Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 1.
Published in final edited form as: Cell Microbiol. 2019 Jan 15;21(3):e12988. doi: 10.1111/cmi.12988

Listeriolysin O: a phagosome-specific cytolysin revisited

Brittney N Nguyen 1, Bret N Peterson 1, Daniel A Portnoy 2,3,*
PMCID: PMC6779163  NIHMSID: NIHMS1515399  PMID: 30511471

Abstract

Listeriolysin O (LLO) is an essential determinant of Listeria monocytogenes pathogenesis that mediates the escape of L. monocytogenes from host cell vacuoles, thereby allowing replication in the cytosol without causing appreciable cell death. As a member of the cholesterol-dependent cytolysin (CDC) family of pore-forming toxins, LLO is unique in that it is secreted by a facultative intracellular pathogen, whereas all other CDCs are produced by pathogens that are largely extracellular. Replacement of LLO with other CDCs results in strains that are extremely cytotoxic and 10,000-fold less virulent in mice. LLO has structural and regulatory features that allow it to function intracellularly without causing cell death, most of which map to a unique N-terminal region of LLO referred to as the PEST-like sequence. Yet, while LLO has unique properties required for its intracellular site of action, extracellular LLO, like other CDCs, affects cells in a myriad of ways. Because all CDCs form pores in cholesterol-containing membranes that lead to rapid Ca2+ influx and K+ efflux, they consequently trigger a wide range of host cell responses, including MAPK activation, histone modification, and caspase-1 activation. There is no debate that extracellular LLO, like all other CDCs, can stimulate multiple cellular activities, but the primary question we wish to address in this perspective is whether these activities contribute to L. monocytogenes pathogenesis.

Keywords: Cholesterol, toxin, bacteria, intracellular pathogen, Listeria, Macrophage, inflammasome

1. Introduction to Cholesterol-Dependent Cytolysins

Cholesterol-dependent cytolysins (CDCs) represent the largest family of pore-forming toxins (PFTs) and the subset of PFTs that form the largest pores (Bischofberger, Iacovache, & van der Goot, 2012). To date, more than 50 CDCs have been identified in Firmicutes, Actinobacteria, and most recently in Proteobacteria (Hotze et al., 2013; Tweten, Hotze, & Wade, 2015). With the exception of those produced by Proteobacteria, CDCs are produced by primary and opportunistic Gram-positive pathogens, and many have important roles in pathogenesis. Among CDCs that have demonstrated contributions to pathogenesis are perfringolysin (PFO) of Clostridium perfringens, pneumolysin (PLY) of Streptococcus pneumoniae, streptolysin O (SLO) of Streptococcus pyogenes, anthrolysin (ALO) of Bacillus anthracis, and listeriolysin O (LLO) of Listeria monocytogenes. LLO is distinct in that it is the only CDC produced by an intracellular pathogen and has specialized features that make it suitable for its intracellular localization.

The hallmarks of CDCs are their requirement of membrane cholesterol for pore-forming activity, and their extremely large pores—which can be 30 – 40 nm in diameter (Gilbert, 2010; Mulvihill, Van Pee, Mari, Muller, & Yildiz, 2015). CDCs also contain a singular conserved cysteine that makes them highly sensitive to oxidation; CDCs were once classified as ‘thiol-activated (oxygen-sensitive) cytolysins’ because they required reducing agents for maximal activity (Morgan, Andrew, & Mitchell, 1996; Smyth & Duncan, 1978).

CDCs are secreted via a Sec-dependent pathway as monomers 50–70 kDa in mass and consist of four distinct domains. Secreted monomers bind to cell membranes and oligomerize into arc and ring prepore assemblies, which may contain up to 50 subunits. Following membrane binding, α-helical regions in domain 3 of each monomer refold into two β-hairpins that insert into the membrane and form a β-barrel pore (Christie, Johnstone, Tweten, Parker, & Morton, 2018; Leung et al., 2014; Mulvihill et al., 2015). Domain 4 contains the signature undecapeptide sequence (ECTGLAWEWWR) that is the most highly conserved region in the primary CDC sequence and is required for coupling of cholesterol binding to domain 3 rearrangement (Dowd & Tweten, 2012). The cholesterol recognition/binding motif, which consists of a threonine-leucine pair, is also located in domain 4 (Farrand, LaChapelle, Hotze, Johnson, & Tweten, 2010). Both incomplete ring oligomers (arcs or slits) and complete rings perforate cell membranes, though pores formed by arcs are considerably smaller and may only function as ion channels, while rings allow the translocation of fully folded proteins (Palmer et al., 1998). There is also evidence that CDCs translocate proteins in vivo, thereby acting as secretion systems (Madden, Ruiz, & Caparon, 2001).

Although cholesterol is required for CDC activity and is generally considered the CDC receptor, a number of CDCs use human CD59 as a receptor and consequently have increased specificity for human cell membranes. However, these CDCs still require cholesterol for pore formation. These include intermedilysin (ILY) of Streptococcus intermedius, vaginolysin (VLY) of Gardnerella vaginalis, and lectinolysin (LLY) of Streptococcus mitis. The use of CD59 as a receptor may be attributed to a proline residue in place of a tryptophan in the undecapeptide (Lawrence et al., 2016). Additionally, CDCs also have conserved lectin-binding properties (Shewell et al., 2014). Using glycan array analysis, it was shown that PLY and SLO had affinities for different glycan structures and that binding these glycans altered the hemolytic activity of these toxins. Like cholesterol, the functional domain responsible for glycan binding is domain 4. While glycan binding has not yet been reported for LLO, many of the modeled carbohydrate binding sites within domain 4 are conserved between LLO and CDCs from extracellular pathogens. Future experiments should investigate the roles of glycosylation with respect to cellular tropism and pathogen lifestyle.

Although the structure and mechanism of pore formation of CDCs are largely conserved, several CDCs have variations in their structure that contribute to changes in function. SLO has 60 amino acids at its N-terminus that mediate specific translocation of NAD+ glycohydrolase (SPN) into keratinocytes (Madden et al., 2001). Translocation of SPN induces cell death, following depletion of cellular NAD+, and significantly increases the virulence of S. pyogenes (Chandrasekaran & Caparon, 2016; Zhu et al., 2017). PLY lacks a signal peptide and may be released by cell lysis or by another export mechanism (Lemon & Weiser, 2015; Price, Greene, & Camilli, 2012). PLY also localizes to the cell wall, and its cell wall localization is dependent on SecY2A2, an accessory Sec system (Bandara et al., 2017). LLO has a 26-amino acid addition (known as the PEST-like sequence) near its N-terminus that reduces the intracellular toxicity of LLO and is necessary for L. monocytogenes to survive intracellularly following escape from phagocytic vacuoles (Schnupf & Portnoy, 2007). The role of the LLO PEST-like sequence in pathogenesis will be discussed in depth below.

2. Cellular Responses to CDC-Mediated Pore Formation

CDCs can induce a wide range of effects in cells, including activation of membrane damage responses and alteration of immune cell function. Among other things, CDCs can activate MAPKs, caspase-1, and TLR4, modulate SUMOylation, induce mitochondrial fragmentation, cause T cell apoptosis, and enhance bacterial internalization (Cajnko, Mikelj, Turk, Podobnik, & Anderluh, 2014; Cassidy & O’Riordan, 2013; Seveau, 2014). These responses are usually common to membrane insult by a range of PFTs and are often the direct result of Ca2+ influx and/or K+ efflux. As a result of these numerous and diverse effects, LLO has been called the ‘Swiss-army knife of Listeria’ (Hamon, Ribet, Stavru, & Cossart, 2012; Osborne & Brumell, 2017). However, the role of these cellular responses in pathogenesis is not clear.

For more details on cellular responses to pore formation, refer to the following references: Cajnko et al., 2014, Cassidy & O’Riordan, 2013, Seveau, 2014, and Gonzalez, Bischofberger, Pernot, van der Goot, & Frêche, 2008.

2.1. Mitogen-activated Protein Kinase (MAPK) activation

MAPKs are involved in the initiation of signaling cascades that activate cellular responses to many stimuli. Cell membrane damage by PFTs causes the rapid efflux of intracellular K+, and activation of the MAP kinases p38, extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK), mitogen- and stress-activated kinase 1 and 2 (MSK1/2), and cAMP response-element binding protein (CREB). Activation of p38 and ERK are required for recovery of intracellular K+ levels following treatment of cells with sublytic concentrations of LLO and aerolysin, a non-CDC PFT that forms 2 nm pores (Cabezas et al., 2017; Gonzalez et al., 2011). In Caenorhabditis elegans, p38 and JNK MAPK pathways, and importantly one downstream target, activator protein 1 (AP-1), provide protection against PFT toxicity (Kao et al., 2011). Thus, the restoration of ion homeostasis is one effect of MAPK activation in response to membrane perforation by PFTs.

2.2. Histone Modification

Histone modification has been observed in response to multiple pathogens and their CDCs. L. monocytogenes infection causes phosphorylation or dephosphorylation of Ser10 in histone H3 and acetylation or deacetylation of histone H4, depending on the experimental conditions (Hamon et al., 2007; Schmeck et al., 2005). In human umbilical vein endothelial cells, L. monocytogenes infection caused phosphorylation of Ser10 in histone H3, leading to increased expression of numerous cytokines in a p38 MAPK-dependent manner (Schmeck et al., 2005). Conversely, in human cervical epithelial cells (HeLa cells), L. monocytogenes infection or LLO alone caused dephosphorylation of Ser10 in histone H3, deacetylation of histone H4, and transcriptional repression of cxcl2, a cytokine involved in inflammation and neutrophil chemotaxis (Hamon et al., 2007). Treatment of HeLa cells with aerolysin, PFO, and PLY also results in dephosphorylation of Ser10 in histone H3, and dephosphorylation is dependent on K+ efflux (Hamon & Cossart, 2011). Part of the Pseudomonas aeruginosa Type III secretion system, the PopB-PopD translocon, can form 4 nm pores on cell membranes that also result in K+ efflux-dependent dephosphorylation of Ser10 in histone H3 in HeLa cells (Dortet, Lombardi, Cretin, Dessen, & Filloux, 2018). Thus, K+ efflux resulting from membrane pore formation may have the ability to alter transcriptional profiles in ways that affect inflammation, although a role in vivo has not been demonstrated.

