Abstract
Mycobacterium tuberculosis has succeeded in infecting one third of the human race though inhibition or evasion of innate and adaptive immunity. The pathogen is a facultative intracellular parasite that uses the niche provided by mononuclear phagocytes for its advantage. Complex interactions determine whether the bacillus will or will not be delivered to acidified lysosomes, whether the host phagocyte will survive infection or die, and whether the timing and mode of cell death works to the advantage of the host or the pathogen. Here we discuss cell death and autophagy in TB. These fundamental processes of cell biology feature in all aspects of TB pathogenesis and may be exploited to the treatment or prevention of TB disease.
Keywords: Tuberculosis, Phagocyte, Apoptosis, Necrosis, Autophagy
1. Introduction
Programmed cell death and autophagy are fundamental processes of cell biology intimately involved in the interaction between Mycobacterium tuberculosis (Mtb) and the phagocytes it infects, including macrophages, dendritic cells (DC) and neutrophils. The remarkable success of Mtb as a human pathogen results from its capacity to evade the innate antimicrobial effector mechanisms of mononuclear phagocytes (MPs) and leverage the intracellular environment as a replication niche. Infected MPs are faced with a pathogen surviving in phagosomes that fail to incorporate the molecular machinery needed to reduce vacuolar pH and generate free radicals of oxygen or nitrogen, and that fail to fuse with lysosomes to expose bacilli to damaging hydrolases [1]. Plan B for the infected MP is to undergo programmed cell death, which eliminates the intracellular sanctuary and exerts other potentially host-protective effects described in section 3.1.2. Alternatively, a variety of extracellular signals may activate the autophagic machinery of infected MP to drive Mtb into lethal autolysosomes as described in section 5. These responses set the stage for what are now recognized as a very complex series of measures and countermeasures culminating in the survival or death of the infecting pathogen or its host cell, the progression or resolution of immune pathology, and outcome of tuberculosis (TB) disease.
2. Overview of programmed cell death
A requirement for regulated cell death to support tissue development and homeostasis was conceived by Karl Vogt in 1842 but the term apoptosis to describe a morphologically distinct form of non-traumatic cell death and the understanding of its biochemical mechanisms did not emerge until the late 20th century [2]. Apoptosis is a tightly regulated process of cellular deconstruction. It minimizes inflammation and bystander injury by containing the dismembered nuclear and cytoplasmic contents of dying cells within membrane-bound vesicles called apoptotic bodies that are engulfed by other phagocytes in a process called efferocytosis (section 3.1.2). Binding of apoptotic bodies to specific receptors on MPs responding to “find me” and “eat me” signals induces the expression of anti-inflammatory cytokines including transforming growth factor-β and interleukin (IL)-10 to further insure the silent elimination of cellular corpses [3, 4]. The ultrastructural morphology of apoptosis is characterized by cell shrinkage and chromatin condensation (pyknosis), nuclear fragmentation (karyorrhexis), and blebbing of the outer cell membrane that culminates in apoptotic body formation. Chromosomal DNA is cleaved at inter-nucleosomal boundaries, demonstrated by laddering of DNA bands on gel electrophoresis. Phosphatidylserine (PtdSer), a membrane component that in viable cells is held facing the cytosolic side of the plasma membrane by the enzyme flippase, translocates to the outward-facing surface in apoptotic cells. Exposure of PtdSer on the cell surface plays an important role in membrane stability and clearance of apoptotic bodies (section 3.1.2).
Necrosis is a much different death, defined by the loss of outer cell membrane integrity with release of cytoplasmic and nuclear contents to the extracellular space. Necrosis was originally thought to result only from accidental events (e.g. freezing or crushing) but regulated mechanisms of necrosis were later identified (section 2.2) [5]. The ultrastructural morphology of necrosis is characterized by cytoplasmic swelling (onicosis), cytoplasmic vacuolization and swelling of organelles including mitochondria and cell nuclei [6]. These changes result from ATP depletion and the failure of plasma membrane ion pumps to maintain a stable osmotic gradient. Necrosis can also result from direct plasma membrane damage, which disrupts the cells without onicosis. Rupture of the plasma membrane provokes inflammation by releasing damage-associated molecular patterns (DAMPs) such as heat shock proteins, high-mobility group box 1, S100 proteins, extracellular genomic and mitochondrial DNA, ATP, monosodium urate, and heparin sulfate [7, 8, 8-12]. Binding of DAMPs to their cognate receptors activates an innate inflammatory response and sends “endogenous adjuvant” signals that can stimulate DC to promote T cell activation [9]. The diversity of protein and non-protein DAMPs ensures redundancy in immune stimulation but most converge on common pathways involving inflammasomes, IL-1 and leukotriene (LT)B4 [9, 13].
2.1 Apoptosis signaling and execution
Three major pathways of apoptosis initiation (extrinsic, intrinsic and perforin/granzyme) converge on a common execution mechanism with degradation of chromosomal DNA and nuclear and cytoskeletal proteins. Both steps in this process involve caspases; a family of cysteine-dependent aspartate-directed proteases constitutively expressed as zymogens. Caspases operate in a cascade for the rapid induction of apoptosis, which is energy-dependent but independent of transcription [14]. Initiator caspases-8, -9, and -10 are activated by dimerization following recruitment to signaling complexes (sections 2.1.1 and 2.1.2) [15]. Activated initiator caspases then cleave and activate the pre-formed dimeric zymogens of the executioner caspases-3 and -7. Activation of executioner caspases is necessarily a tightly regulated event but can in some circumstances be mediated by proteases other than initiator caspases.
2.1.1 Extrinsic apoptosis
The extrinsic pathway begins with ligand binding to tumor necrosis factor receptor (TNFR) family proteins containing a death domain in their cytoplasmic tail which serves as the site for signal complex formation [16]. The receptor/ligand pairs most relevant to TB are TNF-α/TNFR1 and Fas ligand/Fas [17, 18]. TNF-α binding trimerizes TNFR1 allowing recruitment of TNFR1-associated death domain (TRADD), receptor interacting protein kinase (RIPK)1, TNFR-associated factor (TRAF)2, TRAF5, cellular inhibitor of apoptosis protein (cIAP) 1, and cIAP2 to form membrane-associated complex I. Signals from complex I activate NFκB that upregulates pro-survival genes [19]. Apoptosis is initiated from TNFR1 following dissociation of complex I constituents to form cytoplasmic complexes. Complex IIA contains TRADD, Fas-associated death domain (FADD) and caspase-8. Its formation is opposed by cellular FLICE-like inhibitory protein (cFLIP) that is induced by NFκB [20]. The TRADD-independent complex IIB (also called the ripoptosome) forms when TNFR1 is activated but cIAP1 is inhibited by mimetics of second mitochondria-derived activator of caspases (Smac). Formation of complex IIB also requires deubiquitination of RIPK1 by cylindromatosis. FasL binding to Fas recruits FADD which in turn recruits procaspase-8 and/or cFLIP via death-effector domain interactions. This forms a membrane-associated death inducing signal complex but a secondary cytosolic complex of FADD, cFLIP, and caspase-8 can be released to further amplify apoptosis initiation [19].
2.1.2 Intrinsic apoptosis
The intrinsic apoptosis pathway is induced by diverse intracellular stresses such as DNA damage, starvation, and oxidative stress that lead to outer mitochondrial membrane (OMM) permeabilization. Cytochrome c released from the mitochondrial inter-membrane space binds the cytosolic protein apoptotic protease activating factor (Apaf-1) to form a multimeric signaling complex called the apoptosome. The apoptosome recruits and activates procaspase-9, which in turn activates executioner caspases [21]. Smac and the serine protease HtrA2/OMI are also released from the inter-membrane space; they amplify intrinsic apoptosis by relieving the constitutive caspase repression mediated by cIAP family members. Mitochondrial permeability is controlled by Bcl-2 family proteins that either promote or inhibit apoptosis [22, 23]. Pro-apoptotic Bax and Bak form pores in the OMM to release cytochrome c. This is opposed by anti-apoptotic family members (e.g. Bcl-2, Bclx-L, Mcl-1) but further promoted by other pro-apoptotic Bcl-2 proteins. Cell fate is determined by the integrated activities of pro- and anti-apoptotic Bcl-2 family proteins. Activated caspase-9 cleaves pro-apoptotic Bid into an enzymatically active truncated form (tBid), which orchestrates the activities of Bax to accelerate cytochrome c release. Caspase-8 can also cleave Bid, providing a means for crosstalk between the extrinsic and intrinsic apoptosis pathways [24].
2.1.3 Perforin/granzyme mediated apoptosis
A third apoptosis induction pathway is mediated by serine proteases of the granzyme family contained, along with perforin, in granules of cytotoxic T lymphocytes (CTL) and natural killer (NK) cells [25]. Perforin creates pores in the plasma membrane of target cells through which the granzymes are introduced. Granzyme B cleaves the initiator caspases -8 and -10 and the executioner caspases -3 and -7, and it has other substrates relevant to apoptosis induction including inhibitor of caspase-activated DNase and Bid [26, 27]. The former mediates DNA fragmentation while the latter links perforin/granzyme to the mitochondrial pathway. Granzymes A and C are also implicated in apoptosis although their roles are less well characterized [28]. While perforin/granzymes is the primary mechanism for killing, CTL can also cause apoptosis by engaging Fas on target cells with FasL to trigger the extrinsic pathway [29].
2.2 Regulated necrosis
The field of programmed cell death has become increasingly complex since the discovery of apoptosis. Much recent interest has focused on pathways of regulated necrosis, which currently comprise necroptosis, pyroptosis, pyronecrosis, ETosis, cyclophilin D (CYPD)-dependent necrosis, parthanatos, and autophagic cell death [5]. Necroptosis is the best characterized pathway while pyroptosis, pyronecrosis and ETosis have been most closely identified in the context of infection. Necroptosis occurs when TNFR1 signaling is activated but caspase-8 is inhibited by drugs or virus-encoded anti-apoptotic proteins. This results in formation of a complex called the necrosome, comprising RIPK1, RIPK3, FADD, and caspase-8 [30, 31]. The necrosome recruits and activates mixed lineage kinase domain like which then translocates to the plasma membrane where it mediates TNF- and Ca2+-dependent necrotic cell death (necroptosis) through several terminal mechanisms including energy depletion, reactive oxygen species (ROS) production, lysosomal membrane permeabilization (LMP), and lipid peroxidation [5].
2.2.1 Pyroptosis and pyronecrosis
Pyroptosis and pyronecrosis were discovered in the context of intracellular bacterial infection (e.g. Francisella tularensis, Listeria monocytogenes, and Shigella flexneri [32-34]) although non-infectious triggers have also been identified [35]. These death subroutines share dependence on inflammasome constituents and terminal plasma membrane pore formation resulting onicosis. A common trigger for pyroptosis and pyronecrosis is NOD-like receptor (NLR) activation by pathogen-associated molecular patterns (PAMPs) from intracellular microbes. Distinguishing characteristics are the dependence of pyroptosis on caspase-1 or caspase-11 (human caspase-4) while pyronecrosis is caspase-independent and requires the inflammasome component apoptosis-associated speck-like protein containing a CARD (ASC) and the lysosomal protease cathepsin B. Pyroptosis and pyronecrosis share rapid kinetics of lethality and considerable overlap in triggers. As an example, S. flexneri at low multiplicity of infection (MOI) induces macrophage pyroptosis dependent on the inflammasome constituent NLRC4 but higher MOI triggers pyronecrosis via NLRP3 [36]. These death modes are highly inflammatory, with abundant production of IL-1β and IL-18 that is perpetuated by the release of aggregated ASC multimers (ASC specks) which continue processing pro-IL-1β in the extracellular space and even following phagocytosis by naïve macrophages [37, 38].
2.2.2 CYPD-dependent necrosis
Mitochondrial permeability transition (MPT) occurs when the inner membrane (IMM) becomes permeable to solutes <1500 Da [39]. This is a sudden event caused by opening of a channel in IMM called the permeability transition pore (PTP) [40]. The mitochondrial matrix protein CYPD is a key regulator of PTP formation although not an essential component of the pore. Binding of cyclosporin A to CYPD inhibits MPT [23]. Transient MPT is survivable but sustained PTP opening results in Ca2+ efflux, depletion of pyridine nucleotides from the mitochondrial matrix, and matrix swelling that culminates in OMM rupture with release of inter-membrane space components including cytochrome c, Smac, and apoptosis-inducing factor (AIF). The intrinsic apoptosis pathway is activated by cytochrome c and Smac, (another example of crosstalk between death pathways), while AIF mediates caspase-independent DNA fragmentation and chromatin condensation [41]. AIF also plays a role in parthanatos, a regulated necrosis pathway mediated by poly ADP ribose polymerase hyper activation that is reviewed elsewhere [42]. The CYPD pathway is tied to necrosis as result of catastrophic mitochondrial dysfunction. The inducers of CYPD-dependent necrosis are not fully understood but Ca2+, ROS, adenine nucleotides, and ubiquinones have been implicated [5].
2.2.3 ETosis
This mode of programmed necrosis was first identified in neutrophils and features chromatin disgorgement to the extracellular space, forming extracellular traps (NETs) [43]. In contrast to apoptosis, chromatin decondensation is an early step in ETosis and PtdSer does not flip to the outer cell surface. Unfolded genomic DNA presents a web-like anionic polymer that binds bacteria and fungi by electrostatic interactions. NETs also bind proteins including neutrophil elastase, histones, and antimicrobial peptides that kill bound pathogens [44]. Release of extracellular traps has been identified in eosinophils, mast cells, and macrophages, hence the term ETosis [45, 46].
Triggers for ETosis include lipopolysaccharide, interferon (IFN)-γ, bacteria, fungi, and phorbol myristate acetate [47]. NET release in most instances requires NADPH oxidase (NOX) although a NOX-independent pathway stimulated by CXCL2 was reported [48]. The ETosis pathway, including the requirement NOX, is incompletely understood. Upstream signaling via Raf-MEK-ERK activates NOX and ETosis, as does activation of the mammalian target of rapamycin (mTOR) pathway. Inhibitors of ETosis include serine protease inhibitor B1 and the eicosanoid 5-hydroxyeicosatetraenoic acid, a product of 5-lipoxygenase [48, 49]. ETosis is fundamentally a mode of necrotic cell death but there is remarkable evidence for the persistence of post-ETotic, anuclear neutrophil zombies capable of directed migration and phagocytosis [50].
2.3 Detection of apoptosis and necrosis
Many assays are available to discern whether dead and dying cells are undergoing apoptosis or necrosis. The most common of these test the integrity of the outer cell membrane (e.g. propidium iodide [PI] or 7-aminoactinomycin D), presentation of PtdSer on the outer surface of the plasma membrane (e.g annexin V binding), nuclear pyknosis and fragmentation (e.g. 4',6-diamidino-2-phenylindole staining), DNA fragmentation (e.g. terminal deoxynucleotidyl transferase dUTP nick end labeling [TUNEL] assay), caspase activation, and the state of mitochondrial transmembrane potential (ΔΨm). Transmission electron microscopy (TEM) is perhaps the most accurate method to distinguish between apoptotic and necrotic cell death based on distinct ultrastructural features. Each method has strengths and weaknesses depending on cells and experimental conditions being investigated. TUNEL is highly sensitive but has poor specificity while TEM is highly specific but lacks sensitivity since it is difficult to interrogate large numbers of cells, particularly in vivo.
To critically evaluate the literature on cell death in TB it is important to understand the limitations of these assays and the fundamental problem that none can unambiguously define cell fate. The Cell Death Nomenclature Committee has published guidelines for the use and interpretation of cell death assays [51]. The heterogeneity of cell death and unique characteristics of linked to specific cell types and conditions means that only general guidelines can be suggested. These include determining whether the cells in question are truly dead and then interrogating the responsible cell death subroutine using at least two methodologically unrelated assays. The challenge of defining the specific nature and subroutine of cell death is made greater by the extensive crosstalk between pathways so that different death subroutines may be simultaneously activated in a single cell with one or another appearing predominant based on experimental conditions [5, 51, 52]. Common examples of such ambiguities include: caspase activation in viable cells; caspase activation as a secondary effect of proteolysis in necrotic cells; crosstalk between extrinsic and intrinsic apoptosis pathways; secondary necrosis of apoptotic bodies; and the convergent triggers and execution mechanisms of the different programmed necrosis subroutines.
