  67. 67. Smith J, Manoranjan J, Pan M, Bohsali A, Xu J, et al. (2008) Evidence for pore formation in host cell membranes by ESX-1-secreted ESAT-6 and its role in Mycobacterium marinum escape from the vacuole. Infect Immun 76: 5478–5487.
  68. 68. Sorensen AL, Nagai S, Houen G, Andersen P, Andersen AB (1995) Purification and characterization of a low-molecular-mass T-cell antigen secreted by Mycobacterium tuberculosis. Infect Immun 63: 1710–1717.
  69. 69. Harboe M, Oettinger T, Wiker HG, Rosenkrands I, Andersen P (1996) Evidence for occurrence of the ESAT-6 protein in Mycobacterium tuberculosis and virulent Mycobacterium bovis and for its absence in Mycobacterium bovis BCG. Infect Immun 64: 16–22.
  70. 70. Pollock JM, Andersen P (1997) The potential of the ESAT-6 antigen secreted by virulent mycobacteria for specific diagnosis of tuberculosis. J Infect Dis 175: 1251–1254.
  71. 71. Arend SM, de Haas P, Leyten E, Rosenkrands I, Rigouts L, et al. (2005) ESAT-6 and CFP-10 in clinical versus environmental isolates of Mycobacterium kansasii. J Infect Dis 191: 1301–1310.
  72. 72. Meissner F, Molawi K, Zychlinsky A (2008) Superoxide dismutase 1 regulates caspase-1 and endotoxic shock. Nat Immunol 9: 866–872.
  73. 73. Martinon F (2010) Signaling by ROS drives inflammasome activation. Eur J Immunol 40: 616–619.
  74. 74. Meissner F, Seger RA, Moshous D, Fischer A, Reichenbach J, et al. (2010) Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood 116: 1570–1573.
  75. 75. Segal BH, Han W, Bushey JJ, Joo M, Bhatti Z, et al. (2010) NADPH oxidase limits innate immune responses in the lungs in mice. PLoS One 5: e9631.
  76. 76. van Bruggen R, Koker MY, Jansen M, van Houdt M, Roos D, et al. (2010) Human NLRP3 inflammasome activation is Nox1-4 independent. Blood 115: 5398–5400.
  77. 77. van de Veerdonk FL, Smeekens SP, Joosten LA, Kullberg BJ, Dinarello CA, et al. (2010) Reactive oxygen species-independent activation of the IL-1beta inflammasome in cells from patients with chronic granulomatous disease. Proc Natl Acad Sci U S A 107: 3030–3033.
  78. 78. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J (2010) Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol 11: 136–140.
  79. 79. Cassel SL, Eisenbarth SC, Iyer SS, Sadler JJ, Colegio OR, et al. (2008) The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci U S A 105: 9035–9040.
  80. 80. Abdul-Sater AA, Said-Sadier N, Lam VM, Singh B, Pettengill MA, et al. (2010) Enhancement of reactive oxygen species production and chlamydial infection by the mitochondrial Nod-like family member NLRX1. J Biol Chem 285: 41637–41645.
  81. 81. Abdul-Sater AA, Koo E, Hacker G, Ojcius DM (2009) Inflammasome-dependent caspase-1 activation in cervical epithelial cells stimulates growth of the intracellular pathogen Chlamydia trachomatis. J Biol Chem 284: 26789–26796.
  82. 82. Tschopp J, Schroder K (2010) NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat Rev Immunol 10: 210–215.
  83. 83. Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu SS (2004) Calcium, ATP, and ROS: a mitochondrial love-hate triangle. Am J Physiol Cell Physiol 287: C817–833.
  84. 84. Novo E, Parola M (2008) Redox mechanisms in hepatic chronic wound healing and fibrogenesis. Fibrogenesis Tissue Repair 1: 5.
  85. 85. Kowaltowski AJ, de Souza-Pinto NC, Castilho RF, Vercesi AE (2009) Mitochondria and reactive oxygen species. Free Radic Biol Med 47: 333–343.
  86. 86. Yang CS, Shin DM, Kim KH, Lee ZW, Lee CH, et al. (2009) NADPH oxidase 2 interaction with TLR2 is required for efficient innate immune responses to mycobacteria via cathelicidin expression. J Immunol 182: 3696–3705.
  87. 87. Choi HH, Shin DM, Kang G, Kim KH, Park JB, et al. (2010) Endoplasmic reticulum stress response is involved in Mycobacterium tuberculosis protein ESAT-6-mediated apoptosis. FEBS Lett 584: 2445–2454.
  88. 88. Tattoli I, Carneiro LA, Jehanno M, Magalhaes JG, Shu Y, et al. (2008) NLRX1 is a mitochondrial NOD-like receptor that amplifies NF-kappaB and JNK pathways by inducing reactive oxygen species production. EMBO Rep 9: 293–300.
  89. 89. Zhou R, Yazdi AS, Menu P, Tschopp J (2011) A role for mitochondria in NLRP3 inflammasome activation. Nature 469: 221–225.
  90. 90. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, et al. (2011) Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 12: 222–230.
  91. 91. Yu HB, Finlay BB (2008) The caspase-1 inflammasome: a pilot of innate immune responses. Cell Host Microbe 4: 198–208.
  92. 92. McIntire CR, Yeretssian G, Saleh M (2009) Inflammasomes in infection and inflammation. Apoptosis 14: 522–535.
  93. 93. Yazdi AS, Guarda G, D'Ombrain MC, Drexler SK (2010) Inflammatory caspases in innate immunity and inflammation. J Innate Immun 2: 228–237.