2.3. Alteration of Mitochondrial Dynamics

Treatment of HeLa cells with a sublytic concentration of recombinant LLO caused mitochondrial fragmentation, defined by breakage of the mitochondrial network into visually punctate structures. Mitochondria fragmentation occurred quickly after LLO treatment, and was transient; L. monocytogenes-infected cells completely recovered their normal mitochondrial network phenotype overnight, and did not become apoptotic. Mitochondrial fragmentation was dependent on Ca2+ influx, as cells incubated in Ca2+-free media did not undergo mitochondrial fragmentation upon LLO addition (Stavru, Bouillaud, Sartori, Ricquier, & Cossart, 2011). In addition to causing histone modifications, infection of HeLa cells with P. aeruginosa also caused mitochondrial fragmentation, and was dependent on the expression of PopB and PopD (Dortet et al., 2018).

2.4. SUMOylation

SUMOylation is a eukaryotic post-translational modification, similar to ubiquitylation, in which small ubiquitin-like modifier (SUMO) is covalently attached to proteins. Most commonly, SUMOylation of transcriptional regulators leads to transcriptional repression (Gill, 2005). Treatment of HeLa cells with LLO, PFO, PLY, and suilysin (SLY)—a CDC produced by Streptococcus suis—resulted in degradation of Ubc9, an E2 SUMO enzyme, and a reduction in SUMO-conjugated proteins (Li, Lam, Lai, & Au, 2017; Ribet et al., 2010). Interestingly, the patterns of SUMOylated proteins were different for each CDC. Blockage of K+ efflux prevented LLO-induced degradation of Ubc9, but enhanced Ubc9 degradation induced by PFO, PLY, and SLY, suggesting CDCs have different mechanisms for inducing Ubc9 degradation (Li et al., 2017). Overexpression of SUMO 1 and SUMO 2 in HeLa cells limited infection of L. monocytogenes 2-fold (Ribet et al., 2010). Based on the observation that mice deficient for promyelocytic leukemia protein (PML), a known target of SUMO, had a defect in controlling L. monocytogenes, a recent study investigated the relationship between LLO, PML and SUMO during infection (Lunardi et al., 2011; Ribet et al., 2017). The authors reported that treatment with LLO, PFO, and PLY caused deSUMOylation of PML in Chinese hamster ovary (CHO) cells, and that gene expression of many cytokines was reduced in pml−/− mouse embryonic fibroblasts (MEFs) compared to pml+/+ MEFs after L. monocytogenes infection, though the altered gene expression was not dependent on LLO (Ribet et al., 2017). A better understanding of how SUMOylation affects pathogenesis can be acquired by performing studies in mice with conditional deletions of Ubc9 (Demarque et al., 2011; Fritah et al., 2014; Wang et al., 2017).

2.5. Caspase-1 activation

One of the hallmarks of the innate immune system is that activation of host pattern recognition receptors by conserved microbial products, known as pathogen-associated molecular patterns (PAMPs), and aberrant structure or localization of host molecules that result during disease, often referred to as damage-associated molecular patterns (DAMPs), leads to the initiation of immune responses (Brubaker, Bonham, Zanoni, & Kagan, 2015; Jounai, Kobiyama, Takeshita, & Ishii, 2012; Land, 2015; Schaefer, 2014; Vénéreau, Ceriotti, & Bianchi, 2015). An example of a DAMP is extracellular ATP, which binds and activates P2X7, an ATP-gated ion channel. Binding of ATP to P2X7 results in K+ efflux and K+ efflux-dependent NLRP3 inflammasome activation (Ayna et al., 2012; Pétrilli et al., 2007; Walev, Reske, Palmer, Valeva, & Bhakdi, 1995). Caspase-1 cleaves pro-interleukin 1 beta (IL-1β) into active IL-1β downstream of NLRP3 activation, which can occur spontaneously in low concentrations of K+ (He, Zeng, Yang, Motro, & Núñez, 2016; Pétrilli et al., 2007). Many PFTs activate the NLRP3 inflammasome, including the pore-forming component of adenylate cyclase toxin (CyaA) produced by Bordetella pertussis, aerolysin, SLO, LLO and tetanolysin, a CDC produced by Clostridium tetani (Ayna et al., 2012; Chu et al., 2009; Dunne et al., 2010; Gurcel, Abrami, Girardin, Tschopp, & van der Goot, 2006; Harder et al., 2009; Idzko, Ferrari, & Eltzschig, 2014; Walev et al., 1995). Roles for the inflammasome in the pathogenesis of B. pertussis, S. pneumoniae, and L. monocytogenes have been reported, although in the case of L. monocytogenes, excess inflammasome activation reduces virulence (Dunne et al., 2010; Brian T Edelson & Unanue, 2002; Hassane et al., 2017; Karmakar et al., 2015; W. Li et al., 2016; J.-D. Sauer et al., 2011; Tsuji et al., 2004; Witzenrath et al., 2011).

3. Contributions of CDCs to Pathogenesis of Extracellular Pathogens

Many CDCs have clear roles in pathogenesis that are often related to the recruitment of immune cells or disruption of tissue barriers. Though all CDCs function by forming pores on host membranes, there are some differences in how they contribute to pathogenesis. Differences in how CDCs function in vivo are likely due to differences in infection sites and the simultaneous effects of other bacterial factors.

3.1. PFO

Upon introduction of C. perfringens into soft tissue, often by a traumatic injury, C. perfringens can cause clostridial myonecrosis, a necrotic infection of muscle that is largely devoid of infiltrating leukocytes (Soltani, Hotze, Johnson, & Tweten, 2007; Titball, 2005). Although C. perfringens produces many toxins, PFO (also called θ–toxin) is critical for severe pathology. PFO acts synergistically with α-toxin, a phospholipase produced by C. perfringens, in the development of clostridial myonecrosis (Ellemor et al., 1999). α-toxin causes most of the damage to the muscle tissue, while PFO contributes to pathology by inducing leukocyte aggregation in the vasculature—thereby preventing infiltration of leukocytes into the site of infection (Awad, Ellemor, Boyd, Emmins, & Rood, 2001). Deletion of PFO, but not α-toxin, in a mouse muscle model resulted in an almost complete reduction in severe leukocyte accumulation, which was likely caused by the upregulation of leukocyte and endothelial cell adhesion factors (Bryant & Stevens, 1996; Ellemor et al., 1999; Verherstraeten et al., 2015). Treatment of mice with a PFO-neutralizing antibody prior to infection with a lethal dose of intramuscular C. perfringens significantly reduced mortality (Bryant et al., 1993). Additionally, PFO is required for persistence of C. perfringens in a low-dose mouse femoral muscle infection model (O’Brien & Melville, 2004). Disruption of leukocyte migration and subsequent inflammation induced by PFO contributes to the disease progression of clostridial myonecrosis.

3.2. PLY

S. pneumoniae is the causative agent for a number of diseases, including pneumonia, otitis media, meningitis, and sepsis (Mitchell & Dalziel, 2014). PLY has a significant role in the pathogenesis of pneumococcal pneumonia and sepsis (Benton, Everson, & Briles, 1995; Berry, Yother, Briles, Hansman, & Paton, 1989). In a mouse S. pneumoniae upper respiratory tract infection model, S. pneumoniae strains with PLY were shed in nasal secretions at higher levels than PLY-deficient strains, and PLY was required for transmission (Zafar, Wang, Hamaguchi, & Weiser, 2017). In an intranasal mouse infection model of pneumonia and septicemia, mice infected with PLY-negative bacteria did not develop severe disease, whereas mice infected with S. pneumoniae expressing PLY were moribund within 48 hours. Mice infected with the PLY-negative mutant had 4-logs less bacteria in both the lungs and blood (Kadioglu et al., 2000). In addition, mice treated with a PLY-neutralizing antibody prior to infection with a lethal dose of S. pneumoniae administered intranasally or intraperitoneally had significantly increased survival (Del Mar García-Suárez et al., 2004; Musher, Phan, & Baughn, 2001). PLY reduced ciliary beating and caused reduced or disorganized cilia on the epithelial cells of human adenoid organ cultures. This result correlated with increased numbers of S. pneumoniae adhering to the organ cultures, mostly on damaged cells near disrupted tight junctions (Rayner et al., 1995). Therefore, PLY may contribute to dissemination of S. pneumoniae during infection of the lungs by disrupting mucociliary elevator-mediated bacterial clearance and allowing the bacteria to invade deeper tissues through disrupted tight junctions.

PLY-induced IL-1β secretion provides partial protection to the host during S. pneumoniae infections. IL-1β secretion in response to PLY is caspase-1, NLRP3, and ASC-dependent in many cell types (Hassane et al., 2017; Karmakar et al., 2015; Mariathasan et al., 2006). IL-1β−/−, caspase-1/11−/−, ASC−/− and NLRP3−/− mice had 1-log increased bacterial burdens compared to WT mice in a S. pneumoniae keratitis model (Karmakar et al., 2015). In a mouse lung infection model, S. pneumoniae infection caused increased lung permeability, which was exacerbated in mice lacking NLRP3 (Witzenrath et al., 2011). IL-1β contributed to the activation of γδT cells, and their production of IL-17A. IL-17A-deficient, TCRδ-deficient, and neutrophil-depleted mice had significantly reduced survival compared to WT mice (Hassane et al., 2017). Thus, inflammasome activation by PLY leads to the generation of the Th17 response, which provides some protection against S. pneumoniae infection.

3.3. ALO

Bacillus anthracis is capable of causing severe disease in humans after inhalation, ingestion, or cutaneous introduction of bacterial spores. In all cases, the disease begins as a localized infection that can quickly lead to sepsis (Owen, Yang, & Mohamadzadeh, 2015). ALO plays a significant role in pathogenesis (Shannon, Ross, Koehler, & Rest, 2003). Administration of 100 µg of ALO-neutralizing antibody to mice infected with a lethal intravenous dose of B. anthracis significantly increased survival (Nakouzi, Rivera, Rest, & Casadevall, 2008). ALO is required for disruption of tight junctions and the gut epithelial barrier, and for the apical to basolateral translocation of B. anthracis across C2BBE monolayers, suggesting that the function of ALO is to facilitate the early establishment and penetration of B. anthracis into the gut epithelium (Bishop, Lodolce, Kolodziej, Boone, & Tang, 2010). Though significant, the role of ALO in systemic infection is not well characterized.

3.4. Varying Roles for CDCs in Pathogenesis

The role of CDCs in the pathogenesis of extracellular pathogens often involves damaging, but not necessarily killing, cells in and around the site of infection. CDCs cause cell remodeling, such as ciliary rearrangement, which may promote bacterial adhesion; they can disrupt tight junctions, facilitating bacterial translocation through epithelial and endothelial barriers and thus facilitating dissemination; and they can alter the expression of adherence factors, which can lead to recruitment of phagocytes and inflammation, or prevention of immune cell infiltration. Though many of these functions do not result in cell death, host cell lysis can also be a survival strategy for extracellular pathogens. Indeed, both C. perfringens and B. anthracis use their CDC to lyse host cells subsequent to phagocytosis, thereby releasing the bacteria back into the extracellular space and promoting bacterial growth (Heffernan, Thomason, Herring-Palmer, & Hanna, 2007; O’Brien & Melville, 2004). Cell lysis may also provide extracellular pathogens with nutrients. These CDC-mediated effects are well suited to the needs of extracellular pathogens.