3. Cell death in TB
Apoptosis can serve as a defense mechanism for cells confronted with intracellular pathogens that consume host resources for survival and replication. This paradigm was discovered the context of virus infection (reviewed in [53]) but has been extended to a wide spectrum of pathogenic microbes. Apoptosis offers several potential benefits for the host including elimination of a replication niche, exposure of pathogens to humoral immunity, and forcing intracellular pathogens to reestablish residence in naïve host cells. Efferocytosis provides a means to defeat the virulence mechanisms of intracellular pathogens that inhibit vesicular trafficking and maturation. It also delivers pathogens or their antigenic components to DC for efficient priming and cross presentation (section 3.1.2) [54]. The rapid, immunostimulatory demise of cells by programmed necrosis might benefit the host by accelerating neutrophil recruitment to control fast-replicating bacteria [55]. Conversely, apoptosis could be disease-promoting if it eliminates key host defense cells, facilitates penetration of epithelial barriers, or spreads infection through efferocytosis [56, 57].
Macrophage-pathogen interactions play a central role in TB pathogenesis. The bacillus is a facultative intracellular parasite that cannot gain a foothold in new hosts without this replication sanctuary. Once Mtb has established infection in the lung, transmission to new hosts by infectious aerosols requires its transition to the extracellular space. It is therefore unsurprising that cell death in its many forms has been identified in the context of Mtb infection in vitro and in vivo. A recent review on this topic cited 48 manuscripts reporting a spectrum of death modes and subroutines linked to TB including extrinsic, intrinsic and perforin/granzyme-mediated apoptosis, and several type of necrosis [58]. In this review we focus primarily on macrophage cell death resulting directly from Mtb infection but the fates of infected DC and neutrophils will also be discussed (section 3.3). While beyond the scope of this review, CTL-mediated death, activation-induced cell death, bystander death of T cells, and Mtb-induced death of epithelial cells all participate in TB pathogenesis.
3.1 Apoptosis of Mtb-infected macrophages
Before the discovery of an intrinsic apoptosis response to Mtb infection, Molloy et al. [59] reported that treating BCG-infected monocytes with exogenous ATP induced apoptosis and killed bacilli while BCG viability was preserved after H2O2-induced monocyte necrosis. In retrospect, the antimicrobial activity of ATP might have been mediated by autophagy (section 5.2) [60] but this publication introduced the concept of apoptosis has host-protective in TB. Keane et al. [18] were the first to describe apoptosis as an autonomous response of primary human alveolar macrophages infected with live but not heat-killed Mtb. Apoptosis was verified by TEM, internucleosomal laddering of genomic DNA, and TUNEL assay (the latter including human TB lung sections). The extrinsic pathway was triggered TNF-α in an autocrine/paracrine manner and required infection-induced priming for TNFR death signals since uninfected cells were resistant to exogenous TNF-α. The attenuated Mtb strain H37Ra was a much stronger apoptosis inducer than virulent H37Rv. Oddo et al. [17] subsequently reported that exogenous FasL or TNF-α induced apoptosis of Mtb-infected macrophages (defined by annexin V/PI staining and by TUNEL) was accompanied by reduced bacillary viability. No antimicrobial effect occurred with complement-induced necrosis of infected macrophages.
A simple model of apoptosis as a host-protective response in TB emerged from those early reports. Subsequent in vitro studies confirmed the association of apoptotic cell death with reduced Mtb viability [61-65]. The concept of apoptosis evasion as a virulence-associated trait of Mtb was substantiated and several mechanisms of apoptosis evasion were described including downregulation of Fas on infected macrophages, interference with death signals downstream of TNFR1, and shedding of soluble TNFR2 that neutralized TNF-α [17, 66, 67].
Recent findings have underscored the complexity of macrophage apoptosis in TB and its role in host defense. Data from the zebrafish/M. marinum model show that apoptosis can facilitate spread of infection to naïve macrophages in vivo [57]. Accelerated dissemination was also demonstrated in mice infected with a pro-apoptotic mutant of virulent Mtb (section 3.1.1) in experiments that also failed to support a direct antimicrobial effect of macrophage apoptosis in vivo [68].
3.1.1 Mtb genes linked to apoptosis evasion and induction
A major advance in this field was provided by the discovery in 2007 of two Mtb genes (secA2 and nuoG) linked to the suppression macrophage apoptosis, both acting reduce levels of ROS in the mycobacterial vacuole [69, 70]. This jibes with the models of extrinsic apoptosis in Mtb-infected macrophages where ROS play an intermediary role in TNF-α signaling for apoptosis (and necrosis), and intrinsic apoptosis that can also be triggered by ROS [71, 72]. Mutation of secA2 in impairs secretion of bacterial superoxide dismutase (SodA) and confers an apoptosis-inducing phenotype on H37Rv demonstrated by TUNEL and caspase activation in THP-1 cells and mouse bone marrow-derived macrophages (BMM] [70]. This result was anticipated by an earlier study of H37Rv mutants where SodA expression was knocked down with anti-sense RNA, resulting in vivo attenuation with less inflammation and more MP apoptosis after high dose intravenous infection in C57BL/6 mice [73]. The ΔsecA2 mutant strongly induced antigen-specific CD8+ T cell priming in vivo, which was attributed to its pro-apoptotic phenotype (section 3.1.2).
The anti-apoptotic activity of nuoG was revealed in a gain-of-function screen in M. smegmatis. Deletion of nuoG in Mtb H37Rv conferred a pro-apoptotic phenotype on infection of THP-1 cells and BMM in vitro [69]. The nuoG gene encodes a subunit of a type I NADH dehydrogenase that neutralizes ROS generated by host NOX2, thereby inhibiting TNF-α-stimulated apoptosis as well as TNF-α secretion [74]. The mutant RvΔnuoG strain grows normally in broth but it is attenuated in SCID and wild-type BALB/c mice after high dose intravenous challenge [69]. Growth of RvΔnuoG in the lung during the first 3 weeks post-infection matched that of wild-type H37Rv and a complemented mutant strain; its attenuation in terms of bacterial burden was only evident at later times points with a 0.8 log reduction compared to wild-type H37Rv at 20 weeks. Bacterial loads in liver and spleen did not differ at any time point. These data suggest that the host-protective role of macrophage apoptosis in TB may be restricted in time and tissue compartment, following the induction of adaptive immunity and in the period when bacterial burden in the lung is normally held at a plateau level in wild-type mice infected with wild-type Mtb. That notion is supported by data from aerosol infection of protein kinase R (PKR)−/− mice [75]. These mice exhibit increased macrophage apoptosis during TB and have lower lung CFU than wild-type mice at 70 days post-infection but not at 21 days.
The enhanced intracellular survival (eis) gene of Mtb is a more recently described pro-survival factor that acts through an ROS-dependent pathway reminiscent of secA2 and nuoG [76]. The phenotype of RvΔeis differs, however, by increasing a caspase-independent cell death that is not clearly apoptotic in nature. This death is not accompanied by a strong TUNEL signature or caspase activation and it is only partially blocked by the pan-caspase inhibitor z-VAD-fmk. The Eis protein also modulates autophagy, and the autophagy inhibitor 3-methyladeneine enhances the survival of macrophages infected with RvΔeis. Following low dose aerosol challenge of wild-type C57BL/6 mice, lung and spleen bacterial burden was no different between RvΔeis, wild-type H37Rv or a complemented mutant strain but PI-positive cells and inflammation (IL-6 and TNF-α levels) were higher in mice infected with RvΔeis. Despite the common theme of ROS reduction, the data suggest that the Eis protein inhibits a predominantly necrotic rather than apoptotic macrophage death mode. This may be explained by its mechanism of action, based on acetylation of DUSP/MKP-7 and inhibition of JNK-dependent autophagy and ROS generation (section 5.4)[77].
An anti-apoptotic function was proposed for the Mtb protein MPT64 acting in a pathway involving NF-κB, miR21, and Bcl-2 [78]. The Mtb Rv3654c and Rv3655c genes were shown to participate in suppressing extrinsic apoptosis by reducing the availability of caspase-8 which has the effect of promoting regulated necrosis (section 2.2) [79]. Finally, protein kinase E of Mtb was reported to suppress macrophage apoptosis, specifically in the context of nitric oxide stress [80].
Far less is known about Mtb genes required for apoptosis induction. The Mtb 6 kDa early secretory antigenic target (ESAT6) is a pore-forming, virulence-associated gene product exported by the Esx-1 secretion system in a complex with 10-kDa culture filtrate protein [81]. The pore-forming function of ESAT6 is a leading candidate for Mtb-mediated LMP that precedes several forms of infection-induced cell death. There is considerable data linking ESAT6 and the Esx-1 secretion system to regulated necrosis of Mtb-infected macrophages (section 3.2) but ESAT6-mediated caspase upregulation and induction of THP-1 apoptosis (defined by annexin-V binding and sensitivity to pan-caspase inhibition) has been described [82]. It is uncertain whether these data obtained with immortalized monocytic cells reflect death mechanisms in primary macrophages infected Mtb. Others found that M. bovis BCG, which lacks ESAT6/CFP10 and the Esx-1 secretion system, is a potent inducer of apoptosis [61, 83, 84].
3.1.2 Efferocytosis in TB defense
Cells undergoing apoptosis release “find me” signals (e.g. CXC3CL1, lysophosphatidylcholine, sphingosine-1-phosphate, ATP, and UTP) that attract phagocytic cells for the clearance of apoptotic bodies [85]. Which of these or other possible signals operate in TB is presently conjectural but available data hint at a role for CX3CL1 and its receptor CX3CR1 [86]. While ATP/P2X7R interactions are implicated in TB defense [87], P2Y2 is the sensor for the small amounts of nucleotides released from apoptotic cells and so far this receptor has not been linked to TB. Neutrophils could respond to “find me” signals but the non-inflammatory clearance of apoptotic corpses is maintained by lactoferrin released from apoptotic cells. Lactoferrin serves as a “stay away” signal specifically inhibiting neutrophil but not MP migration to the vicinity of apoptotic cells [88].
Caspase-dependent exposure of PtdSer on the outer leaflet of plasma membranes is the best studied “eat me” signal although multiple signals may be required to promote efferocytosis [85]. Apoptotic corpses are recognized by a variety of tethering, signaling and/or phagocytic receptors on responding MPs (e.g. CD36, integrins, scavenger receptors, TAM receptors, TIM4, lectins, and RAGE), some of which require bridging molecules (e.g MFG-E8 and thrombospondin). These interactions initiate efferocytosis primarily by activating the Rho family GTPase Rac via CrkII-Dock180-ELMO or LPR1/MEGF10-GULP-ABCA1/ABCA7 pathways. The internalization process of efferocytosis resembles macropinocytosis more than receptor-mediated phagocytosis and delivers apoptotic bodies to acidified phagolysosomes [89]. Specificity is further ensured by the expression of “don't eat-me” signals on viable sells (e.g. CD47 which binds to SIRPα to inhibit efferocytosis) [90].
The potential for efferocytosis to enhance the innate antimycobacterial properties of MPs was first proposed by Fratazzi et al. [91] in experiments using an apoptosis-inducing strain of M. avium. Adding naïve macrophages to cultures of infected, apoptotic macrophages reduced mycobacterial viability in a contact-dependent manner that was not seen if the infected macrophages were made necrotic. Definitive evidence for efferocytosis-dependent killing of Mtb was provided by Martin et al. [92]. Primary mouse macrophages infected with mCherry-expressing H37Rv in vitro were shown to undergo apoptosis via the intrinsic pathway with subsequent delivery of apoptotic bodies to naïve macrophages. Transfer of Mtb from necrotic macrophages was observed in parallel and both phenomena were demonstrated in vivo after transfer of Rv/mCherry-infected DiO-labeled CD45.2+ macrophages into CD45.1+ recipients. Efferocytosis-dependent killing of H37Rv was demonstrated in vitro by blocking this pathway with anti-TIM4 mAb or by pre-treating naïve macrophages with forskolin or prostaglandin (PG)E2 that inhibit efferocytosis through a mechanism involving increased intracellular cAMP. Bacilli ingested by efferocytosis were delivered to capacious, acidified phagolysosomes that co-localized with LAMP1 and vacuolar H+-ATPase. An antimicrobial effect of efferocytosis in vivo was demonstrated by intraperitoneal transfer of H37Rv-infected arachidonate 5-lipoxyengase (Alox5)−/− macrophages into Rag−/− recipients. Alox5−/− macrophages are more prone to apoptosis following Mtb infection (section 3.2). In this infection/transfer model, recipients treated with anti-TIM4 had roughly 2-fold higher bacterial load in spleen and lung than recipients treated with isotype control mAb. The data clearly demonstrate the potential for efferocytosis-dependent killing of Mtb in TB although results from in vivo challenge with RvΔnuoG [68] suggest that this operates mainly at later stages of TB disease (section 3.1.1).
The potential for efferocytosis to promote cross-presentation of mycobacterial antigens for MHC class-I- and CD1-dependent T cell activation was initially reported by Schaible et al. [54]. Restimulation of CD8+ T cells from donors with latent TB infection co-cultured with Mtb-infected macrophages required the intermediary function of naïve DCs that acquired antigen through efferocytosis. Similar results were achieved when naïve DCs were pulsed with sterile, purified vesicles derived from Mtb-infected MPs. These investigators subsequently demonstrated that immunization with Mtb antigen-containing vesicles can cross-prime naïve CD8+ T cells in vivo, which requires DC homing to lymph nodes [93]. They further showed that apoptotic bodies possess adjuvant activity mediated by Toll-like receptor (TLR)-2, and that CD4+ T cells are also stimulated in vivo. Evidence of efferocytotic antigen transfer to DC and cross-priming occur during TB disease was reported by Divangahi et al. [94] in experiments using intratracheal transfer of Mtb-infected Alox5−/− macrophages. Transfer of these apoptosis-prone macrophages accelerated the activation of TB10.4-specific CD8+ T cells and depended on apoptosis (suppressed by inhibition of caspase-8 and caspase-9) as well as DC. Altogether, the data suggest that the capacity of virulent Mtb to suppress the apoptotic death of infected macrophages could contribute to the delayed priming of adaptive immunity that characterizes the early host response in TB.
3.2 Necrosis of Mtb-infected macrophages
The capacity of virulent Mtb to suppress apoptosis implies an alternative exit strategy for the bacillus to reach the extracellular environment. The first evidence of programmed necrosis in Mtb was described by Duan et al. [95] using in vitro infection of human monocyte-derived macrophages with attenuated H37Ra. They found that some infected macrophages died by apoptosis while others died by necrosis, which correlated with MPT. The induction of MPT and CYPD-dependent necrosis by Mtb was subsequently shown to be inhibited by cyclosporin A [96] and to be more potently induced by H37Rv than H37Ra [97]. It was later shown that PGE2 induced by H37Ra protects against mitochondrial PTP formation and necrosis while lipoxin (LX)A4 induced by H37Rv and Erdman suppresses PGE2 production and promotes necrosis [98]. The direct antimicrobial effect of apoptosis against Mtb observed under in vitro culture conditions was not seen when infected cells died by necrosis. These data supported the concept of programmed necrosis as means for replication-competent bacilli to exit host macrophages for spreading infection.
The apoptotic death of infected macrophages confines Mtb in membrane-bound vesicles but this potentially host-protective outcome is lost if the apoptotic bodies decompose before they are cleared by efferocytosis. Plasma membrane stability is another determinant of the fate of Mtb-infected macrophages. This was first revealed by experiments showing that virulent Mtb H37Rv inhibits the cross-linking of annexin-I bound to PtdSer, which serves to stabilize apoptotic membranes [99]. In a series of carefully conducted, mechanistic studies the capacity of Mtb-infected macrophages to repair membrane damage was shown to be a major determinant of an apoptotic versus necrotic demise [94, 100]. Cell fate in this model hinges on eicosanoid regulation of the lysosome-mediated membrane repair machinery, which is induced by PGE2 via the E2 receptor. Virulent H37Rv promotes elevated LXA4 levels in macrophages, which in turn downregulates cyclooxygenase (COX)2 mRNA thereby reducing PGE2 and its protective effects. Infection of Alox5−/− macrophages that cannot synthesize LXA4 results in increased apoptosis even with H37Rv. Conversely, prostaglandin E synthase (Pges)−/− macrophages cannot produce PGE2 and die by necrosis even with H37Ra infection. These data suggest that virulent and attenuated Mtb strains trigger a common pathway that can terminate in apoptosis if mitochondrial and plasma membrane integrity is protected by PGE2, or progresses to necrosis if LXA4 predominates. How virulent Mtb strains induce LXA4 production and what causes the membrane injury counteracted by repair remains to be determined. The pore forming activity of ESAT6 is one suggested mechanism for LMP as well as plasma membrane microdisruptions [101]. Another candidate is the product of the Mtb Rv3903c gene (CpnT) whose C-terminal fragment induces necrotic cell death with plasma membrane disruption that is not blocked by a pan-caspase inhibitor (therefore not caspase-3 mediated apoptosis or caspase-1 dependent pyroptosis) and not blocked by the RIPK1 inhibitor necrostatin-1 (therefore not necroptosis) [5;102].
