You are on page 1of 16

International Journal of Peptide Research and Therapeutics

https://doi.org/10.1007/s10989-020-10127-2

Human Antimicrobial Peptides: Spectrum, Mode of Action


and Resistance Mechanisms
Bibi Sedigheh Fazly Bazzaz1,2 · Shabnam Seyedi3 · Narjes Hoseini Goki3 · Bahman Khameneh2 

Accepted: 22 October 2020


© Springer Nature B.V. 2020

Abstract
Overuse of antibiotics is one of the important factors that contribute to developing antimicrobial resistance. Many studies
have been conducted to find out promising solutions to overcome the problems. Antimicrobial peptides (AMPs) are funda-
mental components of human innate immunity. They have an important role in the treatment of a wide range of diseases,
including cancer, allergies, and also in warding off invading pathogens. In the case of infectious disease, the AMPs exhibit
broad-spectrum activity against a wide range of pathogens including Gram-positive and -negative bacteria, yeasts, fungi, and
enveloped viruses. These peptides have been isolated from various sources such as microorganisms, plants, invertebrates,
and vertebrates. The peptides show distinct physicochemical and structural properties but most of them are small cationic
peptides with amphipathic properties. In this review, an overview of the classification, antimicrobial activities, mode of
action, and mechanism of resistance of human AMPs will be provided. These peptides are categorized into three main
groups. The defensins are cationic peptides containing six cysteine residues with three intramolecular disulfide bridges.
In humans, two classes of defensins could be found, α-defensins and β-defensins. The second group is cathelicidins that
only one AMP, LL-37, has been found in humans. This peptide is derived from proteolytic digestion of the C-terminal of
human CAP18 protein. The third group is the family of histatins that are small cationic histidine-rich peptides, and mainly
present in human saliva. The last two groups have random coil conformation in hydrophilic environments and α-helices in
a hydrophobic environment.

Keywords  Human antimicrobial peptides · Antibiotic resistance · LL-37 · Histatins · Defensins

Introduction in each year for USA, EU, and India was 23,000, 25,000, and
58,000, respectively (Chaudhary 2016). Additionally, with-
Antibacterial resistance by multidrug-resistant (MDR) path- out developing novel approaches to combat MDR pathogens,
ogens has become a global health challenge and threatens many fields of medicine such as surgery, cancer chemother-
the health of societies. The emergence of resistant infections apy, and transplantation medicine will be affected severely
leads to existing antibacterial drugs becoming less effective (Worthington and Melander 2013). Whenever the antibac-
or even ineffective and therefore, there is an urgent need terial resistance has emerged, the problem has been tackled
for the development of new antibacterial agents (Khameneh with various approaches such as repurposing non-antibiotic
et al. 2016). The death by drug-resistant bacterial infections drugs, modification of the existing antibiotic classes with
limited cross-resistance, developing new classes of anti-
biotics, antibiotic combinations, development of adjuvant
* Bahman Khameneh
Khamenehbagherib@mums.ac.ir therapy, developing a novel formulation of antibiotic, and
using natural compounds (Bazzaz et al. 2019; Farhadi et al.
1
Biotechnology Research Center, Pharmaceutical Technology 2019; Khameneh et al. 2015a, 2019a, b; Soheili et al. 2019;
Institute, Mashhad University of Medical Sciences, Theuretzbacher et al. 2020).
Mashhad, Iran
During the past years, the efforts for discovering the
2
Department of Pharmaceutical Control, School of Pharmacy, novel antimicrobial agents have been focused on develop-
Mashhad University of Medical Sciences, Mashhad, Iran
ing groups of short polypeptides, up to 40 residues, namely
3
School of Pharmacy, Mashhad University of Medical antimicrobial peptides (AMPs) (Fox 2013; Javia et al. 2018).
Sciences, Mashhad, Iran