4. The Role of LLO in Disease

L. monocytogenes is a facultative intracellular pathogen that, in humans, primarily causes self-resolving gastrointestinal infections. In immunocompromised individuals, L. monocytogenes can cause fatal systemic infections and, in pregnant women, placental infections that lead to pregnancy loss and systemic disease that results in death to the neonate (Lecuit, 2007). LLO is required for virulence in most if not all L. monocytogenes animal disease models, including acute systemic infection in mice, neonatal mice, pregnant mice and pregnant guinea pigs (Bakardjiev, Stacy, & Portnoy, 2005; Gaillard, Berche, & Sansonetti, 1986; Kathariou, Metz, Hof, & Goebel, 1987; Le Monnier et al., 2007; W. Li et al., 2016; McKay & Lu, 1991; Portnoy, Jacks, & Hinrichs, 1988). The requirement for LLO in virulence can be recapitulated in tissue culture where it is required for L. monocytogenes to escape from phagosomes. Mutants lacking LLO are unable to escape from the phagosome and consequently unable to grow intracellularly (Tilney, L. G., Portnoy, 1989). In a mouse systemic infection model, LLO-negative mutants are 5-logs less virulent. The requirement for LLO in escape from the phagosome in vivo has been observed in real-time in infected zebrafish (Levraud et al., 2009). Strikingly, replacement of LLO with other CDCs results in strains that can escape from a phagosome but then kill the infected host cell, thereby eliminating the intracellular replicative niche (Decatur & Portnoy, 2000; S. Jones & Portnoy, 1994; Portnoy, Tweten, Kehoe, & Bielecki, 1992; Wei et al., 2005). It is important to note that there are populations of L. monocytogenes that replicate extracellularly in the gut and gallbladder, and LLO is not required for the establishment of infection at these sites. However, while wildtype L. monocytogenes can disseminate from the gut to establish infection in systemic organs, LLO-deficient bacteria cannot efficiently disseminate from the gut to systemic sites (Hardy et al., 2004; G. S. Jones et al., 2015; Roll & Czuprynski, 1990).

4.1. LLO Activity is pH-Dependent

The optimal pH for LLO activity is 5.5, while extracellular CDCs such as PFO and SLO have similar activities at pH 5.5 and pH 7, suggesting that LLO has adapted to the specific setting of the acidified phagosome (Geoffroy, Gaillard, Alouf, & Berche, 1987; Portnoy et al., 1992). An early study into the molecular basis of this low optimal pH found that amino acid L461 was the main determinant, and that this leucine is not conserved in CDCs from extracellular pathogens (Glomski, Gedde, Tsang, Swanson, & Portnoy, 2002). Nonsynonymous mutations of L461 affect LLO activity and cytotoxicity. Mutants with a threonine substitution, the residue common in extracellular pathogen CDCs, were 100-fold less virulent in mice due to their increased cytotoxicity. The pH insensitivity of L461T may be caused by an increase in the rate of oligomerization. Later it was reported that LLO is denatured at neutral pH at temperatures greater than 30 C, and that this was caused by charged amino acids within the transmembrane helices of domain 3 that act as a pH sensor (Schuerch, Wilson-Kubalek, & Tweten, 2005). Thus, while LLO is maximally active in acidified phagosomes, in the host cell cytosol its activity is partially reduced and it has the potential to denature. This mechanism is not solely responsible for limiting the activity of LLO to the phagosome, but it does contribute to reducing LLO-mediated cytotoxicity and preserving the replicative niche.

4.2. The LLO PEST-like Sequence

The most distinctive and single largest contributing feature of LLO for the L. monocytogenes-specific lifestyle is a PEST-like sequence at the amino terminus of the protein (Decatur & Portnoy, 2000; Lety et al., 2001). PEST-like sequences were originally described in eukaryotic proteins with short half-lives and were thought to mediate those short half-lives, but it is now appreciated that they often include another domain known as a polyproline type II (PPII) helix that mediates protein-protein interactions (Köster et al., 2014; Rechsteiner & Rogers, 1996; Rogers, Wells, & Rechsteiner, 1986). Structural and in vitro analyses have indicated that residues in the PPII helix region play a role in oligomerization through intermolecular contacts (Köster et al., 2014). Deletion of 26 amino acids of LLO that include the PEST-like sequence has a minor effect on hemolytic activity; however, the bacteria are extremely cytotoxic in tissue culture and 10,000-fold less virulent in mice (Decatur & Portnoy, 2000).

Intracellular LLO exists in multiple forms, including 58kDa and 55kDa molecular weight species. The lighter species is absent during infection with the PEST-deletion mutant or mutants deficient in actin-based motility, suggesting the PEST-like sequence contributes to subcellular compartmentalization or processing of LLO (Schnupf, Portnoy, & Decatur, 2006). Additionally, independently of the PEST-like sequence, LLO is ubiquitylated and accumulates as a ladder of higher molecular weight species in the presence of proteasome inhibitors. LLO has an N-terminal lysine that serves as a destabilizing signal for the N-end rule pathway, which involves ubiquitylation and proteasomal degradation. Indeed, the short intracellular half-life of LLO was extended by replacing the N-terminal lysine with stabilizing amino acids. However, the half-life extension only marginally affected cellular toxicity or virulence unless combined with mutations in the PEST-like sequence (Schnupf, Zhou, Varshavsky, & Portnoy, 2007). Future studies should aim to identify the precise site or sites of ubiquitylation and their roles in pathogenesis and cell biology.

Consistent with the hypothesis that the LLO PEST-like sequence is important for intermolecular interactions, the PEST-like sequence contains three residues (S44, S48, and T51) that are predicted targets for MAPKs, and one or all of these residues are important for LLO phosphorylation inside of infected host cells (Schnupf, Portnoy, et al., 2006). Studies on phosphorylation of the PEST-like sequence have been confounded by the observation that point mutations in the region result in increased protein production and cytotoxicity, and attenuated virulence (Schnupf, Portnoy, et al., 2006). For example, mutations that change the S44 codon to alanine, thereby preventing phosphorylation, have increased translation of LLO. However, mutations that change the S44 codon to other serine codons also have increased translation— suggesting that the PEST-like sequence acts at the mRNA level to affect translation. Further evidence of translational regulation is supported by the observation that mutations in the 5’ UTR alter protein expression (Schnupf, Hofmann, et al., 2006; Shen & Higgins, 2005). The unexplained effect of mutations in the PEST-like sequence on translation complicates the study of post-translational modifications in the PEST-like sequence.

In addition to the above modifications, LLO is covalently modified by exogenously- and endogenously-produced S-glutathione at its cysteine residue (Portman, Huang, Reniere, Iavarone, & Portnoy, 2017). Modification of this residue may modulate the activity of all extracellular CDCs or restrict their activity to phagosomes containing oxidoreductases. For example, this cysteine has been implicated as a target for the phagosomal thiol-reductase known as GILT (Singh, Jamieson, & Cresswell, 2008). GILT−/− mice and macrophages were more resistant to L. monocytogenes due to a defect in phagosomal escape, presumably because LLO activity was reduced by modification with glutathione or another low molecular weight thiol. Thus, the presence of a host oxidoreductase, such as GILT, can confer cellular specificity to CDC-producing pathogens by activating CDCs in the phagosome and promoting escape. However, mutant L. monocytogenes in which the LLO cysteine is substituted with an alanine have a very small virulence defect (Portman et al., 2017).

Although a lot of work is still required to understand the role of LLO modifications inside host cells, recent work has provided a detailed mechanism describing how its N-terminus uses host cell machinery to promote LLO degradation (Chen et al., 2018). Within cells, LLO localized to puncta within the cytosol while LLO lacking the PEST-like sequence was found on the host plasma membrane. This was due to interaction of the PEST-like sequence with the host Ap2a2 subunit of the clathrin-dependent endocytosis machinery, supporting a model in which LLO prevents cytotoxicity by accelerating the removal of membrane-associated LLO by endocytosis and targeting to autophagosomes. Interestingly, replacement of the LLO PEST-like sequence with the PEST-like sequence of human calcium receptor protein (HCaR), a G protein-coupled receptor that also interacts with Ap2a2, restored much of the virulence defect seen in a PEST deletion mutant. Though there are still some unanswered questions about the individual functions of this region of LLO, it is clear that the PEST-like sequence reduces the cytotoxicity of LLO.

4.3. Contribution of LLO to L. monocytogenes Pathogenesis

As discussed above, LLO has many other putative functions that are shared with CDCs produced by extracellular pathogens. Antibody to CDCs can often dramatically affect pathogenesis, as has been shown for PFO, PLY, ALO, and SLY (Bryant et al., 1993; Del Mar García-Suárez et al., 2004; Musher et al., 2001; Nakouzi et al., 2008; Takeuchi et al., 2014). In the case of L. monocytogenes, pretreatment of mice with 1 mg of LLO neutralizing antibody, 10-times the amount of antibody required to effectively neutralize ALO and PLY in vivo, resulted in reduced bacterial burden (B T Edelson, Cossart, & Unanue, 1999). However, it was later shown that this amount of antibody blocked the activity of LLO inside of cells and prevented vacuolar escape (Asano et al., 2016; B. T. Edelson & Unanue, 2001). These results suggest that LLO is required for pathogenesis of L. monocytogenes because it enables vacuolar escape, and that extracellular LLO has little if any effect on pathogenesis.

5. Future Considerations

Tissue culture models of infection provide a convenient way to study the effects that pathogens exert upon cells, and can shed insight into the host and bacterial factors required for any observed phenotypes. Bacterial mutants or antibody can be used to demonstrate the requirement for specific bacterial gene products in a given phenotype, and host mutants or specific inhibitors can be used to demonstrate host requirements. These are powerful strategies that have been used often to show the role of CDCs in the induction of host responses. For example, the conclusion that CDCs of S. pneumoniae, and L. monocytogenes induce inflammasome-dependent IL-1β secretion results from two distinct findings (1) deletion of the CDCs diminished IL-1β secretion and (2) deletion of inflammasome components also diminished IL-1β secretion from cells. However, once the host and bacterial requirements for an in vitro phenotype have been established, how do we accurately determine if and how the phenotype translates into an effect on pathogenesis in vivo?