The in vivo relevance of excess lipoxins as detrimental to TB defense is supported by genetic data from the mouse and zebrafish models, and in human TB patients. Alox5−/− mice are more resistant to aerosol TB than wild-type controls and make a more robust Th1 biased adaptive immune response [103]. Reminiscent of kinetics of RvΔnuoG growth in vivo, lung bacterial load in Alox5−/− mice is only lower than wild-type ≥21 days post-infection, suggesting that its protective effect is not manifest during the primarily innate phase of TB defense. A zebrafish mutant hypersusceptible to M. marinum mapped to the lta4h locus encoding leukotriene A4 hydrolase, which catalyzes the last step in LTB4 synthesis [104]. Zebrafish with this mutation are deficient in pro-inflammatory LTB4 and over-produce anti-inflammatory lipoxins. Human resistance to TB and multibacillary leprosy is associated with heterozygosity of LTA4H polymorphisms that correlate with differential production of LTB4 [104, 105]. These data suggest that an optimal balance of pro- and anti-inflammatory eicosanoids protects against the adverse effects of poorly controlled bacillary replication on one hand and against damaging immune pathology on the other. Whether these in vivo phenotypes reflect differential regulation of MP apoptosis and necrosis, or some other effects on immunity of these pleiotropic eicosanoids remains to be determined
Inflammasome activation and the induction of pyroptosis is postulated as a defense mechanism that deprives rapidly growing intracellular bacterial pathogens of a growth niche. Many pathogenic bacteria have mechanisms to subvert this response [55]. Slow growing Mtb requires the intracellular niche of MPs, so its replication would be restricted the rapid induction of programmed necrosis of its host cells. In this regard, the Mtb Rv3364c gene product was reported to suppress caspase-1 and pyroptosis in macrophages [106].
Lytic viruses induce host cell necrosis after an optimal period of replication to a “burst size” intracellular load. Similar dynamics were suggested for Mtb, which triggers an atypical necrosis subroutine in macrophages at intracellular loads in the range of 20-40 bacilli [106,108]. Dying cells exhibit early apoptotic features of PtdSer externalization and nuclear pyknosis, but progress within hours to necrosis without nuclear fragmentation or apoptotic vesicle formation. This death is caspase-independent and does not require pro-apoptotic Bcl2 family proteins. This death differs from the necrosis of bioenergetic collapse by the absence of osmotic swelling, and it differs from pyroptosis and pyronecrosis, being independent of caspase-1 and cathepsin B [108]. It is not inhibited by cyclosporin A and is therefore distinct from CYDP-dependent necrosis [107]. Death is initiated upon LMP, followed by lipolytic attack on mitochondrial, nuclear and outer cell membranes. It could therefore be classified as a form of lysosomal cell death but is atypical in its dependence on lipase more than protease activities. The bacterial determinants of this death mechanism remain undefined, but it requires one or more genes of the PhoPR regulon since a ΔphoPR mutant of H37Rv fails to cause LMP or kill macrophages at MOI 25 [108]. In contrast, BCG and a defined RvΔRD1ΔespA mutant lacking the ESX-1 secretion system are fully capable of provoking necrosis if introduced at high MOI. Both of those strains are capable of only limited intracellular replication, so this pro-necrotic phenotype is not evident when they are introduced at low MOI. The dynamics of Mtb burden per cell in MPs after aerosol challenge of mice fits a burst size model of necrosis [109]. Neutrophils are the predominant phagocytes harboring Mtb cell during periods of unrestricted bacillary replication [109, 110]. This is consistent with the inflammatory nature of necrotic cell death which recruits neutrophils and absence of the “stay away” signal lactoferrin that is associated with apoptosis [13, 88].
3.3 Programmed death of other myeloid cells in TB
Macrophages have been the focus of research on cell death in TB but data from the mouse model and human TB indicate that DC and neutrophils are important and in some circumstances the predominant phagocytes harboring Mtb in vivo [109-111]. Neutrophils are readily identified by common laboratory methods but distinguishing between tissue-resident macrophages, recruited macrophages, monocytes, and DC is more challenging and some have questioned whether these cell types represent distinct lineages or simply alternative and plastic phenotypes a common monocytic progenitor [112-114]. Recognizing this controversy, we will refer to DC as defined by the authors of cited publications who used generally accepted methods to isolate and phenotype these cells.
The limited literature on the fate of Mtb-infected DC reflects many of the same challenges to interpreting the often contradictory literature on macrophage cell death. An early study compared the activation and survival of C56BL/6 mouse bone marrow derived DC challenged with Erdman at MOI 10 [115]. Immature DC matured in response to infection and survived >48 hours but (unlike macrophages) were unable to kill intracellular bacilli even with IFN-γ activation. The authors noted that high rates of cell death (trypan staining) in DC and macrophages at later time points. More recently, Ryan et al. [116] reported that human peripheral blood monocyte-derived DC undergo necrosis (PI staining) that was caspase-independent (not rescued by Q-VD-Oph) and lacked nuclear fragmentation but did feature DNA cleavage (nucleosomal particle ELISA) despite no activation of the executioner caspases 3 and 7. The induction of DC death required live bacilli and was triggered by H37Rv and H37a. In contrast, a study using bone marrow-derived DC from C57BL/6 mice and H37Rv (MOI 10) described activation of inflammasome and executioner caspases 3/7 with along with apoptosis (defined by TUNEL) but no necrosis (defined by adenylate kinase release) [117]. Based on these and other data the authors concluded that Mtb induces DC apoptosis dependent on Esx-1 but does not cause pyroptosis or pyronecrosis. We are left to conclude that Mtb can infect and activate DC and that apoptotic or necrotic death might ensue depending on variables likely to include the origin, subtype and activation state of the DCs prior to infection. Necrotic death of DC could plausibly delay immune priming while an apoptotic death might facilitate priming. These are important considerations since there is substantial evidence that DC are the major phagocytes that convey viable bacilli to lung-draining lymph nodes for adaptive immune priming [110, 118]. The kinetics of these events has a major impact on the subsequent outcome of disease at least in mice [119].
3.3.1 Neutrophil cell death in TB
Much recent interest has focused on the roles that neutrophils play in TB [120]. A fascinating dynamic has been proposed for the early events following inhalation of Mtb, where neutrophils accelerate immune priming by undergoing infection-induced apoptosis and passing bacilli to migratory DC in a manner that facilitates DC trafficking to the lymph node [68, 121]. Evidence that virulent Mtb strains inhibit neutrophil apoptosis and thereby delay immune priming was obtained using the pro-apoptotic Rv/ΔnuoG mutant [68]. In contrast, Mtb H37Rv was reported to trigger apoptosis of human peripheral blood neutrophils that was dependent on TLR2 and independent of TNF-α [122]. Adding complexity to this interaction, comparison of two different clinical Mtb isolates identified one strain that strongly induced human neutrophil apoptosis while the other strain did not induce apoptosis [123].
While neutrophils may exert a host protective function as an early event in TB, they also appear to be important mediators of tissue injury at later stages of disease if they are present in excess as occurs with poorly controlled Mtb infection in susceptible hosts [65, 124, 125]. Neutrophil lifespan is prolonged in I/St mice with TB, which may be a factor in the increased neutrophil accumulation and lung damage seen in that highly susceptible mouse strain [126]. NETs may contribute to lung tissue damage [47] and ETosis is another fate that has been described for human neutrophils and macrophages infected with Mtb in vitro [46, 127], in the mice with poorly controlled TB in vivo [109]. While NETs exert antimicrobial activity against a range of bacteria, this activity does not extend to Mtb [127]. Release of traps by human macrophages is accelerated by IFN-γ [46] but NETs were identified in lung lavage of IFN-γ−/− mice with TB [109]. The diverse fates of Mtb-infected neutrophils in TB feature in the host-protective and damage-inducing roles that these cells play in TB pathogenesis.
3.4 Translation opportunities
Manipulation of apoptosis in the context of vaccination holds the most promise compared other therapeutic goals for early translation of knowledge about cell death in TB. The pro-apoptotic recombinant strain BCGΔureC::hly+ expresses the pore-forming listeriolysin of L. monocytogenes and lacks urease C to ensure an optimal intraphagosomal pH for listeriolysin activity. This strain is more immunogenic than the parental BCG for type 1 responses and uniquely induces type 17 as well [128, 129]. These results are attributable to exploitation of the the efferocytotic pathway discussed in section 3.1.2. The human vaccine candidate based on this technology, VPM1002, demonstrated safety in a phase I clinical trial [130] and a phase II trial is currently underway.
Opportunities for adjunctive TB therapies that directly enhance apoptosis are less obvious and could risk killing necessary cells (e.g T cells) and/or generating an excess of apoptotic bodies at risk for secondary necrosis. Nonetheless, a more comprehensive understanding of how different fates for MPs and neutrophils influence the effectiveness of host defense and the quantity and quality of immune pathology in TB is vital to understanding TB pathogenesis and new targets for treatment and diagnosis.
3.5 Summary of cell death in TB
The diverse outcomes and conclusions of published studies on cell death in TB (Table 1) reflect differences in host species and strains, mycobacterial species and strains, cell types and their activation state, conditions of infection, and the likelihood that multiple cell death programs can be simultaneously activated in cells stressed by Mtb infection. These variables determine which fate predominates in particular experimental systems. Results from many studies support the concept that apoptosis of macrophages and neutrophils contributes mainly host-protective effects, while macrophage necrosis is mainly linked to adverse outcomes. However, these are clearly not absolute paradigms since apoptosis can promote disseminated infection and accelerated necrosis might limit intracellular bacillary replication.
Table 1.
Myeloid cell death and host defense in TB
Cell death type | Effects on host-pathogen interaction | Consequences for host defense | References |
---|---|---|---|
Extrinsic and intrinsic MΦ apoptosis | Eliminates replication niche Induces anti-inflammatory cytokines Lactoferrin inhibits neutrophil recruitment Packages Mtb and antigens in apoptotic vesicles |
Lower bacillary load Less immune pathology Less immune pathology Efferocytosis by DC promotes immune priming and cross presentation Efferocytosis by MO kills Mtb Efferocytosis promotes spreading infection |
17;18;59; 61-65 3;4 88 54;93;94 91;92 57;68 |
CYPD-dependent necrosis of MΦ MΦ necrosis due to inhibition of membrane repair MΦ necrosis with high Mtb burden CpnT-mediated MΦ necrosis |
Releases bacteria to extracellular space Induces pro-inflammatory cytokines |
Promotes spreading infection and transmission More immune pathology |
95;96 94;100 107-109 102 |
MΦ ETosis | Unknown | Unknown | 46 |
DC apoptosis | Packages Mtb and antigens in apoptotic vesicles | Unknown | 117 |
DC necrosis | Unknown | Unknown | 116 |
Neutrophil apoptosis NETosis |
Antigen transfer to DC Binds Mtb to extruded DNA but no antimicrobial effect in vitro |
Accelerated immune priming Unknown -restricted spread of Mtb? -increased tissue injury? |
68 109;127 |
MΦ, macrophage.
Given the complex interplay of multiple host and bacterial mechanisms to promote or prevent cell death, more research with in vivo approaches is needed to identify what truly matters in TB disease. A comprehensive model must also integrate a multitude of common factors that are often excluded in reductionist experimental systems. Examples include the effects of metabolic disorders like diabetes and hyperlipidemia where methylglyoxal, oxidized LDL cholesterol, or an excess of intracellular free cholesterol that promote apoptosis or necrosis [131, 132]; cytokines, such IFN-γ that promotes survival of macrophages at low MOI but accelerates necrosis with high bacillary loads [133, 134]; and apoptosis induced by CTL. The best studies will confirm unequivocally that cells thought be to dying are indeed dying and ideally use multiple unrelated assays if making a case for one particular fate.
4. Overview of Autophagy
Autophagy (“self-eating”) is an evolutionarily conserved pathway in cell biology that serves to control cytoplasmic content including organelles, to recycle chemical resources in bulk, and to regulate cellular functions under basal conditions and in response to variety of stresses. Macroautophagy refers to the isolation of cytoplasmic content by the formation of lipid bilayer vacuoles called autophagosomes which fuse with lysosomes to degrade their cargoes. Macroautophagy is the autophagic process most relevant to TB and will be simply called “autophagy” in this review. Other autophagic processes such as microautophagy (direct sequestration of cytoplasm within lysosomes) and chaperone-mediated autophagy that targets proteins with a specific signal sequence for degradation are reviewed elsewhere [135].
Autophagy is controlled primarily by Atg proteins, a family with more than 30 members that were revealed in screens for yeast mutants defective for autophagy [136]. The Atg family and other proteins operate in a complex conjugation cascade; many of the interactions and multiple functions of these players are incompletely understood, particularly in mammalian cell systems. A simplified scheme for autophagy includes four subgroups of core proteins involved in three major steps of vesicle formation. Autophagy is initiated by the Atg1/ULK1 complex comprising Unc-51 like autophagy activating kinases 1 and 2 (ULK1/2), Atg13, FIP2000, and Atg101 [137]. Initiation is under negative and positive regulation by mTOR and AMP kinase (AMPK), respectively, as well as mTOR-independent pathways. The Vps34 complex comprised of the class III phosphatidylinositol-3 kinase Vps34, Beclin 1 (homolog of yeast Atg6), p150 (Vps15 in yeast) and Atg14, also participates in autophagy induction. Together, these two complexes promote vesicle nucleation with formation of the “omegasome”, a cup-shaped protrusion from the endoplasmic reticulum (ER). Although other vesicle nucleation sources have been described, ER is the best understood. An ensuing conjugation cascade directs formation of a “phagophore” from repurposed ER, building a double-layered “isolation membrane” on the ring-like base of omegasome. Elongation of this membrane requires two ubiquitination-like reactions and two protein complexes. The first involves conjugation of the ubiquitin-like protein Atg12 to Atg5, which then form a complex with Atg16L. The second reaction follows cleavage of microtubule associated protein-1 light chain-3 (LC3), yielding cytosolic LC3-I (Atg8) that is converted to LC3-II through conjugation to phosphatidylethanolamine, tethering LC3-II to the isolation membrane. Through mechanisms not yet fully defined, these complexes facilitate expansion and bending of the isolation membrane. Autophagosome formation is completed upon closure of the isolation membrane, sequestering the targeted cytosolic constituents. In the final step, mediated by SNARE proteins, the autophagosome fuses with a lysosome to form an “autolysosome” where digestion of the sequestered vacuolar contents proceeds [138].
The ratio of LC3-II to LC3-I on immunoblots and a punctate as opposed to cytosolic distribution of LC3 identified by fluorescence microscopy are commonly used as assays to quantify autophagy. They are useful but can be misleading; alternative measures of autophagic flux may be required to distinguish between accelerated autophagosome biogenesis versus reduced turnover [139]. In the related but distinct pathway called LC3-associated phagocytosis (LAP), LC3-II is recruited to conventional, single-membrane phagosomes and promotes their fusion with lysosomes. LAP has been implicated in the clearance of apoptotic and necrotic corpses among other settings [140].
Autophagy proceeds at basal levels to maintain the homeostasis of unstressed cells and is further activated by a variety of factors including starvation, hypoxia, extremes of pH or temperature, growth factor withdrawal, oxidative stress, ER stress, and infection [141-143]. Targeting signals (e.g. externalization of cardiolipin to the limiting membrane of damaged mitochondria [144]) initiate autophagosome formation, which is subject to a wide range of regulatory influences, some dependent and some independent of mTOR and AMPK [145]. Among its many functions, autophagy provides a mechanism to counteract the various strategies employed by intracellular pathogens to use host cells to their advantage. The detection and autophagic destruction of intracellular pathogens, also called xenophagy, has been linked to host defense against certain viruses (e.g. HIV and influenza A virus), bacteria (e.g. Shigella, Salmonella, Group A Streptococcus, and Listeria), fungi (e.g. Candida and Cryptococcus), and protozoa (e.g. Toxoplasma) [146-148]. Conversely, certain intracellular pathogens subvert autophagy to promote infection [149]. Xenophagy plays an integral role in innate and adaptive immunity to infection with Mtb that is the focus of this review.