13
Vol.:(0123456789)
International Journal of Peptide Research and Therapeutics

AMPs can be obtained from both natural and synthetic and composition and their mode of action. Moreover, the
sources and show a broad spectrum of targeted organisms essential aspects of human AMPs that have revealed their
including viruses to parasites (Bahar and Ren 2013; Zasloff significance will be discussed.
2002). These peptides are categorized based on different
properties such as similarities in charge, sequence, func-
tional and 3-dimensional structure. Based on the charge of
AMPs, they are grouped into anionic and cationic peptides. Classification of AMPs
Anionic ones are small peptides (721.6–823.8 Da) and pre-
sent in bronchoalveolar lavage, fluid surfactant extracts, and Knowing about the three-dimensional structure of AMPs
airway epithelial cells, while, cationic peptides are mainly leads to a better understanding of their mode of actions.
found in all living species. These latter AMPs contain 12–50 Nuclear magnetic resonance (NMR) has been used exten-
amino acid residues with a net positive charge of + 2 to  + 7. sively for analyzing the structures of most of the AMPs.
They have an excess of basic amino acid residues such as Because of the small size of AMPs in comparison with other
arginine, lysine, and histidine in comparison with acidic resi- biopharmaceuticals, the three-dimensional structures could
dues. Cationic AMPs have a diverse range of cellular targets be obtained by conventional two-dimensional NMR methods
(Bahar and Ren 2013; Brogden et al. 2003; Bulet et al. 2004; (Reddy et al. 2004; Zhang and Falla 2006). Based on the
Huang et al. 2010). NMR data, AMPs could be classified into five groups.
AMPs show their antimicrobial properties via different
mechanisms. They showed weak antimicrobial actions but
have wide and strong immune-modulatory properties. Taken
together, they have been considered as alternative agents for α‑Helical AMPs
traditional antibiotics. AMPs have been successfully used
against resistant infections such as infectious biofilm or Abundant classes of AMPs are those with amphipathic
MDR bacteria (Boparai and Sharma 2020; Park et al. 2011). properties that have α-helical domains. Their particularly
In contrast to an antibiotic peptide that is synthesized successful structural arrangements help them for innating
through a specific metabolic pathway, the amino acid defense. Most of them are short (< 40 residues) and there-
sequence of AMPs is encoded naturally by the genetic fore can be synthesized easily. Because of the structure, they
materials of the host organisms (Boparai and Sharma 2020). can be characterized by circular dichroism (CD) or NMR
Upon pathogen invasion, transcription and translation pro- spectroscopies, simply. It should be noted that these types of
cesses are started and followed by post-translational modi- AMPs are active against a wide range of pathogens, includ-
fications including precursor protein cleavage, C-terminus ing both Gram‐positive and ‐negative bacteria, fungi, and
amidation, and disulfide bond formation for approximately protozoa (Giangaspero et al. 2001; Reddy et al. 2004). Some
50% of the known AMPs. Moreover, glycosylation modifi- of them are illustrated in Fig. 1.
cation may happen in uncommon cases (Toke 2005). These
AMPs were first detected in plant organisms, then in a wide
range of other organisms like humans (Toke 2005). So far,
the number of detected AMPs has reached more than one Cysteine‑Rich AMPs
million (Boparai and Sharma 2020).
AMPs show a broad variety of secondary structures Cysteine-rich AMPs are diverse and widely distributed in
like β-strands, α-helices with extended structures, loops, animal and plant tissues. They have an important role in host
and one or more disulfide bridges (Edwards et al. 2017; defense systems (Dimarcq et al. 1998). The human neutro-
Hwang and Vogel 1998). These observed structural differ- phil peptides HNP-1, -2, and -3 were the first of these AMPs
ences are necessary for their antimicrobial activities in a which isolated from the human granules (Ganz et al. 1985).
broad spectrum (Hancock 2001). Additionally, particular These α-defensins compose of 30 amino acid residues which
crucial factors like peptide self-association to biological are rich in cysteine and could be found in a wide variety of
membranes, amphipathic stereo-geometry, hydrophobic- organisms (Reddy et al. 2004). Most of these AMPs contain
ity, charge, and also size contribute to their antimicro- six cysteine residues and all of them participate in forming
bial activities (Boparai and Sharma 2020). Moreover, the three intramolecular disulfide bonds (Dimarcq et al. 1998).
smaller size of antimicrobial peptides facilitates the fast These disulfide bridges are mostly between C1–C4, C2–C5,
secretion and diffusion of peptide outside the cells that is and C3–C6 (Dimarcq et al. 1998; Hill et al. 1991). Based
important for inducing defense responses against patho- on the NMR data, α-defensin has three-stranded antiparal-
genic fungi and bacteria (Brogden 2005). This article aims lel β-sheets (Pardi et al. 1992). Some of them are illustrated
to review the classification of AMPs based on the structure in Fig. 2.

13
International Journal of Peptide Research and Therapeutics

Fig. 1  Some examples of
α-helical antimicrobial peptides

Fig. 2  Some examples of
cysteine-rich antimicrobial
peptides

β‑Sheet AMPs crab (Limulus polyphemus) peptides, tachyplesins, and


polyphemusin II, which are stabilized via two disulfide
The third group of AMPs is peptides with β-sheets struc- bridges; thanatin from insects (one disulfide bond); and
ture. Most of them are further stabilized by one or more protegrin-1, peptides isolated from porcine leukocytes.
disulfide bonds. Based on their cysteine content and struc- These molecules form an antiparallel β-sheet connected
tural properties, they can be divided into two groups: (i) to a β-turn and are composed of disulfide bonds (Kawano
β-hairpin peptides; and (ii) α-defensin peptides (Koe- et al. 1990; Tamamura et al. 1993). As shown by NMR
hbach and Craik 2019). β-hairpin peptides encompass studies, lactoferricin B, containing 25 amino acids,
approximately 20 residues containing one or two disulfide presents a β-sheet structure that stabilized by a single
bonds. Examples of β-hairpin peptides are Horseshoe disulfide bridge (Hwang et al. 1998). Some of these AMPs
are illustrated in Fig. 3.

13
International Journal of Peptide Research and Therapeutics

Fig. 3  Some examples of
β-sheet antimicrobial peptides

AMPs Rich in Regular Amino Acids conformations could not see in other classes of AMPs. For
example, subtilin is a ribosomally synthesized AMP that
Some of the AMPs contain high numbers of regular amino composes of several unusual amino acids as a result of post-
acid residues and therefore their structures are different from translational modifications (Liu and Hansen 1993). Gramici-
the regular α-helical or β-sheet peptides. For example, hista- din is another example that contains several DH-amino acids
tin, a peptide isolated from human saliva is histidine-rich that result in forming an unusual cyclic β-hairpin (Gibbs
AMPs and also effective against Candida albicans (Xu et al. et al. 1998). Cbf-14, derived from cathelicidin-BF, is effec-
1991). Cathelicidins, a family of endogenous AMPs, are tive against antibiotic-resistant bacteria. In this AMP, lysine
proline-rich and show irregular structures (Tomasinsig and or leucine was substituted with similar unnatural amino
Zanetti 2005). Indolicidin, a cationic AMP that consists of acids such as ornithine (Orn) and norleucine (Ile) to gener-
only 13 amino acids, has the highest tryptophan content, and ate AMPs with enhanced antibacterial activities. The data
tritripticin like indolicidin is rich in tryptophan (Bacalum indicated that the mutant of Cbf-14 possesses potent anti-
et al. 2017; Falla et al. 1996). Bactenecins are a class of bacterial activities against penicillin-resistant bacteria (Kang
arginine-rich AMPs of bovine neutrophil granules (Ciociola et al. 2017).
et al. 2018).