If a host is genetically tractable and the host factor in question is nonessential, it is possible to use a similar combinatorial approach, which can appropriately be called ‘genetics-squared’ (Persson & Vance, 2007). For some of the proposed LLO functions, genetic models can be used to verify the role of the host factors in pathogenesis. For example, Ubc9+/− mice have been used to demonstrate the importance of SUMOylation in control of Shigella flexneri, which is also a facultative intracellular pathogen, and could be used similarly for L. monocytogenes (Fritah et al., 2014). Host gene deletions were used to understand the role of IL-1β in the pathogenesis of S. pneumoniae and L. monocytogenes. Caspase-1/11−/− mice infected with S. pneumoniae had increased bacterial burdens compared to WT mice, effectively demonstrating the role of caspase-1 in control of S. pneumoniae (Karmakar et al., 2015). However, PLY-deficient strains were not used in the in vivo experiments and thus we are left with questions: would the PLY-deficient strain grow better than wildtype S. pneumoniae in WT mice as a result of not activating the inflammasome, and if so, would that benefit still occur in caspase-1-deficient mice? These approaches are not straightforward because of the multiple effects of individual virulence factors but nevertheless should be performed whenever possible. In L. monocytogenes, LLO showed the same capacity for activating caspase-1 as other PFTs in vitro, while infection of caspase-1/11−/− mice yielded opposing results in vivo and, in our hands, had no effect on infection or immunity in mice (Sauer et al., 2011). Thus, similar tissue culture model results do not always translate directly to similar effects on pathogenesis. Furthermore, evaluating the role of LLO on caspase-1 activation—and most phenotypes for that matter—in vivo is difficult to assess because LLO-negative bacteria cannot grow intracellularly.

How, then, can the role of LLO in vivo be validated separately from its essential role in vacuolar escape? One strategy used to validate the significance of extracellular CDCs to pathogenesis is the use of neutralizing antibodies. Treatment of mice with PFO- PLY-, SLY- and ALO-neutralizing antibodies prior to infection with their respective pathogens resulted in a reduction in disease, thereby providing evidence for their role in disease. Many of the proposed functions of LLO, including MAPK activation, histone dephosphorylation, mitochondrial fragmentation, Ubc9 degradation, and caspase-1 activation occur upon addition of purified LLO to cells. It has been proposed that extracellular LLO that is secreted before bacterial invasion could cause the same effects in vivo. We propose the following experimental process to confirm or disprove that extracellular LLO causes these effects in vivo and that they have an effect on pathogenesis. First, these phenotypes must be identified following infection of mice; second, administration of an LLO-neutralizing antibody must reduce or abrogate the phenotypes; and third, the administration of LLO-neutralizing antibody must affect pathogenesis.

Lastly, how can roles for cytosolic LLO be elucidated when deletion of the gene prevents phagosomal escape thereby preventing secretion of cytosolic LLO? Various approaches including inducible promoters have been used to show that LLO was necessary for cell-to-cell spread (Dancz, Haraga, Portnoy, & Higgins, 2002). An alternative approach would be to incorporate an inducible degradation tag such as the auxin-inducible degron, where LLO could be targeted for degradation in the cytosol (Holland, Fachinetti, Han, & Cleveland, 2012; Nishimura, Fukagawa, Takisawa, Kakimoto, & Kanemaki, 2009). However, this would be very difficult to adapt to animal experiments. Another approach has recently been developed in our lab in which the gene encoding LLO was bracketed by loxP sites in a strain that induces the expression of Cre upon reaching the host cytosol. In this strain, LLO mediates escape from a phagosome, but is rapidly deleted upon reaching the cytosol. This system revealed that intracellular LLO has the potential to be cytotoxic, but that cytotoxicity is reduced by subversion of host endocytosis machinery to remove LLO from the host plasma membrane (Chen et al., 2018). Others have proposed that LLO participates in cell-to-cell spread by causing localized damage in membrane protrusions, resulting in markers of apoptosis that allow those protrusions to be recognized and subject to efferocytosis by adjacent macrophages (Czuczman et al., 2014). The foundation of this concept, and other concepts, could be strengthened by using the strain of L. monocytogenes that escapes the vacuole and deletes LLO in the cytosol. Although the strain is not ideal for all experiments, because it is defective in cell-to-cell spread, it provides a valuable starting point for the evaluation of intracellular LLO phenotypes that was not previously available.

The question remains, is LLO a phagosome-specific cytolysin or a multifunctional virulence factor? LLO has an abundance of features throughout its structure that allow it to mediate the escape of L. monocytogenes from a vacuole without causing excess cytotoxicity in the cytosol. Furthermore, it is absolutely required for disease because of its role in vacuolar escape. Thus, we believe that most evidence points to LLO being a phagosome-specific cytolysin. However, LLO may act extracellularly under some circumstances, perhaps in the intestine or during extracellular growth in the gall bladder (Hardy et al., 2004). The notion that LLO can activate many of the same pathways as extracellular CDCs is intriguing, and the tools exist to validate whether or not LLO activates these pathways in the host in ways that affect the outcome of disease.

Primary function in vivo K+ dependent MAPK activation Histone Modification Mitochondrial fragmentation deSUMOylation/Ubc9 degradation Caspase-1 activation Macrophage TLR4 activation Effects on cell adhesion Protein translocation Vacuolar Escape
CDC Organism
Listeriolysin O (LLO) Listeria monocytogenes Escape from the vacuole (Gonzalez et al., 2011; Tang, Rosenshine, Cossart, & Finlay, 1996) (Hamon et al., 2007) (Stavru et al., 2011) (Ribet et al., 2010) (Meixenberger et al., 2010; Sauer et al., 2010) (Park, Ng, Maeda, Rest, & Karin, 2004) (Drevets, 1997; Kayal et al., 1999; Krüll et al., 1997) (Sibelius et al., 1996) (Tilney, L. G., Portnoy, 1989)
Perfringolysin (PFO) Clostridium perfringens Disruption of neutrophil migration (Ellemor et al., 1999) (Hamon et al., 2007) (Ribet et al., 2010) (Park et al., 2004) (Bryant & Stevens, 1996; Rafii, Park, Bryant, Johnson, & Wagner, 2008) (O’Brien & Melville, 2004)
Pneumolysin (PLY) Streptococcus pneumoniae Epithelial barrier disruption (Rayner et al., 1995) Transmission (Zafar et al., 2017) (Aguilar et al., 2009) (Hamon et al., 2007) (Li et al., 2017; Ribet et al., 2010) (Fang et al., 2011; Hassane et al., 2017; Karmakar et al., 2015; Witzenrath et al., 2011) (Malley et al., 2003; Srivastava et al., 2005) (Nel et al., 2017; Thornton & McDaniel, 2005; Zhang et al., 2016)
Streptolysin O (SLO) Streptococcus pyogenes Cell killing by SPN translocation (Zhu et al., 2017) (Stassen et al., 2003) (Li et al., 2017) (Harder et al., 2009; Keyel et al., 2013) (Park et al., 2004) (Bryant et al., 2005; Zhang et al., 2016) (Madden et al., 2001)
Suilysin (SLY) Streptococcus suis (Bi et al., 2015) (Li et al., 2017) (Bi et al., 2015) (Zhang et al., 2016)
Anthrolysin O (ALO) Bacillus anthracis (Ratner et al., 2006) (Park et al., 2004) (Tonello & Zornetta, 2012)
Vaginolysin (VLY) Gardnerella vaginalis (Gelber, Aguilar, Lewis, & Ratner, 2008)
Other Pore-forming molecules
T3SS (Dortet et al., 2018) (Dortet et al., 2018) (McCoy, Koizumi, Higa, & Suzuki, 2010) (Wagner et al., 2018)
Aerolysin Aeromonas hydrophila (Gonzalez et al., 2011) (Hamon & Cossart, 2011) (Gurcel et al., 2006; McCoy et al., 2010)

Acknowledgments

This work was supported by National Institutes of Health grants 1P01 AI063302 (D.A.P.) and 1R01 AI027655 (D.A.P). B.N.N. and B.N.P. were supported by the National Science Foundation Graduate Research Fellowship under Grant No. DGE 1106400.

Footnotes

Declaration of Interests

Daniel A. Portnoy has a consulting relationship with and a financial interest in Aduro Biotech. Both he and the company stand to benefit from the commercialization of the results of this research.