  94. 94. Dinarello CA (1998) Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci 856: 1–11.
  95. 95. Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27: 519–550.
  96. 96. Akhter A, Gavrilin MA, Frantz L, Washington S, Ditty C, et al. (2009) Caspase-7 activation by the Nlrc4/Ipaf inflammasome restricts Legionella pneumophila infection. PLoS Pathog 5: e1000361.
  97. 97. Havell EA, Moldawer LL, Helfgott D, Kilian PL, Sehgal PB (1992) Type I IL-1 receptor blockade exacerbates murine listeriosis. J Immunol 148: 1486–1492.
  98. 98. Satoskar AR, Okano M, Connaughton S, Raisanen-Sokolwski A, David JR, et al. (1998) Enhanced Th2-like responses in IL-1 type 1 receptor-deficient mice. Eur J Immunol 28: 2066–2074.
  99. 99. Yamada H, Mizumo S, Horai R, Iwakura Y, Sugawara I (2000) Protective role of interleukin-1 in mycobacterial infection in IL-1 alpha/beta double-knockout mice. Lab Invest 80: 759–767.
  100. 100. Fremond CM, Togbe D, Doz E, Rose S, Vasseur V, et al. (2007) IL-1 receptor-mediated signal is an essential component of MyD88-dependent innate response to Mycobacterium tuberculosis infection. J Immunol 179: 1178–1189.
  101. 101. Mayer-Barber KD, Barber DL, Shenderov K, White SD, Wilson MS, et al. (2010) Caspase-1 independent IL-1beta production is critical for host resistance to mycobacterium tuberculosis and does not require TLR signaling in vivo. J Immunol 184: 3326–3330.
  102. 102. Kasahara K, Kobayashi K, Shikama Y, Yoneya I, Soezima K, et al. (1988) Direct evidence for granuloma-inducing activity of interleukin-1. Induction of experimental pulmonary granuloma formation in mice by interleukin-1-coupled beads. Am J Pathol 130: 629–638.
  103. 103. Chensue SW, Warmington KS, Berger AE, Tracey DE (1992) Immunohistochemical demonstration of interleukin-1 receptor antagonist protein and interleukin-1 in human lymphoid tissue and granulomas. Am J Pathol 140: 269–275.
  104. 104. Bellamy R, Ruwende C, Corrah T, McAdam KP, Whittle HC, et al. (1998) Assessment of the interleukin 1 gene cluster and other candidate gene polymorphisms in host susceptibility to tuberculosis. Tuber Lung Dis 79: 83–89.
  105. 105. Gomez LM, Camargo JF, Castiblanco J, Ruiz-Narvaez EA, Cadena J, et al. (2006) Analysis of IL1B, TAP1, TAP2 and IKBL polymorphisms on susceptibility to tuberculosis. Tissue Antigens 67: 290–296.
  106. 106. Awomoyi AA, Charurat M, Marchant A, Miller EN, Blackwell JM, et al. (2005) Polymorphism in IL1B: IL1B-511 association with tuberculosis and decreased lipopolysaccharide-induced IL-1beta in IFN-gamma primed ex-vivo whole blood assay. J Endotoxin Res 11: 281–286.
  107. 107. Wilkinson RJ, Patel P, Llewelyn M, Hirsch CS, Pasvol G, et al. (1999) Influence of polymorphism in the genes for the interleukin (IL)-1 receptor antagonist and IL-1beta on tuberculosis. J Exp Med 189: 1863–1874.
  108. 108. McElvania Tekippe E, Allen IC, Hulseberg PD, Sullivan JT, McCann JR, et al. (2010) Granuloma formation and host defense in chronic Mycobacterium tuberculosis infection requires PYCARD/ASC but not NLRP3 or caspase-1. PLoS One 5: e12320.
  109. 109. Walter K, Holscher C, Tschopp J, Ehlers S (2010) NALP3 is not necessary for early protection against experimental tuberculosis. Immunobiology 215: 804–811.
  110. 110. Dorhoi A, Nouailles G, Jorg S, Hagens K, Heinemann E, et al. (2012) Activation of the NLRP3 inflammasome by Mycobacterium tuberculosis is uncoupled from susceptibility to active tuberculosis. Eur J Immunol 42: 374–384.
  111. 111. Atianand MK, Duffy EB, Shah A, Kar S, Malik M, et al. (2011) Francisella tularensis reveals a disparity between human and mouse NLRP3 inflammasome activation. J Biol Chem 286: 39033–39042.
  112. 112. Gavrilin MA, Wewers MD (2011) Francisella Recognition by Inflammasomes: Differences between Mice and Men. Front Microbiol 2: 11.
  113. 113. Abdul-Sater AA, Said-Sadier N, Padilla EV, Ojcius DM (2010) Chlamydial infection of monocytes stimulates IL-1beta secretion through activation of the NLRP3 inflammasome. Microbes Infect 12: 652–661.
  114. 114. Said-Sadier N, Padilla E, Langsley G, Ojcius DM (2010) Aspergillus fumigatus stimulates the NLRP3 inflammasome through a pathway requiring ROS production and the Syk tyrosine kinase. PLoS One 5: e10008.
  115. 115. Yang Y, Yin C, Pandey A, Abbott D, Sassetti C, et al. (2007) NOD2 pathway activation by MDP or Mycobacterium tuberculosis infection involves the stable polyubiquitination of Rip2. J Biol Chem 282: 36223–36229.