ArticlePDF AvailableLiterature Review

The Cholesterol-Dependent Cytolysin Family of Gram-Positive Bacterial Toxins

Authors:

Abstract

The cholesterol-dependent cytolysins (CDCs) are a family of beta-barrel pore-forming toxins secreted by Gram-positive bacteria. These toxins are produced as water-soluble monomeric proteins that after binding to the target cell oligomerize on the membrane surface forming a ring-like pre-pore complex, and finally insert a large beta-barrel into the membrane (about 250 A in diameter). Formation of such a large transmembrane structure requires multiple and coordinated conformational changes. The presence of cholesterol in the target membrane is absolutely required for pore-formation, and therefore it was long thought that cholesterol was the cellular receptor for these toxins. However, not all the CDCs require cholesterol for binding. Intermedilysin, secreted by Streptoccocus intermedius only binds to membranes containing a protein receptor, but forms pores only if the membrane contains sufficient cholesterol. In contrast, perfringolysin O, secreted by Clostridium perfringens, only binds to membranes containing substantial amounts of cholesterol. The mechanisms by which cholesterol regulates the cytolytic activity of the CDCs are not understood at the molecular level. The C-terminus of perfringolysin O is involved in cholesterol recognition, and changes in the conformation of the loops located at the distal tip of this domain affect the toxin-membrane interactions. At the same time, the distribution of cholesterol in the membrane can modulate toxin binding. Recent studies support the concept that there is a dynamic interplay between the cholesterol-binding domain of the CDCs and the excess of cholesterol molecules in the target membrane.
1
Chapter 20 Submitted July 2009
The original publication is available at:
https://link.springer.com/chapter/10.1007%2F978-90-481-8622-8_20
The cholesterol-dependent cytolysins family of Gram-positive bacterial
toxins
Alejandro P. Heuck, Paul C. Moe, and Benjamin B. Johnson
Department of Biochemistry and Molecular Biology, University of Massachusetts,
Amherst, MA 01003, U.S.A. heuck@biochem.umass.edu
Abstract
The cholesterol-dependent cytolysins (CDC) are a family of β-barrel pore-
forming toxins secreted by Gram-positive bacteria. These toxins are produced as water-
soluble monomeric proteins that after binding to the target cell oligomerize on the
membrane surface forming a ring-like pre-pore complex, and finally insert a large
β-barrel into the membrane (about 250 Å in diameter). Formation of such a large
transmembrane structure requires multiple and coordinated conformational changes. The
presence of cholesterol in the target membrane is absolutely required for pore-formation,
and therefore it was long thought that cholesterol was the cellular receptor for these
toxins. However, not all the CDC require cholesterol for binding. Intermedilysin, secreted
by Streptoccocus intermedius only binds to membranes containing a protein receptor, but
forms pores only if the membrane contains sufficient cholesterol. In contrast,
perfringolysin O, secreted by Clostridium perfringens, only binds to membranes
containing substantial amounts of cholesterol. The mechanisms by which cholesterol
regulates the cytolytic activity of the CDC are not understood at the molecular level. The
2
C-terminus of perfringolysin O is involved in cholesterol recognition, and changes in the
conformation of the loops located at the distal tip of this domain affect the
toxin-membrane interactions. At the same time, the distribution of cholesterol in the
membrane can modulate toxin binding. Recent studies support the concept that there is a
dynamic interplay between the cholesterol-binding domain of the CDC and the excess of
cholesterol molecules in the target membrane.
Keywords
Cholesterol, membranes, pore-forming toxins, cholesterol-dependent cytolysins,
membrane structure, cholesterol activity, transmembrane beta-barrel, transmembrane
pore, fluorescence spectroscopy, perfringolysin, lipid cluster.
Abbreviations
CDC, cholesterol-dependent cytolysins; PFO, perfringolysin O; ILY, intermedilysin;
PLY, pneumolysin; SLO, streptolysin O; anthrolysin; ALO; TMH/s, transmembrane β-
hairpin/s; D4, domain 4; L1, L2, and L3, loop 1, loop 2 and loop 3.
1. INTRODUCTION
The cholesterol-dependent cytolysins (CDC) are a growing group of β-barrel
pore-forming toxins secreted by Gram-positive bacteria (Farrand et al., 2008, Gelber et
al., 2008, Heuck et al., 2001, Jefferies et al., 2007, Mosser and Rest, 2006), and the first
members were discovered more than a century ago (see Alouf et al., 2006 for a historical
background on the CDCs). To date, there are complete amino acid sequences for 28
species distributed among the phyla of Firmicutes (genera of Bacillus, Paenibacillus,
Lysinibacillus, Listeria, Streptococcus, and Clostridium), and of Actinobacteria (genera
3
of Arcanobacterium and Gardenella) (Table 1). Most of the CDC have a cleavable signal
sequence and are therefore secreted to the extracellular medium via the general secretion
system (Sec, Harwood and Cranenburgh, 2008). A few exceptions are species of the
genus Streptoccocus (S. pneumoniae, S. mitis, and S. pseudoneumoniae) that produce
CDC without a signal sequence. The secretion mechanism for these CDC is unclear
(Jefferies et al., 2007, Marriott et al., 2008). After secretion to the extracellular medium,
the CDC fold into water-soluble monomeric proteins, travel and bind to the target
membrane, and oligomerize on the membrane surface forming characteristic arcs and
ring-like structures which are responsible for cytolysis. Several reviews have been
published describing the recent advances in the structural and mechanistic studies of the
CDC (Alouf et al., 2006, Giddings et al., 2006, Gilbert, 2005, Rossjohn et al., 2007,
Tweten, 2005). Here, we will focus on the role played by cholesterol during the
transformation of the CDC from a water-soluble monomer to a membrane-inserted
oligomeric complex. Although the cholesterol-dependent inhibition of the activity for
these toxins was one of the first biochemical properties attributed to the family
(Arrhenius, 1907), the molecular mechanism of the cholesterol-toxin interaction remains
as one of the least understood aspects in the study of the CDC toxin family.
TABLE 1
2. MECHANISM OF PORE-FORMATION
The 28 CDC family members listed in Table 1 show a significant degree of amino
acid identity (from 28.1 to 99.6 percent) and similarity (greater than 45.7 percent), with
amino acid sequences ranging from 471 to 665 amino acids in length. A comparison of
the primary structure of these proteins shows that they share a very low degree of
4
similarity at their N-terminus, in part because different species employ distinct signal
sequences for secretion, but also because some of the CDC members possess additional
domains located in this region (e.g., Farrand et al., 2008). If we consider just the
conserved core shared by all CDC and required for pore-formation activity [amino acids
38-500 in perfringolysin O (PFO)], the amino acid identity and similarity among different
members becomes higher than 36.7 and 58 percent, respectively (sequence length of
analyzed sequences range from 462 to 469, Figure 1). Therefore, from the analysis of the
primary structure of these toxins we can anticipate that all the CDC will exhibit similar
activities and three-dimensional structures.
FIGURE 1
The first crystal structure for a CDC was solved for PFO by Rossjohn and
colleagues (1997). The crystal structure for two other CDC, intermedylisin (ILY) and
anthrolysin (ALO), have been solved so far, and all of them share similar secondary and
tertiary structure (Bourdeau et al., 2009, Polekhina et al., 2005). They have a high
β-strand content and their structures have been divided into four domains, with the
C-terminal domain (domain 4 or D4) being the only independent and continuous domain
(Figure 2A) (Polekhina et al., 2006).
PFO secreted by pathogen Clostridium perfringens is a prototypical CDC (Tweten
et al., 2001). To describe the general mechanism of pore-formation for the CDC we will
depict the current knowledge of the PFO cytolytic mechanism which starts with the
binding of the toxin to the target membrane, and concludes with the insertion of a large
transmembrane β-barrel (Figure 2A).
FIGURE 2
5
Upon encountering a cholesterol-containing membrane, PFO oligomerizes and
spontaneously inserts into the bilayer to form a large transmembrane pore (~35-50
monomers per oligomer; approximately 250 Å in diameter, Figure 2), (Czajkowsky et al.,
2004, Dang et al., 2005, Mitsui et al., 1979, Olofsson et al., 1993). The C-terminus of
PFO (D4) encounters the membrane first (Figure 2A, I, Heuck et al., 2000, Nakamura et
al., 1995, Ramachandran et al., 2002). The binding of D4 triggers the structural
rearrangements required to initiate the oligomerization of PFO monomers
(Ramachandran et al., 2004, Soltani et al., 2007a) and formation of a pre-pore complex
on the membrane surface (Figure 2A, II, Heuck et al., 2003, Shepard et al., 2000, Tilley
et al., 2005). Pore formation commences when two amphipathic β-hairpins from each
PFO molecule insert and span the membrane (Figure 2A, III, Hotze et al., 2002,
Shatursky et al., 1999, Shepard et al., 1998). The concerted insertion of two
transmembrane β-hairpins (TMHs) from ~35 PFO monomers then creates a large
transmembrane β-barrel that perforate the membrane (Dang et al., 2005, Tilley et al.,
2005). This general mechanism of pore-formation is followed by most CDC, however,
some variations have been observed for specific members and they will be described in
the following sections.
2.1 Localizing the target membrane
The first step in the CDC cytolytic cascade is the recognition of the target cell
(Figure 2A, I). The CDC bind to the target membrane by recognizing a specific
membrane lipid, cholesterol, or by recognizing a membrane-anchored protein as in the
case of ILY (Giddings et al., 2004). Cholesterol-recognition provides specificity towards
eukaryotic cells in general, and the glycosylphosphatidylinositol-anchored protein CD59
6
provide specificity for human cells. While it has been shown that ILY interacts with the
CD59 receptor forming a 1:1 complex (Lachapelle et al., 2009), the interaction of other
CDC with cholesterol is less understood. Independently of the recognition mechanism, it
appears that all CDC bind to the target membranes via D4 (Nagamune et al., 2004,
Soltani et al., 2007a).
2.2 Grouping forces on the membrane surface: pre-pore formation
After successful recognition of the target membrane, the CDC oligomerize in the
membrane surface to form a membrane-bound pre-pore complex (Figure 2, II).
Formation of a pre-pore complex seems to be a common feature of the β-barrel pore-
forming toxins (Heuck et al., 2001, Miller et al., 1999, Shepard et al., 2000, Walker et al.,
1992). The secreted monomeric proteins do not oligomerize in solution, and it has been
shown that the binding of the toxins to the target membrane is required to trigger the
monomer-monomer association (Abdel Ghani et al., 1999, Lachapelle et al., 2009,
Ramachandran et al., 2004). Although oligomerization has been observed in the absence
of membranes for certain CDC (e.g., pneumolysin, (PLY) Gilbert et al., 1998, Solovyova
et al., 2004), it only occurs when the toxin concentration is relatively high (in the
micromolar range or higher) compared to the concentration needed for efficient
oligomerization when incubated with natural membranes. The difference in efficiency
between oligomerization in solution and at the surface of a cell membrane suggests that
the cells in some way promote the association of toxin monomers. In general,
oligomerization of β-barrel pore-forming toxins requires the exposure of hidden
polypeptide regions involved in the monomer-monomer interaction (Heuck and Johnson,
2005, Heuck et al., 2001). In the CDC, this process is triggered by conformational
7
changes induced by protein-lipid interactions (e.g., PFO, Ramachandran et al., 2004) or
by conformational changes induced by protein-protein interactions (e.g., ILY Lachapelle
et al., 2009).
Ramachandran et al. (2004) have shown that in the water-soluble form of the
toxin, oligomerization is prevented by blocking access to one edge of a core β-sheet in
the monomer (Figure 2B). This blockage prevents its association with the edge of the
core β-sheet in the neighboring monomer, thus impeding formation of an extended
β-sheet. Specifically, premature association of PFO molecules (before they bind to the
appropriate membrane surface) is prevented by the presence of β5, a short polypeptide
loop that hydrogen bonds to β4 in the monomer, and thereby prevents its interaction with
the β1 strand in the adjacent monomer. This feature is conserved in all crystal structures
reported for the CDC (i.e., PFO, ILY, and ALO).
The structural changes associated with converting a CDC from a water-soluble
monomer to a membrane-inserted oligomer extend through much of the molecule. The
binding of D4 to the membrane surface immediately elicits a conformational change in
domain 3, more than 70 Å above the membrane (Abdel Ghani et al., 1999, Heuck et al.,
2000, Ramachandran et al., 2002, Ramachandran et al., 2004, Ramachandran et al.,
2005). This conformational change rotates β5 away from β4 and thereby exposes β4 to
the aqueous medium where it can associate with the always-exposed β1 strand of another
PFO molecule to initiate and promote oligomerization (Figure 2B).
Such an extensive network of structural linkages within a CDC can be
advantageous because it reduces the chance of prematurely entering a structural transition
that exposes a TMH. By allosterically linking different domains or regions of the protein,
8
the system can couple separate interactions (e.g., binding to the membrane and binding to
another subunit) and thereby ensure that pore formation proceeds only when the
necessary criteria are met. Given the important allosteric communication between the
membrane binding domain and the pore-forming domain, it is not surprising that the most
conserved regions on these proteins are located among inter-domain segments, forming
an almost continuous path with its origin at the tip of D4 and terminus at the segments
that form the amphipathic TMHs (Figure 3). Interestingly, while most of the surface
exposed residues of the CDC are not very conserved, the residues at the surface of the D4
tip, involve in membrane interaction, are highly conserved.
FIGURE 3
Establishment of an oligomeric complex in the membrane surface facilitates the
formation of a transmembrane pore because the insertion of a single amphipathic
β-hairpin into a membrane is not energetically favored. In a hydrophobic environment
that lacks hydrogen bond donors or acceptors, isolated β-hairpins cannot achieve the
hydrogen-bond formation necessary to lower the thermodynamic cost of transferring the
polar atoms of the polypeptide backbone into the hydrocarbon interior (White and
Wimley, 1999). However, this energy barrier is circumvented if the β-strands are inserted
as β-sheets and form closed structures such as a β-barrel. For monomeric β-barrel
membrane proteins such as OmpA, a concerted folding mechanism has been observed in
vitro in which the hydrogen bonds formed between adjacent β-chains presumably favor
the insertion of the β-barrel into the membrane (Kleinschmidt, 2006, Tamm et al., 2004).