References

  1. Aguilar JL, Kulkarni R, Randis TM, Soman S, Kikuchi A, Yin Y, & Ratner AJ. (2009). Phosphatase-Dependent Regulation of Epithelial Mitogen-Activated Protein Kinase Responses to Toxin-Induced Membrane Pores. PLoS ONE, 4(11), e8076 10.1371/journal.pone.0008076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Asano K, Sashinami H, Osanai A, Hirose S, Ono HK, Narita K, … Nakane A. (2016). Passive immunization with anti-ActA and anti-listeriolysin O antibodies protects against Listeria monocytogenes infection in mice. Scientific Reports, 6(1), 39628 10.1038/srep39628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Awad MM, Ellemor DM, Boyd RL, Emmins JJ, & Rood JI. (2001). Synergistic effects of alpha-toxin and perfringolysin O in Clostridium perfringens-mediated gas gangrene. Infection and Immunity, 69(12), 7904–10. 10.1128/IAI.69.12.7904-7910.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ayna G, Krysko DV, Kaczmarek A, Petrovski G, Vandenabeele P, & Fésüs L. (2012). ATP Release from Dying Autophagic Cells and Their Phagocytosis Are Crucial for Inflammasome Activation in Macrophages. PLoS ONE, 7(6), e40069 10.1371/journal.pone.0040069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bakardjiev AI, Stacy BA, & Portnoy DA. (2005). Growth of Listeria monocytogenes in the Guinea Pig Placenta and Role of Cell‐to‐Cell Spread in Fetal Infection. The Journal of Infectious Diseases, 191(11), 1889–1897. 10.1086/430090 [DOI] [PubMed] [Google Scholar]
  6. Bandara M, Skehel JM, Kadioglu A, Collinson I, Nobbs AH, Blocker AJ, & Jenkinson HF. (2017). The accessory Sec system (SecY2A2) in Streptococcus pneumoniae is involved in export of pneumolysin toxin, adhesion and biofilm formation. Microbes and Infection, 19(7–8), 402–412. 10.1016/J.MICINF.2017.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Benton KA, Everson MP, & Briles DE. (1995). A pneumolysin-negative mutant of Streptococcus pneumoniae causes chronic bacteremia rather than acute sepsis in mice. Infection and Immunity, 63(2), 448–55. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7822009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Berry AM, Yother J, Briles DE, Hansman D, & Paton JC. (1989). Reduced virulence of a defined pneumolysin-negative mutant of Streptococcus pneumoniae. Infection and Immunity, 57(7), 2037–42. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/2731982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bi L, Pian Y, Chen S, Ren Z, Liu P, Lv Q, … Jiang Y. (2015). Toll-like receptor 4 confers inflammatory response to Suilysin. Frontiers in Microbiology, 6, 644 10.3389/fmicb.2015.00644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bischofberger M, Iacovache I, & van der Goot FG. (2012). Pathogenic pore-forming proteins: function and host response. Cell Host & Microbe, 12(3), 266–75. 10.1016/j.chom.2012.08.005 [DOI] [PubMed] [Google Scholar]
  11. Bishop BL, Lodolce JP, Kolodziej LE, Boone DL, & Tang WJ. (2010). The role of anthrolysin O in gut epithelial barrier disruption during Bacillus anthracis infection. Biochemical and Biophysical Research Communications, 394(2), 254–259. 10.1016/J.BBRC.2010.02.091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Brubaker SW, Bonham KS, Zanoni I, & Kagan JC. (2015). Innate Immune Pattern Recognition: A Cell Biological Perspective. Annual Review of Immunology, 33(1), 257–290. 10.1146/annurev-immunol-032414-112240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bryant AE, Bayer CR, Chen RYZ, Guth PH, Wallace RJ, & Stevens DL. (2005). Vascular Dysfunction and Ischemic Destruction of Tissue in Streptococcus pyogenes Infection: The Role of Streptolysin O–Induced Platelet/Neutrophil Complexes. The Journal of Infectious Diseases, 192(6), 1014–1022. 10.1086/432729 [DOI] [PubMed] [Google Scholar]
  14. Bryant AE, Bergstrom R, Zimmerman GA, Salyer JL, Hill HR, Tweten RK, … Stevens DL. (1993). Clostridium perfringens invasiveness is enhanced by effects of theta toxin upon PMNL structure and function: The role of leukocytotoxicity and expression of CD11/CD18 adherence glycoprotein. FEMS Immunology and Medical Microbiology, 7(4), 321–336. Retrieved from https://www.sciencedirect.com/science/article/pii/0928824493900537 [DOI] [PubMed] [Google Scholar]
  15. Bryant AE, & Stevens DL. (1996). Phospholipase C and Perfringolysin O from Clostridium perfringens Upregulate Endothelial Cell-Leukocyte Adherence Molecule 1 and Intercellular Leukocyte Adherence Molecule 1 Expression and Induce Interleukin-8 Synthesis in Cultured Human Umbilical Vein Endothelial Cells. INFECTION AND IMMUNITY (Vol. 64). Retrieved from http://iai.asm.org/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cabezas S, Ho S, Ros U, Lanio ME, Alvarez C, & van der Goot FG. (2017). Damage of eukaryotic cells by the pore-forming toxin sticholysin II: Consequences of the potassium efflux. Biochimica et Biophysica Acta (BBA) - Biomembranes, 1859(5), 982–992. 10.1016/J.BBAMEM.2017.02.001 [DOI] [PubMed] [Google Scholar]
  17. Cajnko MM, Mikelj M, Turk T, Podobnik M, & Anderluh G. (2014). Membrane Interactions and Cellular Effects of MACPF/CDC Proteins (pp. 119–144). Springer, Dordrecht: 10.1007/978-94-017-8881-6_7 [DOI] [PubMed] [Google Scholar]
  18. Cassidy SKB, & O’Riordan MXD. (2013). More Than a Pore: The Cellular Response to Cholesterol-Dependent Cytolysins. Toxins, 5, 618–636. 10.3390/toxins5040618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chandrasekaran S, & Caparon MG. (2016). The NADase-Negative Variant of the Streptococcus pyogenes Toxin NAD+ Glycohydrolase Induces JNK1-Mediated Programmed Cellular Necrosis. MBio, 7(1), e02215–15. 10.1128/mBio.02215-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen C, Nguyen BN, Mitchell G, Margolis SR, Ma D, & Portnoy DA. (2018). The Listeriolysin O PEST-like Sequence Co-opts AP-2-Mediated Endocytosis to Prevent Plasma Membrane Damage during Listeria Infection. Cell Host and Microbe, 23(6). 10.1016/j.chom.2018.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Christie MP, Johnstone BA, Tweten RK, Parker MW, & Morton CJ. (2018). Cholesterol-dependent cytolysins: from water-soluble state to membrane pore. Biophysical Reviews, 1–12. 10.1007/s12551-018-0448-x [DOI] [PMC free article] [PubMed]
  22. Chu J, Thomas LM, Watkins SC, Franchi L, Núñez G, & Salter RD. (2009). Cholesterol-dependent cytolysins induce rapid release of mature IL-1β from murine macrophages in a NLRP3 inflammasome and cathepsin B-dependent manner. Journal of Leukocyte Biology, 86(5), 1227–1238. 10.1189/jlb.0309164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Czuczman MA, Fattouh R, van Rijn JM, Canadien V, Osborne S, Muise AM, … Brumell JH. (2014). Listeria monocytogenes exploits efferocytosis to promote cell-to-cell spread. Nature, 509(7499), 230–234. 10.1038/nature13168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Dancz CE, Haraga A, Portnoy DA, & Higgins DE. (2002). Inducible control of virulence gene expression in Listeria monocytogenes: temporal requirement of listeriolysin O during intracellular infection. Journal of Bacteriology, 184(21), 5935–45. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/12374827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Decatur AL, & Portnoy DA. (2000). A PEST-like sequence in listeriolysin O essential for Listeria monocytogenes pathogenicity. Science (New York, N.Y.), 290(5493), 992–5. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/11062133 [DOI] [PubMed] [Google Scholar]
  26. Del Mar García-Suárez M, Cima-Cabal MD, Flórez N, García P, Cernuda-Cernuda R, Astudillo A, … Méndez FJ. (2004). Protection against pneumococcal pneumonia in mice by monoclonal antibodies to pneumolysin. Infection and Immunity. 10.1128/IAI.72.8.4534-4540.2004 [DOI] [PMC free article] [PubMed]
  27. Demarque MD, Nacerddine K, Neyret–Kahn H, Andrieux A, Danenberg E, Jouvion G, … Dejean A. (2011). Sumoylation by Ubc9 Regulates the Stem Cell Compartment and Structure and Function of the Intestinal Epithelium in Mice. Gastroenterology, 140(1), 286–296. 10.1053/j.gastro.2010.10.002 [DOI] [PubMed] [Google Scholar]
  28. Dortet L, Lombardi C, Cretin F, Dessen A, & Filloux A. (2018). Pore-forming activity of the Pseudomonas aeruginosa type III secretion system translocon alters the host epigenome. Nature Microbiology, 3(3), 378–386. 10.1038/s41564-018-0109-7 [DOI] [PubMed] [Google Scholar]
  29. Dowd KJ, & Tweten RK. (2012). The Cholesterol-Dependent Cytolysin Signature Motif: A Critical Element in the Allosteric Pathway that Couples Membrane Binding to Pore Assembly. PLoS Pathogens, 8(7), e1002787 10.1371/journal.ppat.1002787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Drevets DA (1997). Listeria monocytogenes infection of cultured endothelial cells stimulates neutrophil adhesion and adhesion molecule expression. Journal of Immunology (Baltimore, Md. : 1950), 158(11), 5305–13. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/9164950 [PubMed] [Google Scholar]
  31. Dunne A, Ross PJ, Pospisilova E, Masin J, Meaney A, Sutton CE, … Mills KHG. (2010). Inflammasome activation by adenylate cyclase toxin directs Th17 responses and protection against Bordetella pertussis. Journal of Immunology (Baltimore, Md. : 1950), 185(3), 1711–9. 10.4049/jimmunol.1000105 [DOI] [PubMed] [Google Scholar]
  32. Edelson BT, Cossart P, & Unanue ER. (1999). Cutting edge: paradigm revisited: antibody provides resistance to Listeria infection. Journal of Immunology (Baltimore, Md. : 1950), 163(8), 4087–90. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/10510340 [PubMed] [Google Scholar]
  33. Edelson BT, & Unanue ER. (2001). Intracellular antibody neutralizes Listeria growth. Immunity, 14(5), 503–512. 10.1016/S1074-7613(01)00139-X [DOI] [PubMed] [Google Scholar]
  34. Edelson BT, & Unanue ER. (2002). MyD88-dependent but Toll-like receptor 2-independent innate immunity to Listeria: no role for either in macrophage listericidal activity. Journal of Immunology (Baltimore, Md. : 1950), 169(7), 3869–75. 10.4049/JIMMUNOL.169.7.3869 [DOI] [PubMed] [Google Scholar]