AMPs with Unnatural Amino Acids The Mode of AMPs Action

Some of the AMPs contain unnatural amino acids such as It was shown that the main mechanism of AMPs is pre-
peptides from bacteria themselves. Nisin, a lantibiotic, is a sumably to include the establishment of membrane pore or
notable example of AMPs from this group. It is produced ion channel, without stereo-special interactions with chiral
by Lactococcus lactis and contains rare amino acids like receptors (Toke 2005). These observations were also con-
lanthionine, 3-methyllanthionine, dehydroalanine, and dehy- firmed by others that showed D enantiomers of melittin,
drobutyrine (de Vos et al. 1993; Zhang et al. 2014). Nisin Q, magainin 2 amide, and cecropin A were assayed and discov-
consisting of 34 amino acids, is a ribosomally-synthesized ered to show the identical hemolytic and antibacterial effica-
AMP and contains post-translationally modified residues cies as their L counterparts that naturally occurred. Further-
such as lanthionine and dehydroalanine (Fukao et al. 2008). more, the compounds all generated one channel conductance
Leucocin A is another peptide that composes of 37 amino in lipid bilayers, with the L, as well as D analogs, inducing
acids and isolated from Leuconostoc gelidum. It can form the identical extent of conductivity (Boparai and Sharma
an amphiphilic conformation and could interact with cell 2020; Wade et al. 1990). AMPs attain dynamic interchanges
membranes (Fregeau Gallagher et al. 1997). These AMPs in their topologies and structures upon interacting with cell
undergo post-translational modifications and after that, their membranes in microorganisms (Sansom 1998).

13
International Journal of Peptide Research and Therapeutics

The outer surface of eukaryotic cells is composed of external section of membranes. In other words, the AMPs
sphingomyelin phospholipids and zwitterionic phosphati- cumulate and impel the lipid monolayers for continuous
dylcholine, whereas the outer surface of prokaryotic cells bending through the pore so that both lipid head groups as
is negatively charged, because of the existence of teichoic well as inserted peptides line the water core (Rahnamaeian
acid or lipopolysaccharides (Dolis et al. 1997). The elec- 2011). The related mechanisms were illustrated in Fig. 4.
trostatic interactions of AMPs with the negatively charged The permeabilizing non-membrane peptides are mostly
molecules on membranes seem to be the preliminary mecha- represented by the capability for translocating across the
nism responsible for antimicrobial activities (Boparai and cell membranes without permeabilizing the membrane,
Sharma 2020). AMPs apply their activities in host cells whereas the permeabilizing membrane peptides are cati-
through translocating across the cell membranes and pre- onic peptides with the ability to form the transient pores
vent fundamental cellular processes like cell wall synthe- on the biological membranes (Pushpanathan et al. 2013).
sis, enzymatic activities, as well as nucleic acid and protein Particular AMPs inducing trans-membrane pores on the
synthesis (Brogden 2005). Particular other factors like outer membrane of target cells comprise LL-37 (Harder et al.
membrane fluidity, molecular architecture, the concentration 2007), magainins (Hallock et al. 2003), melittin (Yang
of negatively charged molecules, as well as outer membrane et al. 2001), and defensin (Schneider et al. 2005a). AMPs
charge and magnitude also are important for the transport of like mersacidin (Brotz et al. 1995), pyrrhocidin (Kragol
AMPs across the biological membranes (Kondejewski et al. et al. 2001), indolicidin (Friedrich et al. 2001), pleuro-
1999). The membrane fluidity was determined to regulate cidin (Patrzykat et al. 2002), HNP-1 (Lee et al. 2002),
the insertion and adsorption of antimicrobial peptides into dermaseptin (Patrzykat et al. 2002), and buforin II (Park
the cell membranes. et al. 2000) get translocated across the biological mem-
According to the mechanisms of action, AMPs are branes and suppress important cellular processes that
classified widely into non-membrane acting and mem- result in cells death. Besides, some AMPs like lactofer-
brane acting peptides. Three mechanisms were suggested rin (Patrzykat et al. 2002), histatin (Kavanagh and Dowd
for pore-forming by membrane acting peptides (Boparai 2004), melittin (Park and Lee 2010), and papiliocin
and Sharma 2020). The barrel-stave mechanism refers (Hwang et al. 2011) apply their antimicrobial activity by
to insert AMPs into the biological membrane hydropho- the formation of reactive oxygen species.
bic substance that flip inward and create pores through
producing trans-membrane helical bundles (Peters et al.
2010). In the carpet-like mechanism, AMPs destruct the
cell membranes assembly via their collaborative activity. Human AMPs
In this path, AMPs self-associate onto the acidic phospho-
lipids-rich areas located at lipid bilayer, and as soon as Human AMPs protect the human from microbial infec-
their concentrations approach specific thresholds, they can tion by various mechanisms. They have been identified
permeate into biological membranes. This phenomenon is in a variety of tissues or surfaces such as eyes, skin, ears,
facilitated via escalating in the positive potential of lipid mouth, lungs, intestines, and also the urinary tract (Wang
bilayers (Mookherjee and Hancock 2007). Toroidal refers 2014). Some of the most important ones with more details
to build toroidal pore in a lipid bilayer by the AMPs. Pore are summarized here. Some important features of these
constructions are administrated through the helix bundles AMPs are summarized in Table 1.
and lipid polar head groups, which vertically orient to the