5. Autophagy in TB
As noted in section 1, the capacity of Mtb to inhibit phagosome biogenesis and survive inside macrophages that eliminate most other phagocytosed bacteria is an essential virulence mechanism. Despite this countermeasure, a proportion of internalized bacilli are directed to acidified compartments and this is accelerated by macrophage activating factors such as IFN-γ. The role of autophagy in this successful outcome was first identified by Gutierrez et al. in 2004 [150]. They reported that induction of autophagy in Mtb-infected macrophages by starvation or by treatment with the mTOR inhibitor rapamycin delivered bacilli to phagolysosomes and increased co-localization of Mtb with LC3 and Beclin 1. They also found that autophagy was induced by treatment with IFN-γ or transfection with the IFN-γ effector LRG-47. Since that initial report, a substantial body of evidence has established the major role that autophagy plays in TB defense.
5.1 Targeting Mtb for autophagic elimination
Following receptor-mediated phagocytosis, most Mtb bacilli reside in early endosome-like vacuoles that resist acidification or fusion with lysosomes. This niche is permissive for some movement of macromolecules to and from the cytoplasm, to other vesicular compartments, and even the extracellular environment [151-153]. Such bidirectional transfer is required for Mtb to acquire nutrients and manipulate host cell functions but also confers vulnerability to host defense [154, 155]. The LMP generated at least in part by ESAT6 (section 3.1.1) enables detection of PAMPs by innate cytosolic sensors. Genomic DNA of Mtb activates the STING-dependent cytosolic pathway, leading to ubiquitination of bacilli and their delivery to autophagosomes in a process dependent on the ubiquitin-autophagy receptors p62 and NDP52, and the serine/threonine protein kinase TBK1 [154]. Data from the zebrafish model of M. marinum infection corroborated the involvement of RD1 and STING in autophagic defense against mycobacteria, and further identified the participation of the autophagy modulator DRAM1 in this response [156]. The contribution of autophagy to TB defense in mammals was demonstrated by the increased susceptibility of Lyz-Cre-Atg5fl/fl mice with targeted deletion of Atg5 in myeloid cells [154, 157].
Redundant pathways for innate recognition and autophagic targeting of intracellular Mtb have been identified. The ubiquitin ligase Parkin, which mediates the autophagic elimination of dysfunctional mitochondria (mitophagy), was shown to play a critical role in the colocalization of ubiquitin and Mtb in macrophages [158]. The dual role of Parkin in mitophagy and xenophagy for intracellular bacteria is unsurprising since mitochondria evolved from intracellular prokaryotes. Once tagged with ubiquitin, bacteria may be targeted by cytoplasmic sequestasome (p62/SQSTM1)-like receptors (SLRs). By virtue of its ubiquitin-associated domain and LC3 interaction region, p62 serves as an adaptor bridging ubiquitinated bacilli and LC3 on the preautophagosome [146]. A role of p62 in recruiting an E3 ligase to ubiquitinate Mtb has also been proposed but the precise targets for ubiquitination on bacterial cells and the full spectrum of mediators for this conjugation reactions are incompletely defined [159].
Innate cytosolic receptors bind PAMPS, providING surveillance for intracellular infection. The NLR subfamily receptor NOD2 binds bacterial peptidoglycans like muramyl dipeptide and is activated in macrophages infected with Mtb [160]. Signaling downstream of NOD2 in Mtb-infected macrophages induces nuclear translocation of NF-κβ, resulting in increased expression of the autophagy-related genes IRGM, LC3, and ATG16L1, the antimicrobial peptide human cathelicidin (hCAP-18/LL-37), and the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α [161].
The capture of Mtb in autophagosomes that subsequently fuse with lysosomes delivers bacilli to a hostile, acidified compartment replete with hydrolytic enzymes and free radicals of oxygen and nitrogen. Microbicidal activity against Mtb in autolysomes was recently shown involve another role for p62, namely the delivery of ribosomal precursor and other ubiquitinated proteins from the cytosol to autolysosomes where they are cleaved to generate “neo-antimicrobial peptides” [162]. Whether the cytosolic precursor proteins are delivered directly to autolysosomes containing Mtb or if killing depends on subsequent fusion of peptide-containing vesicles with Mtb-containing phagosomes remains to be determined.
5.2 Regulation of autophagy in TB
Following inhalation of Mtb by naïve mice, lung bacterial burden rises in a log linear manner for ~3 weeks until bacterial replication is curtailed by adaptive immunity and particularly IFN-γ produced mainly by Th1 cells [163]. Inducible nitric oxide synthase was considered the major host-protective macrophage response to IFN-γ activation but it is now clear that accelerated autophagic flux is another important effect of IFN-γ [150]. Host-protective effects of IFN-γ are mediated in part by members of the immunity-related GTPase family (IRG proteins, also called p47 GTPases). Two pathways for autophagy induction by IFN-γ have been reported. One involves Irgm1, a STAT1-dependent IRG previously called Lrg47 (IRGM in humans) [164]. The alternative IFN-γ autophagy pathway is independent of STAT1 but requires JAK 1/2, PI3K, and p38 mitogen-activated protein kinase (MAPK) [165]. The Th1 cytokine TNF-α also induces autophagy and there is at least indirect evidence for this effect in Mtb-infected macrophages [166]. In contrast, the Th2 cytokines IL-4 and IL-13 inhibit autophagy at least in in mice [167]. There is evidence for and against autophagy induction by Mtb in the absence of exogenous activators like IFN-γ, and different mycobacterial strains may be high or low inducers [168]. Indeed, the suppression of autophagy may be a virulence mechanism of the bacillus (section 5.4). Infection is associated with increased mTOR activity, indicating that innate activation autophagy by mycobacteria is independent of mTOR [168].
Vitamin D receptor (VDR) signaling complements IFN-γ for Mtb clearance by a pathway that may be highly significant in humans but is absent in mice. Shin et al. [169] reported that stimulation of TLR2/1 by the Mtb lipoprotein LpqH induces autophagy in human monocyte cultures supplemented with the pro-vitamin D hormone calcifediol (25-dihydroxyvitamin D3) [169]. Their data support a model where signaling by TLR2/1 and CD14 induces Ca2+ influx through PLC-γ activation. Calcium flux results in AMPK phosphorylation and activation of p38 MAPK that in turn upregulates VD3 1A hydroxylase, which catalyzes the hydroxylation of calcifediol to biologically active calcitriol (1,25-dihydroxyvitamin D3). Calcitriol binding to VDR induces expression of cathelicidin that increases the expression Beclin-1 and Atg5 and promotes autophagic flux [170,171]. The antimicrobial activity of IFN-γ activated human macrophages requires a sufficient level of calcifediol, linking the VDR-stimulated autophagic pathway the protective effect of IFN-γ in human TB [172]. The VDR pathway is not required for autophagy induction by rapamycin or starvation [172]. Discovery of the VDR pathway and its role in TB defense stimulated renewed interest in the therapeutic potential of vitamin D in human TB (section 5.6).
A number of other mediators and pathways have been linked to autophagy induction in Mtb-infected macrophages. Given the links between intracellular and cell surface innate receptors and autophagy, it is unsurprising that the TLR4 ligand LPS and IL-1β (via Myd88 and TBK1) have been linked to the autophagic elimination of Mtb [173,174]. The growing list autophagy activators in Mtb-infected MPs includes the scavenger protein Apoptosis Inhibitor of Macrophages (a target of liver X receptor activation) that was also shown to inhibit apoptosis of THP1 cells challenged with Mtb at low MOI [175]. Extracellular ATP (a DAMP discussed in section 2) promotes autophagy via P2X7 receptor, linking autophagy induction and the host response to necrosis [60]. MicroRNA-155 has been linked to TB defense in several reports including evidence that its induction is dependent on ESAT6, that it regulates apoptosis of M. bovis BCG-infected macrophages, and that it accelerates autophagy and the killing of intracellular Mtb by suppressing the Ras homolog Rheb [176-178]. It is likely that a wider array of pathways and mediators regulating xenophagy in Mtb-infected MPs will be identified in the future. Prioritizing these pathways in terms of their protective effects against TB disease in vivo will help direct translational research on harnessing autophagy for TB treatment (section 5.6).
5.3 Autophagy-mediated regulation of immunity in TB
Autophagy participates in TB defense beyond its effector function to kill intracellular bacilli. Since Mtb inhibits conventional phagosome biogenesis, its capture and degradation in autolysosomes provides an opportunity to generate peptides for loading onto MHC class II molecules. This outcome was demonstrated by Seto et al. [159] in murine bone marrow derived DC and DC2.4 cells infected with Mtb Erdman. The recruitment of MHC II followed the capture of ubiquitinated bacilli in autophagosomes and fusion with lysosomes. In contrast to Erdman, BCG was not ubiquitinated in DC. This is consistent with a requirement for ESAT6 (which is deleted from BCG) to cause LMP. Treating BCG-infected DC with rapamycin, however, increases colocalization of bacilli with autophagosomes and enhances the immunogenicity and protective efficacy of BCG [179].
Autophagy also functions to modulate inflammation through targeted degradation of cytokines such and TNF-α and IL-1β, and by preventing the accumulation of damaged mitochondria that activate the NLRP3 inflammasome via ROS and mtDNA [180-184]. The effect of this regulation to limit inflammation in TB was reported by the Castillo et al. [157]. Pulmonary TB in autophagy-deficient Atg5fl/fl LysM-Cre+ mice challenged with a low aerosol dose of Mtb H37Rv featured increased bacterial burden compared to wild-type controls but also increased neutrophilic inflammation with tissue necrosis, Th17 skewing, and elevated levels of IL-1α, IL-12, IL-17, and CXCL1. The authors concluded that the host-protective effects of autophagy in TB include modulation of the inflammatory response to infection. Bonilla et al. [185] similarly reported increased inflammation and bacillary load in autophagy-deficient Atg7−/− LysM-Cre+ mice challenged with a high intranasal dose of BCG. Cytokine levels were not reported and the authors attributed susceptibility to accelerated phagocytosis of mycobacteria due to increased surface expression of the scavenger (and Mtb) receptors MARCO and MSR1. An excess of p62 in the Atg7−/− macrophages bound KEAP1, a suppressor of the transcription factor Nrf2, resulting in increased expression of Nrf2 regulated genes including scavenger receptors. Parenthetically, these authors observed no difference in autophagy induction between BCG (lacking ESAT6 and the Esx-1 secretion system) and Mtb H37Rv.
5.4 Evasion of autophagy by Mtb
Evasion of the vesicular trafficking systems that deliver most phagocytosed bacilli to acidified phagolysosomes is a key to Mtb virulence and is mediated through inhibition of Ca2+ flux and Rab family GTPases [1]. Similarly, the bacillus has evolved countermeasures to evade xenophagy. Zhang et al. [186] reported that ectopic expression of ESAT6/CFP10 inhibited autophagosome formation in with H37Rv-infected RAW264.7 cells, along with reduced expression of Atg8 and other autophagy-related genes. Romangoli et al. [187] compared autophagic flux in human monocyte-derived DC infected with H37Rv, H37Ra or BGG, finding inhibition of autophagosome-lysosome fusion only in DC infected with H37Rv. The capacity to inhibit autophagic flux was restored by complementation of BCG with the Esx-1 region from Mtb and by complementation of H37Ra with the PhoP gene. They also showed that the Esx-1 dependent autophagic block exerted by H37Rv could be overcome with rapamycin.
The Mtb Eis protein, which inhibits infection-induced macrophage cell death through acetylation of DUSP16/MKP-7 and downregulation of JNK-induced ROS generation (section 3.1.1) inhibits autophagy as well [76]. Macrophage cytolysis triggered by RvΔeis was attributed to autophagic cell death but the relationship between autophagy and cytotoxicity is controversial since autophagy is commonly activated as a prosurvival response in stressed cells that may succumb to one or another subroutine of programmed death [51]. The Mtb cell wall component lipoarabinomannan is another virulence factor associated with autophagy inhibition. Phagocytosis of lipoarabinomannan-coated latex meads by RAW264.7 cells inhibited the accumulation of autophagosomes while beads coated with E. coli LPS or phophatidyl-myo-inositol of M. smegmatis had no suppressive effect [188]. Further research will likely reveal more host and pathogen countermeasures regulating the predominantly host-protective effects of autophagy in TB.
5.5 Genetic associations of autophagy and TB
A body of genetic evidence supports the relevance of autophagy as a biologically significant host defense mechanism in TB. Mice with targeted mutations in the autophagy-related genes Atg5, Atg7, and Irgm1 all demonstrate increased susceptibility to Mtb [154,157,185,189]. In a genome-wide siRNA screen in human (THP-1) cells infected with Mtb H37Rv or a several clinical isolates of different clades, Kumar et al. [190] identified 275 genes functionally involved in control of infection. Seventy-four of these genes defined essential components active against the spectrum of different Mtb isolates and within that subset, more than half were associated with the regulation of autophagy.
Several studies identified association of polymorphic alleles in autophagy-related genes with TB susceptibility or resistance. The complexity of these interactions is highlighted by several studies investigating polymorphisms in IRGM. The -261TT allele of IRGM was negatively associated with TB in a cohort from the west African country of Ghana but this applied only to disease caused by Mtb strains of the Euro-American lineage [191]. The -261TT allele (rs9637876) is predicted to eliminate binding sites for inhibitory transcription factors and therefore increase IGRM expression. In the same Ghanaian population, this allele was not associated with protection from TB caused by M. africanum or M. bovis. Several other studies variously identified associations of polymorphic IRGM alleles with protection or susceptibility to TB in African-American, Iranian, Chinese and Korean populations [191-194]. In some cases opposing effects on TB susceptibility were associated with the same allele in different populations. Analysis of 22 polymorphisms in 14 autophagy genes in an Indonesian population found an association of TB disease with polymorphisms in LAMP1 and MTOR but statistical significance was lost after correction for multiple comparisons [195]. These divergent associations may reflect differences in host genetic background, locally prevalent Mtb strains and/or environmental influences but regardless of the effect the data clearly link the IRGM gene to TB defense.
5.6 Therapeutic opportunities
The goal of TB elimination is hampered by challenges of compliance with prolonged multi-drug antibiotic regimens and the emergence of drug-resistant Mtb strains. This has stimulated interest in the potential for host-directed therapies (HDTs) to amplify endogenous effector mechanisms and accelerate the response to antituberculous chemotherapy [196]. Among the pathways that may be amenable to this approach, autophagy is an attractive candidate based on its anti-mycobacterial and anti-inflammatory effects in TB. Interestingly, the first line anti-TB drugs isoniazid and pyrazinamide were reported to induce autophagy selectively in Mtb-infected but not uninfected macrophages [196]. Activation of autophagy was dependent on ROS and attributed to bacteria-derived hydroxyl radicals produced in response to these drugs. In the fly model of M. marinum infection, these authors showed that that the anti-mycobacterial activity of the antibiotics depended at least in part on autophagy. The anti-protozoal drug nitazoxanide and its metabolite tizoxanide inhibit mammalian target of rapamycin complex 1 (mTORC1) signaling and stimulate autophagy by a mechanism attributed to suppression of the quinone oxidoreductase NQO1 [197]. This drug also kills replicating and non-replicating Mtb in broth culture but its antimicrobial activity was greatest in Mtb-infected macrophages and it was more potent than rapamycin. Drugs with dual antibiotic and autophagy-stimulating effects may particularly attractive candidates for TB treatment.
The best studied non-antibiotic effector of autophagy in the context of TB is vitamin D (section 5.2). Low serum levels of calcifediol are associated with increased TB susceptibility and adverse treatment outcomes in different geographic locations [198-203]. None of these studies established causation, and a cautionary note comes from a case-control study in Greenland where serum calcifediol levels below and above 75-140 nmol/l was associated with increased odds for TB [202]. Two trials have tested vitamin D supplementation in TB. The first of these was a randomized, double-blind, placebo-controlled trial conducted in Guinea-Bissau [204]. Adult patients starting TB treatment received 100,000 IU of cholecalciferol on enrollment and after 5 and 8 months of inclusion. Vitamin D treatment did not reduce the clinical TB severity score (primary outcome) or 12-month mortality. The second study enrolled, new smear-positive adult pulmonary TB patients in the United Kingdom who had corrected serum calcium concentration >2.65 mmol/l for randomization to receive four oral doses of 2.5 mg cholecalciferol or placebo given at baseline and then 14, 28 and 42 days after starting TB treatment. Supplementation was shown to increase serum calcifediol concentrations in the patients receiving intensive-phase TB treatment but did not significantly influence the time to sputum culture conversion (primary endpoint) across the whole cohort. Sputum culture conversion was, however, accelerated by vitamin D supplementation in the subgroup of participants having the tt genotype of the TaqI VDR polymorphism. Data from that trial were re-analyzed a separate publication that included only patients who fulfilled per-protocol analysis criteria [205]. In this patient group, vitamin D supplementation accelerated sputum smear conversion and resolution of lymphopenia and monocytosis, and it suppressed pro-inflammatory cytokines in the circulation and in cultures of Mtb antigen-stimulated whole blood.