Similarly, the formation of a pre-pore complex may be required to allow the concerted,
and perhaps simultaneous, insertion of the β-hairpins from individual monomers, thereby
9
overcoming the energetic barrier of inserting non-hydrogen-bonded β-strands into the
membrane. Whereas it is clear that the formation of a complete ring (or pre-pore
complex) on the membrane surface will minimize the energetic requirements for inserting
a β-barrel into the membrane, it is likely that the insertion of incomplete rings can also
occur if monomer recruitment into the oligomer slows down. In the absence of additional
monomers, the incomplete pre-pore complexes observed in vitro (or metastable arc
structures) will be trapped, and they may have enough time to insert into the membrane
and form a pore (Gilbert, 2005). Insertion of an arc may well form a transmembrane pore
by itself, or in association with other arcs (double arc structures, Palmer et al., 1998). A
minimal number of monomers must be required to overcome the energetic barrier of
inserting an arc-like β-sheet into the membrane. It has been shown that independently of
the toxin/lipid ratio, the pores formed by PFO and streptolysin O (SLO) are at least large
enough to allow the passage of proteins with an approximate diameter of 100 Ǻ (Heuck
et al., 2003).
In summary, a coordinated train of events regulates the proper assembly of the
CDC oligomeric complex at the surface of the target membrane. Formation of these
oligomeric structures facilitates the insertion of numerous TMHs which are required to
form the large transmembrane β-barrel.
2.3 Perforating the membrane: insertion of a large β-barrel
A characteristic of the CDC that distinguishes them from most other β-barrel
pore-forming toxins is the use of two amphipathic β-hairpins per monomer to form the
large transmembrane barrel (Heuck and Johnson, 2005, Heuck et al., 2001, Shatursky et
al., 1999). In the water-soluble monomeric configuration of the CDC these TMHs are
10
folded as short α-helices presumably to minimize the exposure of the hydrophobic
surfaces (Heuck and Johnson, 2005). These helices, located at either side of the central
β-sheet in domain 3, extend and insert into the membrane bilayer (Shatursky et al., 1999,
Shepard et al., 1998). The conversion of short α-helices to amphipathic β-hairpins
constituted a new paradigm for how pore-forming toxins transform from a water-soluble
to membrane-inserted conformation. This structural transformation has been recently
found in eukaryotic pore-forming proteins, as revealed by the structure of the membrane
attack complex/perforin superfamily members (Hadders et al., 2007, Rosado et al., 2007).
After insertion the hydrophobic surfaces of the TMHs are exposed to the non-polar lipid
core of the membrane and the hydrophilic surfaces face the aqueous pore. A concerted
mechanism of insertion ensures that the hydrophilic surfaces of the hairpins remain
exposed to the aqueous medium, and not to the hydrophobic core of the membrane. Such
a coordinated insertion requires the displacement of lipids as the aqueous pore is formed
in the membrane.
The creation of a circular hole, having a radius of nearly 150 Å, in a liposomal
membrane requires the displacement of about 1000 phospholipid molecules in each
leaflet (or about 800 phospholipids plus 800 cholesterol molecules because the average
surface area occupied by one phospholipid molecule plus one cholesterol molecule is ~90
Å2 in a 1:1 phospholipid/cholesterol mixture (Heuck et al., 2001, Lecuyer and
Dervichian, 1969)). Analysis of the release of markers encapsulated in liposomes when
using limiting concentrations of PFO or SLO showed that both, the small markers and the
large markers are released with the same rate. Therefore, it appears that all of these lipid
11
molecules leave the pore formed by these CDC at the same time (Heuck et al., 2003),
though not all agree (Palmer et al., 1998).
A direct comparison of the cytolytic mechanism of PFO and ILY showed that
whereas ILY does not require cholesterol for binding, pore-formation is entirely
dependent on the presence of cholesterol in the target membrane (Giddings et al., 2003).
Employing a series of ILY mutants that block pore formation at different stages, Hotze
and colleagues have shown that ILY remains engaged with its receptor (human CD59)
throughout the assembly of the pre-pore complex, but it is released from CD59 upon the
transition to the membrane-inserted oligomer (Lachapelle et al., 2009). Upon release
from the receptor, ILY is anchored to the membrane via D4 suggesting that this domain
still conserves the cholesterol binding properties of other CDC members (note that
insertion of the ILY β-barrel does not occur if cholesterol is depleted from the
membrane).
After pre-pore formation, the insertion of the PFO TMHs requires the proper
intermonomer β-strand alignment. Ramachandran et al. (2004) suggested that the
π-stacking interaction between Y181 and F318 guides the alignment of the TMHs of
adjacent monomers (Figure 2B). Interestingly, while Y181 is completely conserved in the
28 members of the CDC family, F318 is not. Instead of phenylalanine, this position is
occupied by valine in lectinolysin, vaginolysin, and PLY, by isoleucine in ILY, and
alanine in pyolysin. It will be interesting to determine if a mutation of the conserved
PFO-Y181-equivalent in ILY results in a pre-pore blocked derivative, as observed in
PFO.
3. THE ROLE OF CHOLESTEROL IN MEMBRANE BINDING
12
Among all the different lipids that shape the vast diversity of cell membranes,
cholesterol is a distinguishing feature of mammalian cells. The CDC have evolved to take
advantage of this feature of mammalian membranes, and their ability to perforate the
target membrane is totally dependent on the presence of cholesterol (Giddings et al.,
2003, Palmer, 2004).
In liposomal membranes containing only phosphatidylcholine and cholesterol,
more than 30 mole % cholesterol is required for CDC such as tetanolysin (Alving et al.,
1979), SLO (Rosenqvist et al., 1980), and PFO (Heuck et al., 2000, Ohno-Iwashita et al.,
1992), to bind and create a pore in the bilayer. For PFO, no binding at all is detected
when the cholesterol concentration in the liposomal membrane is less than ~30 mole % of
the total lipids (Flanagan et al., 2009, Heuck et al., 2000, Nelson et al., 2008). Thus, if
cholesterol acts solely as a receptor, and hence as a PFO binding ligand, reducing the
cholesterol concentration in the bilayer should only affect the kinetics of the cytolytic
process. In other words, lowering the amount of cholesterol in the membrane should
result in a longer time required for PFO to form a transmembrane pore. However, the
sharp transition observed in the binding isotherm of PFO suggests that the basis of this
recognition is more complex than a simple encounter frequency between PFO and
individual cholesterol molecules (Heuck et al., 2000).
3.1 Domain 4 and membrane recognition
The initial members of the CDC family were characterized by their sensitivity to
oxygen and cholesterol (Alouf et al., 2006). Toxins isolated from culture supernatants
were inactivated by exposure to oxygen present in the air or when pre-incubated with
cholesterol. While the oxygen-dependent inactivation of the toxins could be reversed by
13
incubation with thiol-based reducing agents, inactivation by pre-incubation with
cholesterol was not reversible. A direct consequence of these findings was that the
discovery of new CDC members was strongly influenced by the search for these two
distinguishing features in the newly encountered hemolytic toxins: inhibition by oxygen
and cholesterol. Therefore, it is not surprising that the first sequences obtained for the
CDC revealed that all of them contained a conserved undecapeptide which was critical
for cholesterol recognition, and a unique cysteine in this segment that was sensitive to
aerobic oxidation. This correlation led researchers to postulate that the conserved
undecapeptide, and attendant cysteine constituted the cholesterol binding site for the
CDC. However, advancements in recombinant DNA technology soon allowed
researchers to show that this unique cysteine was not essential for cholesterol recognition.
First, the replacement of this cysteine with alanine rendered a protein that remained
hemolytic (Michel et al., 1990, Pinkney et al., 1989, Saunders et al., 1989, Shepard et al.,
1998). Second, the sequence of newly discovered CDC members showed that this
cysteine was indeed replaced by alanine during the evolution of different Gram-positive
species (Billington et al., 2001, Nagamune et al., 2000).
New protein homologues of the CDC are being revealed as new genomes are
sequenced, and these new family members are showing more variability in the amino
acid sequence of this segment. The multi-sequence alignment for the 28 CDC sequences
shows that 20% of the CDC contain amino acid substitutions in the undecapeptide. Based
on this newly accumulated evidence, the original view of the conserved undecapeptide as
the cholesterol binding site is being replaced by alternative models for membrane-
binding. It has been shown that one of the CDC, intermedilysin (ILY) recognize the
14
target membrane by the specific binding to a human protein receptor, CD59, and it is
therefore possible that other members may also bind to the target membrane by as yet
unidentified protein receptors (Bourdeau et al., 2009). In addition to the undecapeptide,
other well conserved peptide loops located at the tip of D4 may contribute to the
cholesterol recognition motif (Ramachandran et al., 2002, Soltani et al., 2007a, Soltani et
al., 2007b).
3.1.1 The conserved loops
PFO D4 has a 4 stranded β-sandwich structure that interacts with the membrane
surface only at one end via the distal loops that interconnect the eight β-strands that form
the domain (Figure 4A Ramachandran et al., 2002, Rossjohn et al., 1997, Soltani et al.,
2007a). Superimposition of the D4 α-carbons for PFO, ALO, and ILY reveals that the
global structure of D4 is well conserved among these members. The main differences
arise in the conformation of the undecapeptide, involved in toxin-membrane interaction,
and in the loops that are close to the domain 2-D4 interface (Figure 4A).
FIGURE 4
Three of the four loops located at the distal tip of D4 are highly conserved among
the CDC members: the conserved undecapeptide (also known as the Trp-rich loop), L1,
and L2 (Figure 4B). The L3 loop is less conserved and is located farther away from the
unique cysteine residue. Recent data obtained by Tweten and colleagues suggest that in
addition to the undecapeptide, the other D4 loops (L1-L3) may also play a role in the
cholesterol-dependent recognition of the CDC (Soltani et al., 2007b). Single amino acid
modifications in these loops prevented the binding of PFO to cholesterol-rich liposomes,
and abolished the pre-pore to pore transition for ILY in a cholesterol-dependent manner.
15
Both of these events involve the association of the D4 with the cholesterol-containing
membrane. It has become clear that the three-dimensional arrangement of the
undecapeptide and the L1-L3 loops is important for the association of the CDC with the
cholesterol-containing membrane (Giddings et al., 2003, Polekhina et al., 2005, Soltani et
al., 2007a, Soltani et al., 2007b).
Interestingly, changes in the pH of the medium which affect the conformation of
D4 also influence the cholesterol-toxin interaction. A reduction of the pH from 7.5 to 6.0
induces a conformational change in PFO causing the tryptophan residues to be more
exposed to the aqueous solvent, and also alters the threshold for the minimal cholesterol
concentration required to trigger binding of PFO to liposomal membranes (Nelson et al.,
2008). Since no major changes are expected to occur in the structure of the membrane in
between pH 7.5 and 6.0, one can assume that protonation of certain amino acids in PFO
may alter the D4 conformation, and as a consequence, its ability to recognize cholesterol
in the target membrane. A related effect has been observed for listeriolysin O (LLO), a
CDC recognized for having an optimum acidic pH for activity (Bavdek et al., 2007). The
loss of activity for LLO at neutral pH can be rescued by increasing the concentration of
cholesterol in the membrane.
Given that conformational changes in D4 can alter the cholesterol-dependent
properties of the CDC, one can speculate that the conformational change triggered by the
binding of ILY to the CD59 receptor (Soltani et al., 2007a), may modulate the
cholesterol-dependent association with the membrane required for pore-formation.
Unfortunately, despite the various high-resolution structures available for the
CDC, and the multiple functional data obtained by modification of amino acids located at
16
the D4 loops, it is still unclear how cholesterol modulates the conformational changes
required to anchor the toxin to the membrane and to insert a large transmembrane
β-barrel. Furthermore, is no clear if the binding of PFO (and related CDC) is triggered by
the binding of a single cholesterol molecule (Geoffroy and Alouf, 1983, Nollmann et al.,
2004, Polekhina et al., 2005), or by the recognition of a more complex cholesterol-
arrangement in the bilayer structure (Bavdek et al., 2007, Flanagan et al., 2009, Heuck
and Johnson, 2005, Heuck et al., 2007, Nelson et al., 2008).
3.2 Searching for cholesterol in the membrane
The binding of a protein domain to a membrane surface is in general, a two step
process that involves the initial formation of non-specific collisional complex followed
by the formation of a tightly bound complex. The first step is diffusional and may involve
electrostatic interactions, and the second step stabilizes the initial interaction by
membrane penetration of non-polar amino acids and/or specific interactions between the
protein and the membrane lipids (Cho and Stahelin, 2005). The initial membrane
association locates non-polar amino acids close to the interfacial region of the bilayer,
facilitating their exposure to the hydrophobic core. Non-polar amino acids are not usually
exposed to the protein surface, and therefore conformational changes are required to
expose them to the membrane.
Exposure of the aromatic residues located in the undecapeptide occurs upon
membrane binding, though they do not penetrate deeply into the bilayer core (Heuck et
al., 2003, Nakamura et al., 1998, Sekino-Suzuki et al., 1996). The sensitivity of the
undecapeptide to amino acid changes suggests that the exposure of aromatic amino acids
and membrane binding requires precise conformational changes and/or a particular three-
17
dimensional conformation. A conformational change in the undecapeptide that modulate
cholesterol binding and membrane anchoring has been suggested for PFO (Rossjohn et
al., 1997), however the binding site for cholesterol, if any, remains elusive.
It has become apparent that in addition to the three dimensional structure of the
binding-domain, the arrangement of the cholesterol molecules in the bilayer is also
critical for successful binding. In a membrane, the cholesterol molecule swims in the
non-polar core of the bilayer with an orientation nearly parallel to the acyl chains of the
phospholipids. The non-polar hydrocarbon tail of the molecule orients towards the center
of the bilayer, and the 3-β-OH group locates close to the ester bonds formed by the fatty
acid chains and the glycerol backbone of the phospholipids near the membrane-water
interface. Compared to the phospholipid head groups, the polar group of the cholesterol
molecule is not very exposed to the membrane surface. Therefore, it is not strange that at
relatively low concentrations the cholesterol molecules are not readily available to
interact with water-soluble molecules (e.g., cholesterol oxidase, cyclodextrins) (Lange et
al., 1980).
