Você está na página 1de 9

Review

The expanding scope of antimicrobial


peptide structures and their modes
of action
Leonard T. Nguyen, Evan F. Haney and Hans J. Vogel
Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada, T2N 1N4

Antimicrobial peptides (AMPs) are an integral part of the analogs for structure–function studies. De novo peptide
innate immune system that protect a host from invading design approaches have also been used, including high-
pathogenic bacteria. To help overcome the problem of throughput combinatorial library screening, structure-
antimicrobial resistance, cationic AMPs are currently be- based modeling, predictive algorithms and the introduc-
ing considered as potential alternatives for antibiotics. tion of non-coded modifications to conventional peptide
Although extremely variable in length, amino acid com- chemistry [4,5]. Furthermore, AMPs are being discovered
position and secondary structure, all peptides can adopt a through sequence searches for potential antimicrobial
distinct membrane-bound amphipathic conformation. fragments within large proteins. In fact, several proteins
Recent studies demonstrate that they achieve their anti- have now been shown to exert direct antimicrobial activity
microbial activity by disrupting various key cellular pro- themselves, further widening the scope of the structural
cesses. Some peptides can even use multiple frameworks for AMPs or antimicrobial proteins.
mechanisms. Moreover, several intact proteins or protein Generally, two physical features are common for AMPs: a
fragments are now being shown to have inherent antimi- cationic charge and a significant proportion of hydrophobic
crobial activity. A better understanding of the structure– residues. The former property promotes selectivity for neg-
activity relationships of AMPs is required to facilitate the atively charged microbial cytoplasmic membranes over
rational design of novel antimicrobial agents. zwitterionic mammalian membranes whereas the latter
facilitates interactions with the fatty acyl chains [6]. There
Introduction are also a few anionic AMPs, such as dermcidin, although
The emergence of multidrug-resistant ‘superbug’ bacteria other biological activities seem to be more important for
has created an urgent need for the development of novel these peptides [7]. Many linear AMPs are unstructured in
classes of antimicrobials. Unfortunately, the number of aqueous solution and require a membranous environment
new antibiotics in the pipeline of the big pharmaceutical to adopt a stable amphipathic conformation [8]. Although
companies has been declining because they have shifted the mode of action usually involves disrupting the integrity
their attention towards more lucrative areas of drug de- of the bacterial cytoplasmic membrane in many ways
velopment [1]. Since the initial discovery of antimicrobial (Figure 1), other antimicrobial mechanisms have now been
peptides (AMPs) in insects and animals in the 1980s, they characterized that target key cellular processes including
have been heralded as a promising alternative to today’s DNA and protein synthesis, protein folding, enzymatic ac-
antibiotics [2]. Ubiquitous in nature, AMPs form a key tivity and cell wall synthesis [9–11]. A comprehensive list of
component of an organism’s innate immune system. AMPs mechanisms utilized by AMPs is presented in Table 1.
typically have a broad spectrum of activity against patho- Membrane interactions remain important even for intracel-
genic bacteria and fungi, and often, their killing activities lular-targeting peptides because they must have a means of
extend to enveloped viruses, parasites and sometimes even translocation. The cationicity of the AMPs also promotes
to cancerous cells. Many eukaryotic peptides act on bacte- interactions with negatively charged moieties on other bio-
rial membranes or other generalized targets, in contrast to molecules such as outer membrane lipids, nucleic acids and
most antibiotics, which usually target specific proteins. phosphorylated proteins. During an infection, a host animal
This creates an advantage for AMPs because development may release multiple isoforms or structurally similar AMPs
of microbial resistance by gene mutation is less likely [3]. that act by distinct mechanisms to achieve an overall syn-
AMPs obtained from higher eukaryote organisms are also ergistic effect [12]. Furthermore, for some of the better
frequently referred to as ‘host defense peptides’, a term studied AMPs, such as magainin and indolicidin, it has
that emphasizes their additional immunomodulatory ac- been found that they can have multiple modes of action,
tivities [2]. suggesting that they may work as ‘dirty’ antimicrobials that
Traditionally, the search for novel AMPs involved the rely on a multiple-hit strategy to increase their efficiency
identification of active peptides from natural sources. Such and evade potential resistance mechanisms [3].
studies are followed by the design of synthetic peptide In this review, we aim to illustrate the wide diversity of
structural properties demonstrated by AMPs as well as the
Corresponding author: Vogel, H.J. (vogel@ucalgary.ca). growing number of ways in which they exert their activities.
464 0167-7799/$ – see front matter ß 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2011.05.001 Trends in Biotechnology, September 2011, Vol. 29, No. 9
(Figure_1)TD$IG][ Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Toroidal pore

Disordered toroidal pore


Carpet model

+ Δd
+ +

Membrane
Membrane Conformational thinning/thickening
Barrel stave
adsorption change
+ + + +

0.2V

?
Electroporation
Charged lipid clustering
Bacterial membrane
ΔΨ

O
Non-lytic membrane
depolarization + + H
+ Non-bilayer intermediate

- -
Oxidized
Anion carrier lipid targeting
TRENDS in Biotechnology

Figure 1. Events occurring at the bacterial cytoplasmic membrane following initial antimicrobial peptide (AMP) adsorption. These events are not necessarily exclusive of
each other. In the classical models of membrane disruption, the peptides lying on the membrane reach a threshold concentration and insert themselves across the
membrane to form either peptide-lined pores in the barrel-stave model, solubilize the membrane into micellar structures in the carpet model, or form peptide-and-lipid-
lined pores in the toroidal pore model. In the revised disordered toroidal pore model, pore formation is more stochastic and involves fewer peptides. The thickness of the
bilayer can be affected by the presence of the peptides, or the membrane itself can be remodeled to form domains rich in anionic lipids surrounding the peptides. In more
specific cases, non-bilayer intermediates in the membrane can be induced; peptide adsorption to the membrane can be enhanced by targeting them to oxidized
phospholipids; a peptide may couple with small anions across the bilayer, resulting in their efflux; the membrane potential can be dissipated without other noticeable
damage; or conversely, in the molecular electroporation model, the accumulation of peptide on the outer leaflet increases the membrane potential above a threshold that
renders the membrane transiently permeable to various molecules including the peptides themselves.

Recent structure–function relationship studies will be pre- kink in the middle of the a-helix. The amphipathicity of the
sented within the framework of the three AMP structural peptides is usually segregated along the axis of the a-helix
groups: a-helical peptides, b-sheet peptides and extended such that the peptide can lie parallel to the membrane plane
peptides (Figure 2) [13]. during the initial lipid interactions, with the charged side
facing outward towards the phospholipid head groups and
a-Helical AMPs the hydrophobic side embedded into the acyl tail core. The
The a-helical AMPs, such as magainin from the African properties of the a-helix tails sometimes influence the depth
clawed frog, are amongst the most intensely studied AMPs of membrane insertion and, in turn, the antimicrobial activ-
[14,15]. Numerous structure–function studies have indicat- ity [18].
ed that membrane-bound a-helices with too large a hydro- The lengths of the a-helices are usually sufficient to span
phobic surface become cytotoxic to mammalian cells [16]. the thickness of the membrane bilayers, with mismatches
Although these peptides are characterized by their struc- between the hydrophobic portion of the helices and the lipid
ture, an extremely high propensity for the formation of the acyl core strongly influencing the AMPs’ lateral and orien-
a-helix, which may present a more continuous hydrophobic tational motions in the membranes [19,20]. The peptides are
surface, can become a contributing factor to cytotoxicity [17]. thought to disrupt bacterial membranes through the forma-
Indeed, peptides that remain antimicrobial but have low tion of toroidal pores composed of loosely associated peptides
cytotoxicity require a membranous environment to induce with interdigitating phospholipid head groups among them
proper folding and often have a proline- or glycine-induced [10]. Pore formation is often associated with a local change in

465
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Table 1. Different modes of action that have been described for AMPs
Major target Specific target/mode of action Example peptides a Refs
External proteins Autolysin activation Pep5, nisin [10]
Phospholipase A2 activation Magainin 2, indolicidin [10]
Outer surface lipids LPS permeabilization (Gram-negative) Cecropin P1 [69]
Lipid II (peptidoglycan precursor) Defensins, Nisin and other lantibiotics [34,35]
[70]
Outer membrane proteins (Gram-negative) Outer membrane protein I (OprI) SMAP-29, CAP-18 [71]
LPS-assembly protein D (LptD) inhibition Protegrin I peptidomimetic analogs [33]
Outer membrane protein F (OmpF) HP(2–20) analog [72]
Inner membrane Barrel-stave pore Alamethicin [10]
Detergent micellization Dermaseptin, cecropin [10]
Toroidal pore Magainin 2, melittin [10]
protegrin I [31]
Disordered toroidal pore Magainin analog, melittin [22]
Membrane thinning/thickening PGLa, LL-37 [21]
Charged lipid clustering Magainin analogs, Arg-rich peptides [26]
Non-bilayer intermediate formation Gramicidin S [73]
Oxidized phospholipid targeting Temporin L, indolicidin [27]
Anion carrier Indolicidin [49]
Non-lytic membrane depolarization Bovine lactoferricin, daptomycin [32]
[74]
Electroporation NK-lysin [42]
Integral membrane proteins Proton translocation-related proteins Clavanin A [28]
Nucleic acids DNA (general) Buforin 2, tachyplesin, indolicidin [10]
DNA (covalent interaction) Indolicidin [50]
Branched DNA WRWYCR [43]
RNA (general) Buforin 2 [10]
Intracellular proteins DnaK inhibition, GroEL chaperonin Pyrrhocoricin, drosocin, apidaecin [10]
20S proteasome, SH3-containing proteins PR-39 [46]
[47]
a
Peptides listed are not limited to their associated modes of action here and may employ multiple mechanisms simultaneously.