Some drugs already in clinical use for other indications can stimulate autophagy and therefore might be considered for adjunctive therapy in TB. Rapamycin is used for immunosuppression in organ transplantation and to inhibit endothelial proliferation on coronary stents [206,207]. It potently suppresses mTOR via inhibition of mTORC1 [208]. Systemic immunosuppression is clearly an unacceptable effect for TB patients but this might be mitigated by delivery of rapamycin in inhalable particles [209]. Alternative drugs with less troubling side effects include statins that where shown to have activity against Mtb in vitro and in vivo [201,211]. A high-throughput screen for small molecules that restrict Mtb growth in macrophages identified activity for gefitinib and fluoxetine, both of which also stimulate autophagy [166]. Of these, gefitinib was tested in mice and restricted Mtb growth following aerosol infection of BALB/c mice.
Many other approved drugs are known to induce autophagy although their application to TB has not yet been reported. Among these, the oral anti-diabetic biguanide metformin is an attractive candidate. Metformin activates AMPK and stimulates autophagy as well as cell death in malignant cells and has attracted considerable interest in for cancer treatment [212]. Metformin also appears to stimulate autophagy by an AMPK-independent mechanism linked to increased expression of REDD1, which is a negative regulator of mTOR [213]. Zhang et al. [214] identified eight small molecule regulators of autophagy using an image-based high-throughput screen. Three of the compounds are U.S. Food and Drug Administration-approved anti-psychotic drugs, three are Ca2+ current inhibitors approved for cardiovascular indications (niguldipine, nicardipine, amiodarone), and one (loperamide) is approved for treatment of diarrhea. None of these compounds modulated mTOR phosphorylation, indicating that they induce autophagy through pathways distinct from rapamycin. Minoxidil and clonidine were identified in another screen for autophagy enhancers restricted to drugs previously used in humans without major side effects [215]. As more autophagy-promoting drugs are evaluated against Mtb it will be interesting to learn if particular pathways offer better efficacy advantages as HDTs for TB. At the same time it will be necessary and informative to test whether candidate HDTs adversely influence the balance of protective versus damaging immunity through mechanisms like mTOR inhibition. A cautionary, if unrelated, note is provided by the experience with glyburide in patients infected with Burkholderia pseudomallei (melioidosis). Koh et al. [216] reported that glyburide improved survival in diabetic melioidosis patients independent of glycemic control; an effect attributed to inhibition of inflammasome assembly. A contrasting effect of glyburide was later reported by Liu et al. [217] who found higher rates of septic complications in melioidosis patients treated with (unspecified) sulfonylureas and suppressive effects of glyburide on inflammatory cytokine production by peripheral blood mononuclear cells treated with B. pseudomallei antigen.
5.6 Conclusions
Autophagy provides a means for infected MPs to overcome the key virulence mechanism of Mtb; inhibition of phagosome biogenesis. Its contributions to TB defense include killing of intracellular bacilli, promoting MHC class II restricted antigen presentation, and modulating the inflammatory responses that cause tissue damage and the clinical manifestations of TB disease. Phagocytes use several cytosolic sensors recognize intracellular bacilli and tag them for destruction. Further research will likely identify additional cytosolic and cell surface receptors involved in the detection of Mtb and stimulation of autophagic flux. Autophagy is managed by a complex regulatory network with evidence of mTOR-dependent and independent pathways linked to TB defense. The relative contribution of these different pathways to positive outcomes in TB remains to be determined. Such knowledge might be leveraged for the optimal design of host-directed therapies that stimulate autophagy. The exciting translational opportunities for autophagy in TB will also be advanced by better understanding of the countermeasures that have evolved in the bacillus to escape detection and inhibit autolysosome biogenesis.
HIGHLIGHTS.
Cell Death and Autophagy in TB (Moraco and Kornfeld)
Phagocytes infected with M. tuberculosis may undergo apoptosis or necrosis
Apoptosis promotes immune priming and kills bacilli but can spread infection
Apoptosis can also facilitate spread of M. tuberculosis infection
Autophagy sends M. tuberculosis to acidified lysosomes and regulates inflammation
Apoptosis and autophagy may be leveraged for TB prevention and treatment
ACKNOWLEGEMENTS
Supported in part by NIH grant HL081149 (to H.K.).
Abbreviations
- AIF
apoptosis-inducing factor
- AMPK
AMP kinase
- Apaf-1
apoptotic protease activating factor
- BMM
bone marrow-derived macrophages
- cFLIP
cellular FLICE-like inhibitory protein
- cIAP
cellular inhibitor of apoptosis protein
- COX
cyclooxygenase
- CTL
cytotoxic T lymphocytes
- CYPD
cyclophilin D
- DAMP
damage-associated molecular pattern
- DC
dendritic cell
- FADD
Fas-associated death domain
- HrtA2/Ommi
high temperature requirement
- IL
interleukin
- IMM
inner mitochondrial membrane
- LT
leukotriene
- LX
lipoxin
- MAPK
mitogen-activated protein kinase
- MPT
mitochondrial permeability transition
- Mtb
Mycobacterium tuberculosis
- mTOR
mammalian target of rapamycin
- NET
neutrophil extracellular trap
- NK
natural killer
- OMM
outer mitochondrial membrane
- PG
prostaglandin
- PI
propidium iodide
- PtdSer
phosphatidylserine
- PTP
permeability transition pore
- RIPK
receptor interacting serine/threonine protein kinase
- Smac
second mitochondria-derived activator of caspases
- SLR
SodA, superoxide dismutase
- SLR
sequestasome-like receptor
- TEM
transmission electron microscopy
- TB
tuberculosis
- TLR
Toll-like receptor
- TNF
tumor necrosis factor
- TNFR
tumor necrosis factor receptor
- TRADD
TNFR-associated death domain
- TUNEL
terminal deoxynucleotidyl transferase dUTP nick end-labeling
- VDR
vitamin D receptor
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Vergne I, Chua J, Singh SB, Deretic V. Cell biology of mycobacterium tuberculosis phagosome. Annu Rev Cell Dev Biol. 2004;20:367–394. doi: 10.1146/annurev.cellbio.20.010403.114015. [DOI] [PubMed] [Google Scholar]
- 2.Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007 Jun;35(4):495–516. doi: 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF- beta1 secretion and the resolution of inflammation. J Clin Invest. 2002 Jan;109(1):41–50. doi: 10.1172/JCI11638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chung EY, Kim SJ, Ma XJ. Regulation of cytokine production during phagocytosis of apoptotic cells. Cell Res. 2006 Feb;16(2):154–161. doi: 10.1038/sj.cr.7310021. [DOI] [PubMed] [Google Scholar]
- 5.Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 2014 Feb;15(2):135–147. doi: 10.1038/nrm3737. [DOI] [PubMed] [Google Scholar]
- 6.Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol. 1995 Jan;146(1):3–15. [PMC free article] [PubMed] [Google Scholar]
- 7.Pechkovsky DV, Zalutskaya OM, Ivanov GI, Misuno NI. Calprotectin (MRP8/14 protein complex) release during mycobacterial infection in vitro and in vivo. FEMS Immunol Med Microbiol. 2000 Sep;29(1):27–33. doi: 10.1111/j.1574-695X.2000.tb01501.x. [DOI] [PubMed] [Google Scholar]
- 8.Yamasaki S, Ishikawa E, Sakuma M, Hara H, Ogata K, Saito T. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008 Oct;9(10):1179–1188. doi: 10.1038/ni.1651. [DOI] [PubMed] [Google Scholar]
- 9.Rock KL, Lai JJ, Kono H. Innate and adaptive immune responses to cell death. Immunol Rev. 2011 Sep;243(1):191–205. doi: 10.1111/j.1600-065X.2011.01040.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010 Mar 4;464(7285):104–107. doi: 10.1038/nature08780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Raoof M, Zhang Q, Itagaki K, Hauser CJ. Mitochondrial peptides are potent immune activators that activate human neutrophils via FPR-1. J Trauma. 2010 Jun;68(6):1328–1332. doi: 10.1097/TA.0b013e3181dcd28d. [DOI] [PubMed] [Google Scholar]
- 12.Grover A, Taylor J, Troudt J, Keyser A, Sommersted K, Schenkel A, et al. Mycobacterial infection induces the secretion of high-mobility group box 1 protein. Cell Microbiol. 2008 Jun;10(6):1390–1404. doi: 10.1111/j.1462-5822.2008.01135.x. [DOI] [PubMed] [Google Scholar]
- 13.Lammermann T, Afonso PV, Angermann BR, Wang JM, Kastenmuller W, Parent CA, et al. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature. 2013 Jun 20;498(7454):371–375. doi: 10.1038/nature12175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Creagh EM, Conroy H, Martin SJ. Caspase-activation pathways in apoptosis and immunity. Immunol Rev. 2003 Jun;193:10–21. doi: 10.1034/j.1600-065x.2003.00048.x. [DOI] [PubMed] [Google Scholar]
- 15.Boatright KM, Salvesen GS. Mechanisms of caspase activation. Curr Opin Cell Biol. 2003 Dec;15(6):725–731. doi: 10.1016/j.ceb.2003.10.009. [DOI] [PubMed] [Google Scholar]
- 16.Ware CF. The TNF receptor super family in immune regulation. Immunol Rev. 2011 Nov;244(1):5–8. doi: 10.1111/j.1600-065X.2011.01065.x. [DOI] [PubMed] [Google Scholar]
- 17.Oddo M, Renno T, Attinger A, Bakker T, MacDondal HR, Meylan PAR. Fas ligand-induced apoptosis of infected human alveolar macrophages reduces the viability of intracellular Mycobacterium tuberculosis. J Immunol. 1998;160:5448–5454. [PubMed] [Google Scholar]
- 18.Keane J, Balcewicz-Sablinska K, Remold HG, Chupp GL, Meek BB, Fenton MJ, et al. Infection by Mycobacterium tuberculosis promotes human alveolar macrophage apoptosis. Infect Immun. 1997;65(1):298–304. doi: 10.1128/iai.65.1.298-304.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dickens LS, Powley IR, Hughes MA, MacFarlane M. The ‘complexities’ of life and death: death receptor signalling platforms. Exp Cell Res. 2012 Jul 1;318(11):1269–1277. doi: 10.1016/j.yexcr.2012.04.005. [DOI] [PubMed] [Google Scholar]
- 20.Kreuz S, Siegmund D, Scheurich P, Wajant H. NF-kappaB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling. Mol Cell Biol. 2001 Jun;21(12):3964–3973. doi: 10.1128/MCB.21.12.3964-3973.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Riedl SJ, Salvesen GS. The apoptosome: signalling platform of cell death. Nat Rev Mol Cell Biol. 2007 May;8(5):405–413. doi: 10.1038/nrm2153. [DOI] [PubMed] [Google Scholar]
- 22.Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008 Jan;9(1):47–59. doi: 10.1038/nrm2308. [DOI] [PubMed] [Google Scholar]
- 23.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev. 2007 Jan;87(1):99–163. doi: 10.1152/physrev.00013.2006. [DOI] [PubMed] [Google Scholar]
- 24.Segal M, Niazi S, Simons MP, Galati SA, Zangrilli JG. Bid activation during induction of extrinsic and intrinsic apoptosis in eosinophils. Immunol Cell Biol. 2007 Oct;85(7):518–524. doi: 10.1038/sj.icb.7100075. [DOI] [PubMed] [Google Scholar]
- 25.Pardo J, Bosque A, Brehm R, Wallich R, Naval J, Mullbacher A, et al. Apoptotic pathways are selectively activated by granzyme A and/or granzyme B in CTL-mediated target cell lysis. J Cell Biol. 2004 Nov 8;167(3):457–468. doi: 10.1083/jcb.200406115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Afonina IS, Cullen SP, Martin SJ. Cytotoxic and non-cytotoxic roles of the CTL/NK protease granzyme B. Immunol Rev. 2010 May;235(1):105–116. doi: 10.1111/j.0105-2896.2010.00908.x. [DOI] [PubMed] [Google Scholar]
- 27.Barry M, Heibein JA, Pinkoski MJ, Lee SF, Moyer RW, Green DR, et al. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol. 2000 Jun;20(11):3781–3794. doi: 10.1128/mcb.20.11.3781-3794.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Getachew Y, Stout-Delgado H, Miller BC, Thiele DL. Granzyme C supports efficient CTL-mediated killing late in primary alloimmune responses. J Immunol. 2008 Dec 1;181(11):7810–7817. doi: 10.4049/jimmunol.181.11.7810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chavez-Galan L, Arenas-Del Angel MC, Zenteno E, Chavez R, Lascurain R. Cell death mechanisms induced by cytotoxic lymphocytes. Cell Mol Immunol. 2009 Feb;6(1):15–25. doi: 10.1038/cmi.2009.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Vanlangenakker N, Bertrand MJ, Bogaert P, Vandenabeele P, Vanden Berghe T. TNF-induced necroptosis in L929 cells is tightly regulated by multiple TNFR1 complex I and II members. Cell Death Dis. 2011;2:e230. doi: 10.1038/cddis.2011.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li J, McQuade T, Siemer AB, Napetschnig J, Moriwaki K, Hsiao YS, et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell. 2012 Jul 20;150(2):339–350. doi: 10.1016/j.cell.2012.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Henry T, Monack DM. Activation of the inflammasome upon Francisella tularensis infection: interplay of innate immune pathways and virulence factors. Cell Microbiol. 2007 Nov;9(11):2543–2551. doi: 10.1111/j.1462-5822.2007.01022.x. [DOI] [PubMed] [Google Scholar]
- 33.Cervantes J, Nagata T, Uchijima M, Shibata K, Koide Y. Intracytosolic Listeria monocytogenes induces cell death through caspase-1 activation in murine macrophages. Cell Microbiol. 2008 Jan;10(1):41–52. doi: 10.1111/j.1462-5822.2007.01012.x. [DOI] [PubMed] [Google Scholar]
- 34.Suzuki T, Franchi L, Toma C, Ashida H, Ogawa M, Yoshikawa Y, et al. Differential regulation of caspase-1 activation, pyroptosis, and autophagy via Ipaf and ASC in Shigella-infected macrophages. PLoS Pathog. 2007 Aug 10;3(8):e111. doi: 10.1371/journal.ppat.0030111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zheng Y, Gardner SE, Clarke MC. Cell death, damage-associated molecular patterns, and sterile inflammation in cardiovascular disease. Arterioscler Thromb Vasc Biol. 2011 Dec;31(12):2781–2786. doi: 10.1161/ATVBAHA.111.224907. [DOI] [PubMed] [Google Scholar]
- 36.Ting JP, Willingham SB, Bergstralh DT. NLRs at the intersection of cell death and immunity. Nat Rev Immunol. 2008 May;8(5):372–379. doi: 10.1038/nri2296. [DOI] [PubMed] [Google Scholar]
- 37.Baroja-Mazo A, Martin-Sanchez F, Gomez AI, Martinez CM, Amores-Iniesta J, Compan V, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014 Jun 22; doi: 10.1038/ni.2919. [DOI] [PubMed] [Google Scholar]
- 38.Franklin BS, Bossaller L, De ND, Ratter JM, Stutz A, Engels G, et al. The adaptor ASC has extracellular and ‘prionoid’ activities that propagate inflammation. Nat Immunol. 2014 Jun 22; doi: 10.1038/ni.2913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lemasters JJ, Theruvath TP, Zhong Z, Nieminen AL. Mitochondrial calcium and the permeability transition in cell death. Biochim Biophys Acta. 2009 Nov;1787(11):1395–1401. doi: 10.1016/j.bbabio.2009.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sileikyte J, Blachly-Dyson E, Sewell R, Carpi A, Menabo R, Di LF, et al. Regulation of the Mitochondrial Permeability Transition Pore by the Outer Membrane does not Involve the Peripheral Benzodiazepine Receptor (TSPO). J Biol Chem. 2014 Apr 1; doi: 10.1074/jbc.M114.549634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Joza N, Susin SA, Daugas E, Stanford WL, Cho SK, Li CY, et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature. 2001 Mar 29;410(6828):549–554. doi: 10.1038/35069004. [DOI] [PubMed] [Google Scholar]