3.2.1 Cholesterol availability in membrane bilayers
In multicomponent membranes, the availability of cholesterol at the membrane
surface is regulated by the interactions between cholesterol and other the components of
the membrane (phospholipids, glycolipids, proteins). The more the cholesterol interacts
with the membrane components, the less available it will be to interact with extra-
membranous molecules. Factors that affect the interaction of cholesterol with
phospholipids are the length of the acyl chains, the presence of double bonds in these
18
chains, the size of the polar head-groups, and the ability of the phospholipid to form
hydrogen bonds with the hydroxyl group of cholesterol (Ohvo-Rekilä et al., 2002).
When cholesterol is added to a membrane containing a single phospholipid
species, two phases appear in a concentration-dependent manner (Mouritsen and
Zuckermann, 2004, Sankaram and Thompson, 1991). This suggests that instead of
randomly distributing among the membrane phospholipids, cholesterol associates with
the phospholipids presumably forming stoichiometric complexes (Radhakrishnan and
Mcconnell, 1999). When the phospholipids are in excess, most of the cholesterol
molecules are forming complexes with phospholipids. These complexes are immiscible
in the pure phospholipid phase and therefore a two-phase mixture appears in the
membrane. Increasing the cholesterol concentration will increase the population of the
complexes until they form a single phase containing the complexes with a minor presence
of uncomplexed phospholipids and cholesterol molecules. Beyond this point, the added
cholesterol molecules (free cholesterol) will mix with the complexes until they reach the
solubility limit and precipitate out of the membrane (Mason et al., 2003). Cholesterol
molecules do not form stable bilayers in aqueous solution, so when present in excess they
cannot form a new stable and extended phase. The free cholesterol molecules in excess
have a tendency to “fly” away from the membrane, and outside the membrane they will
be prone to aggregate and form crystals (Harris, 1988).
The formation of phospholipid-cholesterol complexes can explain the low
interaction detected between cyclodextrins and cholesterol when the sterol is present in
low amounts (Mcconnell and Radhakrishnan, 2003). An alternative model to account for
this behavior was proposed by Huang and Feigenson (1999). These authors propose that
19
the hydrophobic effect positions the phospholipid head groups toward the membrane
surface to protect the hydrophobic molecule of cholesterol from the unfavorable contact
with water. When the concentration of cholesterol in the membrane achieves and exceeds
the protective capacity of the head-groups, the tendency for the sterol molecules to “fly”
away will increase.
Both models provide a reasonable explanation for the increased accessibility of
cholesterol at high sterol/phospholipid ratios, and the consensus is that they are not
mutually exclusive (Lange and Steck, 2008, Mesmin and Maxfield, 2009). Binding
(and/or pore-formation) of the CDC occurs at high cholesterol concentration where free
cholesterol become available, and therefore any of these models can be used to explain
the experimental observations.
In more complex lipids mixtures, when more than one phospholipid is present in
the membrane, the total cholesterol content will distribute unevenly between any formed
phases (Goñi et al., 2008, Veatch and Keller, 2002). How much cholesterol is present in
each phase will be governed by the interaction between cholesterol and the components
(lipids and proteins) present in the phases (Epand, 2006).
3.2.2 The role of other lipids
The pioneering work of Ohno-Iwashita and colleagues in the binding of PFO to
membranes showed that the phospholipid composition affects the arrangement of
cholesterol in the membrane. Using a protease-nicked derivate of PFO they showed that
the binding of the toxin was no only influenced by the total amount of cholesterol present
in the membrane, but also by the phospholipid composition. They found that this PFO
derivative preferentially binds to cholesterol-rich membranes composed of phospholipids
20
with 18-carbon acyl chains (Ohno-Iwashita et al., 1992, Ohno-Iwashita et al., 1991). An
effect on cholesterol state in the membrane by ceramides and glycerolipids was also
suggested by Zitzer et al. (2003) based on their studies of SLO pore-formation in
liposomal membranes prepared with different phospholipids. Lipids having a conical
molecular shape appear to effect a change in the energetic state of membrane cholesterol
that in turn augments the interaction of the sterol with the cholesterol-specific cytolysin.
Interestingly, these authors also showed that SLO was active when membranes were
prepared solely with the enantiomeric cholesterol, suggesting that the effect associated
with the presence of cholesterol may be other than a site specific binding event (Zitzer et
al., 2003).
A more systematic analysis of the interaction of PFO D4 with membranes
prepared with different phospholipds and sterols revealed that PFO binding to the bilayer
and the initiation of the sequence of events that culminate in the formation of a
transmembrane pore depend on the availability of free cholesterol at the membrane
surface (Flanagan et al., 2002, Flanagan et al., 2009, Nelson et al., 2008). These studies
also showed that changes in the acyl chain packing of the phospholipids, and cholesterol
in the membrane core do not correlate with PFO binding. Taken together, all these studies
suggest than the binding of PFO (and SLO) to the membrane is triggered when the
concentration of cholesterol exceeds the association capacity of the phospholipids, and
this cholesterol excess is then free to associate with the toxin (Figure 5).
FIGURE 5
The requirement of such high cholesterol content in membranes was initially
associated with the binding of PFO to cholesterol-rich domains (or membrane rafts)
21
(Ohno-Iwashita et al., 2004, Waheed et al., 2001). However, recent results indicate that
this assertion may require further analysis and consideration. It was found that the
incorporation of sphingomyelin, a necessary component for the formation of membrane
rafts, inhibited rather than promoted the binding of PFO to membranes (Flanagan et al.,
2009). No correlation was found between PFO binding, and the amount of the detergent-
resistant fraction in membranes, a fraction usually associated with membrane rafts
(Flanagan et al., 2009). Incorporation of sterols that promote the formation of ordered
membrane domains was not critical to promoting the PFO-membrane interaction (Nelson
et al., 2008). Therefore, one needs to be cautious when employing PFO as a probe to
reveal the presence of membrane rafts in cellular membranes. Rather than recognizing a
particular membrane “raft”, PFO seems to bind to membranes containing free cholesterol
(or where cholesterol has a high chemical activity).
3.2.3 Cholesterol is enough
It was long known that incubation of SLO (Duncan and Schlegel, 1975, Johnson
et al., 1980), PFO (Mitsui et al., 1979), cereolysin (Cowell and Bernheimer, 1978),
alveolysin (Johnson et al., 1980), PLY (Johnson et al., 1980), and LLO (Vazquez-Boland
et al., 1989) with cholesterol dispersed in aqueous solution produced the typical
aggregated sterol-toxin complexes. For PFO and SLO, typical ring- and arc-like
structures were observed after incubation with cholesterol at concentrations above its
solubility limit (i.e., higher than 5 μM Duncan and Schlegel, 1975, Haberland and
Reynolds, 1973, Harris et al., 1998, Mitsui et al., 1979).
To clarify the role of cholesterol in PFO cytolysis, the extent to which the
different steps of the cytolytic mechanism could be elicited solely by the presence of
22
cholesterol was analyzed (Heuck et al., 2007). Using site-directed fluorescence labeling
of PFO in combination with multiple independent fluorescence techniques (Heuck and
Johnson, 2002, Johnson, 2005), it was revealed that a selective interaction between the
undecapeptide and the D4 loops with cholesterol dispersed in aqueous solution is
indistinguishable from the interaction of PFO with cholesterol-containing membranes.
Binding solely to cholesterol aggregates in aqueous solution is sufficient to initiate the
coupled conformational changes that extend throughout the toxin molecule from the tip
of D4 to the TMHs. Moreover, it was found that the topology of D4 bound to cholesterol
aggregates was identical to the one observed in liposomal membranes, and that the
binding of PFO to cholesterol aggregates was sufficient to trigger the conformational
change in domain 3 that has been associated with oligomerization (Heuck et al., 2007,
Ramachandran et al., 2004). As previously observed for SLO in cholesterol micro-
crystals (Harris et al., 1998), oligomerization and formation of typical arc and ring
structures were observed in the presence of cholesterol aggregates. Surprisingly, none of
these changes were produced by epicholesterol, a sterol that differs from cholesterol only
in that the hydroxyl group is directed axially instead of equatorial (Heuck et al., 2007).
Taking advantage of the inability of PFO to recognize epicholesterol, competition
experiments were done to examine how cholesterol packing in the bilayer affects the
interactions with the membrane. More than 48 mole% cholesterol is required for PFO to
bind to POPC-cholesterol liposomes (Flanagan et al., 2009). However, when the
epicholesterol was mixed with cholesterol to maintain the concentration of total sterols
constant at 48 mole%, and to reduce the net amount of cholesterol in the membrane, it
was shown that in this case considerable binding of PFO was found with as little as 19
23
mole% cholesterol. Epicholesterol apparently intercalates in the bilayer and competes
with cholesterol for association with phospholipids, as reported for other membrane
intercalating agents (Lange et al., 2005). These data therefore confirmed that there are at
least two distinctive states of cholesterol in a typical membrane bilayer: one in which
cholesterol is readily accessible for binding to proteins such as PFO (free cholesterol),
and one in which the sterol is associated with surrounding membrane components that
reduce its exposure to the surface (e.g., phospholipid headgroups may obscure access to
sterols associated with phospholipid acyl chains).
The selective binding of PFO to cholesterol aggregates and not to epicholesterol
aggregates, suggests that the failure to bind epicholesterol when incorporated in
membrane bilayers is not related to the packing or association of this sterol with the
phospholipids. This failure is rather caused by the wrong orientation of the hydroxyl
group (Murari et al., 1986), which it may be required for the specific docking of the sterol
molecule to a binding pocket located in D4 (Figure 5B Rossjohn et al., 2007). or
alternatively, to be properly exposed at the surface of a lipid cluster that may act as a
platform for the anchoring of the D4 loops (Figure 5C). Such a cluster may be preformed
on the membrane before binding, or formed as a result of the interaction of D4 with the
bilayer surface. Redistribution of lipids after protein-binding has been observed for LLO
(Gekara et al., 2005), and other proteins (e.g., Heimburg et al., 1999).
The PFO and SLO specific binding to cholesterol aggregates and microcrystals
(Harris et al., 1998, Heuck et al., 2007), together with the need for more than 30 mole%
cholesterol in membranes to trigger binding (Flanagan et al., 2009, Heuck et al., 2000,
Nelson et al., 2008), suggest that the role of cholesterol in the cytolytic mechanism of the
24
CDC may be more complex than solely binding to a specific binding site. An alternative
explanation would be the need of a cluster of cholesterol molecules at the membrane
surface to provide a docking platform for the D4 loops (Gekara et al., 2005, Heimburg et
al., 1999, Heuck and Johnson, 2005). Interestingly, the binding of pore-forming toxins to
lipid clusters have been reported for Staphylococcus aureus α-hemolysin (Valeva et al.,
2006), and the need for small cholesterol clusters have been recently suggested for the
binding of LLO to membranes (Bavdek et al., 2007). Further work is needed to
unambiguously determine the mechanism by which cholesterol specifically anchors the
CDC to the target membrane.
4. CONCLUSIONS AND FUTURE PERSPECTIVES
Recent studies support the concept that there is a complex interplay between the
structural arrangement of the D4 loops and the distribution of cholesterol in the target
membrane (Bavdek et al., 2007, Flanagan et al., 2009, Giddings et al., 2003, Heuck and
Johnson, 2005, Nelson et al., 2008, Polekhina et al., 2005, Ramachandran et al., 2002,
Soltani et al., 2007a, Soltani et al., 2007b). Modifications in the lipid composition alter
the cholesterol arrangement in the membrane, and as a consequence, the binding of the
CDC (Flanagan et al., 2009, Nelson et al., 2008). At the same time, modifications to the
structure of the CDC effected by mutations, changes in the pH of the medium, or other
factors, modifies the threshold for the amount of cholesterol required to trigger binding
(Bavdek et al., 2007, Nelson et al., 2008, Moe & Heuck, unpublished).
The presence of free cholesterol molecules at the membrane surface seems to be
critical to trigger the binding of most CDC. A direct inference from these findings is that
the exposure of cholesterol at the membrane surface may be facilitated by the action of
25
other membrane-damaging toxins secreted by these pathogens like, for example,
phospholipases C. These toxins cleave the head-groups of phospholipids, and
consequently increase the exposure of cholesterol molecules (or availability of free
cholesterol) to the membrane surface. Cooperation between the CDC and different
phospholipases C contribute to the pathogenesis of at least two organisms. A synergic
effect has been reported for the action of PFO and α-toxin in clostridial myonecrosis
(Awad et al., 2001), and both phospholipases C and LLO have been identified as key
factors for the vacuolar dissolution and cell-to-cell spreading mechanism of Listeria
monocytogenes (Alberti-Segui et al., 2007).
Complete understanding of the mechanism of pore formation for the CDC at the
molecular level would require high-resolution structures of the initial (water-soluble
monomer), the final (membrane-inserted oligomer), and any intermediate state involved
in the cytolytic process (including complexes with receptors or lipids). Great progress has
been achieved to this end, but there is much more to be accomplished. A few crystal
structures for monomeric CDC are currently available (PFO, ILY, ALO, Bourdeau et al.,
2009 , Polekhina et al., 2005, Rossjohn et al., 1997), and the low resolution structure for
the pre-pore complex and the membrane-inserted oligomer of PLY have been obtained by
cryo-electron microscopy (Tilley et al., 2005).