  35. Ellemor DM, Baird RN, Awad MM, Boyd RL, Rood JI, & Emmins JJ. (1999). Use of genetically manipulated strains of Clostridium perfringens reveals that both alpha-toxin and theta-toxin are required for vascular leukostasis to occur in experimental gas gangrene. Infection and Immunity, 67(9), 4902–7. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/10456947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Fang R, Tsuchiya K, Kawamura I, Shen Y, Hara H, Sakai S, … Mitsuyama M. (2011). Critical Roles of ASC Inflammasomes in Caspase-1 Activation and Host Innate Resistance to Streptococcus pneumoniae Infection. The Journal of Immunology, 187(9), 4890–4899. 10.4049/jimmunol.1100381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Farrand AJ, LaChapelle S, Hotze EM, Johnson AE, & Tweten RK. (2010). Only two amino acids are essential for cytolytic toxin recognition of cholesterol at the membrane surface. Proceedings of the National Academy of Sciences, 107(9), 4341–4346. 10.1073/pnas.0911581107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Fritah S, Lhocine N, Golebiowski F, Mounier J, Andrieux A, Jouvion G, … Dejean A. (2014). Sumoylation controls host anti-bacterial response to the gut invasive pathogen Shigella flexneri. EMBO Reports, 15(9), 965–72. 10.15252/embr.201338386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gaillard JL, Berche P, & Sansonetti P. (1986). Transposon mutagenesis as a tool to study the role of hemolysin in the virulence of Listeria monocytogenes. Infection and Immunity, 52(1), 50–5. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/3007363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Gelber SE, Aguilar JL, Lewis KLT, & Ratner AJ. (2008). Functional and phylogenetic characterization of Vaginolysin, the human-specific cytolysin from Gardnerella vaginalis. Journal of Bacteriology, 190(11), 3896–903. 10.1128/JB.01965-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Geoffroy C, Gaillard JL, Alouf JE, & Berche P. (1987). Purification, characterization, and toxicity of the sulfhydryl-activated hemolysin listeriolysin O from Listeria monocytogenes. Infection and Immunity, 55(7), 1641–6. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/3110067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Gilbert RJC (2010). Cholesterol-Dependent Cytolysins. Advances in Experimental Medicine and Biology. 10.1007/978-1-4419-6327-7_5 [DOI] [PubMed]
  43. Gill G (2005). Something about SUMO inhibits transcription. Current Opinion in Genetics & Development, 15(5), 536–541. 10.1016/J.GDE.2005.07.004 [DOI] [PubMed] [Google Scholar]
  44. Glomski IJ, Gedde MM, Tsang AW, Swanson J. a., & Portnoy D. a. (2002). The Listeria monocytogenes hemolysin has an acidic pH optimum to compartmentalize activity and prevent damage to infected host cells. Journal of Cell Biology, 156(6), 1029–1038. 10.1083/jcb.200201081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Gonzalez MR, Bischofberger M, Frêche B, Ho S, Parton RG, & van der Goot FG. (2011). Pore-forming toxins induce multiple cellular responses promoting survival. Cellular Microbiology, 13(7), 1026–43. 10.1111/j.1462-5822.2011.01600.x [DOI] [PubMed] [Google Scholar]
  46. Gonzalez MR, Bischofberger M, Pernot L, van der Goot FG, & Frêche B. (2008). Bacterial pore-forming toxins: The (w)hole story? Cellular and Molecular Life Sciences, 65(3), 493–507. 10.1007/s00018-007-7434-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gurcel L, Abrami L, Girardin S, Tschopp J, & van der Goot FG. (2006). Caspase-1 activation of lipid metabolic pathways in response to bacterial pore-forming toxins promotes cell survival. Cell, 126(6), 1135–45. 10.1016/j.cell.2006.07.033 [DOI] [PubMed] [Google Scholar]
  48. Hamon MA, Batsché E, Régnault B, Tham TN, Seveau S, Muchardt C, & Cossart P. (2007). Histone modifications induced by a family of bacterial toxins. Proceedings of the National Academy of Sciences of the United States of America, 104(33), 13467–72. 10.1073/pnas.0702729104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Hamon MA, & Cossart P. (2011). K+ efflux is required for histone H3 dephosphorylation by Listeria monocytogenes listeriolysin O and other pore-forming toxins. Infection and Immunity, 79(7), 2839–46. 10.1128/IAI.01243-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Hamon MA, Ribet D, Stavru F, & Cossart P. (2012). Listeriolysin O: The Swiss army knife of Listeria. Trends in Microbiology, 20(8), 360–368. 10.1016/j.tim.2012.04.006 [DOI] [PubMed] [Google Scholar]
  51. Harder J, Franchi L, Muñoz-Planillo R, Park J-H, Reimer T, & Núñez G. (2009). Activation of the Nlrp3 inflammasome by Streptococcus pyogenes requires streptolysin O and NF-kappa B activation but proceeds independently of TLR signaling and P2X7 receptor. Journal of Immunology (Baltimore, Md. : 1950), 183(9), 5823–9. 10.4049/jimmunol.0900444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hardy J, Francis KP, DeBoer M, Chu P, Gibbs K, & Contag CH. (2004). Extracellular replication of Listeria monocytogenes in the murine gall bladder. Science (New York, N.Y.), 303(5659), 851–3. 10.1126/science.1092712 [DOI] [PubMed] [Google Scholar]
  53. Hassane M, Demon D, Soulard D, Fontaine J, Keller LE, Patin EC, … Paget C. (2017). Neutrophilic NLRP3 inflammasome-dependent IL-1β secretion regulates the γδT17 cell response in respiratory bacterial infections. Mucosal Immunology, 10(4), 1056–1068. 10.1038/mi.2016.113 [DOI] [PubMed] [Google Scholar]
  54. He Y, Zeng MY, Yang D, Motro B, & Núñez G. (2016). NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature, 530(7590), 354–357. 10.1038/nature16959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Heffernan BJ, Thomason B, Herring-Palmer A, & Hanna P. (2007). Bacillus anthracis anthrolysin O and three phospholipases C are functionally redundant in a murine model of inhalation anthrax. FEMS Microbiology Letters, 271(1), 98–105. 10.1111/j.1574-6968.2007.00713.x [DOI] [PubMed] [Google Scholar]
  56. Holland AJ, Fachinetti D, Han JS, & Cleveland DW. (2012). Inducible, reversible system for the rapid and complete degradation of proteins in mammalian cells. Proceedings of the National Academy of Sciences, 109(49), E3350–E3357. 10.1073/pnas.1216880109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hotze EM, Le HM, Sieber JR, Bruxvoort C, McInerney MJ, & Tweten RK. (2013). Identification and characterization of the first cholesterol-dependent cytolysins from Gram-negative bacteria. Infection and Immunity, 81(1), 216–25. 10.1128/IAI.00927-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Idzko M, Ferrari D, & Eltzschig HK. (2014). Nucleotide signalling during inflammation. Nature, 509(7500), 310–317. 10.1038/nature13085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Jones GS, Bussell KM, Myers-Morales T, Fieldhouse AM, Bou Ghanem EN, & D’Orazio SEF. (2015). Intracellular Listeria monocytogenes Comprises a Minimal but Vital Fraction of the Intestinal Burden following Foodborne Infection. Infection and Immunity, 83(8), 3146–3156. 10.1128/IAI.00503-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Jones S, & Portnoy DA. (1994). Characterization of Listeria monocytogenes pathogenesis in a strain expressing perfringolysin O in place of listeriolysin O. Infection and Immunity, 62(12), 5608–13. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7960143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Jounai N, Kobiyama K, Takeshita F, & Ishii KJ. (2012). Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination. Frontiers in Cellular and Infection Microbiology, 2, 168 10.3389/fcimb.2012.00168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kadioglu A, Gingles NA, Grattan K, Kerr A, Mitchell TJ, & Andrew PW. (2000). Host cellular immune response to pneumococcal lung infection in mice. Infection and Immunity, 68(2), 492–501. 10.1128/IAI.68.2.492-501.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kao C-Y, Los FCO, Huffman DL, Wachi S, Kloft N, Husmann M, … Aroian RV. (2011). Global Functional Analyses of Cellular Responses to Pore-Forming Toxins. PLoS Pathogens, 7(3), e1001314 10.1371/journal.ppat.1001314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Karmakar M, Katsnelson M, Malak HA, Greene NG, Howell SJ, Hise AG, … Pearlman E. (2015). Neutrophil IL-1β processing induced by pneumolysin is mediated by the NLRP3/ASC inflammasome and caspase-1 activation and is dependent on K+ efflux. Journal of Immunology (Baltimore, Md. : 1950), 194(4), 1763–75. 10.4049/jimmunol.1401624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kathariou S, Metz P, Hof H, & Goebel W. (1987). Tn916-induced mutations in the hemolysin determinant affecting virulence of Listeria monocytogenes. Journal of Bacteriology, 169(3), 1291–7. 10.1128/JB.169.3.1291-1297.1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kayal S, Lilienbaum A, Poyart C, Memet S, Israel A, & Berche P. (1999). Listeriolysin O-dependent activation of endothelial cells during infection with Listeria monocytogenes: activation of NF-kappaB and upregulation of adhesion molecules and chemokines. Molecular Microbiology, 31(6), 1709–1722. 10.1046/j.1365-2958.1999.01305.x [DOI] [PubMed] [Google Scholar]
  67. Keyel P, Roth R, Yokoyama W, Heuser J, Salter R, Keyel PA, … Salter RD. (2013). Reduction of Streptolysin O (SLO) Pore-Forming Activity Enhances Inflammasome Activation. Toxins, 5(6), 1105–1118. 10.3390/toxins5061105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Köster S, van Pee K, Hudel M, Leustik M, Rhinow D, Kühlbrandt W, … Yildiz Ö. (2014). Crystal structure of listeriolysin O reveals molecular details of oligomerization and pore formation. Nature Communications, 5, 3690 10.1038/ncomms4690 [DOI] [PubMed] [Google Scholar]
  69. Krüll M, Nöst R, Hippenstiel S, Domann E, Chakraborty T, & Suttorp N. (1997). Listeria monocytogenes potently induces up-regulation of endothelial adhesion molecules and neutrophil adhesion to cultured human endothelial cells. Journal of Immunology (Baltimore, Md. : 1950), 159(4), 1970–6. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/9257863 [PubMed] [Google Scholar]
  70. Land WG (2015). The Role of Damage-Associated Molecular Patterns (DAMPs) in Human Diseases: Part II: DAMPs as diagnostics, prognostics and therapeutics in clinical medicine. Sultan Qaboos University Medical Journal, 15(2), e157–70. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/26052447 [PMC free article] [PubMed] [Google Scholar]
  71. Lawrence SL, Gorman MA, Feil SC, Mulhern TD, Kuiper MJ, Ratner AJ, … Parker MW. (2016). Structural Basis for Receptor Recognition by the Human CD59-Responsive Cholesterol-Dependent Cytolysins. Structure, 24(9), 1488–1498. 10.1016/J.STR.2016.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Le Monnier A, Autret N, Join-Lambert OF, Jaubert F, Charbit A, Berche P, & Kayal S. (2007). ActA is required for crossing of the fetoplacental barrier by Listeria monocytogenes. Infection and Immunity, 75(2), 950–7. 10.1128/IAI.01570-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lecuit M (2007). Human listeriosis and animal models. Microbes and Infection, 9(10), 1216–1225. 10.1016/J.MICINF.2007.05.009 [DOI] [PubMed] [Google Scholar]