- 42.Aredia F, Scovassi AI. Poly(ADP-ribose): A signaling molecule in different paradigms of cell death. Biochem Pharmacol. 2014 Jun 26; doi: 10.1016/j.bcp.2014.06.021. [DOI] [PubMed] [Google Scholar]
- 43.Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. 2012 Sep 15;189(6):2689–2695. doi: 10.4049/jimmunol.1201719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Medina E. Neutrophil extracellular traps: a strategic tactic to defeat pathogens with potential consequences for the host. J Innate Immun. 2009;1(3):176–180. doi: 10.1159/000203699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.von Kockritz-Blickwede M, Goldmann O, Thulin P, Heinemann K, Norrby-Teglund A, Rohde M, et al. Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation. Blood. 2008 Mar 15;111(6):3070–3080. doi: 10.1182/blood-2007-07-104018. [DOI] [PubMed] [Google Scholar]
- 46.Wong KW, Jacobs WR., Jr Mycobacterium tuberculosis exploits human interferon gamma to stimulate macrophage extracellular trap formation and necrosis. J Infect Dis. 2013 Jul;208(1):109–119. doi: 10.1093/infdis/jit097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Almyroudis NG, Grimm MJ, Davidson BA, Rohm M, Urban CF, Segal BH. NETosis and NADPH oxidase: at the intersection of host defense, inflammation, and injury. Front Immunol. 2013;4:45. doi: 10.3389/fimmu.2013.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Farley K, Stolley JM, Zhao P, Cooley J, Remold-O'Donnell E. A serpinB1 regulatory mechanism is essential for restricting neutrophil extracellular trap generation. J Immunol. 2012 Nov 1;189(9):4574–4581. doi: 10.4049/jimmunol.1201167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Clark SR, Guy CJ, Scurr MJ, Taylor PR, Kift-Morgan AP, Hammond VJ, et al. Esterified eicosanoids are acutely generated by 5-lipoxygenase in primary human neutrophils and in human and murine infection. Blood. 2011 Feb 10;117(6):2033–2043. doi: 10.1182/blood-2010-04-278887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yipp BG, Petri B, Salina D, Jenne CN, Scott BN, Zbytnuik LD, et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med. 2012 Sep;18(9):1386–1393. doi: 10.1038/nm.2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Galluzzi L, Aaronson SA, Abrams J, Alnemri ES, Andrews DW, Baehrecke EH, et al. Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes. Cell Death Differ. 2009 Aug;16(8):1093–1107. doi: 10.1038/cdd.2009.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Jain MV, Paczulla AM, Klonisch T, Dimgba FN, Rao SB, Roberg K, et al. Interconnections between apoptotic, autophagic and necrotic pathways: implications for cancer therapy development. J Cell Mol Med. 2013 Jan;17(1):12–29. doi: 10.1111/jcmm.12001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Roulston A, Marcellus RC, Branton PE. Viruses and apoptosis. Annu Rev Microbiol. 1999;53:577–628. doi: 10.1146/annurev.micro.53.1.577. [DOI] [PubMed] [Google Scholar]
- 54.Schaible UE, Winau F, Sieling PA, Fischer K, Collins HL, Hagens K, et al. Apoptosis facilitates antigen presentation to T lymphocytes through MHC-I and CD1 in tuberculosis. Nat Med. 2003 Aug;9(8):1039–1046. doi: 10.1038/nm906. [DOI] [PubMed] [Google Scholar]
- 55.Cunha LD, Zamboni DS. Subversion of inflammasome activation and pyroptosis by pathogenic bacteria. Front Cell Infect Microbiol. 2013;3:76. doi: 10.3389/fcimb.2013.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zychlinsky A. Programmed cell death in infectious diseases. Trends in Microbiology. 1993;1:114–117. doi: 10.1016/0966-842x(93)90118-b. [DOI] [PubMed] [Google Scholar]
- 57.Davis JM, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009 Jan 9;136(1):37–49. doi: 10.1016/j.cell.2008.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Parandhaman DK, Narayanan S. Cell death paradigms in the pathogenesis of Mycobacterium tuberculosis infection. Front Cell Infect Microbiol. 2014;4:31. doi: 10.3389/fcimb.2014.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.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;180:1499–1509. doi: 10.1084/jem.180.4.1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Biswas D, Qureshi OS, Lee WY, Croudace JE, Mura M, Lammas DA. ATP-induced autophagy is associated with rapid killing of intracellular mycobacteria within human monocytes/macrophages. BMC Immunol. 2008;9:35. doi: 10.1186/1471-2172-9-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Keane J, Remold HG, Kornfeld H. Virulent Mycobacterium tuberculosis strains evade apoptosis of infected alveolar macrophages. J Immunol. 2000 Feb 15;164(4):2016–2020. doi: 10.4049/jimmunol.164.4.2016. [DOI] [PubMed] [Google Scholar]
- 62.Riendeau CJ, Kornfeld H. THP-1 cell apoptosis in response to Mycobacterial infection. Infect Immun. 2003 Jan;71(1):254–259. doi: 10.1128/IAI.71.1.254-259.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.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(2-3):55–61. doi: 10.1054/tube.2002.0322. [DOI] [PubMed] [Google Scholar]
- 64.Sly LM, Hingley-Wilson SM, Reiner NE, McMaster WR. Survival of Mycobacterium tuberculosis in host macrophages involves resistance to apoptosis dependent upon induction of antiapoptotic Bcl-2 family member Mcl-1. J Immunol. 2003 Jan 1;170(1):430–437. doi: 10.4049/jimmunol.170.1.430. [DOI] [PubMed] [Google Scholar]
- 65.Pan H, Yan BS, Rojas M, Shebzukhov YV, Zhou H, Kobzik L, et al. Ipr1 gene mediates innate immunity to tuberculosis. Nature. 2005 Apr 7;434(7034):767–772. doi: 10.1038/nature03419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Balcewicz-Sablinska MK, Keane J, Kornfeld H, Remold HG. Pathogenic Mycobacterium tuberculosis evades apoptosis of host macrophages by release of TNF-R2, resulting in inactivation of TNF-alpha. J Immunol. 1998 Sep 1;161(5):2636–2641. [PubMed] [Google Scholar]
- 67.Spira A, Carroll JD, Liu G, Aziz Z, Shah V, Kornfeld H, et al. Apoptosis Genes in Human Alveolar Macrophages Infected with Virulent or Attenuated Mycobacterium tuberculosis: A Pivotal Role for Tumor Necrosis Factor. Am J Respir Cell Mol Biol. 2003 Nov;29(5):545–551. doi: 10.1165/rcmb.2002-0310OC. [DOI] [PubMed] [Google Scholar]
- 68.Blomgran R, Desvignes L, Briken V, Ernst JD. Mycobacterium tuberculosis inhibits neutrophil apoptosis, leading to delayed activation of naive CD4 T cells. Cell Host Microbe. 2012 Jan 19;11(1):81–90. doi: 10.1016/j.chom.2011.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Velmurugan K, Chen B, Miller JL, Azogue S, Gurses S, Hsu T, et al. Mycobacterium tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected host cells. PLoS Pathog. 2007 Jul;3(7):e110. doi: 10.1371/journal.ppat.0030110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hinchey J, Lee S, Jeon BY, Basaraba RJ, Venkataswamy MM, Chen B, et al. Enhanced priming of adaptive immunity by a proapoptotic mutant of Mycobacterium tuberculosis. J Clin Invest. 2007 Aug;117(8):2279–2288. doi: 10.1172/JCI31947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shen HM, Pervaiz S. TNF receptor superfamily-induced cell death: redox-dependent execution. FASEB J. 2006 Aug;20(10):1589–1598. doi: 10.1096/fj.05-5603rev. [DOI] [PubMed] [Google Scholar]
- 72.Tsujimoto Y, Shimizu S. Role of the mitochondrial membrane permeability transition in cell death. Apoptosis. 2007 May;12(5):835–840. doi: 10.1007/s10495-006-0525-7. [DOI] [PubMed] [Google Scholar]
- 73.Edwards KM, Cynamon MH, Voladri RK, Hager CC, DeStefano MS, Tham KT, et al. Iron-cofactored superoxide dismutase inhibits host responses to Mycobacterium tuberculosis. Am J Respir Crit Care Med. 2001 Dec 15;164(12):2213–2219. doi: 10.1164/ajrccm.164.12.2106093. [DOI] [PubMed] [Google Scholar]
- 74.Miller JL, Velmurugan K, Cowan MJ, Briken V. The type I NADH dehydrogenase of Mycobacterium tuberculosis counters phagosomal NOX2 activity to inhibit TNF-alpha-mediated host cell apoptosis. PLoS Pathog. 2010 Apr;6(4):e1000864. doi: 10.1371/journal.ppat.1000864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wu K, Koo J, Jiang X, Chen R, Cohen SN, Nathan C. Improved control of tuberculosis and activation of macrophages in mice lacking protein kinase R. PLoS One. 2012;7(2):e30512. doi: 10.1371/journal.pone.0030512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Shin DM, Jeon BY, Lee HM, Jin HS, Yuk JM, Song CH, et al. Mycobacterium tuberculosis eis regulates autophagy, inflammation, and cell death through redox-dependent signaling. PLoS Pathog. 2010;6(12):e1001230. doi: 10.1371/journal.ppat.1001230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kim KH, An DR, Song J, Yoon JY, Kim HS, Yoon HJ, et al. Mycobacterium tuberculosis Eis protein initiates suppression of host immune responses by acetylation of DUSP16/MKP-7. Proc Natl Acad Sci U S A. 2012 May 15;109(20):7729–7734. doi: 10.1073/pnas.1120251109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang Q, Liu S, Tang Y, Liu Q, Yao Y. MPT64 Protein from Mycobacterium tuberculosis Inhibits Apoptosis of Macrophages through NF-kB-miRNA21-Bcl-2 Pathway. PLoS One. 2014;9(7):e100949. doi: 10.1371/journal.pone.0100949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Danelishvili L, Yamazaki Y, Selker J, Bermudez LE. Secreted Mycobacterium tuberculosis Rv3654c and Rv3655c proteins participate in the suppression of macrophage apoptosis. PLoS One. 2010;5(5):e10474. doi: 10.1371/journal.pone.0010474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jayakumar D, Jacobs WR, Jr., Narayanan S. Protein kinase E of Mycobacterium tuberculosis has a role in the nitric oxide stress response and apoptosis in a human macrophage model of infection. Cell Microbiol. 2008 Feb;10(2):365–374. doi: 10.1111/j.1462-5822.2007.01049.x. [DOI] [PubMed] [Google Scholar]
- 81.Renshaw PS, Lightbody KL, Veverka V, Muskett FW, Kelly G, Frenkiel TA, et al. Structure and function of the complex formed by the tuberculosis virulence factors CFP-10 and ESAT-6. EMBO J. 2005 Jul 20;24(14):2491–2498. doi: 10.1038/sj.emboj.7600732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Derrick SC, Morris SL. The ESAT6 protein of Mycobacterium tuberculosis induces apoptosis of macrophages by activating caspase expression. Cell Microbiol. 2007 Jun;9(6):1547–1555. doi: 10.1111/j.1462-5822.2007.00892.x. [DOI] [PubMed] [Google Scholar]
- 83.Haoues M, Refai A, Mallavialle A, Barbouche MR, Laabidi N, Deckert M, et al. Forkhead box O3 (FOXO3) transcription factor mediates apoptosis in BCG-infected macrophages. Cell Microbiol. 2014 Apr 8; doi: 10.1111/cmi.12298. [DOI] [PubMed] [Google Scholar]
- 84.Iyoda T, Takada M, Fukatsu Y, Kumokoshi S, Fujisawa T, Shimada T, et al. A novel mechanism underlying the basic defensive response of macrophages against Mycobacterium infection. J Immunol. 2014 May 1;192(9):4254–4262. doi: 10.4049/jimmunol.1301526. [DOI] [PubMed] [Google Scholar]
- 85.Hochreiter-Hufford A, Ravichandran KS. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb Perspect Biol. 2013 Jan;5(1):a008748. doi: 10.1101/cshperspect.a008748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hingley-Wilson SM, Connell D, Pollock K, Hsu T, Tchilian E, Sykes A, et al. ESX1-dependent fractalkine mediates chemotaxis and Mycobacterium tuberculosis infection in humans. Tuberculosis (Edinb ) 2014 May;94(3):262–270. doi: 10.1016/j.tube.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Amaral EP, Ribeiro SC, Lanes VR, Almeida FM, de Andrade MR, Bomfim CC, et al. Pulmonary Infection with Hypervirulent Mycobacteria Reveals a Crucial Role for the P2X7 Receptor in Aggressive Forms of Tuberculosis. PLoS Pathog. 2014 Jul;10(7):e1004188. doi: 10.1371/journal.ppat.1004188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bournazou I, Pound JD, Duffin R, Bournazos S, Melville LA, Brown SB, et al. Apoptotic human cells inhibit migration of granulocytes via release of lactoferrin. J Clin Invest. 2009 Jan;119(1):20–32. doi: 10.1172/JCI36226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Martin CJ, Peters KN, Behar SM. Macrophages clean up: efferocytosis and microbial control. Curr Opin Microbiol. 2014 Feb;17:17–23. doi: 10.1016/j.mib.2013.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, et al. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005 Oct 21;123(2):321–334. doi: 10.1016/j.cell.2005.08.032. [DOI] [PubMed] [Google Scholar]
- 91.Fratazzi C, Arbeit RD, Carini C, Remold HG. Programmed cell death of Mycobacterium avium serovar 4-infected human macrophages prevents mycobacteria from spreading and induces mycobacterial growth inhibition by freshly added, uninfected macrophages. J Immunol. 1997;158:4320–4327. [PubMed] [Google Scholar]
- 92.Martin CJ, Booty MG, Rosebrock TR, Nunes-Alves C, Desjardins DM, Keren I, et al. Efferocytosis is an innate antibacterial mechanism. Cell Host Microbe. 2012 Sep 13;12(3):289–300. doi: 10.1016/j.chom.2012.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Winau F, Weber S, Sad S, de DJ, Hoops SL, Breiden B, et al. Apoptotic Vesicles Crossprime CD8 T Cells and Protect against Tuberculosis. Immunity. 2006 Jan;24(1):105–117. doi: 10.1016/j.immuni.2005.12.001. [DOI] [PubMed] [Google Scholar]
- 94.Divangahi M, Desjardins D, Nunes-Alves C, Remold HG, Behar SM. Eicosanoid pathways regulate adaptive immunity to Mycobacterium tuberculosis. Nat Immunol. 2010 Aug;11(8):751–758. doi: 10.1038/ni.1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Duan L, Gan H, Golan DE, Remold HG. Critical role of mitochondrial damage in determining outcome of macrophage infection with Mycobacterium tuberculosis. J Immunol. 2002 Nov 1;169(9):5181–5187. doi: 10.4049/jimmunol.169.9.5181. [DOI] [PubMed] [Google Scholar]
- 96.Gan H, He X, Duan L, Mirabile-Levens E, Kornfeld H, Remold HG. Enhancement of antimycobacterial activity of macrophages by stabilization of inner mitochondrial membrane potential. J Infect Dis. 2005 Apr 15;191(8):1292–1300. doi: 10.1086/428906. [DOI] [PubMed] [Google Scholar]
- 97.Chen M, Gan H, Remold HG. A mechanism of virulence: virulent Mycobacterium tuberculosis strain H37Rv, but not attenuated H37Ra, causes significant mitochondrial inner membrane disruption in macrophages leading to necrosis. J Immunol. 2006 Mar 15;176(6):3707–3716. doi: 10.4049/jimmunol.176.6.3707. [DOI] [PubMed] [Google Scholar]
- 98.Chen M, Divangahi M, Gan H, Shin DS, Hong S, Lee DM, et al. Lipid mediators in innate immunity against tuberculosis: opposing roles of PGE2 and LXA4 in the induction of macrophage death. J Exp Med. 2008 Nov 24;205(12):2791–2801. doi: 10.1084/jem.20080767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gan H, Lee J, Ren F, Chen M, Kornfeld H, Remold HG. Mycobacterium tuberculosis blocks crosslinking of annexin-1 and apoptotic envelope formation on infected macrophages to maintain virulence. Nat Immunol. 2008 Oct;9(10):1189–1197. doi: 10.1038/ni.1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Divangahi M, Chen M, Gan H, Desjardins D, Hickman TT, Lee DM, et al. Mycobacterium tuberculosis evades macrophage defenses by inhibiting plasma membrane repair. Nat Immunol. 2009 Aug;10(8):899–906. doi: 10.1038/ni.1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Divangahi M, Behar SM, Remold H. Dying to live: how the death modality of the infected macrophage affects immunity to tuberculosis. Adv Exp Med Biol. 2013;783:103–120. doi: 10.1007/978-1-4614-6111-1_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Dalichanka OA, Sun J, Pavlenok M, Maueroder C, Speer A, Siroy A, et al. An outer membrane channel protein of Mycobacterium tuberculosis with exotoxin activity. Proc Natl Acad Sci (USA) 2014 May;111(18):6750–6755. doi: 10.1073/pnas.1400136111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bafica A, Scanga CA, Serhan C, Machado F, White S, Sher A, et al. Host control of Mycobacterium tuberculosis is regulated by 5-lipoxygenase-dependent lipoxin production. J Clin Invest. 2005 Jun;115(6):1601–1606. doi: 10.1172/JCI23949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tobin DM, Vary JC, Jr., Ray JP, Walsh GS, Dunstan SJ, Bang ND, et al. The lta4h locus modulates susceptibility to mycobacterial infection in zebrafish and humans. Cell. 2010 Mar 5;140(5):717–730. doi: 10.1016/j.cell.2010.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tobin DM, Roca FJ, Oh SF, McFarland R, Vickery TW, Ray JP, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012 Feb 3;148(3):434–446. doi: 10.1016/j.cell.2011.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Danelishvili L, Everman JL, McNamara MJ, Bermudez LE. Inhibition of the Plasma-Membrane-Associated Serine Protease Cathepsin G by Mycobacterium tuberculosis Rv3364c Suppresses Caspase-1 and Pyroptosis in Macrophages. Front Microbiol. 2011;2:281. doi: 10.3389/fmicb.2011.00281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lee J, Remold HG, Ieong MH, Kornfeld H. Macrophage apoptosis in response to high intracellular burden of Mycobacterium tuberculosis is mediated by a novel caspase-independent pathway. J Immunol. 2006 Apr 1;176(7):4267–4274. doi: 10.4049/jimmunol.176.7.4267. [DOI] [PubMed] [Google Scholar]
- 108.Lee J, Repasy T, Papavinasasundaram K, Sassetti C, Kornfeld H. Mycobacterium tuberculosis induces an atypical cell death mode to escape from infected macrophages. PLoS One. 2011;6(3):e18367. doi: 10.1371/journal.pone.0018367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Repasy T, Lee J, Marino S, Martinez N, Kirschner DE, Hendricks G, et al. Intracellular Bacillary Burden Reflects a Burst Size for Mycobacterium tuberculosis In Vivo. PLoS Pathog. 2013 Feb;9(2):e1003190. doi: 10.1371/journal.ppat.1003190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wolf AJ, Linas B, Trevejo-Nunez GJ, Kincaid E, Tamura T, Takatsu K, et al. Mycobacterium tuberculosis infects dendritic cells with high frequency and impairs their function in vivo. J Immunol. 2007 Aug 15;179(4):2509–2519. doi: 10.4049/jimmunol.179.4.2509. [DOI] [PubMed] [Google Scholar]
- 111.Eum SY, Kong JH, Hong MS, Lee YJ, Kim JH, Hwang SH, et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest. 2010 Jan;137(1):122–128. doi: 10.1378/chest.09-0903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hume DA, Ross IL, Himes SR, Sasmono RT, Wells CA, Ravasi T. The mononuclear phagocyte system revisited. J Leukoc Biol. 2002 Oct;72(4):621–627. [PubMed] [Google Scholar]
- 113.Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, et al. In vivo analysis of dendritic cell development and homeostasis. Science. 2009 Apr 17;324(5925):392–397. doi: 10.1126/science.1170540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Naik SH. Demystifying the development of dendritic cell subtypes, a little. Immunol Cell Biol. 2008 Jul;86(5):439–452. doi: 10.1038/icb.2008.28. [DOI] [PubMed] [Google Scholar]
- 115.Bodnar KA, Serbina NV, Flynn JL. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect Immun. 2001 Feb;69(2):800–809. doi: 10.1128/IAI.69.2.800-809.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ryan RC, O'Sullivan MP, Keane J. Mycobacterium tuberculosis infection induces non-apoptotic cell death of human dendritic cells. BMC Microbiol. 2011;11:237. doi: 10.1186/1471-2180-11-237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Abdalla H, Srinivasan L, Shah S, Mayer-Barber KD, Sher A, Sutterwala FS, et al. Mycobacterium tuberculosis infection of dendritic cells leads to partially caspase-1/11-independent IL-1beta and IL-18 secretion but not to pyroptosis. PLoS One. 2012;7(7):e40722. doi: 10.1371/journal.pone.0040722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Khader SA, Partida-Sanchez S, Bell G, Jelley-Gibbs DM, Swain S, Pearl JE, et al. Interleukin 12p40 is required for dendritic cell migration and T cell priming after Mycobacterium tuberculosis infection. J Exp Med. 2006 Jul 10;203(7):1805–1815. doi: 10.1084/jem.20052545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chackerian AA, Alt JM, Perera TV, Dascher CC, Behar SM. Dissemination of Mycobacterium tuberculosis is influenced by host factors and precedes the initiation of T-cell immunity. Infect Immun. 2002 Aug;70(8):4501–4509. doi: 10.1128/IAI.70.8.4501-4509.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lowe DM, Redford PS, Wilkinson RJ, O'Garra A, Martineau AR. Neutrophils in tuberculosis: friend or foe? Trends Immunol. 2012 Jan;33(1):14–25. doi: 10.1016/j.it.2011.10.003. [DOI] [PubMed] [Google Scholar]
- 121.Blomgran R, Ernst JD. Lung neutrophils facilitate activation of naive antigen-specific CD4+ T cells during Mycobacterium tuberculosis infection. J Immunol. 2011 Jun 15;186(12):7110–7119. doi: 10.4049/jimmunol.1100001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Aleman M, Schierloh P, de la Barrera SS, Musella RM, Saab MA, Baldini M, et al. Mycobacterium tuberculosis triggers apoptosis in peripheral neutrophils involving toll-like receptor 2 and p38 mitogen protein kinase in tuberculosis patients. Infect Immun. 2004 Sep;72(9):5150–5158. doi: 10.1128/IAI.72.9.5150-5158.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Romero MM, Balboa L, Basile JI, Lopez B, Ritacco V, de la Barrera SS, et al. Clinical isolates of Mycobacterium tuberculosis differ in their ability to induce respiratory burst and apoptosis in neutrophils as a possible mechanism of immune escape. Clin Dev Immunol. 2012;2012:152546. doi: 10.1155/2012/152546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Martens GW, Arikan MC, Lee J, Ren F, Vallerskog T, Kornfeld H. Hypercholesterolemia impairs immunity to tuberculosis. Infect Immun. 2008 Aug;76(8):3464–3472. doi: 10.1128/IAI.00037-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Dorhoi A, Desel C, Yeremeev V, Pradl L, Brinkmann V, Mollenkopf HJ, et al. The adaptor molecule CARD9 is essential for tuberculosis control. J Exp Med. 2010 Apr 12;207(4):777–792. doi: 10.1084/jem.20090067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Eruslanov EB, Lyadova IV, Kondratieva TK, Majorov KB, Scheglov IV, Orlova MO, et al. Neutrophil responses to Mycobacterium tuberculosis infection in genetically susceptible and resistant mice. Infect Immun. 2005 Mar;73(3):1744–1753. doi: 10.1128/IAI.73.3.1744-1753.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ramos-Kichik V, Mondragon-Flores R, Mondragon-Castelan M, Gonzalez-Pozos S, Muniz-Hernandez S, Rojas-Espinosa O, et al. Neutrophil extracellular traps are induced by Mycobacterium tuberculosis. Tuberculosis (Edinb ) 2009 Jan;89(1):29–37. doi: 10.1016/j.tube.2008.09.009. [DOI] [PubMed] [Google Scholar]
- 128.Farinacci M, Weber S, Kaufmann SH. The recombinant tuberculosis vaccine rBCG DeltaureC::hly(+) induces apoptotic vesicles for improved priming of CD4(+) and CD8(+) T cells. Vaccine. 2012 Dec 14;30(52):7608–7614. doi: 10.1016/j.vaccine.2012.10.031. [DOI] [PubMed] [Google Scholar]
- 129.Desel C, Dorhoi A, Bandermann S, Grode L, Eisele B, Kaufmann SH. Recombinant BCG DeltaureC hly+ induces superior protection over parental BCG by stimulating a balanced combination of type 1 and type 17 cytokine responses. J Infect Dis. 2011 Nov 15;204(10):1573–1584. doi: 10.1093/infdis/jir592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Grode L, Ganoza CA, Brohm C, Weiner J, III, Eisele B, Kaufmann SH. Safety and immunogenicity of the recombinant BCG vaccine VPM1002 in a phase 1 open-label randomized clinical trial. Vaccine. 2013 Feb 18;31(9):1340–1348. doi: 10.1016/j.vaccine.2012.12.053. [DOI] [PubMed] [Google Scholar]
- 131.Rachman H, Kim N, Ulrichs T, Baumann S, Pradl L, Nasser EA, et al. Critical role of methylglyoxal and AGE in mycobacteria-induced macrophage apoptosis and activation. PLoS One. 2006;1:e29. doi: 10.1371/journal.pone.0000029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lommatzsch M, Bratke K, Bier A, Julius P, Kuepper M, Luttmann W, et al. Airway dendritic cell phenotypes in inflammatory diseases of the human lung. Eur Respir J. 2007 Nov;30(5):878–886. doi: 10.1183/09031936.00036307. [DOI] [PubMed] [Google Scholar]
- 133.Park JS, Tamayo MH, Gonzalez-Juarrero M, Orme IM, Ordway DJ. Virulent clinical isolates of Mycobacterium tuberculosis grow rapidly and induce cellular necrosis but minimal apoptosis in murine macrophages. J Leukoc Biol. 2006 Jan;79(1):80–86. doi: 10.1189/jlb.0505250. [DOI] [PubMed] [Google Scholar]
- 134.Lee J, Kornfeld H. Interferon-gamma regulates the death of M. tuberculosis-infected macrophages. J Cell Death. 2010 Mar 3;3:1–11. doi: 10.4137/jcd.s2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Todde V, Veenhuis M, van der Klei IJ. Autophagy: principles and significance in health and disease. Biochim Biophys Acta. 2009 Jan;1792(1):3–13. doi: 10.1016/j.bbadis.2008.10.016. [DOI] [PubMed] [Google Scholar]
- 136.Klionsky DJ, Cregg JM, Dunn WA, Jr., Emr SD, Sakai Y, Sandoval IV, et al. A unified nomenclature for yeast autophagy-related genes. Dev Cell. 2003 Oct;5(4):539–545. doi: 10.1016/s1534-5807(03)00296-x. [DOI] [PubMed] [Google Scholar]
- 137.Mizushima N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol. 2010 Apr;22(2):132–139. doi: 10.1016/j.ceb.2009.12.004. [DOI] [PubMed] [Google Scholar]
- 138.Itakura E, Kishi-Itakura C, Mizushima N. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012 Dec 7;151(6):1256–1269. doi: 10.1016/j.cell.2012.11.001. [DOI] [PubMed] [Google Scholar]
- 139.Barth S, Glick D, Macleod KF. Autophagy: assays and artifacts. J Pathol. 2010 Jun;221(2):117–124. doi: 10.1002/path.2694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Martinez J, Almendinger J, Oberst A, Ness R, Dillon CP, Fitzgerald P, et al. Microtubule-associated protein 1 light chain 3 alpha (LC3)-associated phagocytosis is required for the efficient clearance of dead cells. Proc Natl Acad Sci U S A. 2011 Oct 18;108(42):17396–17401. doi: 10.1073/pnas.1113421108. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 141.Burman C, Ktistakis NT. Autophagosome formation in mammalian cells. Semin Immunopathol. 2010 Dec;32(4):397–413. doi: 10.1007/s00281-010-0222-z. [DOI] [PubMed] [Google Scholar]
- 142.Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010 Apr;22(2):124–131. doi: 10.1016/j.ceb.2009.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ryter SW, Cloonan SM, Choi AM. Autophagy: a critical regulator of cellular metabolism and homeostasis. Mol Cells. 2013 Jul;36(1):7–16. doi: 10.1007/s10059-013-0140-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol. 2013 Oct;15(10):1197–1205. doi: 10.1038/ncb2837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sarkar S. Regulation of autophagy by mTOR-dependent and mTOR-independent pathways: autophagy dysfunction in neurodegenerative diseases and therapeutic application of autophagy enhancers. Biochem Soc Trans. 2013 Oct;41(5):1103–1130. doi: 10.1042/BST20130134. [DOI] [PubMed] [Google Scholar]
- 146.Deretic V. Autophagy in immunity and cell-autonomous defense against intracellular microbes. Immunol Rev. 2011 Mar;240(1):92–104. doi: 10.1111/j.1600-065X.2010.00995.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Nicola AM, Albuquerque P, Martinez LR, Dal-Rosso RA, Saylor C, De JM, et al. Macrophage autophagy in immunity to Cryptococcus neoformans and Candida albicans. Infect Immun. 2012 Sep;80(9):3065–3076. doi: 10.1128/IAI.00358-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Beale R, Wise H, Stuart A, Ravenhill BJ, Digard P, Randow F. A LC3-interacting motif in the influenza A virus M2 protein is required to subvert autophagy and maintain virion stability. Cell Host Microbe. 2014 Feb 12;15(2):239–247. doi: 10.1016/j.chom.2014.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Dupont N, Temime-Smaali N, Lafont F. How ubiquitination and autophagy participate in the regulation of the cell response to bacterial infection. Biol Cell. 2010 Dec;102(12):621–634. doi: 10.1042/BC20100101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, Deretic V. Autophagy Is a Defense Mechanism Inhibiting BCG and Mycobacterium tuberculosis Survival in Infected Macrophages. Cell. 2004 Dec 17;119(6):753–766. doi: 10.1016/j.cell.2004.11.038. [DOI] [PubMed] [Google Scholar]
- 151.Xu S, Cooper A, Sturgill-Koszycki S, van HT, Chatterjee D, Orme I, et al. Intracellular trafficking in Mycobacterium tuberculosis and Mycobacterium avium-infected macrophages. J Immunol. 1994 Sep 15;153(6):2568–2578. [PubMed] [Google Scholar]
- 152.Teitelbaum R, Cammer M, Maitland ML, Freitag NE, Condeelis J, Bloom BR. Mycobacterial infection of macrophages results in membrane-permeable phagosomes. Proc Natl Acad Sci U S A. 1999 Dec 21;96(26):15190–15195. doi: 10.1073/pnas.96.26.15190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Beatty WL, Rhoades ER, Ullrich HJ, Chatterjee D, Heuser JE, Russell DG. Trafficking and release of mycobacterial lipids from infected macrophages. Traffic. 2000 Mar;1(3):235–247. doi: 10.1034/j.1600-0854.2000.010306.x. [DOI] [PubMed] [Google Scholar]
- 154.Watson RO, Manzanillo PS, Cox JS. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell. 2012 Aug 17;150(4):803–815. doi: 10.1016/j.cell.2012.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Manzanillo PS, Shiloh MU, Portnoy DA, Cox JS. Mycobacterium tuberculosis activates the DNA-dependent cytosolic surveillance pathway within macrophages. Cell Host Microbe. 2012 May 17;11(5):469–480. doi: 10.1016/j.chom.2012.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.van der Vaart M, Korbee CJ, Lamers GE, Tengeler AC, Hosseini R, Haks MC, et al. The DNA damage-regulated autophagy modulator DRAM1 links mycobacterial recognition via TLP-MYD88 to authophagic defense. Cell Host Microbe. 2014 Jun 11;15(6):753–767. doi: 10.1016/j.chom.2014.05.005. [DOI] [PubMed] [Google Scholar]
- 157.Castillo EF, Dekonenko A, Arko-Mensah J, Mandell MA, Dupont N, Jiang S, et al. Autophagy protects against active tuberculosis by suppressing bacterial burden and inflammation. Proc Natl Acad Sci U S A. 2012 Nov 13;109(46):E3168–E3176. doi: 10.1073/pnas.1210500109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Manzanillo PS, Ayres JS, Watson RO, Collins AC, Souza G, Rae CS, et al. The ubiquitin ligase parkin mediates resistance to intracellular pathogens. Nature. 2013 Sep 26;501(7468):512–516. doi: 10.1038/nature12566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Seto S, Tsujimura K, Horii T, Koide Y. Autophagy adaptor protein p62/SQSTM1 and autophagy-related gene Atg5 mediate autophagosome formation in response to Mycobacterium tuberculosis infection in dendritic cells. PLoS One. 2013;8(12):e86017. doi: 10.1371/journal.pone.0086017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Yang Y, Yin C, Pandey A, Abbott D, Sassetti C, Kelliher MA. NOD2 pathway activation by MDP or Mycobacterium tuberculosis infection involves the stable polyubiquitination of Rip2. J Biol Chem. 2007 Dec 14;282(50):36223–36229. doi: 10.1074/jbc.M703079200. [DOI] [PubMed] [Google Scholar]