It has become clear that the analysis of complex biological systems, in particular
those involving membranes, benefits from the combination of high-resolution structural
techniques (e.g., X-ray crystallography, nuclear magnetic resonance, electron
microscopy) and spectroscopic analysis of probes incorporated at specific positions in the
proteins (e.g., electron paramagnetic resonance, fluorescence spectroscopy) (Cowieson et
26
al., 2008, Heuck and Johnson, 2002, Hubbell et al., 2000). In addition to providing
structural information, by monitoring the spectral signal of these probes as a function of
time, one can determine the kinetics of the discrete steps of the pore-formation
mechanism (Heuck et al., 2000, Heuck et al., 2003) and the dynamics of the structural
transformations (Columbus and Hubbell, 2002).
Understanding the CDC function in the establishment of the diseases caused by
various Gram-positive pathogens is far from complete (Marriott et al., 2008, Schnupf and
Portnoy, 2007). The actual role of the CDC in bacterial pathogenesis may be more
complex than merely forming a transmembrane pore. For example, it has been proposed
that SLO is involved in protein translocation during Streptococcus pyogenes infection
(Madden et al., 2001, Meehl and Caparon, 2004).
The involvement of protein receptors in the mechanism of certain CDC is another
area that require further investigation. The discovery of the ILY receptor illuminated two
distinct roles for cholesterol in the cytolytic mechanism of the CDC (Giddings et al.,
2003). ALO’s strong preference for targeting the apical side of gut epithelial cells
suggests that a receptor (other than cholesterol) may be present in these cells (Bourdeau
et al., 2009). Clearly, there is yet much to learn about the complex and fascinating roles
played by the CDC in bacterial pathogenesis.
ACKNOWLEDGMENTS
Work in the authors’ laboratory was supported by a Scientist Development Grant
from the American Heart Association to A.P.H.
27
28
Table 1. Homologs in Gram-positive species compose the CDC family. Twenty-eight
CDC family members from divergent phyla have been identified by amino acid sequence.
The protein three letter code for each homolog (as defined in figure 1) is followed by its
phylogenetic relationship to the PFO standard. Because many of the CDC family are
expressed with variable N-terminus, PFO relationship is expressed in bold for the
conserved core only (corresponding to amino acids 38-500 of PFO) and in parentheses
for the full length form. The lengths of the respective polypeptides are presented.
Percentages of identity and similarity were calculated as indicated in Figure 1 legend.
* subsp. equisimilis
29
PHYLUM
Firmicutes
CLASS
Bacilli
ORDER
Bacillales
FAMILY
Bacillaceae
GENUS
Bacillus %Identity %Similarity Length ID
SPECIES
B. anthracis ALO 72 (68) 88 (83) 462 (512) ZP_03017964.1
B. thurigiensis TLO 74 (69) 88 (83) 462 (512) YP_037419
B. cereus CLO 74 (69) 88 (84) 462 (512) YP_002369889.1
B. weihenstephanensis WLO 74 (69) 87 (83) 462 (512) ABY46062
Listeriaceae
Listeria
L. monocytogenes LLO 43 (40) 66 (62) 469 (529) ABH07645
L. seeligeri LSO 45 (41) 67 (63) 469 (530) P31830.1
L. ivanovii ILO 46 (43) 66 (62) 469 (528) AAR97343.1
Planococcaceae
Lysinibacillus
L. sphaericus SPH 76 (72) 90 (87) 463 (506) YP_001699692.1
Paenibacillaceae
Paenibacillus
P. alvei ALV 75 (71) 87 (84) 462 (501) P23564
Brevibacillus
B. brevis BVL 73 (69) 88 (84) 464 (511) YP_002770211.1
Lactobacillales
Streptococcaceae
Streptococcus
S. dysgalactiae* SLOe 67 (56) 83 (70) 463 (571) BAD77791
S. pyogenes SLO 67 (56) 83 (70) 463 (571) NP_268546.1
S. canis SLOc 66 (55) 82 (69) 463 (574) Q53957
S. pseudonemoniae PSY 46 (43) 67 (63) 466 (471) ACJ76900
S. pneumoniae PLY 46 (43) 67 (64) 466 (471) ABO21366.1
S. mitis MLY 46 (43) 67 (63) 466 (471) ABK58695
S. suis SLY 41 (40) 65 (63) 465 (497) ABE66337.1
S. intermedius ILY 41 (37) 65 (59) 469 (532) BAE16324.1
S. mitis (Lectinolysin) LLY 39 (29) 62 (47) 463 (665) BAE72438.1
Clostridia
Clostridiales
Clostridiaceae
Clostridium
C. perfringens PFO 463 (500) NP_561079
C. butyricum BRY 69 (65) 85 (82) 462 (513) ZP_02950902.1
C. tetani TLY 60 (55) 78 (72) 464 (527) NP_782466.1
C. botulinum B BLYb 60 (49) 78 (63) 464 (602) YP_001886995.1
C. botulinum E3 BLYe 60 (48) 77 (60) 464 (602) YP_001921918.1
C. botulinum C BLYc 60 (56) 79 (74) 463 (518) ZP_02620972.1
C. novyi NVL 58 (54) 78 (73) 463 (514) YP_878174.1
Actinobacteria
Actinobacteria
Bifidobacteriales
Bifidobacteriaceae
Gardenella
G. vaginallis VLY 40 (39) 65 (60) 466 (516) ACD3946.1
Actinomycetales
Actinomycetaceae
Arcanobacterium
A. pyogenes PLO 41 (38) 60 (56) 469 (534) AAC45754.1
30
FIGURE LEGENDS
Figure 1. Analysis of the primary structure for the CDC reveals a high degree of identity
and similarity among them. Only the sequence for the conserved core of the CDC was
used for the analysis (corresponding to PFO amino acids 38-500). If more than one
sequence was available for individual species, only one was used in the analysis. The
databank access numbers are provided in Table 1. Sequence relationships were calculated
using the MatGat 2.02 alignment program using the BLOSUM 62 matrix and open and
extension gap penalties of 12 and 1, respectively (Campanella et al., 2003). The identity
scores occupy the upper triangle (in bold) with scores higher than 70% shaded in dark
gray, and those at 50 – 70% in light gray. Similarity scores in the lower triangle where
shaded in dark gray if higher than 80% and in light gray if between 70 – 80%.
Figure 2. Pore formation mechanism for the CDC. Secreted as water-soluble monomeric
proteins, the toxins bind to the target membrane and oligomerize into a ring-like structure
called the pre-pore complex. A poorly understood conformational change then leads to
the insertion of the TMHs into the bilayer to form the aqueous pore. (A) Stages of PFO
pore formation. The defined PFO structural domains are numbered. The membrane
bilayer is depicted with cholesterol molecules (ovals) intercalated between the
phospholipid constituents. Membrane binding is accomplished as D4 interacts with
membrane regions having free cholesterol molecules. Subsequent allosteric
rearrangements within the monomer promote oligomerization and pore-formation. (B)
Conformational changes in domain 3 of PFO are required for monomer–monomer
association and β-barrel pore formation. Each stage corresponds to the stage shown above
31
in (A). The TMH1 is shown as bicolor and the TMH2 in black. The small β5 strand is
shown as a black loop. The aromatic residues involved in the alignment of the β-strands
are shown as open rectangles. Adapted from Ramachandran et al. with permission
(2004).
Figure 3. Comparison of PFO homologs reveals a conserved core backbone. Alignment
and comparison of the composite members of the CDC family reveals conserved regions
that extend from the tip of the membrane recognition domain, D4, through the regions
involved in oligomerization and membrane insertion. (A) Cartoon representation of PFO
with the conserved residues shown in black. (B) Surface representation of PFO the
conserved core highlighted in black. It is postulated that this conserved backbone is
especially adapted to allosterically communicate successful, cholesterol-dependent
membrane binding, and thus permit subsequent conformational adaptations that favor
oligomerization and pore formation. Alignment of the 28 CDC sequences was effected
using the PRALINE multiple sequence alignment tool using a BLOSUM62 matrix with
open and extension gap penalties set at 12 and 1, respectively, a PSI-BLAST pre-profile
processing with iterations set at 3, e-value cut off set at 0.01, non-redundant data bases,
and a DSSP-defined secondary structure search using PSIPRED (Simossis et al., 2005).
PFO structure representation was rendered using PyMol (DeLano Scientific LLC).
Figure 4. The three dimensional structure of D4 is highly conserved in the CDC family.
(A) Comparison of D4 from 3 CDC homologs highlights the conserved architecture of
this C-terminal domain. A cartoon, upper left, clarifies the threading of 2 β-sheets and
32
loops in the β-sandwich and indicates the spatial organization of the undecapeptide, L1,
and L2. The α-backbone for the D4 domains of PFO, ILY, and ALO were superimposed
using PyMol (DeLano Scientific LLC; available at www.pymol.org). (B) Alignment of
the sequence for the 28 CDC family members reveals substantial conservation in loops
L1, L2 and the undecapeptide. While integrity of the undecapeptide was long recognized
for being critical to the cholesterol-dependent activity of these toxins, other loops are also
important. Residues conserved in all sequences are shaded in black, and highly conserved
residues are shaded in gray. Protein names are as in Figure 2, residue numbers correspond
to the PFO sequence. Multiple sequence alignment was effected as indicated in Figure 3.
Figure 5. PFO only binds to membranes containing free cholesterol molecules. Examples
of mechanisms for cholesterol-dependent anchoring of PFO to the membrane surface: (A)
PFO cannot stably bind to the bilayer if there are no free cholesterol molecules available
in the membrane surface. (B) At high cholesterol concentrations free cholesterol
molecules become available (black ovals), and D4 can anchor to the bilayer. In this
example, a single cholesterol molecule binds to D4 and induces the conformational
changes required to expose the D4 loops to the bilayer core. (C) Alternatively, the
interplay between D4 and the membrane result in the redistribution of the lipids at the
surface, clustering the free cholesterol molecules underneath the tip of D4. Anchoring
may be accomplished by the interaction of multiple hydroxyl groups located in the
cholesterol-rich cluster and the conserved amino acids of the loops.
33
REFERENCES
Abdel Ghani, E. M., Weis, S., Walev, I., Kehoe, M., Bhakdi, S. & Palmer, M. (1999)
Streptolysin O: inhibition of the conformational change during membrane binding of the
monomer prevents oligomerization and pore formation. Biochemistry, 38, 15204-15211.
Alberti-Segui, C., Goeden, K. R. & Higgins, D. E. (2007) Differential function of Listeria
monocytogenes listeriolysin O and phospholipases C in vacuolar dissolution following
cell-to-cell spread. Cell. Microbiol., 9, 179-195.
Alouf, J. E., Billington, S. J. & Jost, B. H. (2006) Repertoire and general features of the
family of cholesterol-dependent cytolysins. In Alouf, J. E. & Popoff, M. R. (Eds.) The
Comprehensive Sourcebook of Bacterial Protein Toxins. 3rd ed., pp. 643-658, Oxford,
England. Academic Press
Alving, C. R., Habig, W. H., Urban, K. A. & Hardegree, M. C. (1979) Cholesterol-
dependent tetanolysin damage to liposomes. Biochim. Biophys. Acta, 551, 224-228.
Arrhenius, S. (1907) Immunochemistry. The application of the principles of physical
chemistry to the study of the biological antibodies., New York, The Macmillian
Company.
Awad, M. M., Ellemor, D. M., Boyd, R. L., Emmins, J. J. & Rood, J. I. (2001)
Synergistic effects of alpha-toxin and perfringolysin O in Clostridium perfringens-
mediated gas gangrene. Infect. Immun., 69, 7904-7910.
Bavdek, A., Gekara, N. O., Priselac, D., Gutierrez Aguirre, I., Darji, A., Chakraborty, T.,
Macìœek, P., Lakey, J. H., Weiss, S. & Anderluh, G. (2007) Sterol and pH
interdependence in the binding, oligomerization, and pore formation of listeriolysin O.
Biochemistry, 46, 4425-4437.
34
Billington, S. J., Songer, J. G. & Jost, B. H. (2001) Molecular characterization of the
pore-forming toxin, pyolysin, a major virulence determinant of Arcanobacterium
pyogenes. Vet. Microbiol., 82, 261-274.
Bourdeau, R. W., Malito, E., Chenal, A., Bishop, B. L., Musch, M. W., Villereal, M. L.,
Chang, E. B., Mosser, E. M., Rest, R. F. & Tang, W.-J. (2009) Cellular functions and x-
ray structure of anthrolysin O, a cholesterol-dependent cytolysin secreted by Bacillus
anthracis. J. Biol. Chem., 284, 14645-14656.
Campanella, J., Bitincka, L. & Smalley, J. (2003) MatGAT: An application that generates
similarity/identity matrices using protein or DNA sequences. BMC Bioinf., 4, 29.
Cho, W. & Stahelin, R. V. (2005) Membrane-protein interactions in cell signaling and
membrane trafficking. Annu. Rev. Biophys. Biomol. Struct., 34, 119-151.
Columbus, L. & Hubbell, W. L. (2002) A new spin on protein dynamics. Trends
Biochem. Sci., 27, 288-295.
Cowell, J. L. & Bernheimer, A. W. (1978) Role of cholesterol in the action of cereolysin
on membranes. Arch. Biochem. Biophys., 190, 603-610.
Cowieson, N. P., Kobe, B. & Martin, J. L. (2008) United we stand: combining structural
methods. Curr. Opin. Struct. Biol., 18, 617-622.
Czajkowsky, D. M., Hotze, E. M., Shao, Z. & Tweten, R. K. (2004) Vertical collapse of a
cytolysin prepore moves its transmembrane beta-hairpins to the membrane. EMBO J., 23,
3206-3215.
35
Dang, T. X., Hotze, E. M., Rouiller, I., Tweten, R. K. & Wilson-Kubalek, E. M. (2005)
Prepore to pore transition of a cholesterol-dependent cytolysin visualized by electron
microscopy. J. Struct. Biol., 150, 100-108.
Duncan, J. L. & Schlegel, R. (1975) Effect of streptolysin O on erythrocyte membranes,
liposomes, and lipid dispersions. A protein-cholesterol interaction. J. Cell Biol., 67, 160-
174.
Epand, R. M. (2006) Cholesterol and the interaction of proteins with membrane domains.
Prog. Lipid Res., 45, 279-294.