  74. Lemon JK, & Weiser JN. (2015). Degradation Products of the Extracellular Pathogen Streptococcus pneumoniae Access the Cytosol via Its Pore-Forming Toxin. MBio, 6(1). 10.1128/mBio.02110-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Lety MA, Frehel C, Dubail I, Beretti JL, Kayal S, Berche P, & Charbit A. (2001). Identification of a PEST-like motif in listeriolysin O required for phagosomal escape and for virulence in Listeria monocytogenes. Molecular Microbiology, 39(5), 1124–39. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/11251831 [DOI] [PubMed] [Google Scholar]
  76. Leung C, Dudkina NV, Lukoyanova N, Hodel AW, Farabella I, Pandurangan AP, … Hoogenboom BW. (2014). Stepwise visualization of membrane pore formation by suilysin, a bacterial cholesterol-dependent cytolysin. ELife, 3(domain 2), e04247 10.7554/eLife.04247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Levraud J-P, Disson O, Kissa K, Bonne I, Cossart P, Herbomel P, & Lecuit M. (2009). Real-time observation of listeria monocytogenes-phagocyte interactions in living zebrafish larvae. Infection and Immunity, 77(9), 3651–60. 10.1128/IAI.00408-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Li J, Lam WW, Lai T, & Au SW. (2017). Degradation of nuclear Ubc9 induced by listeriolysin O is dependent on K + efflux. Biochemical and Biophysical Research Communications, 493(2), 1115–1121. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/28911869 [DOI] [PubMed] [Google Scholar]
  79. Li W, Chang Y, Liang S, Zhong Z, Li X, Wen J, … Zhong F. (2016). NLRP3 inflammasome activation contributes to Listeria monocytogenes-induced animal pregnancy failure. BMC Veterinary Research, 12(1), 36 10.1186/s12917-016-0655-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lunardi A, Gaboli M, Giorgio M, Rivi R, Bygrave A, Antoniou M, … Pandolfi PP. (2011). A Role for PML in Innate Immunity. Genes & Cancer, 2(1), 10–9. 10.1177/1947601911402682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Madden JC, Ruiz N, & Caparon M. (2001). Cytolysin-mediated translocation (CMT): a functional equivalent of type III secretion in gram-positive bacteria. Cell, 104(1), 143–52. 10.1016/S0092-8674(01)00198-2 [DOI] [PubMed] [Google Scholar]
  82. Malley R, Henneke P, Morse SC, Cieslewicz MJ, Lipsitch M, Thompson CM, … Golenbock DT. (2003). Recognition of pneumolysin by Toll-like receptor 4 confers resistance to pneumococcal infection. Proceedings of the National Academy of Sciences of the United States of America, 100(4), 1966–71. 10.1073/pnas.0435928100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, … Dixit VM. (2006). Cryopyrin activates the inflammasome in response to toxins and ATP. Nature, 440(7081), 228–232. 10.1038/nature04515 [DOI] [PubMed] [Google Scholar]
  84. McCoy AJ, Koizumi Y, Higa N, & Suzuki T. (2010). Differential regulation of caspase-1 activation via NLRP3/NLRC4 inflammasomes mediated by aerolysin and type III secretion system during Aeromonas veronii infection. Journal of Immunology (Baltimore, Md. : 1950), 185(11), 7077–84. 10.4049/jimmunol.1002165 [DOI] [PubMed] [Google Scholar]
  85. McKay DB, & Lu CY. (1991). Listeriolysin as a virulence factor in Listeria monocytogenes infection of neonatal mice and murine decidual tissue. Infection and Immunity, 59(11), 4286–90. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/1937789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Meixenberger K, Pache F, Eitel J, Schmeck B, Hippenstiel S, Slevogt H, … Opitz B. (2010). Listeria monocytogenes-Infected Human Peripheral Blood Mononuclear Cells Produce IL-1, Depending on Listeriolysin O and NLRP3. The Journal of Immunology, 184(2), 922–930. 10.4049/jimmunol.0901346 [DOI] [PubMed] [Google Scholar]
  87. Mitchell TJ, & Dalziel CE. (2014). The Biology of Pneumolysin (pp. 145–160). Springer, Dordrecht: 10.1007/978-94-017-8881-6_8 [DOI] [PubMed] [Google Scholar]
  88. Morgan PJ, Andrew PW, & Mitchell TJ. (1996). Thiol-activated cytolysins. Reviews in Medical Microbiology.
  89. Mulvihill E, Van Pee K, Mari S. a., Muller DJ, & Yildiz O. (2015). Directly Observing the Lipid-Dependent Self-Assembly and Pore-Forming Mechanism of the Cytolytic Toxin Listeriolysin O. Nano Letters, 15(10), 6965–6973. 10.1021/acs.nanolett.5b02963 [DOI] [PubMed] [Google Scholar]
  90. Musher DM, Phan HM, & Baughn RE. (2001). Protection against Bacteremic Pneumococcal Infection by Antibody to Pneumolysin. The Journal of Infectious Diseases. 10.1086/318833 [DOI] [PubMed]
  91. Nakouzi A, Rivera J, Rest RF, & Casadevall A. (2008). Passive administration of monoclonal antibodies to Anthrolysin O prolong survival in mice lethally infected with Bacillus anthracis. BMC Microbiology, 8(1), 159 10.1186/1471-2180-8-159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Nel JG, Durandt C, Theron AJ, Tintinger GR, Pool R, Richards GA, … Anderson R. (2017). Pneumolysin mediates heterotypic aggregation of neutrophils and platelets in vitro. The Journal of Infection, 74(6), 599–608. 10.1016/j.jinf.2017.02.010 [DOI] [PubMed] [Google Scholar]
  93. Nishimura K, Fukagawa T, Takisawa H, Kakimoto T, & Kanemaki M. (2009). An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nature Methods, 6(12), 917–922. 10.1038/nmeth.1401 [DOI] [PubMed] [Google Scholar]
  94. O’Brien DK, & Melville SB. (2004). Effects of Clostridium perfringens alpha-toxin (PLC) and perfringolysin O (PFO) on cytotoxicity to macrophages, on escape from the phagosomes of macrophages, and on persistence of C. perfringens in host tissues. Infection and Immunity, 72(9), 5204–15. 10.1128/IAI.72.9.5204-5215.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Osborne SE, & Brumell JH. (2017). Listeriolysin O: from bazooka to Swiss army knife. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 372(1726), 20160222 10.1098/rstb.2016.0222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Owen JL, Yang T, & Mohamadzadeh M. (2015). New insights into gastrointestinal anthrax infection. Trends in Molecular Medicine, 21(3), 154–63. 10.1016/j.molmed.2014.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Palmer M, Harris R, Freytag C, Kehoe M, Tranum-Jensen J, & Bhakdi S. (1998). Assembly mechanism of the oligomeric streptolysin O pore: the early membrane lesion is lined by a free edge of the lipid membrane and is extended gradually during oligomerization. The EMBO Journal, 17(6), 1598–1605. 10.1093/emboj/17.6.1598 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Park JM, Ng VH, Maeda S, Rest RF, & Karin M. (2004). Anthrolysin O and other gram-positive cytolysins are toll-like receptor 4 agonists. The Journal of Experimental Medicine, 200(12), 1647–55. 10.1084/jem.20041215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Persson J, & Vance RE. (2007). Genetics-squared: combining host and pathogen genetics in the analysis of innate immunity and bacterial virulence. Immunogenetics, 59(10), 761–778. 10.1007/s00251-007-0248-0 [DOI] [PubMed] [Google Scholar]
  100. Pétrilli V, Papin S, Dostert C, Mayor A, Martinon F, & Tschopp J. (2007). Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death & Differentiation, 14(9), 1583–1589. 10.1038/sj.cdd.4402195 [DOI] [PubMed] [Google Scholar]
  101. Portman JL, Huang Q, Reniere ML, Iavarone AT, & Portnoy DA. (2017). Activity of the Pore-Forming Virulence Factor Listeriolysin O Is Reversibly Inhibited by Naturally Occurring S-Glutathionylation. Infection and Immunity, 85(4), e00959–16. 10.1128/IAI.00959-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Portnoy DA, Jacks PS, & Hinrichs DJ. (1988). Role of hemolysin for the intracellular growth of Listeria monocytogenes. The Journal of Experimental Medicine, 167(4), 1459–71. 10.1084/JEM.167.4.1459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Portnoy DA, Tweten RK, Kehoe M, & Bielecki J. (1992). Capacity of listeriolysin O, streptolysin O, and perfringolysin O to mediate growth of Bacillus subtilis within mammalian cells. Infection and Immunity, 60(7), 2710–7. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/1612739 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Price KE, Greene NG, & Camilli A. (2012). Export Requirements of Pneumolysin in Streptococcus pneumoniae. Journal of Bacteriology, 194(14), 3651–3660. 10.1128/JB.00114-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Rafii F, Park M, Bryant AE, Johnson SJ, & Wagner RD. (2008). Enhanced production of phospholipase C and perfringolysin O (alpha and theta toxins) in a gatifloxacin-resistant strain of Clostridium perfringens. Antimicrobial Agents and Chemotherapy, 52(3), 895–900. 10.1128/AAC.01316-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Ratner AJ, Hippe KR, Aguilar JL, Bender MH, Nelson AL, & Weiser JN. (2006). Epithelial cells are sensitive detectors of bacterial pore-forming toxins. The Journal of Biological Chemistry, 281(18), 12994–8. 10.1074/jbc.M511431200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Rayner CF, Jackson AD, Rutman A, Dewar A, Mitchell TJ, Andrew PW, … Wilson R. (1995). Interaction of pneumolysin-sufficient and -deficient isogenic variants of Streptococcus pneumoniae with human respiratory mucosa. Infection and Immunity, 63(2), 442–7. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7822008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Rechsteiner M, & Rogers SW. (1996). PEST sequences and regulation by proteolysis. Trends in Biochemical Sciences, 21(7), 267–271. 10.1016/S0968-0004(96)10031-1 [DOI] [PubMed] [Google Scholar]
  109. Ribet D, Hamon M, Gouin E, Nahori M-A, Impens F, Neyret-Kahn H, … Cossart P. (2010). Listeria monocytogenes impairs SUMOylation for efficient infection. Nature, 464(April), 1192–1195. 10.1038/nature08963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Ribet D, Lallemand-Breitenbach V, Ferhi O, Nahori M-A, Varet H, de Thé H, & Cossart P. (2017). Promyelocytic Leukemia Protein (PML) Controls Listeria monocytogenes Infection. MBio, 8(1), e02179–16. 10.1128/mBio.02179-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Rogers S, Wells R, & Rechsteiner M. (1986). Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis. Science, 234(4774), 364–368. 10.1126/science.2876518 [DOI] [PubMed] [Google Scholar]