Fig. 4  The pore-forming
mechanisms of antimicrobial
peptides

13

Table 1  Some important features of human antimicrobial peptides


AMPs family Structure Name Source Active against Mode of action References

13
α-Defensins β-Sheet structure, with six HNP (1–4) Neutrophils, late promyelo- Gram-negative and Gram- Mainly membrane per- Agerberth and Guðmundsson
cysteine residues, form cytes, bone marrow, some positive bacteria (Both meabilization and disrupt (2006), Cunliffe (2003), De
three intramolecular surface epithelial cells, intracellular and extracel- bacterial membranes Smet and Contreras (2005),
disulfide bonds, arginine- intestinal epithelial cells lular organisms), Fungi, Ganz (2001), Schneider
rich peptides (The female reproductive yeast, viruses, parasites et al. (2005)
tract)
HD 5,6 Intestinal Paneth cells Their spectrum of activity is
Paneth cells of the normal similar to that of neutrophil
duodenum, jejunum, and defensins
ileum
β-Defensins β-Sheet structure, with six hBD-1 Leukocytes and epithelial Poor antibacterial activity, ‘Carpet model’, bind to Agerberth and Guðmunds-
cysteine residues, form cells, keratinocytes antiviral against HIV, and the negatively charged son (2006), De Smet and
three intramolecular antifungal activity cytoplasmic membranes Contreras (2005), Pazgier
disulfide bonds and disrupt their integ- et al. (2006a), Schneider
rity, leading to leakage of et al. (2005)
intracellular components
and also inhibit the DNA,
RNA, and protein synthe-
sis or form channel-like
pores spanning across the
membranes
hBD-2 Leukocytes and epithelial Gram-negative and Gram-
cells, keratinocytes, the positive bacteria
gingival mucosa, and the Fungi, yeast, antiviral against
tracheal epithelium HIV viruses
hBD-4 Epithelia and in neutrophils, Weak antimicrobial activity
neutrophils and the epi- against E. coli, S. cerevi-
thelia of the thyroid gland, siae, S. aureus, S. pneu-
the lung, the uterus, and moniae, and B. cepacea,
the kidney, testis and the and strong antimicrobial
gastric antrum activity against S. carnosus
and P. aeruginosa
Human cathelicidin Simple, linear, amphipathic, LL37 The large variety of cells Gram-positive and Gram- Membrane perturbation (The Agerberth and Guðmunds-
cysteine-free α-helices (neutrophils, negative bacteria fungi, barrel stave model and the son (2006), De Smet and
structure leukocytes and epithelial and virus (Only little viral carpet model), detergent- Contreras (2005), Dürr et al.
cells, Myelocytes, meta- inactivation: herpes sim- like effect, non-pore (2006), Lee et al. (2011),
myelocytes, etc.), tissues, plex virus) and parasitic carpet-like mechanism, Neville et al. (2006), Oren
body fluids, and profes- pathogens, the yeast cell transmembrane pores et al. (1999), Thennarasu
sional phagocytes induced et al. (2010)
International Journal of Peptide Research and Therapeutics
International Journal of Peptide Research and Therapeutics

Human Defensins

(2017b), Shah et al. (2016)


(2005), Kalmodia et al.
(2019), Khurshid et al.
De Smet and Contreras α-Defensins were the first human group of AMPs to be
characterized which were isolated from human blood.
They are mostly cationic peptides containing between
29 and 35 amino acid residues. They have six cysteine
References

residues that form three disulphide bonds. The cysteine


bridges are between C1–C6, C2–C4, and C3–C5 that form
cyclic peptides with a typical structure of a triple-stranded
β-sheet and a β-hairpin loop (Ryley 2001; Wang 2014).
ing to the Trk1 potassium
against bacteria and viruses to the loss of intracellular

potassium ions (via bind-


membrane and releasing

Based on the source, property, and size, these peptides


membrane, which leads
Disruption of the plasma

components. For yeast


cell by damaging the

α-defensins are categorized into four groups: (I) HNP-1,


HNP-2, and HNP-3; these three AMPs have almost iden-
Mode of action

tical amino acid sequences. In comparison with HNP-2,


transporter)

HNP-1 and HNP-3 contain only one additional residue at


the N-terminus: alanine in HNP-1 and aspartate in HNP-
3. (II) The fourth human neutrophil defensin, HNP-4, has
a distinct peptide sequence with 33 amino acid residues.
(III) HD-5 was obtained from human Paneth cells and
Especially active against

present in the female reproductive tract. (IV) HD-6 ones


fungi but also active

are only expressed in the Paneth cells of human intestines


(Schneider et al. 2005; Wang 2014). The human β-defensin
Active against

family (hBD) was different from α-defensins. The disulfide


bonds of β-defensins are between C1–C5, C2–C4, and
C3–C6. Also, these AMPs have a slightly longer sequence
than α-defensins that leads to form an additional helical
region. The related structures are illustrated in Fig. 5.
Histatin 1, 3, 5 Salivary glands (parotid and
submandibular glands)

Spectrum of Activity

They are active against a wide spectrum of both Gram-


Source

positive and -negative bacteria, mycobacteria, fungi, and


some enveloped viruses (Winter and Wenghoefer 2012).
For example, at concentrations higher than 100 μg/ml,
HNP-1 and HNP-2 could kill the bacteria, including both
Name

intracellular and extracellular microorganisms (Lehrer


et al. 1993). HNP-1 exerts potent in vitro microbicidal
activity against a wide range of human pathogens, such
Small, cationic, histidine-

as Staphylococcus aureus. Based on the evidence, HNP-1


could target and disrupt the bacterial membrane (Xiong
rich polypeptides

et al. 1999). HNP1–4 and HD-5 show antibacterial activi-


ties against Gram-positive bacteria, such as S. aureus, and
also Gram-negative bacteria as Enterobacter aerogenes
Structure

and Escherichia coli (Ericksen et al. 2005). HNP1 was


also highly effective against Mycobacterium tuberculosis
(Sharma et al. 2000, 2001). HNP1–3 by binding to the
Table 1  (continued)

lethal factor of the anthrax pathogen, Bacillus anthracis,


was able to inhibition of its enzymatic activity (Verma
AMPs family

et al. 2007). HD-5 shows potent antimicrobial activities


Histatins

against a wide range of pathogens such as E. coli, Listeria

13
International Journal of Peptide Research and Therapeutics

Fig. 5  The related structures of


human defensins

monocytogenes, Salmonella typhimurium, and C. albi- anti-viral activities and at natural levels they could provide
cans, whereas minimal inhibitory concentration (MIC) a minimal level of defense against viral infections (Park
values are in the nanomolar range (Porter et al. 1997). et al. 2018).
hBD-1, -2, -3 showed antibacterial activities against E.
coli and S. aureus in a dose-dependent manner (Chen et al.
2005). hBD-3 showed a broad spectrum of antimicrobial Mechanism of Action
activities against different pathogenic microbes such as
multiresistant S. aureus, vancomycin-resistant Enterococ- It was assumed that the antibacterial activities the defensins
cus faecium, Pseudomonas aeruginosa, Klebsiella pneu- are composed of a two-stage mode of action. It should be
monia, Streptococcus pneumoniae, and Burkholderia noted that there is the same mechanism of action for both
cepacia. This AMP has also antifungal activities against types of defensins. At first, they bond to the outer membrane
Candida glabrata and synergistic effects with fluconazole of the bacteria that leads to access to the inner (cytoplas-
were observed (Dhople et al. 2006; Harder et al. 2001; mic) membrane. Then, by internalization into the cytoplas-
Inthanachai et al. 2020; Pazgier et al. 2006b). The in vitro mic membrane, they form channels in the bacterial surfaces
activities of hBD-3 alone or combined with other antimi- (Ryley 2001).
crobial agents were investigated and the results showed In more detail, these AMPs interact with the divalent cati-
that it was effective against Streptococcus mutans, Strep- onic binding sites such as ­Ca2+ and ­Mg2+ in the lipopolysac-
tococcus sanguinis, Streptococcus sobrinus, Lactobacillus charide of bacterial surface and displacing these cations.
acidophilus, and Porphyromonas gingivalis. The bacte- In the case of Gram-positive bacteria, AMPs could interact
ricidal activities were enhanced in combination with the with the anionic lipoteichoic acid of the cell resulting in
antimicrobial agents (Maisetta et al. 2003). Antimicrobial access to the cytoplasmic membrane (Malanovic and Lohner
effect of hBD-4 on Fusobacterium nucleatum and P. gingi- 2016). It should be noted that the size of AMPs has direct
valis has been studied and the results showed antibacterial influences on the distortion of the outer layer and then access
activity (Zhai et al. 2019). hBD‐4 exhibits potent anti- to the underlying cytoplasmic layer (White et al. 1995).
bacterial activity against P. aeruginosa (MIC = : 4.1 μg/ The second stage is similar for both Gram-positive and
ml) (Garcia et al. 2001). These AMPs also showed the -negative bacteria. The cationic residues interact with the

13
International Journal of Peptide Research and Therapeutics

negatively charged membrane and then because of high elec- inactive against some bacteria, like B. cepacia, that is natu-
trical potential, the AMPs being inserted into the membrane. rally resistant to cationic peptides (Ryley 2001). In vitro anti-
Following the aggregation of AMPs in the membrane, they bacterial activities, LL-37 against Legionella pneumophila
could form the channels, and finally, leads to membrane per- was tested and the results indicated that the AMP displayed
meabilization and disruption (Ryley 2001). broad-spectrum and in vitro activity against L. pneumoph-
ila (Birteksoz-Tan et al. 2019). Synergic enhancement of
activity was observed between LL-37 and alpha-defensin
Mechanism of Bacterial Resistance against both E. coli and S. aureus (Nagaoka et al. 2000).
In a study, the antibacterial effects of this peptide were
The mechanisms of antibacterial resistance are poorly under- evaluated against methicillin-resistant S. aureus (MRSA)
stood, however, it was assumed that changing in LPS struc- and multidrug-resistant P. aeruginosa. Additionally, the
ture leads to make it less susceptible to AMPs, and therefore antipseudomonal activities of colistin or imipenem com-
binding of peptides is restricted and antibacterial resistance bined to LL-37 were also studied. The results of the study
will be developed (Ryley 2001). revealed the rapid antibacterial effects of LL-37 against
both antibiotic susceptible and resistant bacterial strains.
When antibiotics were combined with LL-37, the MIC val-
Cathelicidins ues of colistin and imipenem decreased up to eight-fold and
four-fold, respectively (Geitani et al. 2019). In a study, the
Cathelicidins, along with defensins belong to the group of in vitro anti-Helicobacter pylori activity of LL-37 in simu-
cationic AMPs with amphipathic properties and recognize lated gastric juice was assessed and the results showed that
as an integral part of the immune system (De Smet and Con- after incubation the antibacterial activity was not retained
treras 2005). They are mainly stored in neutrophil and mac- (Leszczynska et al. 2009). It was shown that LL-37 and
rophage granules and show a direct antimicrobial activity its fragments exerted antibacterial activities against drug-
against a wide range of microbial pathogens via the oxygen- resistant Acinetobacter baumannii strains. This AMP and
independent activities (Bals and Wilson 2003; Tomasinsig two of its fragments also could inhibit biofilm formation
and Zanetti 2005). Despite a large number of  cathelici- by A. baumannii (Feng et al. 2013). Microscopy studies
din family members in animals, to date, only a single catheli- show that the treatment of fluconazole-resistant Candida
cidin, LL-37, has been known in humans (Ryley 2001). This strains with LL-37 causes Candida cells to undergo surface
AMP is encoded by the cathelicidin gene (CAMP) which is changes that indicating surface membrane damage (Durnas
composed of 39 residues, two leucine residues at N-terminal, et al. 2016). LL-37 is also effective against mycobacteria and
and 37 residues long and has a molecular weight of 18 kDa can kill Mycobacterium smegmatis, Mycobacterium bovis
(Agerberth et al. 1995). Subsequently proteolysis of the pre- BCG, and Mycobacterium tuberculosis under in vitro growth
cursor, possibly by elastase digestion, in the neutrophil and conditions, additionally, this AMP reduced the intracellular
missing the terminal residues, a 37 amino acid peptide was survival of the mycobacteria remarkably (Sonawane et al.
released and consequently termed LL-37 (Gudmundsson 2011).
et al. 1996).
LL-37 has an α-helix structure that lacks cysteine. There-
fore, it has a linear structure without disulphide bonds. It is Mechanism of Action
expressed in leukocytes such as neutrophils, monocytes, NK
cells, T cells, and B cells, and also, like hBD-2, in human They act as antimicrobial agents by either directly killing the
epithelial tissue such as testis, skin, and the gastrointestinal pathogen or indirectly by binding to the bacterial exopoly-
and respiratory tracts in the presence of inflammation and saccharides, outer bacterial cell wall components such as
is possibly related to the interleukin-6 production (De Smet the lipopolysaccharide (LPS) layer in the Gram-negative or
and Contreras 2005; Frohm Nilsson et al. 1999). the teichoic acids in the Gram-positive bacteria (Ramana-
than et al. 2002; Xhindoli et al. 2016). Amphipathicity is an
important factor for effective antibacterial properties. This
Spectrum of Activity attitude is more pronounced for α-helical cathelicidins such
as LL-37. These peptides can form ion channels or aque-
LL-37 shows a wide spectrum of antibacterial activities ous pores in the bacterial membrane and rapidly permeabi-
against both Gram-positive and negative bacteria, with the lize the membranes of microbial pathogens (Gennaro et al.
MIC values lower than those of defensins (Turner et al. 1998; Oren et al. 1999). Additionally, it was observed that
1998). However, unlike defensins, it presents little activ- LL-37 could strongly bind to zwitterionic or acidic phos-
ity against C. albicans or herpesviruses. This AMP is also pholipid membranes of the vesicles and leads to leakage of

13
International Journal of Peptide Research and Therapeutics

vesicular content (Oren et al. 1999). Then the peptide forms These AMPs consist of several members, however,
quite sizeable toroidal pores. In more detail, at the first step, among them, histatin 1, 3, and 5 are the most important ones.
LL-37 is electrostatically attracted by membranes followed They have linear structures and composed of 38, 32, and 24
by assembly and partial integration. In the next step and after amino acid residues for histatin 1, 3, and 5, respectively and
full integration into the lipid bilayer, the peptide forms chan- each of them contains seven histidine residues. Histatin 1
nels based on peptide-peptide and peptide-lipid interactions. and 3 are encoded by two highly related genes, HIS1 and
These interactions initiate the conformational changes and HIS2, respectively and others are produced by the cleavage
the formation of fiber-like oligomers on the inner membrane. of these two peptides. For example, histatin 5 was proteo-
The fibers lead to increasing the local concentration of the lytic digestion of histatin 3 (Wang 2014). Histatin 5 has a
peptide that finally will interfere with the bacterial mem- unique secondary structure. This AMP forms a random coil
brane stability. These transient pores allow the peptide to structure in aqueous solvents while in non-aqueous solvents
translocate into the bacterium. At this site, the peptide may the structure converts to α-helix (Helmerhorst et al. 1997;
interfere with internal targets such as DNA and also vital Raj et al. 1998).
processes such as transcription (Brogden 2005; Wang 2008;
Xhindoli et al. 2016). Moreover, in Gram-negative bacteria,
LPS may be translocated apart from the bacterial cell wall to Spectrum of Activity
build holes for the LL-37 translocation into the periplasmic
space (Vandamme et al. 2012). These peptides possess some bactericidal activities and,
more importantly, fungicidal properties. It should be noted
that of all histatins, histatin 5 has the strongest antimicrobial
Mechanism of Bacterial Resistance activity, and most of the research on histatins has focused on
this peptide (Khurshid et al. 2016; Oppenheim et al. 1988).
It was assumed that the changes in the structure of LPS The in vitro study indicated that histatin 5 could inhibit Can-
in Gram-negative bacteria affect the binding properties of dida species (C. albicans, C. glabrata, C. krusei) at a certain
LL-37. For example, surface structures containing phos- concentration (15–30 μM). It was also demonstrated that the
phorylcholine, a component of eukaryote cell membranes, analog of histatin 3 with the shorter amino acid sequences
have been found in some respiratory pathogens such as Hae- has the same candidacidal activity with respect to the full-
mophilus influenza. The Mutants that do not express this length molecule (Raj et al. 1990).
structure were 1000-fold more sensitive to LL-37 than those These AMPs possess their anti-fungal activities in differ-
having this lipid in their membrane (Lysenko et al. 2000). ent ways such as growth inhibitory actions on C. albicans or
It was also shown that Neisseria gonorrhoeae has a type inhibition of the conversion of C. albicans yeast growth into
of efflux pump system that can reduce the susceptibility of hyphal growth (Moffa et al. 2015).
the bacteria to LL-37 (Miyasaki et al. 1990). In Bordetella The antimicrobial activities against other pathogens
pertussis, d-alanine incorporation induces resistance to Cryptococcus neoformans and Aspergillus fumigatus has
the AMPs. It was demonstrated that the dra operon of B. also been reported (Helmerhorst et al. 1999b). Also, hista-
pertussis is responsible for the d-alanylation of the outer tin 5 virucidal activity and only histatin 5 derived peptides
membrane component. Mutation in this operon results in affect HIV-1 (Wang 2014). Inhibitory and bactericidal
increasing sensitivity of bacteria to LL-37, and other AMPs activities of histatins have also been reported against various
(Taneja et al. 2013). bacteria, including S. mutans, P. gingivalis, P. aeruginosa,
Additionally, the antibacterial resistance against AMPs E. coli, and S. aureus (Gusman et al. 2001; MacKay et al.
might be due to the production of degradation enzymes such 1984; Sajjan et al. 2001).
as proteases AMPs. Bacterial strains can promote bacterial
resistance to the AMPs by their proteolytic degradation
properties (Abdi et al. 2019). Mechanism of Action

Unlike other AMPs, it is well-known that all targets of


Histatin histatins are intracellular and the primary mode of actions
is not related to lyse lipid membranes (Puri and Edgerton
Histatins are a family of cationic small AMPs that are largely 2014; Ryley 2001; Wang 2014). It has been found that the
composed of histidine-rich repeats. They have a molecular massive non-lytic release of ATP along with the decrease in
weight of about 3–4 kDa and are produced by the subman- intracellular ATP levels is the main mechanism of actions
dibular, sublingual, and parotid glands and secreted into of histatins (Helmerhorst et al. 1999a; Koshlukova et al.
human saliva (De Smet and Contreras 2005). 1999). It was shown that histatin 5 could bind to the cell