Farrand, S., Hotze, E., Friese, P., Hollingshead, S. K., Smith, D. F., Cummings, R. D.,
Dale, G. L. & Tweten, R. K. (2008) Characterization of a streptococcal cholesterol-
dependent cytolysin with a Lewis y and b Specific Lectin Domain. Biochemistry, 47,
7097-7107.
Flanagan, J. J., Heuck, A. P. & Johnson, A. E. (2002) Cholesterol-phospholipid
interactions play an important role in perfringolysin O binding to membrane. FASEB J.,
16, A929.
Flanagan, J. J., Tweten, R. K., Johnson, A. E. & Heuck, A. P. (2009) Cholesterol
exposure at the membrane surface is necessary and sufficient to trigger perfringolysin O
binding. Biochemistry, 48, 3977-3987.
Gekara, N. O., Jacobs, T., Chakraborty, T. & Weiss, S. (2005) The cholesterol-dependent
cytolysin listeriolysin O aggregates rafts via oligomerization. Cell Microbiol, 7, 1345-56.
Gelber, S. E., Aguilar, J. L., Lewis, K. L. T. & Ratner, A. J. (2008) Functional and
phylogenetic characterization of vaginolysin, the human-specific cytolysin from
Gardnerella vaginalis. J. Bacteriol., 190, 3896-3903.
36
Geoffroy, C. & Alouf, J. E. (1983) Selective purification by thiol-disulfide interchange
chromatography of alveolysin, a sulfhydryl-activated toxin of Bacillus alvei. Toxin
properties and interaction with cholesterol and liposomes. J. Biol. Chem., 258, 9968-
9972.
Giddings, K. S., Johnson, A. E. & Tweten, R. K. (2003) Redefining cholesterol's role in
the mechanism of the cholesterol-dependent cytolysins. Proc. Natl. Acad. Sci. U.S.A.,
100, 11315-11320.
Giddings, K. S., Johnson, A. E. & Tweten, R. K. (2006) Perfringolysin O and
Intermedilysin: Mechanisms of Pore Formation by the Cholesterol-Dependent
Cytolysins. IN Alouf, J. E. & Popoff, M. R. (Eds.) The Comprehensive Sourcebook of
Bacterial Protein Toxins. 3rd ed., pp. 671-679, Oxford, England, Academic Press.
Giddings, K. S., Zhao, J., Sims, P. J. & Tweten, R. K. (2004) Human CD59 is a receptor
for the cholesterol-dependent cytolysin intermedilysin. Nat. Struct. Mol. Biol., 11, 1173-
1178.
Gilbert, R. J. (2005) Inactivation and activity of cholesterol-dependent cytolysins: what
structural studies tell us. Structure (Camb), 13, 1097-1106.
Gilbert, R. J. C., Rossjohn, J., Parker, M. W., Tweten, R. K., Morgan, P. J., Mitchell, T.
J., Errington, N., Rowe, A. J., Andrew, P. W. & Byron, O. (1998) Self-interaction of
pneumolysin, the pore-forming protein toxin of Streptococcus pneumoniae. J. Mol. Biol.,
284, 1223-1237.
Goñi, F. M., Alonso, A., Bagatolli, L. A., Brown, R. E., Marsh, D., Prieto, M. & Thewalt,
J. L. (2008) Phase diagrams of lipid mixtures relevant to the study of membrane rafts.
Biochim. Biophys. Acta, Mol. Cell. Biol. Lipids, 1781, 665-684.
37
Haberland, M. E. & Reynolds, J. A. (1973) Self-association of cholesterol in aqueous
solution. Proc. Natl. Acad. Sci. U. S. A., 70, 2313-2316.
Hadders, M. A., Beringer, D. X. & Gros, P. (2007) Structure of C8 -MACPF reveals
mechanism of membrane attack in complement immune defense. Science, 317, 1552-
1554.
Harris, J. R. (1988) Electron microscopy of cholesterol. Micro Microsc. Acta, 19, 19-31.
Harris, J. R., Adrian, M., Bhakdi, S. & Palmer, M. (1998) Cholesterol-streptolysin O
interaction: An EM study of wild-type and mutant streptolysin O. J. Struct. Biol., 121,
343-355.
Harwood, C. R. & Cranenburgh, R. (2008) Bacillus protein secretion: an unfolding story.
Trends Microbiol., 16, 73-79.
Heimburg, T., Angerstein, B. & Marsh, D. (1999) Binding of peripheral proteins to
mixed lipid membranes: Effect of lipid demixing upon binding. Biophys. J., 76, 2575-
2586.
Heuck, A. P., Hotze, E. M., Tweten, R. K. & Johnson, A. E. (2000) Mechanism of
membrane insertion of a multimeric -barrel protein: perfringolysin O creates a pore
using ordered and coupled conformational changes. Mol. Cell, 6, 1233-1242.
Heuck, A. P. & Johnson, A. E. (2002) Pore-forming protein structure analysis in
membranes using multiple independent fluorescence techniques. Cell Biochem. Biophys.,
36, 89-101.
38
Heuck, A. P. & Johnson, A. E. (2005) Membrane recognition and pore formation by
bacterial pore-forming toxins. IN Tamm, L. K. (Ed.) Protein-Lipid Interactions. From
Membrane Domains to Cellular Networks., pp.165-188, Weinheim, Wiley-VCH.
Heuck, A. P., Savva, C. G., Holzenburg, A. & Johnson, A. E. (2007) Conformational
changes that effect oligomerization and initiate pore formation are triggered throughout
perfringolysin O upon binding to cholesterol. J. Biol. Chem., 282, 22629-22637.
Heuck, A. P., Tweten, R. K. & Johnson, A. E. (2001) beta-Barrel pore-forming toxins:
intriguing dimorphic proteins. Biochemistry, 40, 9065-9073.
Heuck, A. P., Tweten, R. K. & Johnson, A. E. (2003) Assembly and topography of the
prepore complex in cholesterol-dependent cytolysins. J. Biol. Chem., 278, 31218-31225.
Hotze, E. M., Heuck, A. P., Czajkowsky, D. M., Shao, Z., Johnson, A. E. & Tweten, R.
K. (2002) Monomer-monomer interactions drive the prepore to pore conversion of a beta
-barrel-forming cholesterol-dependent cytolysin. J. Biol. Chem., 277, 11597-11605.
Huang, J. & Feigenson, G. W. (1999) A microscopic interaction model of maximum
solubility of cholesterol in lipid bilayers. Biophys. J., 76, 2142-2157.
Hubbell, W. L., Cafiso, D. S. & Altenbach, C. (2000) Identifying conformational changes
with site-directed spin labeling. Nat. Struct. Mol. Biol., 7, 735-739.
Jefferies, J., Nieminen, L., Kirkham, L.-A., Johnston, C., Smith, A. & Mitchell, T. J.
(2007) Identification of a secreted cholesterol-dependent cytolysin (Mitilysin) from
Streptococcus mitis. J. Bacteriol., 189, 627-632.
Johnson, A. E. (2005) Fluorescence approaches for determining protein conformations,
interactions and mechanisms at membranes. Traffic, 6, 1078-1092.
39
Johnson, M. K., Geoffroy, C. & Alouf, J. E. (1980) Binding of cholesterol by sulfhydryl-
activated cytolysins. Infect. Immun., 27, 97-101.
Kleinschmidt, J. H. (2006) Folding kinetics of the outer membrane proteins OmpA and
FomA into phospholipid bilayers. Chem. Phys. Lipids, 141, 30-47.
Lachapelle, S., Tweten, R. K. & Hotze, E. M. (2009) Intermedilysin-receptor interactions
during assembly of the pore complex: assembly intermediates increase host cell
susceptibility to complement-mediated lysis. J. Biol. Chem., 284, 12719-12726.
Lange, Y., Cutler, H. B. & Steck, T. L. (1980) The effect of cholesterol and other
intercalated amphipaths on the contour and stability of the isolated red cell membrane. J.
Biol. Chem., 255, 9331-9337.
Lange, Y. & Steck, T. L. (2008) Cholesterol homeostasis and the escape tendency
(activity) of plasma membrane cholesterol. Prog. Lipid Res., 47, 319-332.
Lange, Y., Ye, J. & Steck, T. L. (2005) Activation of membrane cholesterol by
displacement from phospholipids. J. Biol. Chem., 280, 36126-36131.
Lecuyer, H. & Dervichian, D. G. (1969) Structure of aqueous mixtures of lecithin and
cholesterol. J. Mol. Biol., 45, 39-57.
Madden, J. C., Ruiz, N. & Caparon, M. (2001) Cytolysin-mediated translocation (CMT):
a functional equivalent of type III secretion in gram-positive bacteria. Cell, 104, 143-152.
Marriott, H. M., Mitchell, T. J. & Dockrell, D. H. (2008) Pneumolysin: a double-edged
sword during the host-pathogen interaction. Curr. Mol. Med., 8, 497-509.
40
Mason, P. R., Tulenko, T. N. & Jacob, R. F. (2003) Direct evidence for cholesterol
crystalline domains in biological membranes: role in human pathobiology. Biochim.
Biophys. Acta, 1610, 198-207.
Mcconnell, H. M. & Radhakrishnan, A. (2003) Condensed complexes of cholesterol and
phospholipids. Biochim. Biophys. Acta, 1610, 159-73.
Meehl, M. A. & Caparon, M. G. (2004) Specificity of streptolysin O in cytolysin-
mediated translocation. Mol. Microbiol., 52, 1665-1676.
Mesmin, B. & Maxfield, F. R. (2009) Intracellular sterol dynamics. Biochim. Biophys.
Acta, Mol. Cell. Biol. Lipids, 1791, 636-645.
Michel, E., Reich, K. A., Favier, R., Berche, P. & Cossart, P. (1990) Attenuated mutants
of the intracellular bacterium Listeria monocytogenes obtained by single amino acid
substitutions in listeriolysin O. Mol. Microbiol., 4, 2167-2178.
Miller, C. J., Elliott, J. L. & Collier, R. J. (1999) Anthrax protective antigen: prepore-to-
pore conversion. Biochemistry, 38, 10432-10441.
Mitsui, K., Sekiya, T., Okamura, S., Nozawa, Y. & Hase, J. (1979) Ring formation of
perfringolysin O as revealed by negative stain electron microscopy. Biochim. Biophys.
Acta, 558, 307-313.
Mosser, E. & Rest, R. (2006) The Bacillus anthracis cholesterol-dependent cytolysin,
Anthrolysin O, kills human neutrophils, monocytes and macrophages. BMC Microbiol.,
6, 56.
Mouritsen, O. G. & Zuckermann, M. J. (2004) What's so special about cholesterol?
Lipids, 39, 1101-1113.
41
Murari, R., Murari, M. P. & Baumann, W. J. (1986) Sterol orientations in
phosphatidylcholine liposomes as determined by deuterium NMR. Biochemistry, 25,
1062-1067.
Nagamune, H., Ohkura, K., Sukeno, A., Cowan, G., Mitchell, T. J., Ito, W., Ohnishi, O.,
Hattori, K., Yamato, M., Hirota, K., Miyake, Y., Maeda, T. & Kourai, H. (2004) The
human-specific action of intermedilysin, a homolog of streptolysin O, is dictated by
domain 4 of the protein. Mol. Microbiol., 48, 677-692.
Nagamune, H., Whiley, R. A., Goto, T., Inai, Y., Maeda, T., Hardie, J. M. & Kourai, H.
(2000) Distribution of the intermedilysin gene among the anginosus group streptococci
and correlation between intermedilysin production and deep-seated infection with
Streptococcus intermedius. J. Clin. Microbiol., 38, 220-226.
Nakamura, M., Sekino-Suzuki, N., Mitsui, K.-I. & Ohno-Iwashita, Y. (1998)
Contribution of tryptophan residues to the structural changes in perfringolysin O during
interaction with liposomal membranes. J. Biochem., 123, 1145-1155.
Nakamura, M., Sekino, N., Iwamoto, M. & Ohno-Iwashita, Y. (1995) Interaction of
.theta.-toxin (perfringolysin O), a cholesterol-binding cytolysin, with liposomal
membranes: change in the aromatic side chains upon binding and insertion. Biochemistry,
34, 6513-6520.
Nelson, L. D., Johnson, A. E. & London, E. (2008) How interaction of perfringolysin O
with membranes is controlled by sterol structure, lipid structure, and physiological low
pH: insights into the origin of perfringolysin O-lipid raft interaction J. Biol. Chem., 283,
4632-4642.
42
Nollmann, M., Gilbert, R., Mitchell, T., Sferrazza, M. & Byron, O. (2004) The role of
cholesterol in the activity of pneumolysin, a bacterial protein toxin. Biophys. J., 86, 3141-
3151.
Ohno-Iwashita, Y., Iwamoto, M., Ando, S. & Iwashita, S. (1992) Effect of lipidic factors
on membrane cholesterol topology - mode of binding of -toxin to cholesterol in
liposomes. Biochimica et Biophysica Acta, 1109, 81-90.
Ohno-Iwashita, Y., Iwamoto, M., Mitsui, K.-I., Ando, S. & Iwashita, S. (1991) A
cytolysin, -toxin, preferentially binds to membrane cholesterol surrounded by
phospholipids with 18-carbon hydrocarbon chains in cholesterol-rich region. J. Biochem.,
110, 369-375.
Ohno-Iwashita, Y., Shimada, Y., Waheed, A., Hayashi, M., Inomata, M., Nakamura, M.,
Maruya, M. & Iwashita, M. (2004) Perfringolysin O, a cholesterol-binding cytolysin, as a
probe for lipid rafts. Anaerobe, 125-134.
Ohvo-Rekilä, H., Ramstedt, B., Leppimäki, P. & Peter Slotte, J. (2002) Cholesterol
interactions with phospholipids in membranes. Prog. Lipid Res., 41, 66-97.
Olofsson, A., Hebert, H. & Thelestam, M. (1993) The projection structure of
Perfringolysin O (Clostridium perfringens -toxin). FEBS Lett., 319, 125-127.
Palmer, M. (2004) Cholesterol and the activity of bacterial toxins. FEMS Microbiol. Lett.,
238, 281-289.