  112. Roll JT, & Czuprynski CJ. (1990). Hemolysin is required for extraintestinal dissemination of Listeria monocytogenes in intragastrically inoculated mice. Infection and Immunity, 58(9), 3147–50. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/2117581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Sauer J-D, Pereyre S, Archer KA, Burke TP, Hanson B, Lauer P, & Portnoy DA. (2011). Listeria monocytogenes engineered to activate the Nlrc4 inflammasome are severely attenuated and are poor inducers of protective immunity. Proceedings of the National Academy of Sciences, 108(30), 12419–12424. 10.1073/pnas.1019041108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Sauer JD, Witte CE, Zemansky J, Hanson B, Lauer P, & Portnoy DA. (2010). Listeria monocytogenes triggers AIM2-mediated pyroptosis upon infrequent bacteriolysis in the macrophage cytosol. Cell Host and Microbe, 7(5), 412–419. 10.1016/j.chom.2010.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Schaefer L (2014). Complexity of danger: the diverse nature of damage-associated molecular patterns. The Journal of Biological Chemistry, 289(51), 35237–45. 10.1074/jbc.R114.619304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Schmeck B, Beermann W, van Laak V, Zahlten J, Opitz B, Witzenrath M, … Hippenstiel S. (2005). Intracellular bacteria differentially regulated endothelial cytokine release by MAPK-dependent histone modification. Journal of Immunology (Baltimore, Md. : 1950), 175(5), 2843–50. 10.4049/JIMMUNOL.175.5.2843 [DOI] [PubMed] [Google Scholar]
  117. Schnupf P, Hofmann J, Norseen J, Glomski IJ, Schwartzstein H, & Decatur AL. (2006). Regulated translation of listeriolysin O controls virulence of Listeria monocytogenes. Molecular Microbiology, 61(4), 999–1012. 10.1111/j.1365-2958.2006.05286.x [DOI] [PubMed] [Google Scholar]
  118. Schnupf P, & Portnoy D. a. (2007). Listeriolysin O: a phagosome-specific lysin. Microbes and Infection, 9(10), 1176–1187. 10.1016/j.micinf.2007.05.005 [DOI] [PubMed] [Google Scholar]
  119. Schnupf P, Portnoy D. a., & Decatur AL. (2006). Phosphorylation, ubiquitination and degradation of listeriolysin O in mammalian cells: Role of the PEST-like sequence. Cellular Microbiology, 8(2), 353–364. 10.1111/j.1462-5822.2005.00631.x [DOI] [PubMed] [Google Scholar]
  120. Schnupf P, Zhou J, Varshavsky A, & Portnoy DA. (2007). Listeriolysin O secreted by Listeria monocytogenes into the host cell cytosol is degraded by the N-end rule pathway. Infection and Immunity, 75(11), 5135–47. 10.1128/IAI.00164-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Schuerch DW, Wilson-Kubalek EM, & Tweten RK. (2005). Molecular basis of listeriolysin O pH dependence. Proceedings of the National Academy of Sciences of the United States of America, 102(35), 12537–42. 10.1073/pnas.0500558102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Seveau S. (2014). Multifaceted Activity of Listeriolysin O, the Cholesterol-Dependent Cytolysin of Listeria monocytogenes (pp. 161–195). Springer, Dordrecht: 10.1007/978-94-017-8881-6_9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Shannon JG, Ross CL, Koehler TM, & Rest RF. (2003). Characterization of anthrolysin O, the Bacillus anthracis cholesterol-dependent cytolysin. Infection and Immunity, 71(6), 3183–9. 10.1128/IAI.71.6.3183-3189.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Shen A, & Higgins DE. (2005). The 5’ untranslated region-mediated enhancement of intracellular listeriolysin O production is required for Listeria monocytogenes pathogenicity. Molecular Microbiology, 57(5), 1460–1473. 10.1111/j.1365-2958.2005.04780.x [DOI] [PubMed] [Google Scholar]
  125. Shewell LK, Harvey RM, Higgins MA, Day CJ, Hartley-Tassell LE, Chen AY, … Jennings MP. (2014). The cholesterol-dependent cytolysins pneumolysin and streptolysin O require binding to red blood cell glycans for hemolytic activity. Proceedings of the National Academy of Sciences, 111(49), E5312–E5320. 10.1073/pnas.1412703111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Sibelius U, Chakraborty T, Krögel B, Wolf J, Rose F, Schmidt R, … Grimminger F. (1996). The listerial exotoxins listeriolysin and phosphatidylinositol-specific phospholipase C synergize to elicit endothelial cell phosphoinositide metabolism. Journal of Immunology (Baltimore, Md. : 1950), 157(9), 4055–60. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8892639 [PubMed] [Google Scholar]
  127. Singh R, Jamieson A, & Cresswell P. (2008). GILT is a critical host factor for Listeria monocytogenes infection. Nature, 455(7217), 1244–1247. 10.1038/nature07344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Smyth CJ, & Duncan JL. (1978). Thiol-activated (oxygen-labile) cytolysins In Jeljaszewicz J & Wadstrom T. (Eds.), Bacterial toxins and cell membranes (pp. 129–183). New York, NY: Academic Press. [Google Scholar]
  129. Soltani CE, Hotze EM, Johnson AE, & Tweten RK. (2007). Structural elements of the cholesterol-dependent cytolysins that are responsible for their cholesterol-sensitive membrane interactions (Vol. 104). Retrieved from www.pnas.orgcgidoi10.1073pnas.0708104105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Srivastava A, Henneke P, Visintin A, Morse SC, Martin V, Watkins C, … Malley R. (2005). The apoptotic response to pneumolysin is Toll-like receptor 4 dependent and protects against pneumococcal disease. Infection and Immunity, 73(10), 6479–87. 10.1128/IAI.73.10.6479-6487.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Stassen M, Müller C, Richter C, Neudörfl C, Hültner L, Bhakdi S, … Schmitt E. (2003). The streptococcal exotoxin streptolysin O activates mast cells to produce tumor necrosis factor alpha by p38 mitogen-activated protein kinase- and protein kinase C-dependent pathways. Infection and Immunity, 71(11), 6171–7. 10.1128/IAI.71.11.6171-6177.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Stavru F, Bouillaud F, Sartori A, Ricquier D, & Cossart P. (2011). Listeria monocytogenes transiently alters mitochondrial dynamics during infection. Proceedings of the National Academy of Sciences of the United States of America, 108(9), 3612–7. 10.1073/pnas.1100126108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Takeuchi D, Akeda Y, Nakayama T, Kerdsin A, Sano Y, Kanda T, … Oishi K. (2014). The Contribution of Suilysin to the Pathogenesis of Streptococcus suis Meningitis. The Journal of Infectious Diseases, 209(10), 1509–1519. 10.1093/infdis/jit661 [DOI] [PubMed] [Google Scholar]
  134. Tang P, Rosenshine I, Cossart P, & Finlay BB. (1996). Listeriolysin O activates mitogen-activated protein kinase in eucaryotic cells. Infection and Immunity, 64(6), 2359–61. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8675352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Thornton J, & McDaniel LS. (2005). THP-1 monocytes up-regulate intercellular adhesion molecule 1 in response to pneumolysin from Streptococcus pneumoniae. Infection and Immunity, 73(10), 6493–8. 10.1128/IAI.73.10.6493-6498.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Tilney LG, Portnoy D. (1989). Actin filaments and the growth, movement, and spread of the intracellular bacterial parasite, Listeria monocytogenes. The Journal of Cell Biology, 109(4), 1597–1608. 10.1083/jcb.109.4.1597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Titball RW (2005). Gas gangrene: an open and closed case. Microbiology, 151(9), 2821–2828. 10.1099/mic.0.28248-0 [DOI] [PubMed] [Google Scholar]
  138. Tonello F, & Zornetta I. (2012). Bacillus anthracis Factors for Phagosomal Escape. Toxins, 4(7), 536–553. 10.3390/toxins4070536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Tsuji NM, Tsutsui H, Seki E, Kuida K, Okamura H, Nakanishi K, & Flavell RA. (2004). Roles of caspase-1 in Listeria infection in mice. International Immunology, 16(2), 335–343. 10.1093/intimm/dxh041 [DOI] [PubMed] [Google Scholar]
  140. Tweten RK, Hotze EM, & Wade KR. (2015). The Unique Molecular Choreography of Giant Pore Formation by the Cholesterol-Dependent Cytolysins of Gram-Positive Bacteria. Annual Review of Microbiology, 69(1), 323–340. 10.1146/annurev-micro-091014-104233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Vénéreau E, Ceriotti C, & Bianchi ME. (2015). DAMPs from Cell Death to New Life. Frontiers in Immunology, 6, 422 10.3389/fimmu.2015.00422 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Verherstraeten S, Goossens E, Valgaeren B, Pardon B, Timbermont L, Haesebrouck F, … Van Immerseel F. (2015). Perfringolysin O: The Underrated Clostridium perfringens Toxin? Toxins, 7(5), 1702–1721. 10.3390/toxins7051702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Wagner S, Grin I, Malmsheimer S, Singh N, Torres-Vargas CE, & Westerhausen S. (2018). Bacterial type III secretion systems: A complex device for delivery of bacterial effector proteins into eukaryotic host cells. FEMS Microbiology Letters. 10.1093/femsle/fny201 [DOI] [PMC free article] [PubMed]
  144. Walev I, Reske K, Palmer M, Valeva A, & Bhakdi S. (1995). Potassium-inhibited processing of IL-1 beta in human monocytes. The EMBO Journal, 14(8), 1607–14. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7737113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Wang A, Ding X, Demarque M, Liu X, Pan D, Xin H, … Dong C. (2017). Ubc9 Is Required for Positive Selection and Late-Stage Maturation of Thymocytes. Journal of Immunology (Baltimore, Md. : 1950), 198(9), 3461–3470. 10.4049/jimmunol.1600980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Wei Z, Schnupf P, Poussin MA, Zenewicz LA, Shen H, & Goldfine H. (2005). Characterization of Listeria monocytogenes expressing anthrolysin O and phosphatidylinositol-specific phospholipase C from Bacillus anthracis. Infection and Immunity, 73(10), 6639–46. 10.1128/IAI.73.10.6639-6646.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Witzenrath M, Pache F, Lorenz D, Koppe U, Gutbier B, Tabeling C, … Opitz B. (2011). The NLRP3 inflammasome is differentially activated by pneumolysin variants and contributes to host defense in pneumococcal pneumonia. Journal of Immunology (Baltimore, Md. : 1950), 187(1), 434–40. 10.4049/jimmunol.1003143 [DOI] [PubMed] [Google Scholar]
  148. Zafar MA, Wang Y, Hamaguchi S, & Weiser JN. (2017). Host-to-Host Transmission of Streptococcus pneumoniae Is Driven by Its Inflammatory Toxin, Pneumolysin. Cell Host & Microbe, 21(1), 73–83. 10.1016/j.chom.2016.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Zhang S, Zheng Y, Chen S, Huang S, Liu K, Lv Q, … Yuan Y. (2016). Suilysin-induced Platelet-Neutrophil Complexes Formation is Triggered by Pore Formation-dependent Calcium Influx. Scientific Reports, 6(1), 36787 10.1038/srep36787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Zhu L, Olsen RJ, Lee JD, Porter AR, DeLeo FR, & Musser JM. (2017). Contribution of Secreted NADase and Streptolysin O to the Pathogenesis of Epidemic Serotype M1 Streptococcus pyogenes Infections. The American Journal of Pathology, 187(3), 605–613. 10.1016/J.AJPATH.2016.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES