You are on page 1of 15

Send Orders for Reprints to reprints@benthamscience.

ae
Current Topics in Medicinal Chemistry, 2016, 16, 25-39 25

Antimicrobial Peptide Structure and Mechanism of Action: A Focus on the


Role of Membrane Structure

Tzong-Hsien Lee, Kristopher N. Hall and Marie-Isabel Aguilar*

Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Vic,
3800, Australia

Abstract: Antimicrobial peptides (AMPs) are showing increasing promise as potential candidate anti-
bacterial drugs in the face of the rapidly emerging bacterial resistance to conventional antibiotics in
recent years. The target of these peptides is the microbial membrane and there are numerous models to
explain their mechanism of action ranging from pore formation to general membrane disruption. The
interaction between the AMP and the target membrane is critical to the specificity and activity of these
peptides. However, a precise understanding of the relationship between antimicrobial peptide structure
and their cytolytic function in a range of organisms is still lacking. This is a result of the complex na-
ture of the interactions of AMPs with the cell membrane, the mechanism of which can vary considerably between differ-
ent classes of antimicrobia peptides. A wide range of biophysical techniques have been used to study the influence of a
number of peptide and membrane properties on the cytolytic activity of these peptides in model membrane systems. Cen-
tral to characterisation of this interaction is a quantitative analysis of the binding of peptide to the membrane and the co-
herent dynamic changes in membrane structure. Recently, dual polarization interferometry has been used to perform an in
depth analysis of antimicrobial peptide induced membrane perturbation and with new mass-structure co-fitting kinetic
analysis have allowed a real-time label free analysis of binding affinity and kinetics. We review these studies which
describe multi-step mechanisms which are adopted by various AMPs in nature and may advance our approach to the de-
velopment of a new generation of effective antimicrobial therapeutics.
Keywords: Antimicrobial peptides, dual polarisation interferometry, membrane bilayer anisotropy, membrane structure plastic-
ity, supported lipid bilayer.

1. INTRODUCTION only been characterised for a small portion (about 13%) of


AMPs primarily determined by solution nuclear magnetic
Antibiotics were one of the greatest discoveries of the
resonance (NMR) spectroscopy. Based on the Antimicrobial
20th century and have been used extensively with great suc-
Peptide Database, 13.8% are predicted to be helical, 4% β-
cess to treat microbial infections since their introduction in
strand and 4% mixed. In contrast, the updated Collection of
the 1950s. Unfortunately their effectiveness has diminished
over time due to the steady increase in resistant microbial Antimicrobial Peptide (CAMP) database consists of 682
strains. Factors such as increased selective pressure from the high resolution AMP structures of which 18.7% are helical,
use of antibiotics, increased disease transmission and a de- 18.9% are β-strand, 60.1% are mixed or coil [7]. Thus, a
cline in development of novel antimicrobial therapeutics has high resolution structure is only available for a low propor-
seen the rise of once treatable infections becoming untreat- tion of AMPs with the majority of those being helical. The
able. For example, the emergence of increased resistance to diversity of AMP structures also suggests that the action of
antibiotics such as methicillin and drugs of last resort such as these peptides on the target cells can be complex and beyond
vancomycin of strains such as enterococci, staphylococci (eg the pore mechanisms based on the helical structure which
Staphylococcus aureus) pose a serious global health problem have been widely adopted in the design and development of
and urgent action is required [1-4]. AMPs exhibiting antimicrobial activities. Furthermore, given
that only a small proportion of AMPs are helical in structure,
Antimicrobial peptides (AMPs) are ancient weapons used we know even less about the majority of AMPs and the reli-
by organisms to combat microbial infections and have that ance on pore mechanisms as the sole or dominant mecha-
have remained effective, surviving the slow process of evo- nism may not be valid. Ultimately, this may restrict the de-
lution over time [5, 6]. With more than 5000 sequences char- sign and development of novel antimicrobial agents as it is
acterised to date [7], these peptides show a marked variation becoming increasingly evident that the mechanism of action
in sequence, length and structure. The 3D structures have is far more complex than a defined pore or carpet mecha-
nism.
*Address correspondence to this author at the Department of Biochemistry
Structure activity relationship studies of antimicrobial
and Molecular Biology, Monash University, Wellington Rd, Clayton, Vic.
3800, Australia; Tel: +61-3-9905-3723; Fax: +61-3-9902-9500; peptides have revealed that there are many factors that affect
E-mail: Mibel.Aguilar@monash.edu the specificity and biological activity of these peptides. Fac-

1873-5294/16 $58.00+.00 © 2016 Bentham Science Publishers


26 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

tors such as charge, secondary structure, hydrophobicity, crease in hemolytic activity and a significant drop in mem-
amphipathicity and hydrophobic moment are all critical to brane binding [3, 16, 17].
function and are so interdependent that altering one property Membrane charge density also plays critical role in me-
will often result in significant changes to one or more of the diating the binding and selectivity of cationic AMPs. Parti-
others. What many of these studies have in common is the tion of cationic AMPs into zwitterionic lipids is generally
focus on the structure of the AMP during membrane binding. weak with a high dissociation constant. The presence of
However, the membrane also undergoes substantial changes negatively charged lipids such as phosphatidylglycerol (PG)
in structure but has, to date, remained relatively poorly un- and cardiolipin in microbial membranes mediate an electro-
derstood. In this review, the details of mechanisms by which static interaction with the cationic peptides with a remarka-
AMPs exert their activity are explored with a specific focus bly reduced dissociation rate. However, while the contribu-
on the analysis of membrane structure changes during AMP tion of electrostatic interactions forms the basis of selective
binding: from the initial interaction of AMPs with their peptide binding to the bacterial membrane, this alone cannot
membrane target, conformational changes upon interaction be used to explain the selectivity towards bacteria over host
with the membrane, accumulation of critical concentration cells. As described above, enhanced electrostatic interaction
and self-association, direct mechanisms of action such as is accompanied by a non-linear concentration-dependent cell
pore formation or general disruption to downstream effects toxicity, once a charge threshold is exceeded. In addition,
that lead to cell death. Target selectivity and possible resis- there have been some rare cases where an indirect or inverse
tance is discussed and then finer details of factors that modu- relationship between charge and activity has been observed
late the biological activity of these peptides such as: charge, [9, 18].
secondary structure, hydrophobicity, amphipathicity, hydro-
phobic moment and polar angle are discussed in detail. Fi- 2.2. Secondary Structure
nally, we focus on studies using multi-parameter-based opti-
Various structures have been characterized for AMPs
cal biosensors that provide new information on the structure
which can be generally classified into four major structural
of the membrane and the concept of membrane plasticity
groups: helical peptides, β-strand/sheet peptides, mixed heli-
during AMP binding.
cal/sheet peptides and extended non-helical/sheet peptides.
Most AMPs undergo a conformational transition from flexi-
2. PHYSICOCHEMICAL PARAMETERS THAT ble unstructured in solution to a specific structured or more
MODULATE ANTIMICROBIAL PEPTIDE ACTIVITY rigid conformation upon interacting with a membrane. In the
AND SPECIFICITY case of helical peptides, amino acid substitutions that signifi-
It is well known that the biological activity and specific- cantly disrupt the helical structures can lead to a dramatic
ity of AMPs are determined by numerous physicochemical decrease in activity. For example; studies with proline sub-
parameters of both peptides and membrane lipids [8]. The stitutions with an active rabbit CAP18 fragment resulted in
exact relationships between these factors such as the se- complete loss of activity due to disruption of peptide struc-
quence length, amino acid composition and structural char- ture [19]. Furthermore, many studies have shown that all-D
acteristics of AMPs and the bilayer composition and proper- AMPs exhibit activity that is generally comparable to that of
ties of the target cells for their mechanisms of action are not the parent peptides [20, 21]. However, the introduction of
well understood. A better understanding of the relationship single or double D-amino acid substitutions which disrupt
between these factors is critical to designing novel peptides the secondary structure can have a dramatic effect on peptide
with increased potency and tailored species specificity. activity. An example of this is in a study by Oren and Shai
[22], who showed that single-D substitutions within melittin
partly altered the secondary structure, essentially eliminating
2.1. Charge
hemolysis without having a great effect on the antimicrobial
The majority of antimicrobial peptides characterized to action. Other investigations [23] into linear and cyclic ana-
date carries a net positive charge, ranging from +1 to as logues of melittin and magainin showed that the cyclized
much as +9 and can contain highly defined cationic domains. peptides were less efficient in initial binding compared to the
The initial interaction with the membrane is primarily elec- linear peptides. When similar amounts were eventually
trostatic and there is a strong correlation between cationic bound however, the peptides exhibited similar ability to
charge and biological activity with increasing charge corre- permeabilize the membrane. Interestingly the cyclic melittin
lating with increased activity and also with increased peptide analogue showed increased antibacterial activity with de-
concentration bound to the membrane interface [9-14]. There creased hemolysis whereas the cyclic magainin analogue
is often an optimum charge for activity however; with showed increased hemolytic lysis and decreased antibacterial
greater charge beyond this value resulting in decreased activ- activity. Chen et al [24] have also shown that substituting
ity. For example, a charge dependence was demonstrated for residues of low helical propensity for those of high helical
analogues of magainin 2, with activity increasing with propensity with magainin analogues can increase activity. A
greater charge while hydrophobicity and structure remained series of peptides derived from Australian frogs have pro-
constant. However, any further addition of cationic residues vided an important family of related antimicrobial peptides
beyond a threshold point effectively decreased the peptide which have been analysed in terms of effect of net charge,
selectivity for microbial targets with enhanced hemolysis length, and number of helical turns [25]. In particular, the
[15]. The effect of positive charge on activity has also been membrane binding properties of aurein 1.2, citropin 1.1,
shown for melittin analogues in which removal of the cati- maculatin 1.1 and caerin 1.1 have been studied using a range
onic C-terminal tail of melittin resulted in a five-fold de- of techniques [26, 27]. These studies have further demon-
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 27