membrane thickness [21]. Recent molecular dynamics (MD) appears to be applicable mostly to the high concentrations
simulations with a magainin analog and with melittin have used in vitro, and is generally viewed as an extreme
suggested that a re-evaluation of this model is in order extension of the toroidal pore model [10].
[22,23]. In the ‘disordered toroidal pore’, lipid molecules An interesting membrane perturbing effect induced by
are still curved inwards, but pore formation is more stochas- some a-helical AMPs is that of lipid segregation, where
tic and only one or two peptides are located near the center of peptide-induced clustering of anionic lipids in bacterial
the water-permeable pore, with more peptides positioned at membranes causes slow leakage of intracellular contents
the mouth of the pore on the external leaflet. As they and/or membrane depolarization [26]. Alternatively, the
translocate to the interior of the cell, the peptides remain phase boundary effects between domains of differently
mostly parallel to the bilayer plane, and only a small tilt is charged lipids may compromise the overall membrane
observed. Interestingly, the peptides undergo partial stability. Moreover, many bacterial membranes are mostly
unfolding during translocation and their a-helicity is not composed of phosphatidylglycerol and phosphatidyletha-
always maintained. In other MD simulations, similar tilted nolamine lipids, where the latter does not like to form
orientations for a-helical peptides were observed and the stable bilayers after peptide-induced demixing. In the case
notion of imperfect amphipathicity was introduced [24]. of Gram-positive bacteria such as Staphylococcus aureus,
This refers to the presence of some polar or charged residues which have cytoplasmic membranes that are mainly com-
in the hydrophobic face of an AMP to pull lipid head groups posed of the negatively charged lipids phosphatidylglycerol
into the membrane interior, thereby facilitating pore forma- and cardiolipin, lipid segregation is not expected and this is
tion. A perfect hydrophobic face on a peptide would favor a reflected in lower toxicity of some AMPs against these
fixed parallel orientation on a flat membrane surface. Im- species [26].
perfect amphipathicity is also a highlighted feature in the The targeting of oxidized lipids represents a unique
proposed interfacial activity model [25], which is based on a membrane-associated means for AMPs to enhance their
recent review of AMP literature. It closely resembles the activity. The a-helical AMPs temporin B and L intercalate
disordered toroidal pore model, with the key difference of the more efficiently into membranes containing an oxidized
interfacial activity model describing membrane disruption phosphatidylcholine lipid, probably via Schiff base forma-
without a need for peptide self-assembly. tion between the peptide amino groups and lipid aldehyde
The carpet model, where AMPs act more in a detergent- groups [27]. With the release of reactive oxygen species
like manner to break off the membrane lipids into micelle- during phagocytosis, it is possible that lipid oxidation could
like structures, has also been described. This model increase bacterial membrane susceptibility to AMPs.

466
(Figure_2)TD$IG][ Review Trends in Biotechnology September 2011, Vol. 29, No. 9

(a)

LL-37 Bovine
Magainin 2 lactoferrampin

(b)

Bovine lactoferricin Protegrin I


Human β-defensin-3

(c)

RRWQWR
Indolicidin
Tritrpticin
TRENDS in Biotechnology

Figure 2. An overview of the major structural classes of antimicrobial peptides (AMPs). (a) a-Helical peptides, (b) b-sheet peptides and (c) extended peptides. All of these
structures were solved by solution NMR spectroscopy in the presence of detergent micelles, except for the b-sheet peptides, which were studied in aqueous solution.
Positively charged side chains are colored in blue, negatively charged side chains in red and remaining side chains in grey. PDB IDs: magainin 2, 2MAG; LL-37, 2K6O; bovine
lactoferricin, 1LFC; protegrin 1, 1PG1; human b-defensin-3, 1KJ5; tritrpticin, 1D6X; indolicidin, 1G89.

Finally, the Phe-, His- and Gly-rich peptide clavanin A Additional evidence suggests that non-lytic mecha-
has a unique dual pH-dependent mode of action [28]. At nisms are also employed by certain b-sheet peptides. Al-
neutral pH, it permeabilizes membranes like regular a- though tachyplesin from horseshoe crabs is generally
helical AMPs. At slightly acidic pH, clavanin A is still recognized as a membrane active peptide, it is also capable
membrane disruptive. However, this does not occur via of binding to the minor groove of DNA, which may interfere
lipid interactions, but rather through inhibitory interac- with DNA–protein interactions [10]. The b-hairpin peptide
tions with proteins involved in maintaining the pH gradi- bovine lactoferricin can act synergistically with other an-
ent across the membrane. timicrobial agents by affecting the transmembrane poten-
In contrast to most a-helical peptides, buforin II, a tial and proton-motive force, resulting in inhibition of ATP-
peptide derivative from histones of the Asian toad Bufo dependent multi-drug efflux pumps [32]. Following trans-
bufo gargarizans, does not cause membrane disruption [9] location, bovine lactoferricin localizes to the cytoplasm
although its cell entry can be affected by membrane lipid where it can also inhibit DNA, RNA and protein synthesis.
composition [29]. After translocation, buforin II accumu- A series of peptidomimetic analogs of protegrin I featuring
lates in the cytoplasm where it is known to exert its stabilized b-hairpins are no longer membrane active, but
antimicrobial activity by adapting an extended or polypro- employ a new bactericidal mechanism involving a specific
line II conformation to accommodate interactions with protein target that affects outer membrane biosynthesis
nucleic acids [30]. [33]. Although these peptide analogs are remarkably po-
tent, their spectrum of activity is narrowed to Pseudomo-
b-Sheet AMPs nas aeruginosa strains, which express the susceptible
The b-sheet AMPs include several b-hairpin peptides in homolog of this b-barrel protein. Finally, although the
addition to the defensin mini-proteins. Many of these human defensins are membrane active, some of them
peptides disrupt bacterial membranes via the formation can also bind to the peptidoglycan precursor lipid II to
of toroidal pores as observed for the porcine peptide prote- inhibit cell wall biosynthesis enzymes in Staphylococci
grin I. This peptide forms oligomeric transmembrane b- [34,35].
barrels in anionic membranes, but forms b-sheet aggre- The b-hairpin or b-sheet conformations of these pep-
gates on the surface of cholesterol-containing membranes tides are often stabilized by disulfide bridges between
[31]. conserved Cys residues. However, these covalent bonds

467
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

do not appear necessary for antimicrobial activity. Linear they accumulate in the cytoplasm. The hexapeptide
derivatives of tachyplesin and lactoferricin retain antimi- WRWYCR, which was identified as a DNA-repair inhibitor
crobial activity while losing hemolytic activity [32,36]. that binds to Holliday junctions, was subsequently shown
Solid state NMR studies of tachyplesin and two linearized to have broad bactericidal activity [43]. Given the sequence
analogs, in which the four Cys residues were substituted by similarity of this peptide to those mentioned above, their
either Phe or Ala residues, show that all three peptides are efficacy may be due to this mechanism, where trapping of
positioned at the membrane interface and adopt similar b- the replication fork prevents recombination and DNA
sheet conformations on anionic membranes [37]. However, repair.
this does not correlate with their activities: the Cys-to-Phe The 13-residue Arg- and Trp-rich tritrpticin and indo-
peptide is equally active as tachyplesin whereas the Cys- licidin peptides from porcine and bovine leukocytes, re-
to-Ala peptide is inactive. Peptide dynamics, rather than spectively, can cause membrane leakage [42]. In a
their structures, provide a reasonable explanation for membranous environment, the structures of both peptides
these functional differences. Whereas the active tachyple- feature multiple Pro-induced turns that give them amphi-
sin peptide shows enough mobility in the liquid-crystalline pathic boat-like conformations. These peptides are suffi-
phase of the membrane to disturb lipid packing, the inac- ciently small that simple residue substitutions can have
tive analog is immobilized on the membrane and stays large consequences for their structural and functional
highly aggregated. properties. Replacing both Pro residues with Ala in tritrp-
At 45 residues, human b-defensin-3 (HBD-3) is rather ticin transforms it into an a-helical peptide with slightly
large as an AMP, containing an a-helical turn followed by a improved membrane leakage and antimicrobial activity,
three-stranded antiparallel b-sheet. This scaffold is held but also higher cytotoxicity [44]. The role of the two Pro
together by three disulfide bonds in a defined pattern that residues is distinct, with Pro5 having more of a structural
distinguishes the b-defensins from the a-defensins. None- role preventing the induction of a-helical structure. Ironi-
theless, rearranging the disulfide pattern in HBD-3 or cally, the characteristic Arg and Trp side chains do not
reducing them does not alter the antimicrobial activity, seem an optimal choice of residues for tritrpticin. In ana-
nor does it abolish the overall formation of the native logs with Arg-to-Lys and Trp-to-Phe substitutions, the
secondary structure elements [38,39]. Although antimicro- antimicrobial activity is maintained and the slight hemo-
bial activity may be unaffected, chemotactic activities are lytic activity of tritrpticin is abolished. The reduced hydro-
decreased for the analogs with non-native disulfide lin- gen bonding capabilities of Lys compared to the Arg
kages. guanidinium group seem to increase membrane selectivity
Interestingly, reducing the disulfides in HBD-1 creates for the peptide. Replacement of the three Trp residues with
a flexible unstructured peptide with improved activities Phe created a tritrpticin variant that was no longer able to
against Candida albicans and some Gram-positive bacte- adopt a single well-defined conformation in a membrane
ria [40]. The presence of reduced HBD-1 could be of great [44]. Interestingly, this peptide required a few hours to
physiological relevance in the anaerobic environment of take effect whereas native tritrpticin completes bacterial
the colon. killing in less than one hour [45]. The fact that multiple
mechanisms are in play is further substantiated by the
Extended AMPs synergistic effects observed between the Trp-to-Phe and
Extended AMPs, which do not fold into regular secondary membrane active Pro-to-Ala peptides [45]. The Pro- and
structure elements, often contain high proportions of cer- Arg-rich porcine PR-39 peptide interacts with cytoplasmic
tain amino acids, specifically Arg, Trp or Pro residues. The proteins such as the 20S proteasome and SH3-domain
His-rich salivary histatin peptides are sometimes consid- proteins, and it is believed that this contributes to its
ered to be extended AMPs, however they are not bacterio- antimicrobial mode of action [46,47].
static nor bactericidal despite being fungicidal and anti- Indolicidin has been a template for many analogs with
parasitic [41]. Many of the extended peptides are not improved biological properties, the most successful one
membrane active. The Pro-rich insect-derived pyrrhocor- being omiganan, which was developed by Migenix
icin, drosocin and apidaecin peptides penetrate across the (renamed to Biowest Therapeutics) and has entered phase
membrane and exert their antimicrobial activities by mak- III clinical trials [48]. A number of contributing mecha-
ing interactions with intracellular proteins such as the nisms may explain the efficiency of indolicidin. This
heat-shock protein DnaK and GroEL to inhibit the DnaK includes the activation of phospholipase A2 secreted by
ATPase activity and chaperone-assisted protein folding, human lacrimal fluid to hydrolyze anionic membranes [10]
respectively [10]. or a direct membrane action either through lipid segrega-
Trp and Arg side chains are prominent in peptides with tion, the targeting of oxidized phospholipids or acting as a
less than 15 residues [42]. Two of the shortest active small anion carrier across the membrane [27,49]. More-
peptides include RRWQWR and RAWVAWR, which have over, at the intracellular level, both covalent and non-
been identified as the active antimicrobial fragments from covalent DNA–indolicidin interactions can interfere with
bovine lactoferricin and human lysozyme, respectively. In DNA-binding enzymes and induce filamentation in Escher-
addition, combinatorial peptide library screening has iden- ichia coli [50]. It is unclear how the short and unique
tified the hexameric sequence RRWWRF as having potent sequence of indolicidin, which includes five Trp residues
broad spectrum activity [4]. Although these peptides can and three Pro-induced turns, can exhibit all these activities
fold into defined amphipathic molecules in the presence of and which of these proposed mechanisms contributes most
a membrane, they are not membrane active and, instead, to its activity in vivo.

468
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Protein-derived antimicrobial fragments and Whether they are naturally released proteolysis pro-
antimicrobial proteins ducts or designed synthetic peptides, many AMPs have
AMPs can be derived from large proteins. In addition, been derived from proteins such as casein [53], lysozyme
several cationic proteins have been discovered to possess and ovotransferrin [54], myoglobin and hemoglobin [55],
direct nonenzymatic antimicrobial activity themselves, histones [9], growth factors [56], and thrombin [57]. Thus,
some of which are shown in Figure 3. The primary function scanning the proteome for related sequences with appro-
of these antimicrobial proteins is usually immunity-relat- priate combinations of cationic and hydrophobic residues
ed, therefore they show significant functional overlap with may uncover additional AMPs. Some of these proteins such
AMPs (Table 2). as lysozyme and histones already have intrinsic antimi-
Lactoferrin (Lfin) is a multifunctional 80 kDa glycopro- crobial activity and, as is the case for Lfin, their peptide
tein present in biofluids, most notably in milk and neutro- fragments are expected to have different modes of action
phil granules. The protein is involved in several [58].
immunoregulatory processes that primarily take advan- Recently, more than 20 human chemokine proteins have
tage of its strong antimicrobial properties. This activity is been shown to be directly antimicrobial, with macrophage-
due in part to its ability to sequester iron, which prevents inflammatory protein-3a (MIP-3a) possessing the most
bacterial growth. However, Lfin also has an iron-indepen- potent and broadest spectrum of activity (Figure 3b)
dent mechanism arising from direct interactions with [59]. In the case of the chemokine CCL28, the antimicrobial
bacterial cell components [51]. Three cationic antimicrobi- mechanism has been identified as membrane permeation
al fragments have been identified in different parts of the [60]. Conversely, a few defensins are now known to possess
Lfin sequence: lactoferricin, which is a naturally occurring chemotactic activity [61]. A common and unifying feature
pepsin digestion product, lactoferrampin and kaliocin of the antimicrobial chemokines is the presence of a large
(Figure 3a) [18,32,52]. The structural differences for the uninterrupted cationic surface. Some chemokines tend to
isolated peptides and the corresponding regions in Lfin are dimerize under physiological conditions, further extending
significant; in particular, bovine lactoferricin and kaliocin the cationic surface [62]. For example, HBD-3 has a stron-
form amphipathic b-hairpin peptides, whereas lactofer- ger tendency to dimerize, which may contribute to its high
rampin requires a membraneous environment to adopt salt-insensitive activity compared to other b-defensins
its native a-helical structure. The intact protein and the [63]. The C-terminal a-helices of several antimicrobial
peptides probably kill bacteria in a different manner, chemokines have multiple positively charged residues
especially lactoferricin, which is substantially more active and when synthetic peptides of this region are studied,
than intact Lfin. they behave like membrane-active a-helical AMPs [64].
[(Figure_3)TD$IG]

(a)

(b)

TRENDS in Biotechnology

Figure 3. The structures of immunity-related proteins with direct antimicrobial activity and antimicrobial fragments. (a) Human lactoferrin (PDB ID 1FCK) with segments
corresponding to its antimicrobial fragments: lactoferricin in blue, kaliocin in red and lactoferrampin in yellow (left panel). The electrostatic potential surface plot of
lactoferrin is shown in the right panel. (b) Macrophage inflammatory protein-3a (PDB ID 2JYO) with the MIP-3a51–70 peptide segment colored in blue (left panel). The
electrostatic potential surface plot of the monomeric unit of MIP-3a is shown in the middle panel and the same plot for the MIP-3a dimer is in the right panel, showing the
stronger cationic surface around the self-associating a-helices.

469
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Table 2. A selection of human AMPs and antimicrobial proteins highlighting their multifunctionality in the immune system
Main sources Indirect antimicrobial and additional functions a Refs
AMPs
LL-37 Leukocytes, epithelia Endotoxin neutralization, chemotaxis, reactive [61]
oxygen species (ROS) formation, wound healing,
mast cell degranulation
Defensins Leukocytes, epithelia Endotoxin neutralization, opsonization, chemotaxis, [61]
ROS formation, wound healing, complement inhibition
Dermcidin Skin Chemotaxis [7]
Histatins Saliva Wound healing [75]
Lactoferricin Digested milk protein Endotoxin neutralization, complement [32]
inhibition, transcription factor
Antimicrobial proteins
Chemokines Leukocytes Chemotaxis [59]
Neutrophil serprocidins Neutrophils Protease, ROS formation, opsonization, [61]
(azurocidin, cathepsin G, elastase) macrophage activation
Bactericidal/permeability Neutrophils Endotoxin neutralization [61]
increasing protein
Lipocalins Neutrophils, secretory fluids Siderophore sequestering [76]
Lysozyme Secretory fluids, neutrophils Peptidoglycan digestion [61]
Lactoferrin Secretory fluids, neutrophils Iron sequestering, endotoxin neutralization, [61]
phagocytosis stimulation
RNAse 7 Skin Ribonuclease [77]
Psoriasin Skin Neutrophil activation, ROS formation [77]
Histones Nuclei Chromatin regulation [78]
Prion protein Brain, other tissues Unknown [79]
a
These are usually secondary functions for AMPs and primary functions for antimicrobial proteins.

This property may explain the bactericidal properties of bacterial cell. Consequently, the design and prediction of
several intact chemokines; however, some of the a-helical peptide analog characteristics is not straightforward and all
peptides do not retain activity and, furthermore, some peptides and analogs need to be studied on a case-by-case
chemokines do not have cationic C-terminal regions de- basis. Apart from their biological activities, other aspects
spite displaying activity. For the platelet chemokine neu- need to be considered when developing these peptides for
trophil activating protein-2 (NAP-2), the proteolytic systemic use, including in vivo stability [66], side effects and
removal of the two C-terminal Ala-Asp residues leads to production costs. Based on our current understanding of the
a drastic increase in the antimicrobial potency. This was structure–function relationships for different classes of
attributed to the negatively charged Asp side chain inter- AMPs, some non-peptide and peptidomimetic compounds
fering with a positive patch on the protein surface that have been developed and have advantageous properties
includes side chains from the C-terminal a-helix as well as because they combine stability with high antimicrobial
the b-sheet loops [65]. potency [67,68]. Overall, detailed structure–function and
biophysical studies can rationalize why a certain peptide
Concluding remarks is more active or behaves differently from a related peptide
Widely different structural scaffolds seem to support the or an analog. Such comprehensive understanding will be
activity of AMPs and proteins, with amphipathicity remain- essential to guide the design of better AMPs.
ing as the only overarching physical property. A combina-
tion of a well-defined cationic region together with an Acknowledgments
imperfect hydrophobic surface area frequently seems to Research on AMPs in the H.J. Vogel laboratory is supported by a grant
result in broad spectrum activity. In addition, the dynamic from the Novel Alternatives for Antibiotics Program of the Canadian
Institute for Health Research. H.J.V. is the holder of a Scientist award
properties of these peptides can affect their biological prop-
from the Alberta Heritage Foundation for Medical Research.
erties, with structural rigidity generally hindering activity.
Initially, AMPs were considered as a uniform group of
References
molecules that ‘punch holes’ in bacterial membranes. In 1 Williams, K.J. and Bax, R.P. (2009) Challenges in developing new
recent years, it has become apparent that their mechanism antibacterial drugs. Curr. Opin. Investig. Drugs 10, 157–163
of action is much more complex and diverse. Current re- 2 Hancock, R.E. and Sahl, H.G. (2006) Antimicrobial and host-defense
search efforts now also include antimicrobial fragments peptides as new anti-infective therapeutic strategies. Nat. Biotechnol.
24, 1551–1557
from intact proteins and proteins with direct antimicrobial
3 Peschel, A. and Sahl, H.G. (2006) The co-evolution of host cationic
activity, forcing structural biology researchers to reevaluate antimicrobial peptides and microbial resistance. Nat. Rev. Microbiol. 4,
the common properties within this expanding family. Clear- 529–536
ly, host defense systems have developed a broad arsenal and 4 Blondelle, S.E. and Lohner, K. (2010) Optimization and high-
use a multi-pronged strategy to combat invading pathogens. throughput screening of antimicrobial peptides. Curr. Pharm. Des.
16, 3204–3211
Many AMPs seem to be capable of killing bacterial cells in
5 Rotem, S. and Mor, A. (2009) Antimicrobial peptide mimics for
multiple ways, making it a difficult task to unravel all the improved therapeutic properties. Biochim. Biophys. Acta 1788,
molecular events that occur when a peptide encounters a 1582–1592

470
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

6 Lohner, K. (2001) The role of membrane lipid composition in cell 33 Srinivas, N. et al. (2010) Peptidomimetic antibiotics target outer-
targeting of antimicrobial peptides. In Development of Novel membrane biogenesis in Pseudomonas aeruginosa. Science 327,
Antimicrobial Agents: Emerging Strategies (Lohner, K., ed.), pp. 1010–1013
149–165, Horizon Scientific Press 34 Sass, V. et al. (2010) Human beta-defensin 3 inhibits cell wall
7 Harris, F. et al. (2009) Anionic antimicrobial peptides from eukaryotic biosynthesis in staphylococci. Infect. Immun. 78, 2793–2800
organisms. Curr. Protein Pept. Sci. 10, 585–606 35 de Leeuw, E. et al. (2010) Functional interaction of human neutrophil
8 Hwang, P.M. and Vogel, H.J. (1998) Structure–function relationships peptide-1 with the cell wall precursor lipid II. FEBS Lett. 584,
of antimicrobial peptides. Biochem. Cell Biol. 76, 235–246 1543–1548
9 Nicolas, P. (2009) Multifunctional host defense peptides: intracellular- 36 Ramamoorthy, A. et al. (2006) Deletion of all cysteines in tachyplesin I
targeting antimicrobial peptides. FEBS J. 276, 6483–6496 abolishes hemolytic activity and retains antimicrobial activity and
10 Brogden, K.A. (2005) Antimicrobial peptides: pore formers or metabolic lipopolysaccharide selective binding. Biochemistry 45, 6529–6540
inhibitors in bacteria? Nat. Rev. Microbiol. 3, 238–250 37 Doherty, T. et al. (2008) Dynamic structure of disulfide-removed linear
11 Papo, N. and Shai, Y. (2003) Can we predict biological activity of analogs of tachyplesin-I in the lipid bilayer from solid-state NMR.
antimicrobial peptides from their interactions with model Biochemistry 47, 1105–1116
phospholipid membranes? Peptides 24, 1693–1703 38 Wu, Z. et al. (2003) Engineering disulfide bridges to dissect
12 Mangoni, M.L. and Shai, Y. (2009) Temporins and their synergism antimicrobial and chemotactic activities of human beta-defensin 3.
against Gram-negative bacteria and in lipopolysaccharide Proc. Natl. Acad. Sci. U.S.A. 100, 8880–8885
detoxification. Biochim. Biophys. Acta 1788, 1610–1619 39 Kluver, E. et al. (2005) Structure–activity relation of human beta-
13 Bhattacharjya, S. and Ramamoorthy, A. (2009) Multifunctional host defensin 3: influence of disulfide bonds and cysteine substitution on
defense peptides: functional and mechanistic insights from antimicrobial activity and cytotoxicity. Biochemistry 44, 9804–9816
NMR structures of potent antimicrobial peptides. FEBS J. 276, 40 Schroeder, B.O. et al. (2011) Reduction of disulphide bonds unmasks
6465–6473 potent antimicrobial activity of human b-defensin 1. Nature 469,
14 Zasloff, M. (2002) Antimicrobial peptides of multicellular organisms. 419–423
Nature 415, 389–395 41 Luque-Ortega, J.R. et al. (2008) Human antimicrobial peptide histatin
15 Haney, E.F. et al. (2009) Solution NMR studies of amphibian 5 is a cell-penetrating peptide targeting mitochondrial ATP synthesis
antimicrobial peptides: linking structure to function? Biochim. in Leishmania. FASEB J. 22, 1817–1828
Biophys. Acta 1788, 1639–1655 42 Chan, D.I. et al. (2006) Tryptophan- and arginine-rich antimicrobial
16 Zelezetsky, I. and Tossi, A. (2006) Alpha-helical antimicrobial peptides peptides: structures and mechanisms of action. Biochim. Biophys. Acta
- using a sequence template to guide structure–activity relationship 1758, 1184–1202
studies. Biochim. Biophys. Acta 1758, 1436–1449 43 Su, L.Y. et al. (2010) An antimicrobial peptide that targets DNA repair
17 Takahashi, D. et al. (2010) Structural determinants of host defense intermediates in vitro inhibits Salmonella growth within murine
peptides for antimicrobial activity and target cell selectivity. Biochimie macrophages. Antimicrob. Agents Chemother. 54, 1888–1899
92, 1236–1241 44 Schibli, D.J. et al. (2006) Structure–function analysis of tritrpticin
18 Haney, E.F. et al. (2009) Novel lactoferrampin antimicrobial peptides analogs: potential relationships between antimicrobial activities,
derived from human lactoferrin. Biochimie 91, 141–154 model membrane interactions, and their micelle-bound NMR
19 Holt, A. and Killian, J.A. (2010) Orientation and dynamics of structures. Biophys. J. 91, 4413–4426
transmembrane peptides: the power of simple models. Eur. Biophys. 45 Yang, S.T. et al. (2006) Different modes in antibiotic action of tritrpticin
J. 39, 609–621 analogs, cathelicidin-derived Trp-rich and Pro/Arg-rich peptides.
20 Ramadurai, S. et al. (2010) Influence of hydrophobic mismatch and Biochim. Biophys. Acta 1758, 1580–1586
amino acid composition on the lateral diffusion of transmembrane 46 Anbanandam, A. et al. (2008) Molecular basis for proline- and arginine-
peptides. Biophys. J. 99, 1447–1454 rich peptide inhibition of proteasome. J. Mol. Biol. 384, 219–227
21 Lohner, K. (2009) New strategies for novel antibiotics: peptides 47 Zanetti, M. (2004) Cathelicidins, multifunctional peptides of the innate
targeting bacterial cell membranes. Gen. Physiol. Biophys. 28, 105–116 immunity. J. Leukoc. Biol. 75, 39–48
22 Sengupta, D. et al. (2008) Toroidal pores formed by antimicrobial 48 Melo, M.N. et al. (2006) Omiganan pentahydrochloride in the front line
peptides show significant disorder. Biochim. Biophys. Acta 1778, of clinical applications of antimicrobial peptides. Recent Pat. Antiinfect.
2308–2317 Drug Discov. 1, 201–207
23 Leontiadou, H. et al. (2006) Antimicrobial peptides in action. J. Am. 49 Rokitskaya, T.I. et al. (2011) Indolicidin action on membrane
Chem. Soc. 128, 12156–12161 permeability: carrier mechanism versus pore formation. Biochim.
24 Mihajlovic, M. and Lazaridis, T. (2010) Antimicrobial peptides bind Biophys. Acta 1808, 91–97
more strongly to membrane pores. Biochim. Biophys. Acta 1798, 50 Marchand, C. et al. (2006) Covalent binding of the natural
1494–1502 antimicrobial peptide indolicidin to DNA abasic sites. Nucleic Acids
25 Wimley, W.C. (2010) Describing the mechanism of antimicrobial Res. 34, 5157–5165
peptide action with the interfacial activity model. ACS Chem. Biol. 51 Nibbering, P.H. et al. (2001) Human lactoferrin and peptides derived
5, 905–917 from its N terminus are highly effective against infections with
26 Epand, R.M. and Epand, R.F. (2011) Bacterial membrane lipids in the antibiotic-resistant bacteria. Infect. Immun. 69, 1469–1476
action of antimicrobial agents. J. Pept. Sci. 17, 298–305 52 Yount, N.Y. et al. (2007) The gamma-core motif correlates with
27 Mattila, J.P. et al. (2008) Oxidized phospholipids as potential antimicrobial activity in cysteine-containing kaliocin-1 originating
molecular targets for antimicrobial peptides. Biochim. Biophys. Acta from transferrins. Biochim. Biophys. Acta 1768, 2862–2872
1778, 2041–2050 53 Lopez-Exposito, I. et al. (2006) Identification of antibacterial peptides
28 van Kan, E.J. et al. (2002) Clavanin permeabilizes target membranes from bovine kappa-casein. J. Food Prot. 69, 2992–2997
via two distinctly different pH-dependent mechanisms. Biochemistry 54 Pellegrini, A. (2003) Antimicrobial peptides from food proteins. Curr.
41, 7529–7539 Pharm. Des. 9, 1225–1238
29 Fleming, E. et al. (2008) Effect of lipid composition on buforin II 55 Mak, P. et al. (2000) Antimicrobial peptides derived from heme-
structure and membrane entry. Proteins 73, 480–491 containing proteins: hemocidins. Antonie Van Leeuwenhoek 77, 197–207
30 Lan, Y. et al. (2010) Structural contributions to the intracellular 56 Malmsten, M. et al. (2007) Antimicrobial peptides derived from growth
targeting strategies of antimicrobial peptides. Biochim. Biophys. factors. Growth Factors 25, 60–70
Acta 1798, 1934–1943 57 Papareddy, P. et al. (2010) Proteolysis of human thrombin generates
31 Tang, M. and Hong, M. (2009) Structure and mechanism of beta- novel host defense peptides. PLoS Pathog. 6, e1000857
hairpin antimicrobial peptides in lipid bilayers from solid-state 58 Hunter, H.N. et al. (2005) The interactions of antimicrobial peptides
NMR spectroscopy. Mol. Biosyst. 5, 317–322 derived from lysozyme with model membrane systems. Biochim.
32 Gifford, J.L. et al. (2005) Lactoferricin: a lactoferrin-derived peptide Biophys. Acta 1668, 175–189
with antimicrobial, antiviral, antitumor and immunological 59 Yang, D. et al. (2003) Many chemokines including CCL20/MIP-3alpha
properties. Cell Mol. Life Sci. 62, 2588–2598 display antimicrobial activity. J. Leukoc. Biol. 74, 448–455