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. EMBO J., 17, 1598-1605.
43
Pinkney, M., Beachey, E. & Kehoe, M. (1989) The thiol-activated toxin streptolysin O
does not require a thiol group for cytolytic activity. Infect. Immun., 57, 2553-2558.
Polekhina, G., Feil, S. C., Tang, J., Rossjohn, J., Giddings, K. S., Tweten, R. K. &
Parker, M. W. (2006) Comparative three-dimensional structure of cholesterol-dependent
cytolysins. In Alouf, J. E. & Popoff, M. R. (Eds.) The Comprehensive Sourcebook of
Bacterial Protein Toxins. Third ed., pp. 659-670, Oxford, England, Academic Press.
Polekhina, G., Giddings, K. S., Tweten, R. K. & Parker, M. W. (2005) Insights into the
action of the superfamily of cholesterol-dependent cytolysins from studies of
intermedilysin. Proc. Natl. Acad. Sci. U.S.A., 102, 600-605.
Radhakrishnan, A. & Mcconnell, H. M. (1999) Condensed complexes of cholesterol and
phospholipids. Biophys. J., 77, 1507-1517.
Ramachandran, R., Heuck, A. P., Tweten, R. K. & Johnson, A. E. (2002) Structural
insights into the membrane-anchoring mechanism of a cholesterol-dependent cytolysin.
Nat. Struct. Mol. Biol., 9, 823-827.
Ramachandran, R., Tweten, R. K. & Johnson, A. E. (2004) Membrane-dependent
conformational changes initiate cholesterol-dependent cytolysin oligomerization and
intersubunit beta-strand alignment. Nat. Struct. Mol. Biol., 11, 697-705.
Ramachandran, R., Tweten, R. K. & Johnson, A. E. (2005) The domains of a cholesterol-
dependent cytolysin undergo a major FRET-detected rearrangement during pore
formation. Proc. Natl. Acad. Sci. U.S.A., 102, 7139-7144.
Rosado, C. J., Buckle, A. M., Law, R. H. P., Butcher, R. E., Kan, W.-T., Bird, C. H.,
Ung, K., Browne, K. A., Baran, K., Bashtannyk-Puhalovich, T. A., Faux, N. G., Wong,
W., Porter, C. J., Pike, R. N., Ellisdon, A. M., Pearce, M. C., Bottomley, S. P., Emsley, J.,
44
Smith, A. I., Rossjohn, J., Hartland, E. L., Voskoboinik, I., Trapani, J. A., Bird, P. I.,
Dunstone, M. A. & Whisstock, J. C. (2007) A common fold mediates vertebrate defense
and bacterial attack. Science, 317, 1548-1551.
Rosenqvist, E., Michaelsen, T. E. & Vistnes, A. I. (1980) Effect of streptolysin O and
digitonin on egg lecithin/cholesterol vesicles. Biochim. Biophys. Acta, 600, 91-102.
Rossjohn, J., Feil, S. C., Mckinstry, W. J., Tweten, R. K. & Parker, M. W. (1997)
Structure of a cholesterol-binding, thiol-activated cytolysin and a model of its membrane
form. Cell, 89, 685-692.
Rossjohn, J., Polekhina, G., Feil, S. C., Morton, C. J., Tweten, R. K. & Parker, M. W.
(2007) Structures of perfringolysin O suggest a pathway for activation of cholesterol-
dependent cytolysins. J. Mol. Biol., 367, 1227-1236.
Sankaram, M. B. & Thompson, T. E. (1991) Cholesterol-induced fluid-phase
immiscibility in membranes. Proc. Natl. Acad. Sci. U.S.A., 88, 8686-8690.
Saunders, F. K., Mitchell, T. J., Walker, J. A., Andrew, P. W. & Boulnois, G. J. (1989)
Pneumolysin, the thiol-activated toxin of Streptococcus pneumoniae, does not require a
thiol group for in vitro activity. Infect. Immun., 57, 2547-2552.
Schnupf, P. & Portnoy, D. A. (2007) Listeriolysin O: a phagosome-specific lysin.
Microbes Infect., 9, 1176-1187.
Sekino-Suzuki, N., Nakamura, M., Mitsui, K.-I. & Ohno-Iwashita, Y. (1996)
Contribution of individual tryptophan residues to the structure and activity of -toxin
(perfringolysin O), a cholesterol-binding cytolysin. Eur. J. Biochem., 241, 941-947.
45
Shatursky, O., Heuck, A. P., Shepard, L. A., Rossjohn, J., Parker, M. W., Johnson, A. E.
& Tweten, R. K. (1999) The mechanism of membrane insertion for a cholesterol-
dependent cytolysin: A novel paradigm for pore-forming toxins. Cell, 99, 293-299.
Shepard, L. A., Heuck, A. P., Hamman, B. D., Rossjohn, J., Parker, M. W., Ryan, K. R.,
Johnson, A. E. & Tweten, R. K. (1998) Identification of a membrane-spanning domain of
the thiol-activated pore-forming toxin Clostridium perfringens perfringolysin O: an
alpha-helical to beta-sheet transition identified by fluorescence spectroscopy.
Biochemistry, 37, 14563-14574.
Shepard, L. A., Shatursky, O., Johnson, A. E. & Tweten, R. K. (2000) The mechanism of
pore assembly for a cholesterol-dependent cytolysin: formation of a large prepore
complex precedes the insertion of the transmembrane beta-hairpins. Biochemistry, 39,
10284-10293.
Simossis, V. A., Kleinjung, J. & Heringa, J. (2005) Homology-extended sequence
alignment. Nucl. Acids Res., 33, 816-824.
Solovyova, A. S., Nollmann, M., Mitchell, T. J. & Byron, O. (2004) The solution
structure and oligomerization behavior of two bacterial toxins: pneumolysin and
perfringolysin O. Biophys. J., 87, 540-552.
Soltani, C. E., Hotze, E. M., Johnson, A. E. & Tweten, R. K. (2007a) Specific protein-
membrane contacts are required for prepore and pore assembly by a cholesterol-
dependent cytolysin. J. Biol. Chem., 282, 15709-15716.
Soltani, C. E., Hotze, E. M., Johnson, A. E. & Tweten, R. K. (2007b) Structural elements
of the cholesterol-dependent cytolysins that are responsible for their cholesterol-sensitive
membrane interactions. Proc. Natl. Acad. Sci. U.S.A., 104, 20226-20231.
46
Tamm, L. K., Hong, H. & Liang, B. (2004) Folding and assembly of beta-barrel
membrane proteins. Biochim. Biophys. Acta, 1666, 250-263.
Tilley, S. J., Orlova, E. V., Gilbert, R. J., Andrew, P. W. & Saibil, H. R. (2005) Structural
basis of pore formation by the bacterial toxin pneumolysin. Cell, 121, 247-256.
Tweten, R. K. (2005) Cholesterol-dependent cytolysins, a family of versatile pore-
forming toxins. Infect. Immun., 73, 6199-6209.
Tweten, R. K., Parker, M. W. & Johnson, A. E. (2001) The cholesterol-dependent
cytolysins. Curr. Top. Microbiol. Immunol., 257, 15-33.
Valeva, A., Hellmann, N., Walev, I., Strand, D., Plate, M., Boukhallouk, F., Brack, A.,
Hanada, K., Decker, H. & Bhakdi, S. (2006) Evidence that clustered phosphocholine
head groups serve as sites for binding and assembly of an oligomeric protein pore. J.
Biol. Chem., 281, 26014-26021.
Vazquez-Boland, J. A., Dominguez, L., Rodriguez-Ferri, E. F., Fernandez-Garayzabal, J.
F. & Suarez, G. (1989) Preliminary evidence that different domains are involved in
cytolytic activity and receptor (cholesterol) binding in listeriolysin O, the Listeria
monocytogenes thiol-activated toxin. FEMS Microbiol. Lett., 53, 95-99.
Veatch, S. L. & Keller, S. L. (2002) Organization in lipid membranes containing
cholesterol. Phys. Rev. Lett., 89, 268101.
Waheed, A., Shimada, Y., Heijnen, H. F. G., Nakamura, M., Inomata, M., Hayashi, M.,
Iwashita, S., Slot, J. W. & Ohno-Iwashita, Y. (2001) Selective binding of perfringolysin
O derivative to cholesterol-rich membrane microdomains (rafts). Proc. Natl. Acad. Sci.
U.S.A., 98, 4926-4931.
47
Walker, B., Krishnasastry, M., Zorn, L. & Bayley, H. (1992) Assembly of the oligomeric
membrane pore formed by Staphylococcal alpha-hemolysin examined by truncation
mutagenesis. J. Biol. Chem., 267, 21782-21786.
White, S. H. & Wimley, W. C. (1999) Membrane protein folding and stability: physical
principles. Annu. Rev. Biophys. Biomol. Struct., 28, 319-365.
Zitzer, A., Westover, E. J., Covey, D. F. & Palmer, M. (2003) Differential interaction of
the two cholesterol-dependent, membrane-damaging toxins, streptolysin O and Vibrio
cholerae cytolysin, with enantiomeric cholesterol. FEBS Lett., 553, 229-231.
PHYLUM
Firmicutes
CLASS
Bacilli
ORDER
Bacillales
FAMILY
Bacillaceae
GENUS
Bacillus %Identity %Similarity Length ID
SPECIES
B. anthracis ALO 72 (68) 88 (83) 462 (512) ZP_03017964.1
B. thurigiensis TLO 74 (69) 88 (83) 462 (512) YP_037419
B. cereus CLO 74 (69) 88 (84) 462 (512) YP_002369889.1
B. weihenstephanensis WLO 74 (69) 87 (83) 462 (512) ABY46062
Listeriaceae
Listeria
L. monocytogenes LLO 43 (40) 66 (62) 469 (529) DQ838568.1
L. seeligeri LSO 45 (41) 67 (63) 469 (530) P31830.1
L. ivanovii ILO 46 (43) 66 (62) 469 (528) AAR97343.1
Planococcaceae
Lysinibacillus
L. sphaericus SPH 76 (72) 90 (87) 463 (506) YP_001699692.1
Paenibacillaceae
Paenibacillus
P. alvei ALV 75 (71) 87 (84) 462 (501) P23564
Brevibacillus
B. brevis BVL 73 (69) 88 (84) 464 (511) YP_002770211.1
Lactobacillales
Streptococcaceae
Streptococcus
S. dysgalactiae* SLOe 67 (56) 83 (70) 463 (571) BAD77791
S. pyogenes SLO 67 (56) 83 (70) 463 (571) NP_268546.1
S. canis SLOc 66 (55) 82 (69) 463 (574) Q53957
S. pseudonemoniae PSY 46 (43) 67 (63) 466 (471) ACJ76900
S. pneumoniae PLY 46 (43) 67 (64) 466 (471) ABO21366.1
S. mitis MLY 46 (43) 67 (63) 466 (471) ABK58695
S. suis SLY 41 (40) 65 (63) 465
(497) ABE66337.1
S. intermedius ILY 41 (37) 65 (59) 469 (532) B212797.1
S. mitis (Lectinolysin) LLY 39 (29) 62 (47) 463 (665) BAE72438.1
Clostridia
Clostridiales
Clostridiaceae
Clostridium
C. perfringens PFO 463 (500) NP_561079
C. butyricum BRY 69 (65) 85 (82) 462 (513) ZP_02950902.1
C. tetani TLY 60 (55) 78 (72) 464 (527) NP_782466.1
C. botulinum B BLYb 60 (49) 78 (63) 464 (602) YP_001886995.1
C. botulinum E3 BLYe 60 (48) 77 (60) 464 (602) YP_001921918.1
C. botulinum C BLYc 60 (56) 79 (74) 463 (518) ZP_02620972.1
C. novyi NVL 58 (54) 78 (73) 463 (514) YP_878174.1
Actinobacteria
Actinobacteria
Bifidobacteriales
Bifidobacteriaceae
Gardenella
G. vaginallis VLY 40 (39) 65 (60) 466 (516) EU522488.1
Actinomycetales
Actinomycetaceae
Arcanobacterium
A. pyogenes PLO 41 (38) 60 (56) 469 (534) U84782.2
Perfringolysin O
76 72 74 74 74 75 73 69 60 60 60 60 58 67 66 67 39 46 46 46 41 41 46 45 43 40 41
Sphaericolysin
SPH 90 80 82 82 82 77 80 69 58 57 57 56 54 66 65 65 37 42 42 42 41 37 45 45 43 37 38
Anthrolysin O
ALO 88 93 97 97 97 73 76 65 57 56 56 58 55 62 61 62 37 41 42 42 40 39 44 43 41 41 39
Cereolysin O
CLO 88 93 99 98 98 75 76 67 57 57 57 58 54 63 62 63 37 41 41 41 40 39 44 43 42 40 39
Thuringiensilysin O
TLO 88 93 99 99 98 75 77 66 57 57 57 57 54 63 62 63 37 41 41 42 40 39 44 43 42 40 39
Weihenstephanensilysin
WLO 87 93 99 99 99 75 77 66 56 57 57 57 54 63 62 63 37 41 41 42 40 39 43 43 42 40 39
Alveolysin
ALV 87 89 88 88 88 88 72 65 60 57 58 57 53 64 63 64 39 41 41 41 40 40 44 43 42 40 39
Brevilysin
BVL 88 93 91 91 92 92 87 68 58 57 57 58 57 64 63 64 39 42 42 42 40 39 46 44 42 39 40
Butyriculysin
BRY 85 86 84 84 84 84 81 84 58 55 55 57 56 64 63 63 37 43 43 43 40 39 45 44 42 40 39
Tetanolysin O
TLY 78 79 77 78 77 77 77 76 75 82 81 81 77 55 55 55 42 45 45 45 45 42 50 50 48 43 44
Botulinolysin B
BLYb 77 77 75 76 76 75 75 75 74 93 95 77 73 55 55 55 42 46 46 46 45 43 51 49 48 43 44
Botulinolysin E3
BLYe 77 77 75 75 75 75 75 74 75 93 98 74 72 54 54 54 42 44 45 44 45 42 51 49 49 43 43
Botulinolysin C
BLYc 79 79 78 79 78 78 77 77 77 93 90 90 83 54 54 54 42 46 46 47 46 46 49 49 48 45 43
Novyilysin
NVL 77 78 79 79 79 79 76 77 78 90 88 87 93 53 52 52 42 44 44 45 45 44 50 50 47 44 44
Streptolysin O
SLO 83 81 79 80 80 79 80 81 80 74 73 73 74 75 99 100 39 42 42 42 41 38 43 43 41 39 39
Streptolysin O c
SLOc 82 80 79 79 79 79 79 81 79 74 73 72 74 74 99 99 39 41 41 41 40 37 44 43 42 39 39
Streptolysin O e
SLOe 83 81 79 80 80 79 80 81 80 74 73 73 74 75 100 99 39 41 41 42 41 37 44 43 42 39 39
Lectinolysin
LLY 62 61 61 61 61 61 62 62 63 65 64 64 66 64 64 63 64 51 51 51 46 54 40 41 40 59 41
Pneumolysin
PLY 67 66 65 66 66 65 66 66 66 68 67 67 69 67 65 64 65 73 99 99 51 53 43 43 43 53 41
Mitilysin
MLY 67 66 65 66 66 65 66 66 66 67 67 67 69 67 65 64 65 73 100 100 50 53 44 43 43 53 41
Pseudopneumolysin
PSY 67 66 65 66 66 65 66 66 66 67 67 67 69 67 65 64 65 73 100 100 50 53 44 43 43 53 40
Suilysin
SLY 65 65 66 66 66 65 65 66 66 69 68 68 69 69 65 64 65 68 73 74 74 47 48 45 46 52 45
Intermedilysin