strated that conformation has a strong effect on both the ini- phipathic structures with hydrophobic residues down one
tial AMP binding with the membrane as well as the subse- side of the helix and cationic/hydrophilic residues down the
quent disruption process. other side as shown in Fig. (1).
While the degree of helicity can influence peptide activ-
2.3. Hydrophobicity ity [24], it also has been shown that a high degree of helicity
Hydrophobicity is essential for interaction with mem- and amphipathicity with a segregated hydrophobic domain,
branes as it controls the extent to which the peptide can par- correlates with increased hemolysis and toxicity to cells
tition into the membrane layer. AMPs typically contain ap- composed of zwitterionic neutral lipids (36). The hydropho-
proximately 50% hydrophobic residues. Similarly to charge, bic moment was also studied in the role of hydrophobicity of
studies have shown that increasing hydrophobicity to opti- magainin 2 analogues. Increasing hydrophobic moment also
mal percentage can increase activity against microbial cell had little effect on anionic PG liposomes, but had a dramatic
membranes [28]. Further increasing the hydrophobicity be- effect on lipid mixtures of PC/PG (3:1) [30]. The same study
yond this optimum point has been correlated with a loss of also showed an increase in binding. The β -sheet AMPs are
antimicrobial activity and an increase in mammalian cell also usually amphipathic, with a number of β -strands (fre-
toxicity [29]. For example, studies with magainin 2 ana- quently anti-parallel) organized to create both hydrophilic
logues showed that increasing mean hydrophobicity had lit- and hydrophobic surfaces. They are usually stabilized by a
tle effect on purely anionic PG liposomes, but even slight series of disulphide bonds with as many as eight cysteines in
increases in hydrophobicity showed a dramatic increase in some peptides (eg plant defensins [36]) or by cyclization of
ability to lyse zwitterionic/anionic mixtures PC/PG 3:1 [30], the backbone (eg protegrins [37], gramicidin [38] or the cy-
indicating the substantial influence that hydrophobicity has clotides [39]). The polar angle (θp) is closely related to am-
on the membrane interaction. phipathicity and the hydrophobic moment and is defined as
the relative angular proportion of the polar to non-polar resi-
2.4. Amphipathicity, Hydrophobic Moment and Polar due sides of the amphipathic helix. Fig. (2) shows a hypo-
Angle thetical example. If one side of the helix is hydrophobic and
the other side hydrophilic then the polar angle would be 180°
Charge, structure and hydrophobicity have been shown to (Fig. 2a). If one third of the helix is hydrophilic and the two
be important, but it is also the way that these three factors thirds hydrophobic then polar angle would be 120° (Fig. 2b)
come together and form an amphipathic structure that is of- and so on.
ten of greater importance [13, 31-33]. Almost all AMPs form
some kind of amphipathic structure upon interaction with Hp(2-20) is an AMP derived from the N-terminus of
their target membrane. Amphipathicity refers to the relative Helicobacter pylori ribosomal protein L1 (RpL1), and pos-
proportion and topographic location of hydrophilic and hy- sesses broad-spectrum activity against bacteria, fungi and
drophobic domains within the peptide. A quantitative meas- protozoa at low micromole concentration without causing
ure of this is the hydrophobic moment (µH)which is the sum hemolysis [40]. The importance of various structural charac-
of the amino acid vectorial hydrophobicities [34]. Hydro- teristics of Hp(2-20), including sequence, length, conforma-
phobic moment also has a proportional relationship to activ- tion and amphipathicity/ hydrophobicity on the antimicrobial
ity with increasing hydrophobic moment increasing both activity has been assayed via antifungal, antibacterial and
bacterial membrane disruption and hemolytic activity [6, haemolytic tests of a range of peptide analogues [41, 42].
35]. An amphipathic α-helix is one of the most efficient am- One such analogue, HPA3, which has a double

Fig. (1). a) An α-helical representation of an amphipathic structure with charged (red) areas aligning down one side of the helix and hydro-
phobic resides (blue) down the other side. b) Clustering of cationic and hydrophobic residues of several antimicrobial peptides of different
structural classes. Again red residues are positively charged and blue are hydrophobic amino acids. Other amino acids are not shown. Ma-
gainin 2 is depicted in its α-helical form. (b) Is modified from [6].
28 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

Fig. (2). A helical wheel diagram showing two hypothetical amphipathic peptides with different polar angles. Charged hydrophilic residues
are red and hydrophobic residues are blue. a) One side of the helix has hydrophilic residues and the other hydrophobic. This gives a polar
angle of 180˚. b) One third of the helix is hydrophilic and the two thirds hydrophobic, giving a polar angle of 120°.

substitution of Q17W and D19W, showed 4-16 fold in- goes from an aqueous environment to that of a membrane
creased activity against bacteria and 2-4 fold higher potency mimic [48]. With such a stable structure in solution it is
at inhibiting fungal growth [24, 26]. The enhanced antimi- likely that these AMPs will maintain the same stable con-
crobial activity was attributed to the higher overall hydro- formation when they interact with the membrane surface.
phobicity and amphipathicity and longer linear helical struc- These structures are usually amphipathic and this has been
ture as determined by CD and NMR spectroscopy in mem- found to be important for lytic activity [18]. Increasingly, it
brane-mimetic environments [25, 26]. is emerging that antimicrobial activity is not dependent on
specific peptide structure but more on the amino acid com-
3. MECHANISM OF ACTION OF ANTIMICROBIAL position and physicochemical properties. Moreover, the ac-
PEPTIDES tivity can depend more on the balance of electrostatic inter-
actions in the initial steps and hydrophobic interaction in the
The mechanism of AMP action has been extensively later steps during binding-disruption process.
studied on various membrane models with much fewer stud-
ies focussing on the action of AMPs on whole microbial
3.2. Concentration Dependant Threshold
cells with the use of membrane potential sensitive dyes and
fluorescently labelled peptides. These studies have collec- Once the AMPs have targeted and bound to the mem-
tively shown the primary interaction of AMPs with a mem- brane a second stage of membrane interaction requires a cer-
brane and can be divided into two mechanistic classes: tain ‘critical’ concentration in order to proceed [49, 50]. As
membrane disruptive/permeabilizing and non-membrane the concentration increases in the local area the peptides
disruptive/permeabilizing [43]. The proposed models for the have the potential to self-associate and penetrate deeper into
mechanism of action extend from classical pore formation to the membrane core and to exert the peptide’s effect via a
structural rearrangements such as molecular shape or interfa- variety of mechanisms. Factors such as the composition of
cial activity models [43]. Since more than one proposed the lipid head groups and the fluidity of the membrane as
model may be relevant for a specific AMP, it is therefore well as the propensity of the peptide to self-assemble or mul-
important to understand the mechanistic steps that occur se- timerize will influence the determination of such critical
quentially on the membrane leading to disruption. concentration and the subsequent kinetic steps [51].

3.1. Initial Attraction and Interaction 3.3. Self Association and Multimerization
AMPs are usually unstructured in solution and the initial Once the membrane-bound AMPs reached the critical
interaction of AMPs with the membrane is brought about via concentration, some AMPs self-associate or multimerize to
electrostatic attraction between cationic residues within the achieve the final membrane disruption / cell inactivation as
AMP and the negatively charged lipids in the bacterial discussed in the barrel-stave/toroidal pore, aggregation and
membrane target [9, 11, 15, 44]. Lysine and arginine have amyloid models [52, 53]. The AMPs create complex struc-
been shown to interact strongly with phosphate groups in tures through peptide-peptide and peptide membrane interac-
lipid bilayers [45]. This observation, along with the fact that tions associated with specific AMP mechanisms of action.
most bacteria have a strong electrochemical gradient and that The likelihood of the peptide forming these super structures
electrostatic forces are relatively strong over long molecular is dependent on amino acid composition and conformation of
distances likely contributes to the membrane targeting initial the peptide in its monomeric form. Examples of AMPs that
attraction of many AMPs [8]. Once in proximity to the target have been shown to form oligomers that play a role in their
membrane, coil-helix transitions have been characterised for activity are cathelicidin LL-37 [53], alpha-defensin-6 [52]
most AMPs [46, 47] as depicted in Fig. (3a). On the other and plant-derived kalata B2 [54]. The effectiveness of the
hand, β -sheet AMPs are typically much more structured in complex formed depends on the ability of peptides to align
solution. They are stabilized by disulphide bonds or cycliza- hydrophobic and hydrophilic domains toward the membrane
tion of the peptide backbone. The cyclic β-sheet AMP tachy- or adjacent peptide in the complex. In this way they are able
plesin is an example that has little change in structure as it to achieve more as a whole than the individual peptide can
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 29

achieve by itself such as deeper penetration into the mem- ously or interchange sequentially, and that the mode of ac-
brane core. tion can be affected by multiple factors such as the physico-
chemical properties described above, the solution concentra-
tion of peptides, membrane-bound peptide/lipid ratio, com-
position and structure of both peptide and membrane and the
kinetics of binding and structural changes of all interacting
partners. Among all various models (Table 1), the barrel-
stave/toroidal pore models and carpet-like mechanisms have
been extensively applied to the mechanisms of AMPs action
on membrane and are discussed below and depicted sche-
matically in Fig. (3).

Table 1. Proposed models for the membrane interactions of


α -AMPs (adapted from [43]).

Representative
Antibacterial Mechanism
α-AMPs

Non-permeabilizing

1 Translocating via a transient pore Buforin II

2 Translocating via sinking raft δ-lysin

Permeabilizing

1 Barrel-stave pore Almethicin

2 Toroidal pore Magainin 2

3 Huge toroidal pore Lacticin Q

4 Disordered toroidal pore Melittin

5 Aggregation model Magainins

6 Interfacial activity model Magainin 2

7 Chaotic or non-stoichiometric model Magainin 2

Aurein 1.2,
8 Carpet mechanism
Cecropin
Fig. (3). A schematic of the main classical models depicting the
mechanisms of helical AMPs action. (a) peptides are unstructured 9 Detergent model
in solution; (b) initial binding to membrane interface mediated by
10 Membrane discrimination model V13KL
the electrostatic interaction with structural changes in peptides; (c)
once the membrane-bound peptides reached a critical concentration, 11 Shai-Huang-Matsazuki (SHM) model
they proceed through a stochastic cooperative transition in a multi-
step process destabilizing the membrane via either forming (d) 12 Membrane thinning/thickening model LL-37
barrel stave pore; (e) toroidal pore or (f) carpet-like complex with Magainin
extensive surface coverage up to a point resulting in (g) the mem- 13 Charged lipid clustering
analogues
brane disintegration. (e-f) The formation of toroidal pore can also
proceed through a carpet-like process once a critical concentration Synthetic α-
14 Sand in a gearbox model
of extensive surface coverage is reached (h) The overall outcome of AMPs
these membrane-destabilizing processes is leading to cell death. 15 Oxidized phospholipid targeting Temporin L

3.4. Models of Antimicrobial Action 16 Electroporation NK-lysin

Various molecular models have now been used to de- 17 Tilted peptide mechanism Aurein 1.2
scribe the action of AMPs on a membrane, and are listed in
18 Amyloid formation Temporin B
Table 1. These models range from structurally defined
mechanisms such as barrel-stave, toroidal and detergent-like 19 Leaky slit model
carpet models to structurally less defined mechanisms, such
as lipid segregation into domains, interfacial activity and
3.4.1. Pore Formation
formation of non-lamellar phases and the non-permeabilizing
sinking raft model. With such a wide variety of models it is There are two main peptide induced pore formation mod-
possible that multiple modes of action may occur simultane- els. One is the ”barrel stave” mechanism and the other, the
30 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

”toroidal wormhole” mechanism. The barrel-stave model destabilized and no longer can be maintained (Fig. 3f-g). The
was the first to be proposed to account for the single channel collapse of membrane structure into micelles at the final
conductance induced by alamethicin in black lipid mem- stage of the carpet mechanism where the peptides saturate
branes [55]. In this model, the AMP binds to the membrane the membrane surface has also been described as a detergent-
surface through electrostatic attractions where it then under- like model while the carpet-like mechanism is mostly con-
goes a conformational change adopting an amphipathic sidered as the prerequisite step of the toroidal pore forma-
structure. When a critical threshold of peptide concentration tion. Studies have shown that AMPs such as aurein 1.2 [63],
is reached, the individual peptides self-assemble and create a cecropin [36] and indolicidin [64] act in this manner.
larger structure made up of a variable amount of peptides
that inserts more deeply into the membrane forming a ring 3.5. Downstream Biological Effects
like a “barrel” pore (Fig. 3d). “Stave” refers to the individual Irrespective of the mode of action; the downstream effect
spokes within the barrel. In this pore structure, the hydro- of these mechanisms leads to cellular inactivation and death.
phobic domains face outward toward the hydrophobic re- The integrity of the cytoplasmic membrane is of vital impor-
gions of the membrane core and the hydrophilic regions fac- tance in regulating many functions of microbes. An AMP
ing inward toward the aqueous pore. This allows for minimal that undergoes either a pore formation or membrane perme-
exposure of the hydrophilic residues to the hydrophobic tails ablization mechanism above could lead to a loss of one or
of the lipids and the hydrophobic regions of the complex to more functions of the membrane - such as loss of metabolites
avoid the hydrophilic aqueous pore [56, 57]. The “toroidal and ion gradients, membrane depolarization with phosphol-
pore” or “wormhole” mechanism is similar to the “barrel- ipid flip-flopping and a scrambling of the usual distribution
stave” mode of pore formation. This model has been pro- of lipids between the leaflets of the membrane, or loss of
posed primarily from studies of α-helical AMPs such as the respiration - all of which would lead to cellular death [44,
magainins and PGLa [58-60]. The peptides are initially at- 64-69]. This can often happen rapidly and may be only a
tracted to the membrane and undergo similar secondary matter of minutes after exposure [70]. As mentioned previ-
structural changes as the barrel stave model. The helices ously, this is in stark contrast to traditional antibiotics that
formed are initially orientated parallel to the membrane with can take days to be effective. Human defensins have been
the hydrophobic side of the helix binding and displacing the shown to penetrate both the outer and inner membranes of
phospholipid head groups. This causes a breach in the hy- Gram-positive bacteria with cell death attributed to the
drophobic region and induces a positive curvature strain in breaching of the inner membrane [71]. Defensins have also
the membrane. This strain, along with membrane thinning been shown to destabilize the electrostatic gradient of S.
makes the surface more vulnerable to the AMPs by destabi- aureus [72]. Moreover, in E. coli, the inhibition of RNA,
lizing the integrity of the membrane [8]. Once a critical con- DNA and protein synthesis coincided with the membrane
centration of peptide is reached, they self-associate and ori- permeabilization. These assays indicate that permeabilization
ent perpendicular to the membrane surface with the hydro- leading to loss of other downstream cellular function is es-
phobic residues no longer exposed to the lipid head groups sential for bacterial killing [71, 73]
and form the toroidal pore complex as shown in Fig. (3e).
This differs from the barrel stave pore as the peptides still There is also a growing body of evidence that there may
interact with the lipid head groups and are not situated within be additional or complementary mechanisms where mem-
the membrane hydrophobic core. This arrangement is not as brane permeabilization alone is not enough to cause micro-
stable as a barrel stave pore and is therefore more transient. bial death. AMPs have also been linked to the inhibition of
The stability of the pore has been shown to be affected by vital intracellular functions of microbes [71, 74, 75]. For
peptide charge, with a greater number of positive side chains example, Nisin and Pep 5 were shown to liberate hyrolases
causing repulsion and producing pores with shorter half-lives that degrade the cell wall [76]. Thrombin-induced platelet
[3]. Upon disintegration of the pore complex, some AMPs microbicidal proteins are believed to cause inhibition of
have been shown to translocate to the luminal side of model DNA and/or RNA synthesis, with studies showing cell death
membranes [16, 61] and suggests that this mechanism may occurring at least half an hour after membrane permeabiliza-
also be a way in which peptides can access potential intracel- tion of S. aureus [77]. Microcin has also been found to in-
lular targets. hibit DNA replication by targeting DNA gyrase in E. coli.
[78]. Loss of cell viability of C.albicans has also been ob-
3.4.2. Carpet-Like Mechanism served after internalization by the antifungal peptide tenecin-
AMPs do not necessarily have to act via pore formation, 3 without any membrane permeabilization[79]. Pyrrhoco-
and in many cases they are believed to act through non- ricin has been shown to inhibit the ATPase actions of DnaK
specific membrane permeabilization. In a similar way to the and prevent chaperone assisted protein folding [80]. These
other two models described here, the peptides are initially studies suggest that depending on individual factors, AMP-
attracted to specific target membranes through electrostatic induced cell death may occur via several independent or co-
forces. Upon binding there is a conformational change and operative "multi-hit" [81] mechanisms of action. Having
the hydrophobic regions of the peptide may or may not insert multiple and complementary ways of killing a wide variety
into the membrane core. Upon a critical concentration, they of microbes would be useful in suppressing the ability of the
effectively cover the surface of the membrane in a “carpet- individual pathogen to avoid cell death.
like” manner [3, 31, 60, 62]. There is no further quaternary 3.6. Antimicrobial Peptide Selectivity
structure formation and once the critical concentration has
been reached, this causes a change in the membrane energet- If the high potency of AMPs is to be exploited for the
ics and fluidity until eventually the membrane structure is development of novel therapeutics, it must be selective i.e. it
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 31

must show high antimicrobial activity and low host toxicity However, while the different mechanisms described
[82, 83]. AMPs use fundamental differences between pro- above involve changes in the membrane structure, the spe-
karyotic and eukaryotic membranes as a way of selective cific changes in the membrane structural properties and mo-
toxicity. Bacterial membranes are organized with their outer lecular organization of membrane lipids are not often explic-
most membrane layer heavily populated with net negatively itly described in these mechanisms. Moreover, the various
charged acid phospholipids such as PG. This is quite differ- mechanistic actions of AMPs on membrane destabilization
ent to the membranes of animals and plants that consist have been mainly established for helical peptides with physi-
mainly of neutral zwitter ionic phospholipids such as phos- cal properties and structural characteristics discussed in Sec-
phatidylcholine (PC) which decreases the binding strength of tion 2. These well-characterized structural properties may
the cationic AMPs [67, 84]. The negatively charged lipids not be adopted by non-helical AMPs.
that eukaryotic cells possess are usually found more on the Delineating the mechanisms of AMPs action thus re-
inner leaflet side of the membrane facing the cytoplasm, and
quires the dynamic changes in membrane structure to be
as such not exposed to the extracellular space. The presence
characterised quantitatively in response to the peptide bind-
of cholesterol and other sterols that are not present in bacte-
ing and structural changes. There is a significant gap in our
rial membranes are also believed to inhibit AMP activity by
understanding of membrane-mediated interactions particu-
maintaining membrane integrity, and making permeabliza-
larly in view of the role of lipid molecular organisation. This
tion more difficult [84] and these factors may prove useful in is partly due to the limited understanding of the influence of
drug targeting.
structure, molecular organisation and physicochemical prop-
Overall, the interaction of AMPs with a membrane in- erties of biomembranes on the mechanism of AMP action. In
volves structural and topological changes in both the pep- particular, the quantitative analysis of these properties is
tides and the membrane. Complex factors determine the ac- central to characterising the mechanistic steps of AMP bind-
tivities of AMPs in destabilising membrane structure and ing to membrane and will be discussed below.
function and the ability of AMPs to discriminate between
pathogen and host cell membranes. Modifications in the 4. EXPLORING REAL TIME CHANGES IN LIPID
membrane surface charge, packing order and acylation have MOLECULAR ORGANISATION ASSOCIATED
been identified in the AMPs-resistant bacteria [85-87]. Cor- WITH AMP MEMBRANE BINDING USING
relating the changes in membrane packing ordering as a WAVEGUIDE-BASED OPTICAL BIOSENSORS
function of the membrane-bound peptide mass with those
modifications in the membrane properties will provide more In principle, the binding of AMPs to the membrane re-
information for their impact on the interaction of AMPs and sulting in membrane damage involves a degree of change in
dynamic changes in membrane structures and more impor- membrane packing organization of lipid molecules. The
tantly to assist the development of peptide-based therapeu- changes in membrane packing order have been studied by
tics against drug-resistant microbial infection. the incorporation of probes in lipid bilayer or use a probe-
labeled (tagged) lipids with the fluorescence spectroscopy
and NMR [88-90]. However, these changes in the membrane
3.7. Changes in Membrane Structure and Packing – The
Real Issue order must be correlated with the amount of peptide bound to
the membrane to differentiate between the molecular
Even with the present knowledge of the molecular basis changes associated with surface bound and inserted peptides.
of antimicrobial and cytolytic peptide mechanism of action, Label-free optical biosensors have been widely used for real-
the design of new peptides with a narrow spectrum of activ- time qualitative and quantitative measurement of pep-
ity against microbial infection with minimal toxicity to the tide/protein-membrane interactions [91]. While the com-
host cells is still a major challenge. This is partially due to monly used biosensor instruments such as SPR determine
the fact that the target sites of action for both bacterial and mass-only measurements [91, 92], simultaneous membrane
mammalian cells are the membrane lipids, so complete structural information can be obtained by waveguide-based
specificity represents a demanding task to achieve. However, instruments as described below.
the design of peptides having physicochemical properties
that result in optimal therapeutic results has been the goal of 4.1. Simultaneous Measurement of Membrane Anisot-
a number of studies. At the heart of this problem is the need ropy (Birefringence) and Peptide Binding
to measure target membrane properties in terms of structure The first step in characterising the structure of a mem-
and stability during AMP binding. brane is the accurate measurement of the bilayer properties
As membrane destabilization is one of the main actions such as thickness, density and mass. The successful deposi-
for AMPs, the molecular organization of membrane lipids, in tion of defect–free lipid bilayers of various compositions
addition to the membrane surface charge and curvature, play onto the planar sensor chip can be characterised using optical
an important role in the selectivity of AMPs. Therefore, to waveguide-based sensors, such as dual-polarisation interfer-
fully understand the mechanisms of membrane destabiliza- ometry (DPI) [93-96]. In particular, the structural properties
tion by AMPs, it is important to elucidate how membrane of the lipid bilayer in terms of changes in the optical thick-
properties that control and maintain membrane shape, struc- ness and mass can be monitored in real time for lipid bilayer
ture and dynamic stability are affected by the binding of formation. However, a single parameter such as mass or
AMPs. Thus, characterizing the membrane changes associ- thickness alone is not sufficient to represent the dynamic
ated with peptide binding becomes important for understand- changes in the structure of the lipid bilayers. What is now
ing their mechanism of action. possible is the measurement of the disordering of the bilayer,
32 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

that is, the changes in the molecular packing characteristics of mass and structural parameters can also assist in delineat-
of the lipid chain surrounding the membrane-adsorbed pep- ing mechanisms of interaction that are difficult if not impos-
tides. Owing to the liquid crystal properties of lipid mole- sible to observe by other means. The ability to measure bire-
cules self-assembled into uni-axial aligned bilayers, one of fringence is thus the most significant feature of the DPI in-
the unique structural properties of phospholipid bilayers is strument in terms of investigating membrane-mediated
that the ordered orientation of lipid molecules in a membrane events.
creates a unique optical anisotropic property, which shows
Due to the direct action of AMPs on the cell membrane,
differences in refractive index (RI) for two orthogonal po-
the main challenge is to understand the exact interplay be-
larisations, ie, ne and no in a uniaxial optical axis, and shown
tween factors that control AMP structure and binding and
schematically in Fig. (4). The difference between these two
cell membrane organsiation that underpin phospholipid
RIs for a lipid film is defined as the birefringence (∆nf),
where ∆n f = n e − n o . Furthermore, the degree of molecular selectivity and toxicity to cells, particularly in terms of the
order, S, of the uniaxial lipid bilayer as defined by the ratio changes in bilayer structure. The ability to characterise
of the principal polarizabilities of the bilayer to the molecu- both mass and birefringence simultaneously using DPI al-
lar polarizabilities is proportional to the birefringence values lows the analysis of the relationship between the two prop-
[97]. Thus, the birefringence values represent an averaged erties to be measured in the interactions between AMPs and
measurement of lipid molecular orientation order and the lipids (Fig. 4). In the first instance, the real-time changes of
lipid acyl chain packing order. High ∆nf values are obtained the mass response yields information on binding which is
for a fully aligned lipid bilayer with fully saturated acyl analogous to other optical biosensors such as SPR. How-
chains whereas low ∆nf values are determined for those bi- ever, the simultaneous dependence of birefringence and
layers with unsaturated acyl chains assembled in a random mass on time (as depicted in Fig. (4) provides new informa-
and disordered system. A typical bilayer has a birefringence tion on the simultaneous changes in membrane structure
of 0.015-0.025 refractive index units as determined by DPI during the binding event. More significantly, the combina-
[93, 95, 96, 98, 99]. Thus, simultaneous measurement of tion of these plots to give the birefringence vs. mass plots
both mass and birefringence with high sensitivity can there- (as illustrated in Figs. (5 and 6) reveals an enormous
fore characterise very subtle dynamic changes in orientation amount of new information. In particular, a number of tran-
and packing order of lipid molecules as a function of mem- sitions can be described which can be used to evaluate pep-
brane-bound peptide mass. The simultaneous measurement tide behaviour and mechanism of action (discussed below
in Section 5).

Fig. (4). The dual polarisation interferometer (DPI) consists of a dual slab waveguide guiding two orthogonal polarisations light through
high-refractive index sensing and reference waveguide. (A) The two polarization modes, TM and TE, pass through the waveguide forming
two evanescent fields which can effectively result in phase shift for each TM and TE by the molecules binding onto the surface. Such
changes in phase shift are detected as changes in the interference fringe pattern at the far-field. Thus, changes in TM/TE phase shift can be
calculated as the layer thickness and RI for characterizing the lipid bilayer formation and the dynamic impact of peptide binding on the lipid
bilayer structure. Birefringence is a measure of molecular packing order. (A) A fully aligned/ordered bilayer has larger optical birefringence
(∆nf) than (B) a disordered unilamellar supported lipid bilayer which can effect from a peptide insertion. Thus, the changes in the packing,
alignment and degree of order of lipid molecules assembled on the surface can be determined from quantitative analysis of changes in bire-
fringence. (B) The simultaneous measurement of birefringence and mass provides novel method in delineating the mechanisms of the impact
of bound peptides on membrane structure.
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 33

4.2. Lipid Bilayer Perturbation and Disruption by AMPs threshold point in the mass-birefringence plot has been ob-
served for aurein 1.2 binding to E coli lipid extract and
Both the initial binding of an AMP, and any subsequent
DMPE/DMPG [95], HPA3P binding to DMPC and
processes, may affect both the mass and structural ordering
DMPC/DMPG [94], citropin 1.1, maculatin 1.1 and caerin
of the membrane and analysis of mass-birefringence plots,
1.1 binding to DMPC (Fig. 5) [102], and a series of peptides
derived from the phase changes of DPI containing both mass with antimicrobial activity including peptides derived from
and structural axis reveals the changes in membrane ordering
various coagulation factors binding to DOPE/DOPG [104],
that occur during peptide binding. This graphical analysis of
C-terminal peptides from S1 peptidases to DOPE/DOPG
the correlation between binding and structural changes has
[104], novicidin to DOPC/DOPG [105] and KYE28, KYE21
been used to evaluate the accumulated impact of a number of
and NLF binding to DOPE/DOPG [106].
peptides to effect progressive changes in membrane structure
and has greatly assisted in describing the membrane pertur- By comparison, biphasic binding-disorder plots can be
bation process and disruption by AMPs. observed with changes in peptide sequence or changes in
lipid composition and gel-liquid phase. For example, the
The binding of AMPs to a membrane always involves
binding of HPA3 and maculatin 1.1 to a gel-phase DMPC
some increase in mass (at least initially) and may also be
bilayer can be compared where HPA3 displayed a simple
accompanied by changes in birefringence and the depend-
linear plot while maculatin 1.1 displayed a biphasic plot. In
ence of birefringence on the mass of bound peptide can be a particular, for maculatin 1.1, linear behaviour with bilayer
simple linear relationship or can lead to a series of more
disordering [26, 89, 94, 96] was preceded by mass increases
complex binding-disorder profiles as shown in Figs. (5 and
but no bilayer disorder at the point of initial binding, which
6). The simpler plots can range from a horizontal to shallow
was then followed by an abrupt non-linear disordering upon
slope (corresponding to surface binding that has only a small
further increases in mass. This biphasic change (correspond-
effect on birefringence), through to a near vertical decrease
ing to the transition from a surface bound to an inserted state
in birefringence (corresponding to a substantial disruption of above a threshold concentration) was caused by a proline-
the membrane structure), and several other cases that fall
induced helical kink in maculatin 1.1, as analogues in which
between the two extremes [26, 89, 94-96, 100-103].
proline was replaced required much higher concentrations of
The simplest plot of AMP binding to membranes results peptide to achieve this membrane disruption. Moreover, the
in an essentially linear decrease in the bilayer birefringence biphasic mass-birefringence plots were not apparent in the
with increasing peptide mass bound to membrane, as shown fluid state (ie at higher temperatures) suggesting that the
for citropin 1.1 binding to DMPC (Fig. 5) and also seen for kinked peptide structure was able to insert more easily into
HPA binding to DMPC and DMPC/DMPG (80:20) [94, the more fluid bilyaer. In contrast, the proline-substituted
102]. This linear decrease in birefringence immediately after HPA3 analogue, HPA3P, showed a simple linear decrease in
peptide binding is a characteristic pattern for peptides incor- bilayer order with the fluid and the gel-DMPC/DMPG bi-
porated into the membrane without a threshold of coverage layer [96]. These studies clearly demonstrate that the role of
on the membrane surface [94]. This simple profile without a proline in various AMPs in bilayer perturbation can be de-

Fig. (5). Schematic profiling the different effects of Australia tree frog AMPs, aurein1.2, citropin 1.1, maculatin1.1 and caerin1.2, on the
structure of DMPC and DMPC/DMPG bilayers which can be delineated by studying the correlation of bilayer disordering to the peptide
mass bound to the bilayer. The red-dotted arrows in birefringence (∆nf)–mass plots indicate the direction of changes in birefringence with
changes in mass during the association phase. (Adapted from [102])
34 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

lineated by the changes in birefringence which is not possi- and after membrane lysis obtained from DPI showed appar-
ble by mass-only measurements. These distinctively differ- ent differences for these two peptides which were previously
ent features in binding to DMPC/DMPG compared to similar described as acting by the same carpet mechanism. The bire-
binding behaviour with DMPC between HPA3 and macu- fringence-mass plots for magainin2 are shown in Fig. (6A-
latin 1.1 also demonstrate the importance in differentiating D) and can be compared to the data for aurein 1.2 in Fig. (5).
the membrane structural changes from the peptide binding, Firstly, the P:L ratio determined at the point of disruption for
since in the absence of the birefringence data, the main con- an anionic membrane was markedly lower for aurein 1.2
clusion would be that both peptides have similar binding than for magainin 2 (Table 2). Moreover, an alanine-
mechanisms. substituted analogue, Ala8,13,18-mag2 with enhanced bacteri-
cidal activity also showed disruption of the anionic mem-
The birefringence-mass profiles can also provide signifi-
cant new insight that redefines current models of AMP ac- brane at a P:L ratio similar to magainin 2. However, the
Ala8,13,18-mag2 also disrupted the neutral DMPC membrane
tion [95, 100, 102]. In particular, the sequential steps of
[100] at nearly double the P:L ratio. From the high P:L ratio
binding, insertion and bilayer disruption can be examined in
reflecting the saturated surface coverage at the point of
real time at continuous increments of peptide:lipid (P:L) ra-
membrane disruption, a “carpet mechanism” would be sim-
tio. While many studies focus on defining the critical con-
ply attributed to the mechanisms of action of these frog pep-
centration for membrane destruction, characterising the steps
associated with different extents of bilayer perturbation at a tides. However, the drop in birefringence was reversible for
aurein 1.2 after its disruption of both neutral DMPC and ani-
distinctive P:L threshold provides a much clearer under-
onic DMPC/DMPG membranes [95, 100, 102]. In contrast, a
standing of the action of AMPs on the membrane. Aurein 1.2
partially reversible birefringence drop with permanent disor-
and magainin 2 are two examples of naturally isolated AMPs
dering was obtained for Ala8,13,18-mag2 after disrupting the
from the dorsal secretion of an Australian tree frog and an
membranes. Such partially reversible birefringence changes
African frog, respectively. Despite the difference in sequence
and in particular the length, with 13 and 23 amino acids for with membrane disruption are consistent with peptide inser-
tion and bilayer expansion as evident from AFM studies
aurein 1.2 and magainin 2, respectively, both peptides adopt
[100]. Both fluorescence dye release and AFM results also
an amphipathic helix in an anionic membrane environment,
showed that aurein 1.2 and magainin 2 and Ala8,13,18-mag2
while no structure was found for magainin2 in a neutral
disrupt the membrane differently [95, 100, 102]. Overall,
membrane environment. The carpet mechanism-linked de-
these experiments clearly demonstrate that AMP action con-
tergent model has been proposed for aurein 1.2 and a carpet-
induced pore formation for magainin peptides lyse mem- sists of complex sequential intermediates that define the
steps leading to cell death. The specific models describing
branes based on similar critical peptide:lipid (P:L) ratios
the end-point configuration (eg, barrel stave, toroidal, and
measured by conventional means. However, information on
carpet-like) can mislead the interpretation of mechanism
membrane structure changes immediately prior to, during

Fig. (6). Birefringence versus mass plots for the interaction of magainin 2 with (A) DMPC and (B) DMPC/DMPG or Ala8,13,18-mag2 with
(C) DMPC and (D) DMPC/DMPG. (E) Interconversion of membrane structure intermediates as exemplified by the interaction of magainin 2
and the analogue Ala8,13,18-Mag2 with lipid bilayers as a template for antimicrobial peptide action. This model describes alternate pathways
of intermediate membrane states that occur in the presence of increasing amounts of bound peptide (PLX) and the prevalence of each state is
dependent on the peptide and the lipid composition. (Adapted from [100]).
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 35

without details on membrane structural changes. In particu- tion, and the surrounding membrane composition. For exam-
lar, the reversible packing disorder-order behaviour observed ple, as discussed above, antimicrobial peptide action is con-
for different bilayers is quite distinctive for various AMPs, trolled by the nature of the bacterial and mammalian mem-
behaviour which is not accessible by other spectroscopic brane [6].
techniques. The analysis of membrane disordering has there- The data that is now available from techniques such as
fore extended our understanding of how the membrane un-
DPI provides the opportunity to categorise membrane-
dergoes a reversible structural change upon exposure to an-
mediated events in terms of patterns of membrane structure
timicrobial peptides and how the bilayer can either recover
changes. In particular mass-birefringence plots, which reveal
or, at a critical peptide concentration, begins to disintegrate.
the changes in membrane ordering that occur during peptide
binding, now allow very detailed and subtle differences in
Table 2. Molar ratio of peptide to lipid determined at the the interactions between lipids and peptides to be visualised.
point where mass loss and membrane disruption. This birefringence analysis provides novel information on
the impact of peptides on the changes in organisation occur-
ring within a lipid bilayer, and assists in describing the be-
DMPC DMPC/DMPG (4:1) haviour of membrane-active peptides when interacting with a
lipid membrane. Overall a number of transitions can be de-
Aurein 1.2 1:14 1:23 scribed based on the birefringence vs. mass plots (as illus-
Magainin 2 - 1:15 trated in Figs. 5 and 6), which can be used to evaluate pep-
tide behaviour and mechanism of action and are the centre-
8,13,18
Ala -Magainin 2 1:8 1:13 piece of this new membrane biophysical methodology. The
molecular events corresponding to these four transitions are
Together with DPI, the experimental design allows both schematically drawn in Fig. (5) in the context of aurein 1.2,
the acquisition of kinetic data on bilayer formation via vesi- citropin 1.1, maculatin 1.1 and caerin 1.1 [89, 95, 96, 100,
cle adsorption [93] and subsequent analysis of peptide- 102].
membrane interaction, throughout the entire sequence of Based on the multiple stages of membrane structure
events involving peptide binding, insertion and membrane changes evident in Fig. (5), specific models can be defined
destruction. DPI provides simultaneous quantification of real for the molecular mechanism of action in terms of a series of
time changes in the thickness, mass/density and birefrin- intermediate states, each representing an ensemble of
gence of the membrane. While optical birefringence is a closely-related bilayer structures. In most cases, three gen-
well-known property for aligned bilayer membranes [93, 97, eral membrane-inserted states are outlined corresponding to
107-109], changes to these properties during peptide interac- (1) surface-bound peptides (2) partially inserted peptides and
tions have been less studied [94, 108, 110]. Since the bire- (3) significantly inserted peptides. Different degrees of bi-
fringence quantifies the degree of alignment and uniaxial layer disorder reflect the lipid perturbation by each of these
packing of the lipid molecules on the planar surface, changes states. The understanding of structural changes in a mem-
in birefringence as a function of peptide binding to the mem- brane also allow the description of alternate pathways of
brane provide a unique insight into the mechanism of bind- intermediate membrane states that occur in the presence of
ing, and the rate and concentration dependent changes in increasing amounts of bound peptide and the prevalence of
lipid packing and membrane destabilisation. Thus, emphasis each state which is dependent on the peptide and the lipid
can now be placed on examining the geometric and dynamic composition. This mass-structural change model, based on
arrangements within a lipid matrix and the functional role of birefringence changes at critical mass loadings, allows the
lipid molecular ordering in peptide and lipid interactions. properties of membrane-active peptides to be analysed in
Overall, these studies have shown that DPI analysis of AMP sequential steps of surface binding, insertion, membrane
binding data correlates well with previous biosensor [27, 94, opening and bilayer lysis, and in more detail than has been
95, 100] and spectroscopic studies such as NMR[89], neu- previously possible in terms of dynamic structural changes.
tron reflectometry (NR) [26] and AFM imaging [100]and In particular, the molecular events prior to membrane lysis
provides new data on the dynamic, geometric changes in can be fully characterised in relation to the physicochemical
membrane structure leading to a more detailed understanding properties of membrane and provides new criteria that can
of the multiple steps associated with peptide-membrane in- guide the design of selective antibacterial peptides. An im-
teractions. portant element to this endeavour will be the ability to derive
affinity and rate constants for the different states and initial
5. MEMBRANE PLASTICITY: MEMBRANE ORDER- success has been reported for the description of three mem-
ING PROFILES GIVE NEW INSIGHTS INTO MULTI- brane states [101, 112].
STATE KINETIC PROCESSES
Characterizing a series of intermediate states that corre-
Membrane bilayer components alter the physiological spond to different degrees of bilayer disruption in terms of
profiles of a vast number of membrane proteins, either antimicrobial peptide action can also provide a new basis for
through specific interactions with the protein itself or altera- understanding the membrane lysis in terms of membrane
tion of membrane physical properties such as curvature, lat- plasticity and its ability to recover from peptide assault.
eral pressure, and bilayer thickness [51, 111]. For a large Based on the membrane plasticity model of the multiple
number of cases, the available evidence points to an impor- stages of membrane structure changes, the ability of a dam-
tant link between physicochemical properties for AMP ac- aged membrane to recover from the effect of peptide binding
36 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

may also assist in the understanding of mechanisms of mem- cessful. While a majority of studies focus on peptide struc-
brane repair. Significantly, these results also demonstrate ture and how it changes in response to binding to a mem-
that subtle differences in peptide structural properties can brane, it is now clear that the membrane itself undergoes
modulate the mechanism of peptide action on membranes substantial structure change during AMP binding, but the
and provides new approaches towards the design of peptides details and extent of this membrane structure change is very
with selective membrane activity. poorly understood.
A very significant outcome of these studies is therefore This review therefore discusses the recent studies which
the ability to track the changes in membrane structure disor- provide new information on the changes in membrane struc-
der and correlate these changes with the amount of bound ture based on measurements of bilayer birefringence. The
peptide. In terms of the different structural states adopted by simultaneous measurement of this parameter with bound
the bilayer, it has been previously shown that the presence of mass significantly expands the potential of optical biosensors
DMPG in DMPC leads to the formation of an isotropic state to provide new insights into membrane-mediated cellular
and that the negative charges on the membrane surface re- events. Compared to reasonably similar overall profiles ob-
sulted in a greater susceptibility of the bilayer to peptide- tained by SPR, the wide variety of membrane structure
induced disordering [89, 95, 96, 100, 102]. Based on the changes evident by DPI could not be anticipated and has
multiple stages of membrane structure changes such as those opened up exciting opportunities for more detailed under-
evident in Figs. (5 and 6), we propose a new membrane plas- standing of a vast range of membrane–mediated biological
ticity model for antimicrobial peptide action in terms of a events. The examples described above demonstrate how
series of intermediate states as follows and depicted sche- knowledge of the membrane structure changes significantly
matically in Fig. (6E). This model describes alternate path- informs our understanding of a wide range of biological
ways of intermediate membrane states that occur in the pres- processes. So the question is what are the specific structural
ence of increasing amounts of bound peptide (PLX) and the features of each phospholipid bilayer that influence the rela-
prevalence of each state is dependent on the peptide and the tive binding and disruptive properties by each peptide?
lipid composition. For magainin 2 on DMPC, the pathway Moreover, what are the specific physical properties of each
comprises PLA – PLB – PLC - PLA′ states corresponding to bilayer that influence its ability to survive or succumb to a
reversible changes in membrane order, while on peptide assault? The answers to these questions will no
DMPC/DMPG, magainin 2 binding follows the PLA – PLB – doubt emerge as we move forward in the quest to establish
PLC – PLC’ pathway characterised by reversible changes in new AMP design criteria to tackle the on-going crisis of an-
membrane order with a net loss in mass. By comparison, the tibiotic resistance.
binding of Ala8,13,18-Mag2 to DMPC can be described by the
PLA – PLB – PLD – PLE – PLF – PLG pathway and on
CONFLICT OF INTEREST
DMPC/DMPG Ala8,13,18-Mag2 follows the PLA – PLB – PLD
– PLE – PLF – PLG – PLH pathway characterised by the induc- The authors confirm that this article content has no con-
tion of irreversible membrane disorder and is associated with flict of interest.
insertion, pore formation, membrane expansion and mass
loss [100].This model, which is based on birefringence ACKNOWLEDGEMENTS
changes, now allows the properties of membrane-active pep-
tides to be analysed in terms of surface binding, insertion, The financial support of the National Health & Medical
membrane opening and bilayer lysis and in much more detail Research Council (#1044327) and the Australian Research
than has been previously possible. In particular, the molecu- Council (DP1110101866) is gratefully acknowledged.
lar events prior to membrane lysis can be fully characterised
in terms of bilayer structural changes and provides new crite- REFERENCES
ria that can guide the design of selective antibacterial pep- [1] Cohen, M. L. Epidemiology of drug resistance: implications for a
tides. For example, the different pathways are associated post-antimicrobial era. Science,1992, 257, 1050-1055.
with both reversible and irreversible transitions and the abil- [2] Cohen, M. L. Changing patterns of infectious disease. Nature,
ity to differentiate and more importantly to enhance one 2000, 406, 762-767.
[3] Taubes, G. The bacteria fight back. Science, 2008, 321, 356-361.
pathway over the other provides the definition of selectivity [4] Kumarasamy, K. K.; Toleman, M. A.; Walsh, T. R.; Bagaria, J.;
control; ie can a peptide be designed to induce the irreversi- Butt, F.; Balakrishnan, R.; Chaudhary, U.; Doumith, M.; Giske, C.
ble pathway in a target bacterial membrane but induce re- G.; Irfan, S.; Krishnan, P.; Kumar, A. V.; Maharjan, S.; Mushtaq,
versible and recoverable changes in the host cell membrane? S.; Noorie, T.; Paterson, D. L.; Pearson, A.; Perry, C.; Pike, R.;
Rao, B.; Ray, U.; Sarma, J. B.; Sharma, M.; Sheridan, E.; Thiruna-
rayan, M. A.; Turton, J.; Upadhyay, S.; Warner, M.; Welfare, W.;
CONCLUSION Livermore, D. M.; Woodford, N., Emergence of a new antibiotic
resistance mechanism in India, Pakistan, and the UK: a molecular,
Society is currently facing a public health crisis with the biological, and epidemiological study. Lancet Infect Dis., 2010, 10,
increasing resistance to antibiotics. While peptides offer 597-602.
enormous potential as a new class of antibiotics, we have yet [5] Zasloff, M. Antimicrobial peptides in health and disease. N. Engl.
to see an AMP reach the market for internal use. A major J. Med., 2002, 347, 1199-1200.
[6] Zasloff, M., Antimicrobial peptides of multicellular organisms.
issue facing researchers is the difficulty in untangling the Nature, 2002, 415, 389-395.
factors that govern membrane selectivity of AMPs, which in [7] Waghu, F. H.; Gopi, L.; Barai, R. S.; Ramteke, P.; Nizami, B.;
turn prevents a clear definition of the mechanism of action of Idicula-Thomas, S. CAMP: Collection of sequences and structures
these peptides. Without this information, the quest to design of antimicrobial peptides. Nucleic Acids Res., 2014, 42, D1154-
1158.
highly selective non-toxic AMPs will continue to be unsuc-
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 37

[8] Yeaman, M. R.; Yount, N. Y., Mechanisms of antimicrobial pep- timicrobial peptides with improved activity and selectivity against
tide action and resistance. Pharmacol. Rev., 2003, 55, 27-55. Vibrio spp. Int. J. Antimicrob. Agents, 2008, 32, 130-138.
[9] Bessalle, R.; Haas, H.; Goria, A.; Shalit, I.; Fridkin, M., Augmenta- [30] Wieprecht, T.; Dathe, M.; Beyermann, M.; Krause, E.; Maloy, W.
tion of the antibacterial activity of magainin by positive-charge L.; MacDonald, D. L.; Bienert, M. Peptide hydrophobicity controls
chain extension. Antimicrob. Agents Chemother.,1992, 36, 313- the activity and selectivity of magainin 2 amide in interaction with
317. membranes. Biochemistry,1997, 36, 6124-6132.
[10] Dathe, M.; Wieprecht, T.; Nikolenko, H.; Handel, L.; Maloy, W. [31] Shai, Y. Mechanism of the binding, insertion and destabilization of
L.; MacDonald, D. L.; Beyermann, M.; Bienert, M., Hydrophobic- phospholipid bilayer membranes by alpha-helical antimicrobial and
ity, hydrophobic moment and angle subtended by charged residues cell non-selective membrane-lytic peptides. Biochim. Biophys.
modulate antibacterial and haemolytic activity of amphipathic heli- Acta,1999, 1462, 55-70.
cal peptides. FEBS Lett., 1997, 403, 208-212. [32] Resende, J. M.; Verly, R. M.; Aisenbrey, C.; Cesar, A.; Bertani, P.;
[11] Matsuzaki, K.; Nakamura, A.; Murase, O.; Sugishita, K.; Fujii, N.; Pilo-Veloso, D.; Bechinger, B. Membrane interactions of phyl-
Miyajima, K., Modulation of magainin 2-lipid bilayer interactions loseptin-1, -2, and -3 peptides by oriented solid-state NMR spec-
by peptide charge. Biochemistry, 1997, 36, 2104-2111. troscopy. Biophys. J., 2014, 107, 901-911.
[12] Bonucci, A.; Balducci, E.; Martinelli, M.; Pogni, R., Human neu- [33] Zhu, X.; Zhang, L.; Wang, J.; Ma, Z.; Xu, W.; Li, J.; Shan, A.
trophil peptide 1 variants bearing arginine modified cationic side Characterization of antimicrobial activity and mechanisms of low
chains: effects on membrane partitioning. Biophys. Chem., 2014, amphipathic peptides with different alpha-helical propensity. Acta
190-191, 32-40. Biomater., 2015, 18, 155-167.
[13] Fillion, M.; Valois-Paillard, G.; Lorin, A.; Noel, M.; Voyer, N.; [34] Eisenberg, D. Three-dimensional structure of membrane and sur-
Auger, M. Membrane interactions of synthetic peptides with antim- face proteins. Annu. Rev. Biochem.,1984, 53, 595-623.
icrobial potential: effect of electrostatic interactions and amphiphil- [35] Pathak, N.; Salas-Auvert, R.; Ruche, G.; Janna, M. H.; McCarthy,
icity. Probiotics Antimicrob. Proteins, 2015, 7, 66-74. D.; Harrison, R. G. Comparison of the effects of hydrophobicity,
[14] Juba, M. L.; Porter, D. K.; Williams, E. H.; Rodriguez, C. A.; amphiphilicity, and alpha-helicity on the activities of antimicrobial
Barksdale, S. M.; Bishop, B. M., Helical cationic antimicrobial peptides. Proteins, 1995, 22, 182-186.
peptide length and its impact on membrane disruption. Biochim. [36] Sitaram, N.; Nagaraj, R. Interaction of antimicrobial peptides with
Biophys. Acta, 2015, 1848, 1081-1091. biological and model membranes: structural and charge require-
[15] Dathe, M.; Nikolenko, H.; Meyer, J.; Beyermann, M.; Bienert, M., ments for activity. Biochim. Biophys. Acta, 1999, 1462, 29-54.
Optimization of the antimicrobial activity of magainin peptides by [37] Harwig, S. S.; Swiderek, K. M.; Lee, T. D.; Lehrer, R. I. Determi-
modification of charge. FEBS Lett., 2001, 501, 146-150. nation of disulphide bridges in PG-2, an antimicrobial peptide from
[16] Wang, G.; Mishra, B.; Lau, K.; Lushnikova, T.; Golla, R.; Wang, porcine leukocytes. J. Pept. Sci., 1995, 1, 207-215.
X., Antimicrobial peptides in 2014. Pharmaceuticals (Basel),2015, [38] Mou, J.; Czajkowsky, D. M.; Shao, Z. Gramicidin A aggregation in
8, 123-150. supported gel state phosphatidylcholine bilayers. Biochemistry,
[17] Hall, K.; Lee, T. H.; Aguilar, M. I., The role of electrostatic inter- 1996, 35, 3222-3226.
actions in the membrane binding of melittin. J. Mol. Recog., 2011, [39] Craik, D. J.; Daly, N. L.; Bond, T.; Waine, C. Plant cyclotides: A
24, 108-118. unique family of cyclic and knotted proteins that defines the cyclic
[18] Blondelle, S. E.; Houghten, R. A., Design of model amphipathic cystine knot structural motif. J. Mol. Biol., 1999, 294, 1327-1336.
peptides having potent antimicrobial activities. Biochemistry, 1992, [40] Putsep, K.; Branden, C. I.; Boman, H. G.; Normark, S. Antibacte-
31, 12688-12694. rial peptide from H. pylori. Nature,1999, 398, 671-672.
[19] Tossi, A.; Scocchi, M.; Skerlavaj, B.; Gennaro, R. Identification [41] Park, Y.; Hahm, K. S., Effects of N- and C-terminal truncation of
and characterization of a primary antibacterial domain in CAP18, a HP (2-20) from Helicobacter pylori ribosomal protein L1 (RPL1)
lipopolysaccharide binding protein from rabbit leukocytes. FEBS on its anti-microbial activity. Biotechnol. Lett., 2005, 27, 193-199.
Lett., 1994, 339, 108-112. [42] Park, Y.; Park, S. C.; Park, H. K.; Shin, S. Y.; Kim, Y.; Hahm, K.
[20] Bessalle, R.; Kapitkovsky, A.; Gorea, A.; Shalit, I.; Fridkin, M. S. Structure-activity relationship of HP (2-20) analog peptide: en-
All-D-magainin: chirality, antimicrobial activity and proteolytic re- hanced antimicrobial activity by N-terminal random coil region de-
sistance. FEBS Lett., 1990, 274, 151-155. letion. Biopolymers, 2007, 88, 199-207.
[21] Juvvadi, P.; Vunnam, S.; Merrifield, E. L.; Boman, H. G.; Merri- [43] Wang, G. Improved methods for classification, prediction, and
field, R. B. Hydrophobic effects on antibacterial and channel- design of antimicrobial peptides. Methods Mol. Biol., 2015, 1268,
forming properties of cecropin A-melittin hybrids. J. Pept. Sci., 43-66.
1996, 2, 223-232. [44] Melo, M. N.; Ferre, R.; Castanho, M. A. Antimicrobial peptides:
[22] Oren, Z.; Shai, Y. Selective lysis of bacteria but not mammalian linking partition, activity and high membrane-bound concentra-
cells by diastereomers of melittin: structure-function study. Bio- tions. Nat. Rev. Microbiol., 2009, 7, 245-250.
chemistry, 1997, 36, 1826-1835. [45] Mavri, J.; Vogel, H. J. Ion pair formation of phosphorylated amino
[23] Unger, T.; Oren, Z.; Shai, Y. The effect of cyclization of magainin acids and lysine and arginine side chains: a theoretical study. Pro-
2 and melittin analogues on structure, function, and model mem- teins, 1996, 24, 495-501.
brane interactions: implication to their mode of action. Biochemis- [46] Bello, J.; Bello, H. R.; Granados, E. Conformation and aggregation
try, 2001, 40, 6388-6397. of melittin: dependence on pH and concentration. Biochemistry,
[24] Chen, H. C.; Brown, J. H.; Morell, J. L.; Huang, C. M. Synthetic 1982, 21, 461-465.
magainin analogues with improved antimicrobial activity. FEBS [47] Dathe, M.; Wieprecht, T. Structural features of helical antimicro-
Lett., 1988, 236, 462-466. bial peptides: their potential to modulate activity on model mem-
[25] Bowie, J. H.; Separovic, F.; Tyler, M. J. Host-defense peptides of branes and biological cells. Biochim. Biophys. Acta, 1999, 1462,
Australian anurans. Part 2. Structure, activity, mechanism of action, 71-87.
and evolutionary significance. Peptides, 2012, 37, 174-188. [48] Oishi, O.; Yamashita, S.; Nishimoto, E.; Lee, S.; Sugihara, G.;
[26] Fernandez, D. I.; Le Brun, A. P.; Lee, T. H.; Bansal, P.; Aguilar, Ohno, M. Conformations and orientations of aromatic amino acid
M. I.; James, M.; Separovic, F. Structural effects of the antimicro- residues of tachyplesin I in phospholipid membranes. Biochemistry,
bial peptide maculatin 1.1 on supported lipid bilayers. Eur. Bio- 1997, 36, 4352-4359.
phys. J., 2013, 42, 47-59. [49] Huang, H. W. Molecular mechanism of antimicrobial peptides: the
[27] Gehman, J. D.; Luc, F.; Hall, K.; Lee, T. H.; Boland, M. P.; Pukala, origin of cooperativity. Biochim. Biophys. Acta, 2006, 1758, 1292-
T. L.; Bowie, J. H.; Aguilar, M. I.; Separovic, F. Effect of antimi- 1302.
crobial peptides from Australian tree frogs on anionic phospholipid [50] Hall, K.; Lee, T.-H.; Daly, N. L.; Craik, D. J.; Aguilar, M.-I.,
membranes. Biochemistry, 2008, 47, 8557-8565. Gly6 of kalata B1 is critical for the selective binding to phosphati-
[28] Chen, Y.; Guarnieri, M. T.; Vasil, A. I.; Vasil, M. L.; Mant, C. T.; dylethanolamine membranes. Biochim. Biophys. Acta, 2012, 1818,
Hodges, R. S. Role of peptide hydrophobicity in the mechanism of 2354-2361.
action of alpha-helical antimicrobial peptides. Antimicrob. Agents [51] Holthuis, J. C.; Menon, A. K. Lipid landscapes and pipelines in
Chemother., 2007, 51, 1398-1406. membrane homeostasis. Nature, 2014, 510, 48-57.
[29] Chou, H. T.; Kuo, T. Y.; Chiang, J. C.; Pei, M. J.; Yang, W. T.; Yu,
H. C.; Lin, S. B.; Chen, W. J. Design and synthesis of cationic an-
38 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 Lee et al.

[52] Chairatana, P.; Nolan, E. M. Molecular basis for self-assembly of a [75] Sharma, S.; Verma, I.; Khuller, G. K. Biochemical interaction of
human host-defense peptide that entraps bacterial pathogens. J. Am. human neutrophil peptide-1 with Mycobacterium tuberculosis
Chem. Soc., 2014, 136, 13267-13276. H37Ra. Arch Microbiol., 1999, 171, 338-342.
[53] Xhindoli, D.; Pacor, S.; Guida, F.; Antcheva, N.; Tossi, A. Native [76] Bierbaum, G.; Sahl, H. G. Induction of autolysis of staphylococci
oligomerization determines the mode of action and biological ac- by the basic peptide antibiotics Pep 5 and nisin and their influence
tivities of human cathelicidin LL-37. Biochem. J., 2014, 457, 263- on the activity of autolytic enzymes. Arch Microbiol., 1985, 141,
275. 249-254.
[54] Rosengren, K. J.; Daly, N. L.; Harvey, P. J.; Craik, D. J. The self- [77] Xiong, Y. Q.; Bayer, A. S.; Yeaman, M. R. Inhibition of intracellu-
association of the cyclotide kalata B2 in solution is guided by hy- lar macromolecular synthesis in Staphylococcus aureus by throm-
drophobic interactions. Biopolymers, 2013, 100, 453-460. bin-induced platelet microbicidal proteins. J. Infect. Dis., 2002,
[55] Baumann, G.; Mueller, P. A molecular model of membrane excit- 185, 348-356.
ability. J. Supramol. Struct., 1974, 2, 538-557. [78] del Castillo, F. J.; del Castillo, I.; Moreno, F. Construction and
[56] Breukink, E.; de Kruijff, B. The lantibiotic nisin, a special case or characterization of mutations at codon 751 of the Escherichia coli
not? Biochim. Biophys. Acta,1999, 1462, 223-234. gyrB gene that confer resistance to the antimicrobial peptide mi-
[57] Ehrenstein, G.; Lecar, H. Electrically gated ionic channels in lipid crocin B17 and alter the activity of DNA gyrase. J. Bacteriol.,
bilayers. Q. Rev. Biophys., 1977, 10, 1-34. 2001, 183, 2137-2140.
[58] Hara, T.; Kodama, H.; Kondo, M.; Wakamatsu, K.; Takeda, A.; [79] Kim, D. H.; Lee, D. G.; Kim, K. L.; Lee, Y. Internalization of
Tachi, T.; Matsuzaki, K. Effects of peptide dimerization on pore tenecin 3 by a fungal cellular process is essential for its fungicidal
formation: Antiparallel disulfide-dimerized magainin 2 analogue. effect on Candida albicans. Eur. J. Biochem., 2001, 268, 4449-
Biopolymers, 2001, 58, 437-446. 4458.
[59] Hara, T.; Mitani, Y.; Tanaka, K.; Uematsu, N.; Takakura, A.; Ta- [80] Kragol, G.; Lovas, S.; Varadi, G.; Condie, B. A.; Hoffmann, R.;
chi, T.; Kodama, H.; Kondo, M.; Mori, H.; Otaka, A.; Nobutaka, Otvos, L., Jr. The antibacterial peptide pyrrhocoricin inhibits the
F.; Matsuzaki, K. Heterodimer formation between the antimicrobial ATPase actions of DnaK and prevents chaperone-assisted protein
peptides magainin 2 and PGLa in lipid bilayers: a cross-linking folding. Biochemistry, 2001, 40, 3016-3026.
study. Biochemistry, 2001, 40, 12395-12399. [81] Zhang, L.; Dhillon, P.; Yan, H.; Farmer, S.; Hancock, R. E. Interac-
[60] Matsuzaki, K. Magainins as paradigm for the mode of action of tions of bacterial cationic peptide antibiotics with outer and cyto-
pore forming polypeptides. Biochim. Biophys. Acta, 1998, 1376, plasmic membranes of Pseudomonas aeruginosa. Antimicrob.
391-400. Agents Chemother., 2000, 44, 3317-3321.
[61] Yang, L.; Weiss, T. M.; Lehrer, R. I.; Huang, H. W. Crystallization [82] Fernandez, D. I.; Gehman, J. D.; Separovic, F., Membrane interac-
of antimicrobial pores in membranes: magainin and protegrin. Bio- tions of antimicrobial peptides from Australian frogs. Biochim.
phys. J., 2000, 79, 2002-2009. Biophys. Acta, 2009, 1788, 1630-1638.
[62] Shai, Y.; Oren, Z. From "carpet" mechanism to de-novo designed [83] Matsuzaki, K. Control of cell selectivity of antimicrobial peptides.
diastereomeric cell-selective antimicrobial peptides. Peptides, Biochim. Biophys. Acta, 2009, 1788, 1687-1692.
2001, 22, 1629-1641. [84] Matsuzaki, K.; Sugishita, K.; Fujii, N.; Miyajima, K. Molecular
[63] Fernandez, D. I.; Le Brun, A. P.; Whitwell, T. C.; Sani, M. A.; basis for membrane selectivity of an antimicrobial peptide, ma-
James, M.; Separovic, F. The antimicrobial peptide aurein 1.2 dis- gainin 2. Biochemistry, 1995, 34, 3423-3429.
rupts model membranes via the carpet mechanism. Phys. Chem. [85] Bauer, M. E.; Shafer, W. M. On the in vivo significance of bacterial
Chem. Phys., 2012, 14, 15739-15751. resistance to antimicrobial peptides. Biochim. Biophys. Acta, 2015.
[64] Rozek, A.; Friedrich, C. L.; Hancock, R. E., Structure of the bovine [86] Joo, H. S.; Otto, M. Mechanisms of resistance to antimicrobial
antimicrobial peptide indolicidin bound to dodecylphosphocholine peptides in staphylococci. Biochim. Biophys. Acta, 2015.
and sodium dodecyl sulfate micelles. Biochemistry, 2000, 39, [87] LaRock, C. N.; Nizet, V. Cationic antimicrobial peptide resistance
15765-15774. mechanisms of streptococcal pathogens. Biochim. Biophys.
[65] Hancock, R. E.; Chapple, D. S. Peptide antibiotics. Antimicrob. Acta,2015.
Agents Chemother., 1999, 43, 1317-1323. [88] Bagatolli, L. A. To see or not to see: lateral organization of bio-
[66] Matsuzaki, K.; Murase, O.; Fujii, N.; Miyajima, K. An antimicro- logical membranes and fluorescence microscopy. Biochim. Bio-
bial peptide, magainin 2, induced rapid flip-flop of phospholipids phys. Acta, 2006, 1758, 1541-1556.
coupled with pore formation and peptide translocation. Biochemis- [89] Fernandez, D. I.; Lee, T. H.; Sani, M. A.; Aguilar, M. I.; Separovic,
try, 1996, 35, 11361-11368. F. Proline facilitates membrane insertion of the antimicrobial pep-
[67] Vaara, M. Agents that increase the permeability of the outer mem- tide maculatin 1.1 via surface indentation and subsequent lipid dis-
brane. Microbiol. Rev., 1992, 56, 395-411. ordering. Biophys. J., 2013, 104, 1495-1507.
[68] Westerhoff, H. V.; Juretic, D.; Hendler, R. W.; Zasloff, M. Ma- [90] Owen, D. M.; Rentero, C.; Magenau, A.; Abu-Siniyeh, A.; Gaus,
gainins and the disruption of membrane-linked free-energy trans- K. Quantitative imaging of membrane lipid order in cells and or-
duction. Proc. Natl. Acad. Sci. U. S. A., 1989, 86, 6597-6601. ganisms. Nat. Protoc., 2012, 7, 24-35.
[69] Wu, M.; Maier, E.; Benz, R.; Hancock, R. E. Mechanism of inter- [91] Mozsolits, H.; Aguilar, M. I. Surface plasmon resonance spectros-
action of different classes of cationic antimicrobial peptides with copy: an emerging tool for the study of peptide-membrane interac-
planar bilayers and with the cytoplasmic membrane of Escherichia tions. Biopolymers, 2002, 66, 3-18.
coli. Biochemistry,1999, 38, 7235-7242. [92] Mozsolits, H.; Wirth, H. J.; Werkmeister, J.; Aguilar, M. I. Analy-
[70] Tossi, A.; Tarantino, C.; Romeo, D. Design of synthetic antimicro- sis of antimicrobial peptide interactions with hybrid bilayer mem-
bial peptides based on sequence analogy and amphipathicity. Eur. brane systems using surface plasmon resonance. Biochim. Biophys.
J. Biochem., 1997, 250, 549-558. Acta, 2001, 1512, 64-76.
[71] Lehrer, R. I.; Barton, A.; Daher, K. A.; Harwig, S. S.; Ganz, T.; [93] Mashaghi, A.; Swann, M.; Popplewell, J.; Textor, M.; Reimhult, E.
Selsted, M. E. Interaction of human defensins with Escherichia Optical anisotropy of supported lipid structures probed by
coli. Mechanism of bactericidal activity. J. Clin. Invest.,1989, 84, waveguide spectroscopy and its application to study of supported
553-561. lipid bilayer formation kinetics. Anal Chem., 2008, 80, 3666-3676.
[72] Yeaman, M. R.; Bayer, A. S.; Koo, S. P.; Foss, W.; Sullam, P. M. [94] Lee, T. H.; Hall, K. N.; Swann, M. J.; Popplewell, J. F.; Unabia, S.;
Platelet microbicidal proteins and neutrophil defensin disrupt the Park, Y.; Hahm, K. S.; Aguilar, M. I. The membrane insertion of
Staphylococcus aureus cytoplasmic membrane by distinct mecha- helical antimicrobial peptides from the N-terminus of Helicobacter
nisms of action. J. Clin. Invest.,1998, 101, 178-187. pylori ribosomal protein L1. Biochim. Biophys. Acta, 2010, 1798,
[73] Suarez-Carmona, M.; Hubert, P.; Delvenne, P.; Herfs, M. Defens- 544-557.
ins: "Simple" antimicrobial peptides or broad-spectrum molecules? [95] Lee, T. H.; Heng, C.; Swann, M. J.; Gehman, J. D.; Separovic, F.;
Cytokine Growth Factor Rev.,2015, 26, 361-370. Aguilar, M. I. Real-time quantitative analysis of lipid disordering
[74] Park, C. B.; Kim, H. S.; Kim, S. C. Mechanism of action of the by aurein 1.2 during membrane adsorption, destabilisation and ly-
antimicrobial peptide buforin II: buforin II kills microorganisms by sis. Biochim. Biophys. Acta, 2010, 1798, 1977-1986.
penetrating the cell membrane and inhibiting cellular functions. [96] Hirst, D. J.; Lee, T. H.; Swann, M. J.; Unabia, S.; Park, Y.; Hahm,
Biochem. Biophys. Res. Commun., 1998, 244, 253-257. K. S.; Aguilar, M. I. Effect of acyl chain structure and bilayer
Antimicrobial Peptide Structure and Mechanism of Action Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 1 39

phase state on binding and penetration of a supported lipid bilayer [104] Singh, S.; Kasetty, G.; Schmidtchen, A.; Malmsten, M. Membrane
by HPA3. Eur. Biophys. J., 2011, 40, 503-514. and lipopolysaccharide interactions of C-terminal peptides from S1
[97] Salamon, Z.; Tollin, G. Optical anisotropy in lipid bilayer mem- peptidases. Biochim. Biophys. Acta, 2012, 1818, 2244-2251.
branes: coupled plasmon-waveguide resonance measurements of [105] Nielsen, S. B.; Otzen, D. E., Impact of the antimicrobial peptide
molecular orientation, polarizability, and shape. Biophys. J., 2001, Novicidin on membrane structure and integrity. J. Colloid Interface
80, 1557-1567. Sci., 2010, 345, 248-256.
[98] Ouberai, M. M.; Wang, J.; Swann, M. J.; Galvagnion, C.; Guil- [106] Singh, S.; Papareddy, P.; Kalle, M.; Schmidtchen, A.; Malmsten,
liams, T.; Dobson, C. M.; Welland, M. E., alpha-Synuclein senses M., Importance of lipopolysaccharide aggregate disruption for the
lipid packing defects and induces lateral expansion of lipids leading anti-endotoxic effects of heparin cofactor II peptides. Biochim.
to membrane remodeling. J. Biol. Chem., 2013, 288, 20883-20895. Biophys. Acta, 2013, 1828, 2709-2719.
[99] Singh, S.; Kalle, M.; Papareddy, P.; Schmidtchen, A.; Malmsten, [107] Ramsden, J. J., Molecular orientation in lipid bilayers. Phil. Mag.
M., Lipopolysaccharide interactions of C-terminal peptides from B., 1999, 79, 381-386.
human thrombin. Biomacromolecules, 2013, 14, 1482-1492. [108] Mishima, K., Birefringence studies on effects of additives on bi-
[100] Hall, K.; Lee, T. H.; Mechler, A. I.; Swann, M. J.; Aguilar, M. I., layer lipid membranes. 2006; Vol. 3, pp 55-83.
Real-time measurement of membrane conformational states in- [109] Den Engelsen, D. Optical anisotropy in ordered systems of lipids.
duced by antimicrobial peptides: balance between recovery and ly- Surf. Sci., 1976, 56, 272-280.
sis. Sci. Rep., 2014, 4, 5479. [110] Salamon, Z.; Lindblom, G.; Tollin, G. Plasmon-waveguide reso-
[101] Hirst, D. J.; Lee, T. H.; Swann, M. J.; Aguilar, M. I. Combined nance and impedance spectroscopy studies of the interaction be-
mass and structural kinetic analysis of multistate antimicrobial pep- tween penetratin and supported lipid bilayer membranes. Biophys.
tide-membrane interactions. Anal Chem., 2013, 85, 9296-9304. J., 2003, 84, 1796-1807.
[102] Lee, T. H.; Heng, C.; Separovic, F.; Aguilar, M. I. Comparison of [111] De Kroon, A. I.; Rijken, P. J.; De Smet, C. H. Checks and balances
reversible membrane destabilisation induced by antimicrobial pep- in membrane phospholipid class and acyl chain homeostasis, the
tides derived from Australian frogs. Biochim. Biophys. Acta, 2014, yeast perspective. Prog. Lipid Res., 2013, 52, 374-394.
1838, 2205-2215. [112] Hirst, D. J.; Lee, T. H.; Pattenden, L. K.; Thomas, W. G.; Aguilar,
[103] Lee, T. H.; Hirst, D. J.; Aguilar, M. I. New insights into the mo- M. I. Helix 8 of the angiotensin II type 1a receptor interacts with
lecular mechanisms of biomembrane structural changes and inter- phosphatidylinositol phosphates and modulates membrane inser-
actions by optical biosensor technology. Biochim. Biophys. Acta, tion. Sci. Rep., 2015.
2015.

Received: March 31, 2015 Revised: June 03, 2015 Accepted: June 08, 2015

You might also like