471
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

60 Hieshima, K. et al. (2003) CCL28 has dual roles in mucosal immunity 70 Hsu, S.T. et al. (2004) The nisin-lipid II complex reveals a
as a chemokine with broad-spectrum antimicrobial activity. J. pyrophosphate cage that provides a blueprint for novel antibiotics.
Immunol. 170, 1452–1461 Nat. Struct. Mol. Biol. 11, 963–967
61 Soehnlein, O. (2009) Direct and alternative antimicrobial mechanisms 71 Lin, Y.M. et al. (2010) Outer membrane protein I of Pseudomonas
of neutrophil-derived granule proteins. J. Mol. Med. 87, 1157–1164 aeruginosa is a target of cationic antimicrobial peptide/protein. J. Biol.
62 Chan, D.I. et al. (2008) Human macrophage inflammatory protein Chem. 285, 8985–8994
3alpha: protein and peptide nuclear magnetic resonance solution 72 Apetrei, A. et al. (2010) Unimolecular study of the interaction between
structures, dimerization, dynamics, and anti-infective properties. the outer membrane protein OmpF from E. coli and an analogue of
Antimicrob. Agents Chemother. 52, 883–894 the HP(2-20) antimicrobial peptide. J. Bioenerg. Biomembr. 42,
63 Schibli, D.J. et al. (2002) The solution structures of the human beta- 173–180
defensins lead to a better understanding of the potent bactericidal 73 Haney, E.F. et al. (2010) Induction of non-lamellar lipid phases by
activity of HBD3 against Staphylococcus aureus. J. Biol. Chem. 277, antimicrobial peptides: a potential link to mode of action. Chem. Phys.
8279–8289 Lipids 163, 82–93
64 Nguyen, L.T. et al. (2010) Structure–function studies of chemokine- 74 Jeu, L. and Fung, H.B. (2004) Daptomycin: a cyclic lipopeptide
derived carboxy-terminal antimicrobial peptides. Biochim. Biophys. antimicrobial agent. Clin. Ther. 26, 1728–1757
Acta 1798, 1062–1072 75 Oudhoff, M.J. et al. (2008) Histatins are the major wound-closure
65 Nguyen, L.T. et al. (2011) Exploring platelet chemokine antimicrobial stimulating factors in human saliva as identified in a cell culture
activity: NMR backbone dynamics studies of NAP-2 and TC-1. assay. FASEB J. 22, 3805–3812
Antimicrob. Agents Chemother. 55, 2074–2083 76 Fluckinger, M. et al. (2004) Human tear lipocalin exhibits
66 Nguyen, L.T. et al. (2010) Serum stabilities of short tryptophan- and antimicrobial activity by scavenging microbial siderophores.
arginine-rich antimicrobial peptide analogs. PLoS ONE 5, e12684 Antimicrob. Agents Chemother. 48, 3367–3372
67 Lai, X.Z. et al. (2008) Ceragenins: cholic acid-based mimics of 77 Schroder, J.M. and Harder, J. (2006) Antimicrobial skin peptides and
antimicrobial peptides. Acc. Chem. Res. 41, 1233–1240 proteins. Cell Mol. Life Sci. 63, 469–486
68 Haug, B.E. et al. (2008) Synthetic antimicrobial peptidomimetics with 78 Parseghian, M.H. and Luhrs, K.A. (2006) Beyond the walls of the
therapeutic potential. J. Med. Chem. 51, 4306–4314 nucleus: the role of histones in cellular signaling and innate
69 Strauss, J. et al. (2010) Binding, inactivation, and adhesion forces immunity. Biochem. Cell Biol. 84, 589–604
between antimicrobial peptide cecropin P1 and pathogenic E. coli. 79 Pasupuleti, M. et al. (2009) Antimicrobial activity of human prion
Colloids Surf. B Biointerfaces 75, 156–164 protein is mediated by its N-terminal region. PLoS ONE 4, e7358

472

Você também pode gostar