ILY 64 62 64 64 64 64 64 63 64 65 66 66 68 67 62 61 62 77 74 74 74 68 42 42 42 60 42
Ivanolysin
ILO 66 64 65 64 64 64 64 66 66 70 70 70 71 71 65 65 65 64 67 67 67 71 66 79 81 42 43
Seeligeriolysin O
LSO 67 66 65 65 66 65 67 67 65 71 70 71 71 70 66 66 66 64 66 66 66 69 64 91 85 41 43
Listeriolysin O
LLO 66 65 63 64 64 63 65 66 64 72 71 71 71 70 66 66 66 65 67 67 67 69 66 94 95 40 43
Vaginolysin
VLY 63 62 64 64 64 64 63 62 62 64 65 64 66 67 62 61 62 77 73 73 73 69 79 63 62 63 43
Pyolysin
PLO 60 58 58 59 59 59 60 61 61 61 60 60 63 63 60 60 60 62 61 61 61 64 63 63 62 62 60
PFO
CLO
ALO
SPH
PLO
VLY
LLO
LSO
ILO
ILY
SLY
PSY
MLY
PLY
LLY
SLOe
SLOc
SLO
NVL
BLYc
BLYe
BLYb
TLY
BRY
BVL
ALV
WLO
TLO
PFO
I
II III
1
23
4
A
B
β1
β2β3
β4
β5
conserved loops
domain 4
AB
ILY
PFO
ACOOH
L1
L2
L3
undecapeptide
B
PFO H S G A Y V A
Q
FDKTAHYECTGLAWEWWRGTTLYP
ALO H Y G A Y V A
Q
FDKTAHYECTGLAWEWWRGTTLYP
CLO H Y G A Y V A
Q
FDKTAHYECTGLAWEWWRGTTLYP
WLO H Y G A Y V A
Q
FDKTAHYECTGLAWEWWRGTTLYP
TLO H Y G A Y V A
Q
FDKTAHYECTGLAWEWWRGTTLYP
SPH H Y G A Y V A
Q
FDKTAHFECTGLAWEWWRGTTLYP
BLYe H S G A Y V A
Q
FDKTAHFECTGLAWEWWRGTTLYP
BLYb H S G A Y V A
Q
FDKTAHFECTGLAWEWWRGTTLYP
ALV H S G A Y V A
Q
FDRSAHFECTGLAWEWWRGTTLYP
TLY H S G A Y V A
Q
FDRTAHFECTGLAWEWWRGTTLYP
NVL H R G A Y V A K F G R T A H F E C T G L A W E W W R G T T L Y P
BLYc H S G A Y V A
Q
FDRTAHFECTGLAWEWWRGTTLYP
ILO HSGAYVARF DKLAHF ECTGLAWEWWR GTTLYP
LLO H S G G Y V A
Q
FSKLAHFECTGLAWEWWRGTTLYP
PLY H S G A Y V A
Q
YDLTAHFECTGLAWEWWRGTTLYP
MLY H S G A Y V A
Q
YDLTAHFECTGLAWEWWRGTTLYP
PSY H S G A Y V A
Q
YDLTAHFECTGLAWEWWRGTTLYP
SLY H S G A Y V A K Y N L T S H W E C T G L A W E W W R G T T L Y P
SLO H
Q
GAYVA
Q
Y SKTSPF ECTGLAWEWWR GSTLSP
SLOc H
Q
GAYVA
Q
Y SKTSPF ECTGLAWEWWR GSTLSP
SLOe H
Q
GAYVA
Q
Y SKTSPF ECTGLAWEWWR GSTLSP
BVL H Y G W Y V A
Q
FDRTAPFECTGLAWEWWRGTTLNP
LSO H S G G Y V A
Q
FSKLAHFECTGLFWEWWRGTTLYP
BRY H Y G A Y V A
Q
FDKTAHFECTGLSWEWWRGTTLYP
VLY H R G A Y V A R Y Y R T A H F E K T G L V W E P W R G T T L W P
LLY H K G A Y V A R Y N R T S G F E K T G L V W E P W R G T T L N P
ILY H D G A F V A R F N R G A H Y G A T G L A W E P W R G T T L H P
PLO HGGGYVAKF ARTLGF EATGLAWDPWW GTT LNP
L2
398-406
L3
434-439
undecapeptide
458-468
L1
488-493
ALO
L1
L2
undecapeptide
COOH
A
B
C
... It seems that the main task of PLO's cytolytic activity is to obtain free iron and other growth factors, which are crucial for bacterial replication in host cells [25]. In addition, the ability to lyse phagocytic cells protects bacteria from host immune responses. ...
Article
Full-text available
Trueperella pyogenes is an important opportunistic pathogenic bacterium widely distributed in the environment. Pyolysin (PLO) is a primary virulence factor of T. pyogenes and capable of lysing many different cells. PLO is a member of the cholesterol-dependent cytolysin (CDC) family of which the primary structure only presents a low level of homology with other members from 31% to 45%. By deeply studying PLO, we can understand the overall pathogenic mechanism of CDC family proteins. This study established a mouse muscle tissue model infected with recombinant PLO (rPLO) and its single-point mutations, rPLO N139K and rPLO F240A, and explored its mechanism of causing inflammatory damage. The inflammatory injury abilities of rPLO N139K and rPLO F240A are significantly reduced compared to rPLO. This study elaborated on the inflammatory mechanism of PLO by examining its unit point mutations in detail. Our data also provide a theoretical basis and practical significance for future research on toxins and bacteria.
... This allows for comparisons of mitochondrial properties among different cell types, without the plasma membrane permeability barrier. 71,150,154 Unlike human kidney (HEK293) and liver (HepG2) cell lines, mitochondria in permeabilized β cells (INS1E) exhibit a progressive decline in proton leak respiration. 150 This phenomenon does not arise from a malfunction in the respiratory chain but was attributed to substrate unavailability for respiration. ...
Article
Full-text available
Pancreatic β cells play an essential role in the control of systemic glucose homeostasis as they sense blood glucose levels and respond by secreting insulin. Upon stimulating glucose uptake in insulin‐sensitive tissues post‐prandially, this anabolic hormone restores blood glucose levels to pre‐prandial levels. Maintaining physiological glucose levels thus relies on proper β‐cell function. To fulfill this highly specialized nutrient sensor role, β cells have evolved a unique genetic program that shapes its distinct cellular metabolism. In this review, the unique genetic and metabolic features of β cells will be outlined, including their alterations in type 2 diabetes (T2D). β cells selectively express a set of genes in a cell type‐specific manner; for instance, the glucose activating hexokinase IV enzyme or Glucokinase ( GCK ), whereas other genes are selectively “disallowed”, including lactate dehydrogenase A ( LDHA ) and monocarboxylate transporter 1 ( MCT1 ). This selective gene program equips β cells with a unique metabolic apparatus to ensure that nutrient metabolism is coupled to appropriate insulin secretion, thereby avoiding hyperglycemia, as well as life‐threatening hypoglycemia. Unlike most cell types, β cells exhibit specialized bioenergetic features, including supply‐driven rather than demand‐driven metabolism and a high basal mitochondrial proton leak respiration. The understanding of these unique genetically programmed metabolic features and their alterations that lead to β‐cell dysfunction is crucial for a comprehensive understanding of T2D pathophysiology and the development of innovative therapeutic approaches for T2D patients.
... In these studies, the plasma membrane permeabilizer, a recombinant mutant of cholesterol-dependent cytolysin derived from Clostridium perfringens, underwent oligomerization to create pores that specifically penetrated the cytoplasmic membrane. As a result, the permeabilizer facilitated the transportation of solutes and large proteins weighing >200 kDa [41][42][43]. Thus, the cells were permeabilized and treated with pyruvate, a complex I-linked substrate. ...
Article
Full-text available
Simple Summary Glycolysis and oxidative phosphorylation play important roles in the progression and growth of cancers. The development of natural products and their semisynthetic derivatives for cancer treatment is a longstanding focus of our research interests. We developed compounds known as diaminobutoxy-substituted isoflavonoids (DBIs) that effectively stimulated Adenosine 5′ Monophosphate-activated Protein Kinase (AMPK) and suppressed the growth of colorectal cancer cells by specifically targeting mitochondrial complex I. We now report a new DBI analog, namely, DBI-2, with promising properties for cancer treatment. The combination of DBI-2 and BAY-876, a glucose transporter 1 inhibitor, exhibited synergistic effects on colorectal cancer cells. Furthermore, the therapeutic effectiveness of DBI-2 in colorectal cancer cell xenograft mouse models was enhanced by implementing a ketogenic diet, an outcome that indicated this drug/diet combination is a potentially promising combination strategy for cancer therapy. Abstract Cancer cells undergo a significant level of “metabolic reprogramming” or “remodeling” to ensure an adequate supply of ATP and “building blocks” for cell survival and to facilitate accelerated proliferation. Cancer cells preferentially use glycolysis for ATP production (the Warburg effect); however, cancer cells, including colorectal cancer (CRC) cells, also depend on oxidative phosphorylation (OXPHOS) for ATP production, a finding that suggests that both glycolysis and OXPHOS play significant roles in facilitating cancer progression and proliferation. Our prior studies identified a semisynthetic isoflavonoid, DBI-1, that served as an AMPK activator targeting mitochondrial complex I. Furthermore, DBI-1 and a glucose transporter 1 (GLUT1) inhibitor, BAY-876, synergistically inhibited CRC cell growth in vitro and in vivo. We now report a study of the structure–activity relationships (SARs) in the isoflavonoid family in which we identified a new DBI-1 analog, namely, DBI-2, with promising properties. Here, we aimed to explore the antitumor mechanisms of DBIs and to develop new combination strategies by targeting both glycolysis and OXPHOS. We identified DBI-2 as a novel AMPK activator using an AMPK phosphorylation assay as a readout. DBI-2 inhibited mitochondrial complex I in the Seahorse assays. We performed proliferation and Western blotting assays and conducted studies of apoptosis, necrosis, and autophagy to corroborate the synergistic effects of DBI-2 and BAY-876 on CRC cells in vitro. We hypothesized that restricting the carbohydrate uptake with a KD would mimic the effects of GLUT1 inhibitors, and we found that a ketogenic diet significantly enhanced the therapeutic efficacy of DBI-2 in CRC xenograft mouse models, an outcome that suggested a potentially new approach for combination cancer therapy.
... The ΔprsA mutant was significantly attenuated in mice and prone to be cleared in macrophage or whole blood bactericidal environment [28]. PrsA protein of SS2 was non-haemolytic, suggesting that PrsA may damage cellular integrity differently from the pore-forming cholesteroldependent haemolysin as previously described [29]. The present study aimed to investigate PrsA-induced cell death modes, activation signalling pathways, and PrsA structural domains that control cell death. ...
Article
Full-text available
Streptococcus suis type 2 (SS2), a major emerging/re-emerging zoonotic pathogen found in humans and pigs, can cause severe clinical infections, and pose public health issues. Our previous studies recognized peptidyl-prolyl isomerase (PrsA) as a critical virulence factor promoting SS2 pathogenicity. PrsA contributed to cell death and operated as a pro-inflammatory effector. However, the molecular pathways through which PrsA contributes to cell death are poorly understood. Here in this study, we prepared the recombinant PrsA protein and found that pyroptosis and necroptosis were involved in cell death stimulated by PrsA. Specific pyroptosis and necroptosis signalling inhibitors could significantly alleviate the fatal effect. Cleaved caspase-1 and IL-1β in pyroptosis with phosphorylated MLKL proteins in necroptosis pathways, respectively, were activated after PrsA stimulation. Truncated protein fragments of enzymatic PPIase domain (PPI), N-terminal (NP), and C-terminal (PC) domains fused with PPIase, were expressed and purified. PrsA flanking N- or C-terminal but not enzymatic PPIase domain was found to be critical for PrsA function in inducing cell death and inflammation. Additionally, PrsA protein could be anchored on the cell surface to interact with host cells. However, Toll-like receptor 2 (TLR2) was not implicated in cell death and recognition of PrsA. PAMPs of PrsA could not promote TLR2 activation, and no rescued phenotypes of death were shown in cells blocking of TLR2 receptor or signal-transducing adaptor of MyD88. Overall, these data, for the first time, advanced our perspective on PrsA function and elucidated that PrsA-induced cell death requires its flanking N- or C-terminal domain but is dispensable for recognizing TLR2. Further efforts are still needed to explore the precise molecular mechanisms of PrsA-inducing cell death and, therefore, contribution to SS2 pathogenicity.