13
International Journal of Peptide Research and Therapeutics

wall proteins and glycans of C. albicans and then is taken AMPs Challenges
up by the fungal polyamine transporters in the cells in an
energy-dependent manner. Inside the fungal cells, the AMP Although AMPs often present antibacterial activities,
may affect the mitochondrial functions and leads to oxidative own several unfavorable features for clinical applications,
stress. However, cell death is the result of other processes including (I) the pharmacokinetic profiles of antimicro-
such as volume dysregulation and ion imbalance caused bial peptides are not well known, and only a few clinical
by osmotic stress. Additionally, the metal-binding ability research have been performed with AMPs; (II) sensitivity
of histatin 5 is the other mentioned mechanism (Edgerton to proteolysis process derived by bacterial proteases; and
and Koshlukova 2000; Khurshid et  al. 2017a; Komatsu (III) cytotoxicity to the eukaryotic cell, which could result
et al. 2011; Puri and Edgerton 2014). Taken together, the in neurotoxicity, nephrotoxicity, and hemolysis (Craik
key steps in the histatin 5 antifungal activity involve a bio- et  al. 2013; Grassi et  al. 2017). Nevertheless, several
energetic collapse of C. albicans, consequently decreasing types of research have been conducted for improving our
the mitochondrial ATP synthesis. knowledge of the above-mentioned challenges (Chou et al.
The other reported fungistatic and fungicidal mechanisms 2008). In comparison to conventional antibiotics, AMPs
include disruption of the plasma membrane, which leads are also expensive for being synthesized on mass scales
to the loss of intracellular constituents (Oppenheim et al. (Otvos and Wade 2014). Various strategies have been per-
1988). It was also found that these AMPs were also effective suaded to overcome these limitations. For this scenario,
in killing the yeast cells by damaging their membranes and employing nanocarriers, developing pegylated AMPs, and
releasing potassium ions. These effects were related to the covalent immobilization of AMPs on surfaces have been
binding to the Trk1 potassium transporter and then the loss mentioned (Long et al. 2017; Manteghi et al. 2020).
of intracellular components (Swidergall and Ernst 2014). Covalent attachment of the polyethylene glycol polymer
The other modifications caused by histatin 5 in C. albicans to peptides and proteins (PEGylation) could improve the
included organelles in disarray, intracellular membrane dis- bioavailability of AMPs and enhance their pharmacoki-
arrangement, and central cavities with deformed structures netic properties such as bio-distribution and rate of clear-
displaced to the cell periphery (Isola et al. 2007). ance. Additionally, the proteolytic degradation of AMPs
It was found that the histatins, like other AMPs, could might be decreased by protecting their C- and N-terminus
interact with extracellular actin and the extracellular actin (Gong et al. 2015; Gonzalez-Valdez et al. 2012; Khameneh
might regulate histatin anti-fungal activities in the oral cav- et al. 2015b).
ity. In a study, it was demonstrated that both histatin 3 and The progress in nanotechnology has permitted various
5 interact with actin, however, and affect the actin structure nanoparticles to work as a favorable approach for minimiz-
(Blotnick et al. 2017). ing the unfavorable features of synthetic as well as natural
AMPs (Umerska et al. 2017). The researchers have sug-
gested that AMPs in nanoparticles represent increased effi-
ciency, decreased degradation, and lower toxicity (Wang
Mechanism of Microbial Resistance et al. 2017). As a result, nanoparticles could contribute to
the manufacturing of AMPs and their applications in the
It was shown that cellular accumulation of the peptides is industry. Nano-carriers are considered as a drug delivery
necessary for anti-fungal activities and also that accumu- system that presents many profits, like improvement of
lation of histatins depends on the availability of cellular drug pharmacokinetic profile, treatment selectivity, and
energy. Therefore, respiratory mutants of the microorgan- the protection of AMPs against extracellular degradations
isms which have undergone mutations in mitochondrial (Sadat et al. 2016). Over the last 50 years, multiple drug
DNA exhibited resistance against histatins (Gyurko et al. delivery systems have been used for encapsulating drugs
2000). Therefore, it was assumed that the altered membrane and other biomolecules, including polymeric nanopar-
energetics is an important factor for developing resistance ticles, dendrimers, micelles, and liposomes (Malaekeh-
(Yeaman and Yount 2003). Additionally, histatin 5 is trans- Nikouei et al. 2020).
ported out of C. albicans cells by the Flu1 and Mrr1 efflux Nanobiotechnology has proposed two major proce-
pumps (Hampe et al. 2017; Li et al. 2013). The multidrug dures to encapsulate AMPs. The passive delivery, as non-
resistance transporters CgTpo1_1 and CgTpo1_2 have criti- directed procedures, includes traditional nano-delivery
cal roles in the virulence of C. glabrata infections such as systems that do not own surface modifications for guiding
resistance to histatin 5 (Santos et al. 2017). It was demon- the nano-carriers; this could be manipulated by control
strated that the upregulation of CgCDR efflux pumps could the nano-carriers shape as well as size (Makowski et al.
develop the resistance to histatin 5 in C. glabrata (Helmer- 2019). Active targeting, as directed delivery, includes
horst et al. 2006).