- 161.Juarez E, Carranza C, Hernandez-Sanchez F, Leon-Contreras JC, Hernandez-Pando R, Escobedo D, et al. NOD2 enhances the innate response of alveolar macrophages to Mycobacterium tuberculosis in humans. Eur J Immunol. 2012 Apr;42(4):880–889. doi: 10.1002/eji.201142105. [DOI] [PubMed] [Google Scholar]
- 162.Ponpuak M, Davis AS, Roberts EA, Delgado MA, Dinkins C, Zhao Z, et al. Delivery of cytosolic components by autophagic adaptor protein p62 endows autophagosomes with unique antimicrobial properties. Immunity. 2010 Mar 26;32(3):329–341. doi: 10.1016/j.immuni.2010.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.North RJ, Jung YJ. Immunity to tuberculosis. Annu Rev Immunol. 2004;22:599–623. doi: 10.1146/annurev.immunol.22.012703.104635. [DOI] [PubMed] [Google Scholar]
- 164.Singh SB, Davis AS, Taylor GA, Deretic V. Human IRGM induces autophagy to eliminate intracellular mycobacteria. Science. 2006 Sep 8;313(5792):1438–1441. doi: 10.1126/science.1129577. [DOI] [PubMed] [Google Scholar]
- 165.Matsuzawa T, Kim BH, Shenoy AR, Kamitani S, Miyake M, MacMicking JD. IFN-gamma elicits macrophage autophagy via the p38 MAPK signaling pathway. J Immunol. 2012 Jul 15;189(2):813–818. doi: 10.4049/jimmunol.1102041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Stanley SA, Barczak AK, Silvis MR, Luo SS, Sogi K, Vokes M, et al. Identification of host-targeted small molecules that restrict intracellular Mycobacterium tuberculosis growth. PLoS Pathog. 2014 Feb;10(2):e1003946. doi: 10.1371/journal.ppat.1003946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Harris J, De Haro SA, Master SS, Keane J, Roberts EA, Delgado M, et al. T helper 2 cytokines inhibit autophagic control of intracellular Mycobacterium tuberculosis. Immunity. 2007 Sep;27(3):505–517. doi: 10.1016/j.immuni.2007.07.022. [DOI] [PubMed] [Google Scholar]
- 168.Zullo AJ, Lee S. Mycobacterial induction of autophagy varies by species and occurs independently of mammalian target of rapamycin inhibition. J Biol Chem. 2012 Apr 13;287(16):12668–12678. doi: 10.1074/jbc.M111.320135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Shin DM, Yuk JM, Lee HM, Lee SH, Son JW, Harding CV, et al. Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol. 2010 Nov;12(11):1648–1665. doi: 10.1111/j.1462-5822.2010.01497.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, et al. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe. 2009 Sep 17;6(3):231–243. doi: 10.1016/j.chom.2009.08.004. [DOI] [PubMed] [Google Scholar]
- 171.Campbell GR, Spector SA. Vitamin D inhibits human immunodeficiency virus type 1 and Mycobacterium tuberculosis infection in macrophages through the induction of autophagy. PLoS Pathog. 2012;8(5):e1002689. doi: 10.1371/journal.ppat.1002689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Fabri M, Stenger S, Shin DM, Yuk JM, Liu PT, Realegeno S, et al. Vitamin D is required for IFN-gamma-mediated antimicrobial activity of human macrophages. Sci Transl Med. 2011 Oct 12;3(104):104ra102. doi: 10.1126/scitranslmed.3003045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Xu Y, Fattah EA, Liu XD, Jagannath C, Eissa NT. Harnessing of TLR-mediated autophagy to combat mycobacteria in macrophages. Tuberculosis (Edinb ) 2013 Dec;93(Suppl):S33–S37. doi: 10.1016/S1472-9792(13)70008-8. [DOI] [PubMed] [Google Scholar]
- 174.Pilli M, Arko-Mensah J, Ponpuak M, Roberts E, Master S, Mandell MA, et al. TBK-1 promotes autophagy-mediated antimicrobial defense by controlling autophagosome maturation. Immunity. 2012 Aug 24;37(2):223–234. doi: 10.1016/j.immuni.2012.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Sanjurjo L, Amezaga N, Vilaplana C, Caceres N, Marzo E, Valeri M, et al. The scavenger protein apoptosis inhibitor of macrophages (AIM) potentiates the antimicrobial response against Mycobacterium tuberculosis by enhancing autophagy. PLoS One. 2013;8(11):e79670. doi: 10.1371/journal.pone.0079670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Ghorpade DS, Leyland R, Kurowska-Stolarska M, Patil SA, Balaji KN. MicroRNA-155 is required for Mycobacterium bovis BCG-mediated apoptosis of macrophages. Mol Cell Biol. 2012 Jun;32(12):2239–2253. doi: 10.1128/MCB.06597-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Kumar R, Halder P, Sahu SK, Kumar M, Kumari M, Jana K, et al. Identification of a novel role of ESAT-6-dependent miR-155 induction during infection of macrophages with Mycobacterium tuberculosis. Cell Microbiol. 2012 Oct;14(10):1620–1631. doi: 10.1111/j.1462-5822.2012.01827.x. [DOI] [PubMed] [Google Scholar]
- 178.Wang J, Yang K, Zhou L, Minhaowu, Wu Y, Zhu M, et al. MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog. 2013;9(10):e1003697. doi: 10.1371/journal.ppat.1003697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Jagannath C, Lindsey DR, Dhandayuthapani S, Xu Y, Hunter RL, Jr., Eissa NT. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Nat Med. 2009 Mar;15(3):267–276. doi: 10.1038/nm.1928. [DOI] [PubMed] [Google Scholar]
- 180.Saitoh T, Fujita N, Jang MH, Uematsu S, Yang BG, Satoh T, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008 Nov 13;456(7219):264–268. doi: 10.1038/nature07383. [DOI] [PubMed] [Google Scholar]
- 181.Kleinnijenhuis J, Oosting M, Plantinga TS, van der Meer JW, Joosten LA, Crevel RV, et al. Autophagy modulates the Mycobacterium tuberculosis-induced cytokine response. Immunology. 2011 Nov;134(3):341–348. doi: 10.1111/j.1365-2567.2011.03494.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Harris J, Hartman M, Roche C, Zeng SG, O'Shea A, Sharp FA, et al. Autophagy controls IL-1beta secretion by targeting pro-IL-1beta for degradation. J Biol Chem. 2011 Mar 18;286(11):9587–9597. doi: 10.1074/jbc.M110.202911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011 Jan 13;469(7329):221–225. doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
- 184.Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011 Mar;12(3):222–230. doi: 10.1038/ni.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Bonilla DL, Bhattacharya A, Sha Y, Xu Y, Xiang Q, Kan A, et al. Autophagy regulates phagocytosis by modulating the expression of scavenger receptors. Immunity. 2013 Sep 19;39(3):537–547. doi: 10.1016/j.immuni.2013.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhang L, Zhang H, Zhao Y, Mao F, Wu J, Bai B, et al. Effects of Mycobacterium tuberculosis ESAT-6/CFP-10 fusion protein on the autophagy function of mouse macrophages. DNA Cell Biol. 2012 Feb;31(2):171–179. doi: 10.1089/dna.2011.1290. [DOI] [PubMed] [Google Scholar]
- 187.Romagnoli A, Etna MP, Giacomini E, Pardini M, Remoli ME, Corazzari M, et al. ESX-1 dependent impairment of autophagic flux by Mycobacterium tuberculosis in human dendritic cells. Autophagy. 2012 Sep;8(9):1357–1370. doi: 10.4161/auto.20881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Shui W, Petzold CJ, Redding A, Liu J, Pitcher A, Sheu L, et al. Organelle membrane proteomics reveals differential influence of mycobacterial lipoglycans on macrophage phagosome maturation and autophagosome accumulation. J Proteome Res. 2011 Jan 7;10(1):339–348. doi: 10.1021/pr100688h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.MacMicking JD, Taylor GA, McKinney JD. Immune control of tuberculosis by IFN-gamma-inducible LRG-47. Science. 2003 Oct 24;302(5645):654–659. doi: 10.1126/science.1088063. [DOI] [PubMed] [Google Scholar]
- 190.Kumar D, Nath L, Kamal MA, Varshney A, Jain A, Singh S, et al. Genome-wide analysis of the host intracellular network that regulates survival of Mycobacterium tuberculosis. Cell. 2010 Mar 5;140(5):731–743. doi: 10.1016/j.cell.2010.02.012. [DOI] [PubMed] [Google Scholar]
- 191.Intemann CD, Thye T, Niemann S, Browne EN, Amanua CM, Enimil A, et al. Autophagy gene variant IRGM -261T contributes to protection from tuberculosis caused by Mycobacterium tuberculosis but not by M. africanum strains. PLoS Pathog. 2009 Sep;5(9):e1000577. doi: 10.1371/journal.ppat.1000577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.King KY, Lew JD, Ha NP, Lin JS, Ma X, Graviss EA, et al. Polymorphic allele of human IRGM1 is associated with susceptibility to tuberculosis in African Americans. PLoS One. 2011;6(1):e16317. doi: 10.1371/journal.pone.0016317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Bahari G, Hashemi M, Taheri M, Naderi M, Eskandari-Nasab E, Atabaki M. Association of IRGM polymorphisms and susceptibility to pulmonary tuberculosis in Zahedan, Southeast Iran. ScientificWorldJournal. 2012;2012:950801. doi: 10.1100/2012/950801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Song JH, Kim SY, Chung KS, Moon CM, Kim SW, Kim EY, et al. Association between genetic variants in the IRGM gene and tuberculosis in a Korean population. Infection. 2014 Aug;42(4):655–660. doi: 10.1007/s15010-014-0604-6. [DOI] [PubMed] [Google Scholar]
- 195.Songane M, Kleinnijenhuis J, Alisjahbana B, Sahiratmadja E, Parwati I, Oosting M, et al. Polymorphisms in autophagy genes and susceptibility to tuberculosis. PLoS One. 2012;7(8):e41618. doi: 10.1371/journal.pone.0041618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Hawn TR, Matheson AI, Maley SN, Vandal O. Host-directed therapeutics for tuberculosis: can we harness the host? Microbiol Mol Biol Rev. 2013 Dec;77(4):608–627. doi: 10.1128/MMBR.00032-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Kim JJ, Lee HM, Shin DM, Kim W, Yuk JM, Jin HS, et al. Host cell autophagy activated by antibiotics is required for their effective antimycobacterial drug action. Cell Host Microbe. 2012 May 17;11(5):457–468. doi: 10.1016/j.chom.2012.03.008. [DOI] [PubMed] [Google Scholar]
- 198.Ho-Pham LT, Nguyen ND, Nguyen TT, Nguyen DH, Bui PK, Nguyen VN, et al. Association between vitamin D insufficiency and tuberculosis in a Vietnamese population. BMC Infect Dis. 2010;10:306. doi: 10.1186/1471-2334-10-306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Talat N, Perry S, Parsonnet J, Dawood G, Hussain R. Vitamin d deficiency and tuberculosis progression. Emerg Infect Dis. 2010 May;16(5):853–855. doi: 10.3201/eid1605.091693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Hong JY, Kim SY, Chung KS, Kim EY, Jung JY, Park MS, et al. Association between vitamin D deficiency and tuberculosis in a Korean population. Int J Tuberc Lung Dis. 2014 Jan;18(1):73–78. doi: 10.5588/ijtld.13.0536. [DOI] [PubMed] [Google Scholar]
- 201.Mehta S, Mugusi FM, Bosch RJ, Aboud S, Urassa W, Villamor E, et al. Vitamin D status and TB treatment outcomes in adult patients in Tanzania: a cohort study. BMJ Open. 2013;3(11):e003703. doi: 10.1136/bmjopen-2013-003703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nielsen NO, Skifte T, Andersson M, Wohlfahrt J, Soborg B, Koch A, et al. Both high and low serum vitamin D concentrations are associated with tuberculosis: a case-control study in Greenland. Br J Nutr. 2010 Nov;104(10):1487–1491. doi: 10.1017/S0007114510002333. [DOI] [PubMed] [Google Scholar]
- 203.Nnoaham KE, Clarke A. Low serum vitamin D levels and tuberculosis: a systematic review and meta-analysis. Int J Epidemiol. 2008 Feb;37(1):113–119. doi: 10.1093/ije/dym247. [DOI] [PubMed] [Google Scholar]
- 204.Wejse C, Gomes VF, Rabna P, Gustafson P, Aaby P, Lisse IM, et al. Vitamin D as supplementary treatment for tuberculosis: a double-blind, randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2009 May 1;179(9):843–850. doi: 10.1164/rccm.200804-567OC. [DOI] [PubMed] [Google Scholar]
- 205.Coussens AK, Wilkinson RJ, Hanifa Y, Nikolayevskyy V, Elkington PT, Islam K, et al. Vitamin D accelerates resolution of inflammatory responses during tuberculosis treatment. Proc Natl Acad Sci U S A. 2012 Sep 18;109(38):15449–15454. doi: 10.1073/pnas.1200072109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Saunders RN, Metcalfe MS, Nicholson ML. Rapamycin in transplantation: a review of the evidence. Kidney Int. 2001 Jan;59(1):3–16. doi: 10.1046/j.1523-1755.2001.00460.x. [DOI] [PubMed] [Google Scholar]
- 207.Butzal M, Loges S, Schweizer M, Fischer U, Gehling UM, Hossfeld DK, et al. Rapamycin inhibits proliferation and differentiation of human endothelial progenitor cells in vitro. Exp Cell Res. 2004 Oct 15;300(1):65–71. doi: 10.1016/j.yexcr.2004.07.002. [DOI] [PubMed] [Google Scholar]
- 208.Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 2004 Jun;36(6):585–595. doi: 10.1038/ng1362. [DOI] [PubMed] [Google Scholar]
- 209.Gupta A, Pant G, Mitra K, Madan J, Chourasia MK, Misra A. Inhalable particles containing rapamycin for induction of autophagy in macrophages infected with Mycobacterium tuberculosis. Mol Pharm. 2014 Apr 7;11(4):1201–1207. doi: 10.1021/mp4006563. [DOI] [PubMed] [Google Scholar]
- 210.Parihar SP, Guler R, Khutlang R, Lang DM, Hurdayal R, Mhlanga MM, et al. Statin therapy reduces the mycobacterium tuberculosis burden in human macrophages and in mice by enhancing autophagy and phagosome maturation. J Infect Dis. 2014 Mar 1;209(5):754–763. doi: 10.1093/infdis/jit550. [DOI] [PubMed] [Google Scholar]
- 211.Skerry C, Pinn ML, Bruiners N, Pine R, Gennaro ML, Karakousis PC. Simvastatin increases the in vivo activity of the first-line tuberculosis regimen. J Antimicrob Chemother. 2014 Sep;69(9):2453–2457. doi: 10.1093/jac/dku166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Aldea M, Craciun L, Tomuleasa C, Berindan-Neagoe I, Kacso G, Florian IS, et al. Repositioning metformin in cancer: genetics, drug targets, and new ways of delivery. Tumour Biol. 2014 Jun;35(6):5101–5110. doi: 10.1007/s13277-014-1676-8. [DOI] [PubMed] [Google Scholar]
- 213.Ben S, I, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, et al. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 2011 Jul 1;71(13):4366–4372. doi: 10.1158/0008-5472.CAN-10-1769. [DOI] [PubMed] [Google Scholar]
- 214.Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, et al. Small molecule regulators of autophagy identified by an image-based high-throughput screen. Proc Natl Acad Sci U S A. 2007 Nov 27;104(48):19023–19028. doi: 10.1073/pnas.0709695104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqi FH, et al. Novel targets for Huntington's disease in an mTOR-independent autophagy pathway. Nat Chem Biol. 2008 May;4(5):295–305. doi: 10.1038/nchembio.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Koh GCKW, Maude RR, Schreiber MF, Limmathurotsakul D, Wiersinga WJ, Wuthiekanun V, et al. Glyburide is anti-inflammatory and associated with reduced mortality in melioidosis. Clin Infect Dis. 2011;52(6):717–725. doi: 10.1093/cid/ciq192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Liu X, Foo G, Lim WP, Ravikumar S, Sim HS, Win MS, et al. Sulfonylurea usage in melioidosis is associated with severe disease and suppressed immune response. PLoS Negl Trop Dis. 2014;8(4):e2795. doi: 10.1371/journal.pntd.0002795. [DOI] [PMC free article] [PubMed] [Google Scholar]