... Listeriolysin O belongs to the cholesterol-dependent cytolysin (CDC) family (Heuck et al., 2010). The proteins of the CDCs family have four regions. ...
Article
Full-text available
Listeria monocytogenes causes listeriosis, a disease characterized by a high mortality rate (up to 30%). Since the pathogen is highly tolerant to changing conditions (high and low temperature, wide pH range, low availability of nutrients), it is widespread in the environment, e.g., water, soil, or food. L. monocytogenes possess a number of genes that determine its high virulence potential, i.e., genes involved in the intracellular cycle (e.g., prfA , hly , plcA , plcB , inlA , inlB ), response to stress conditions (e.g., sigB , gadA , caspD , clpB , lmo1138 ), biofilm formation (e.g., agr, luxS ), or resistance to disinfectants (e.g., emrELm , bcrABC , mdrL ). Some genes are organized into genomic and pathogenicity islands. The islands LIPI-1 and LIPI-3 contain genes related to the infectious life cycle and survival in the food processing environment, while LGI-1 and LGI-2 potentially ensure survival and durability in the production environment. Researchers constantly have been searching for new genes determining the virulence of L. monocytogenes . Understanding the virulence potential of L. monocytogenes is an important element of public health protection, as highly pathogenic strains may be associated with outbreaks and the severity of listeriosis. This review summarizes the selected aspects of L. monocytogenes genomic and pathogenicity islands, and the importance of whole genome sequencing for epidemiological purposes.
... Filipin is a fluorescent polyene macrolide secreted by Streptomyces filipinens (1,31,32). PFO is a protein J o u r n a l P r e -p r o o f secreted by Clostridium perfringens (33,34). Upon binding to its sterol target, PFO re-orients and oligomerizes within the bilayer where it forms cytolytic pores composed of rings of 30 or more monomers (34)(35)(36). ...
Article
Full-text available
Almost all the cholesterol in cellular membranes is associated with phospholipids in simple stoichiometric complexes. This limits the binding of sterol ligands such as filipin and Perfringolysin O (PFO) to a small fraction of the total. We offer a simple mathematical model that characterizes this complexity. It posits that the cholesterol accessible to ligands has two forms: active cholesterol, which is that not complexed with phospholipids; and extractable cholesterol, that which ligands can capture competitively from the phospholipid complexes. Simulations based on the model match published data for the association of PFO oligomers with liposomes, plasma membranes and the isolated endoplasmic reticulum. The model shows how the binding of a probe greatly underestimates cholesterol abundance when its affinity for the sterol is so weak that it competes poorly with the membrane phospholipids. Two examples are the under-staining of plasma membranes by filipin and the failure of domain D4 of PFO to label their cytoplasmic leaflets. Conversely, the exaggerated staining of endolysosomes suggests that their cholesterol, being uncomplexed, is readily available. The model is also applicable to the association of cholesterol with intrinsic membrane proteins. For example, it supports the hypothesis that the sharp threshold in the regulation of homeostatic ER proteins by cholesterol derives from the cooperativity of their binding to the sterol weakly held by the phospholipid. § Thus, the model explicates the complexity inherent in the binding of ligands like PFO and filipin to the small accessible fraction of membrane cholesterol.
Article
Full-text available
Bacterial protein toxins are secreted by certain bacteria and are responsible for mild to severe diseases in humans and animals. They are among the most potent molecules known, which are active at very low concentrations. Bacterial protein toxins exhibit a wide diversity based on size, structure, and mode of action. Upon recognition of a cell surface receptor (protein, glycoprotein, and glycolipid), they are active either at the cell surface (signal transduction, membrane damage by pore formation, or hydrolysis of membrane compound(s)) or intracellularly. Various bacterial protein toxins have the ability to enter cells, most often using an endocytosis mechanism, and to deliver the effector domain into the cytosol, where it interacts with an intracellular target(s). According to the nature of the intracellular target(s) and type of modification, various cellular effects are induced (cell death, homeostasis modification, cytoskeleton alteration, blockade of exocytosis, etc.). The various modes of action of bacterial protein toxins are illustrated with representative examples. Insights in toxin evolution are discussed.
Article
A novel, selective, practical cholesterol sensing system based on a fusion protein and a solvatochromic molecule was developed to distinguish cholesterol exposed in different phases of the plasma membrane.
Article
CD4+ T cell-mediated immunity against Streptococcus pneumoniae (pneumococcus) can protect against recurrent bacterial colonization and invasive pneumococcal diseases (IPDs). Although such immune responses are common, the pertinent antigens have remained elusive. We identified an immunodominant CD4+ T cell epitope derived from pneumolysin (Ply), a member of the bacterial cholesterol-dependent cytolysins (CDCs). This epitope was broadly immunogenic as a consequence of presentation by the pervasive human leukocyte antigen (HLA) allotypes DPB1∗02 and DPB1∗04 and recognition via architecturally diverse T cell receptors (TCRs). Moreover, the immunogenicity of Ply427-444 was underpinned by core residues in the conserved undecapeptide region (ECTGLAWEWWR), enabling cross-recognition of heterologous bacterial pathogens expressing CDCs. Molecular studies further showed that HLA-DP4-Ply427-441 was engaged similarly by private and public TCRs. Collectively, these findings reveal the mechanistic determinants of near-global immune focusing on a trans-phyla bacterial epitope, which could inform ancillary strategies to combat various life-threatening infectious diseases, including IPDs.
Article
Streptococcus pneumoniae is a major, encapsulated gram‐positive pathogen that causes diseases including community‐acquired pneumonia, meningitis, and sepsis. This pathogen colonizes the nasopharyngeal epithelia asymptomatically but can often migrate to sterile tissues and cause life‐threatening invasive infections (invasive pneumococcal disease: IPD). Although multivalent pneumococcal polysaccharides and conjugate vaccines are available and effective, they also have major shortcomings with respect to the emergence of vaccine‐resistant serotypes. Therefore, alternative therapeutic approaches are needed, and the molecular analysis of host‐pathogen interactions and their applications to pharmaceutical development and clinical practice has recently received increased attention. In this review, we introduce pneumococcal surface virulence factors involved in pathogenicity and highlight recent advances in understanding regarding host autophagy recognition mechanisms against intracellular S. pneumoniae and pneumococcal evasion from autophagy. This article is protected by copyright. All rights reserved.
Article
IntroductionClassification of Bacterial PFTs α-PFTsβ-PFTsA General Mechanism of Pore Formation?Membrane Recognition Recognition of Specific Membrane LipidsRecognition of Membrane-anchored Proteins or CarbohydratesThe Role of Membrane Lipid DomainsOligomerization on the Membrane Surface Oligomerization Triggered by Lipid-induced Conformational ChangesOligomerization Following Proteolytic Activation of ToxinsMembrane Penetration and Pore FormationUnresolved IssuesReferences α-PFTsβ-PFTs Recognition of Specific Membrane LipidsRecognition of Membrane-anchored Proteins or CarbohydratesThe Role of Membrane Lipid Domains Oligomerization Triggered by Lipid-induced Conformational ChangesOligomerization Following Proteolytic Activation of Toxins
Chapter
The cholesterol-dependent cytolysins (CDCs), also called cholesterol-binding cytolysins, constitute a group of 50- to 60-kDa single-chain, pore-forming bacterial protein toxins previously designated "sulfhydryl" (or "thiol-activated)" cytolysins. This group of structurally, antigenically, and functionally related cytolysins constitutes the largest family of bacterial protein toxins produced by Gram-positive bacteria. CDCs are lethal to animals and highly lytic towards eucaryotic cells, including erythrocytes. Their lytic and lethal properties are suppressed by sulfhydryl-group blocking agents and reversibly restored by thiols or other reducing agents. This chapter classifies and lists the general features of this family of cholesterol-dependant cytolysins. Important progress has been made in the past five years with regard to the mechanisms of toxin insertion and oligomerization in target cells in relation to membrane cholesterol and the genetics and structural characteristics of these toxins. However, the role of certain CDCs in disease, particularly in inflammatory processes and probably in sepsis, remains poorly understood. Moreover, the involvement of CDCs in host-signaling pathways still requires further investigation. Finally, the potential use of certain CDCs as potent permeabilization probes in cell biology or as vaccines or novel therapeutical agents remains an important and promising issue.
Article
There is overwhelming evidence that lipid bilayer regions of animal cell membranes are in a liquid state. Quantitative models of these bilayer regions must then be models of liquids. These liquids are highly non-ideal. For example, it has been known for more than 75 years that mixtures of cholesterol and certain phospholipids undergo an area contraction or condensation in lipid monolayers at the air–water interface. In the past 3 years, a thermodynamic model of “condensed complexes” has been proposed to account for this non-ideal behavior. Here we give an overview of the model, its relation to other models, and to modern views of the properties of animal cell membranes.
Article
Negative staining and platinum-carbon shadowing have been used to prepare electron microscope specimens from aqueous colloidal suspensions of cholesterol microscrystals and from crystalline suspensions in methanol and ethanol. Microcrystals prepared by injection of alcoholic solutions of cholesterol into water exhibit angular conformations of varying regularity which contain a number of parallel cholesterol bilayers. The electron optical images of the cholesterol microcrystals, oriented horizontally and ‘on-edge’, obtained by both negative staining and metal shadowing, are in good agreement. Metal shadowing does, however, reveal greater detail within microcrystal clusters than does negative staining, as well as of the bilayer steps at microcrystal edges. The needle-like crystals (from methanol) and plate-like crystals (from ethanol) present considerable difficulties for the negative staining technique, because of their thickness and the consequent depth of the surrounding negative stain. Small crystals are, nevertheless, shown to possess multiple cholesterol bilayers. Platinum-carbon shadowing of cholesterol crystals taken directly from methanol and ethanol provides more satisfactory images than negative staining. The large depth of focus of the transmission electron microscope enables the stacked cholesterol bilayers to be clearly defined at the edges of crystals. The results obtained are discussed in relation to the physicochemical and biological properties of cholesterol, which underlie the fundamental difficulty encountered when fixing and staining cholesterol for thin sectioning, and also the role of cholesterol insolubility in the formation of gallstones.
Article
Perfringolysin O (PFO) is a member of the cholesterol-dependent cytolysin (CDC) family of membrane-penetrating toxins. The CDCs form large homooligomers (estimated to be comprised of up to 50 CDC monomers) that are responsible for generating a large pore in cholesterol-containing membranes of eukaryotic cells. The assembly of the PFO cytolytic complex was examined to determine whether it forms an oligomeric prepore complex on the membrane prior to the insertion of its membrane-spanning beta-sheet. A PFO oligomeric complex was formed on liposomes at both 4 degrees C and 37 degrees C and shown by SDS-agarose gel electrophoresis to be comprised of a large, comparatively homogeneous complex instead of a distribution of oligomer sizes. At low temperature, the processes of oligomerization and membrane insertion could be resolved, and PFO was found to form an oligomer without significant membrane insertion of its beta-hairpins. Furthermore, PFO was found to increase the ion conductivity through a planar bilayer by large and discrete stepwise changes in conductance that are consistent with the insertion of a preassembled pore complex into the bilayer. The combined results of these analyses strongly support the hypothesis that PFO forms a large oligomeric prepore complex on the membrane surface prior to the insertion of its transmembrane beta-sheet.
Article
TO understand the mechanism of membrane lysis by theta-toxin (perfringolysin O) from Clostridium perfringens, a cholesterol-binding, pore-forming cytolysin, we undertook a spectroscopic analysis of the structural changes that occur during the lytic process using lipid vesicles. In particular, the spectra were compared with those obtained using a modified theta-toxin, MC theta, that binds membrane cholesterol without forming oligomeric pores, thus bypassing the oligomerization step. The interaction of theta-toxin liposomes composed of cholesterol and phosphatidylcholine but not with cholesterol-free liposomes caused a remarkable increase in the intensity of the tryptophan fluorescence emission spectra and ellipticity changes in the near- and far-UV CD peaks. A CD peak shift from 292 to 300 nm was specific for theta-toxin, suggesting oligomerization-specific changes occurring around tryptophan residues. Structural changes in the aromatic side chains were detected in the near-UV CD and fluorescence spectra upon MC theta-liposome interaction, although the far-UV CD spectra indicate that the beta-rich secondary structure of MC theta is well-conserved after membrane binding. Quenching of the intrinsic tryptophan fluorescence of MC theta by brominated lecithin/cholesterol liposomes suggests that theta-toxin inserts at least partly into membranes in the absence of oligomerization. These results indicate that regardless of oligomerization, the binding of theta-toxin to cholesterol induces partial membrane insertion and triggers conformational changes accompanied by aromatic side chain rearrangement with retention of secondary structure. The spectral changes depend on the cholesterol/toxin molar ratio and pH, with maxima at pH 5-7, correlating with the optima for binding, suggesting that the cholesterol-induced insertion mechanism is distinct from the acid-induced one.
Article
Perfringolysin O, a bacterial cytolytic toxin, forms unusually large pores in cholesterol-containing membranes by the spontaneous insertion of two of its four domains into the bilayer. By monitoring the kinetics of domain-specific conformational changes and pore formation using fluorescence spectroscopy, the temporal sequence of domain-membrane interactions has been established. One membrane-exposed domain does not penetrate deeply into the bilayer and is not part of the actual pore, but is responsible for membrane recognition. This domain must bind to the membrane before insertion of the other domain into the bilayer is initiated. The two domains are conformationally coupled, even though they are spatially separated. Thus, cytolytic pore formation is accomplished by a novel mechanism of ordered conformational changes and interdomain communication.
Article
The cytolysin Perfringolysin O was applied to lipid layers and the obtained ring-shaped oligomers analyzed by electron microscopy and image processing. The final result shows the periodic repeat of 2.4 nm along the outer rim of the ring. The asymmetric protein unit, corresponding to one monomer, spans the ring from the convex to the concave surface. It shows a clear protein peak close to the outer radius and less density in the middle of the oligomer. The number of monomers in the average ring is 50, and the inner radius of the aggregate is approximately 15 nm.