13
International Journal of Peptide Research and Therapeutics

surface modifications of the nano-carriers with various Data Availability  This review was based on data extracted from pub-
ligands and/or other moieties for permitting interaction lished papers available in all relevant databases without limitation up
to 1st July 2020.
of nano-carriers and the target sites (Biswaro et al. 2018).
However, developing viable drug delivery systems for
Compliance with Ethical Standards 
clinical trials remains a challenge (Wimley and Hristova
2011). There are two procedures of nano-delivery systems Conflict of interest  The authors declare that they have no competing
that have some advantages and disadvantages (Makowski interests.
et al. 2019). Passive systems attend to own fewer agents in
their combination in contrast to active systems (Biswaro
et al. 2018). As passive systems possess fewer agents in
their combination, this causes it easier for preparing them References
(Makowski et al. 2019). Besides, active systems contain
modified surface-carrying ligands and/or other moieties for Abdi M, Mirkalantari S, Amirmozafari N (2019) Bacterial resistance
facilitating its interactions with infected cells and increase to antimicrobial peptides. J Pept Sci 25:e3210
Agerberth B, Guðmundsson G (2006) Host antimicrobial defence
the drug transports at such particular sites (Biswaro et al. peptides in human disease. Antimicrobial peptides and human
2018), while passive systems involve encapsulating pep- disease. Springer, Berlin, pp 67–90
tides without making available extra surface modifications Agerberth B, Gunne H, Odeberg J, Kogner P, Boman HG, Gudmunds-
(Hunter et al. 2012). Given the above-mentioned, the terms son GH (1995) FALL-39, a putative human peptide antibiotic, is
cysteine-free and expressed in bone marrow and testis. Proc Natl
nano-delivery system as well as nano-carriers have been Acad Sci USA 92:195–199
suggested (Makowski et al. 2019). Bacalum M, Janosi L, Zorila F, Tepes AM, Ionescu C, Bogdan E et al
(2017) Modulating short tryptophan- and arginine-rich peptides
activity by substitution with histidine. Biochim Biophys Acta
1861:1844–1854
Bahar AA, Ren D (2013) Antimicrobial peptides. Pharmaceuticals
Conclusion (Basel) 6:1543–1575
Bals R, Wilson JM (2003) Cathelicidins—a family of multifunctional
Clinical overuse of antibiotics unavoidably results in the antimicrobial peptides. Cell Mol Life Sci 60:711–720
Bazzaz BSF, Fakori M, Khameneh B, Hosseinzadeh H (2019) Effects
growing emergence of drug-resistant strains of microbial of omeprazole and caffeine alone and in combination with gen-
pathogens. Consequently, the development of a new class tamicin and ciprofloxacin against antibiotic resistant Staphylo-
of antibiotics is an urgent need. During the last decades, coccus aureus and Escherichia coli strains. J Pharmacopuncture
considerable efforts have been conducted for investigating 22:49–54
Birteksoz-Tan AS, Zeybek Z, Hacioglu M, Savage PB, Bozkurt-Guzel
the possible use of AMPs or synthetic derivatives as thera- C (2019) In vitro activities of antimicrobial peptides and cera-
peutic antibacterial agents. They are implicated in several genins against Legionella pneumophila. J Antibiot 72:291–297
biological processes, and also have an important role in the Biswaro LS, da Costa Sousa MG, Rezende TMB, Dias SC, Franco
innate immune system. The AMPs protect the host against OL (2018) Antimicrobial peptides and nanotechnology, recent
advances and challenges. Front Microbiol 9:855
both systemic and topical infections either alone or in com- Blotnick E, Sol A, Bachrach G, Muhlrad A (2017) Interactions of hista-
bination with conventional antibiotics. Human AMPs are tin-3 and histatin-5 with actin. BMC Biochem 18:3
an important class of these peptides which attract attention Boparai JK, Sharma PK (2020) Mini review on antimicrobial peptides,
for the treatment of various disease and some of them have sources, mechanism and recent applications. Protein Pept Lett
27:4–16
entered clinical trials for use in clinical applications such Brogden KA (2005) Antimicrobial peptides: pore formers or metabolic
as the treatment of diabetic ulcers as topical anti-infective inhibitors in bacteria? Nat Rev Microbiol 3:238–250
agents for the treatment of microbial infections. The ongo- Brogden KA, Ackermann M, McCray PB Jr, Tack BF (2003) Antimi-
ing discovery and development of new AMPs are promis- crobial peptides in animals and their role in host defences. Int J
Antimicrob Agents 22:465–478
ing approaches in the fight against increasingly resistant Brotz H, Bierbaum G, Markus A, Molitor E, Sahl HG (1995) Mode of
pathogens. action of the lantibiotic mersacidin: inhibition of peptidoglycan
biosynthesis via a novel mechanism? Antimicrob Agents Chem-
other 39:714–719
Author Contributions  BK collaborated in the original idea, concept, Bulet P, Stocklin R, Menin L (2004) Anti-microbial peptides: from
design, and writing and drafting the article. SS and NHG contributed invertebrates to vertebrates. Immunol Rev 198:169–184
with data interpretation, writing, and drafting of the article. BSFB Chaudhary AS (2016) A review of global initiatives to fight antibiotic
contributed to all stages of the process and mainly participated in draft- resistance and recent antibiotics discovery. Acta Pharm Sin B
ing the article, writing, and editing the final version to be published. 6:552–556
All the authors read and approved the final version of the manuscript. Chen X, Niyonsaba F, Ushio H, Okuda D, Nagaoka I, Ikeda S et al
(2005) Synergistic effect of antibacterial agents human beta-
defensins, cathelicidin LL-37 and lysozyme against Staphylo-
Funding  The study was not funded by any authority. coccus aureus and Escherichia coli. J Dermatol Sci 40:123–132

13
International Journal of Peptide Research and Therapeutics

Chou HT, Kuo TY, Chiang JC, Pei MJ, Yang WT, Yu HC et al (2008) protein (hCAP18), a peptide antibiotic, is widely expressed in
Design and synthesis of cationic antimicrobial peptides with human squamous epithelia and colocalizes with interleukin-6.
improved activity and selectivity against Vibrio spp. Int J Anti- Infect Immun 67:2561–2566
microb Agents 32:130–138 Fukao M, Obita T, Yoneyama F, Kohda D, Zendo T, Nakayama J et al
Ciociola T, Giovati L, Giovannelli A, Conti S, Castagnola M, Vitali A (2008) Complete covalent structure of nisin Q, new natural nisin
(2018) The activity of a mammalian proline-rich peptide against variant, containing post-translationally modified amino acids.
Gram-negative bacteria, including drug-resistant strains, relies Biosci Biotechnol Biochem 72:1750–1755
on a nonmembranolytic mode of action. Infect Drug Resist Ganz T (2001) Defensins in the urinary tract and other tissues. J Infect
11:969–979 Dis 183:S41–S42
Craik DJ, Fairlie DP, Liras S, Price D (2013) The future of peptide- Ganz T, Selsted ME, Szklarek D, Harwig SS, Daher K, Bainton DF
based drugs. Chem Biol Drug Des 81:136–147 et al (1985) Defensins. Natural peptide antibiotics of human neu-
Cunliffe RJ (2003) α-Defensins in the gastrointestinal tract. Mol Immu- trophils. J Clin Invest 76:1427–1435
nol 40:463–467 Garcia JR, Krause A, Schulz S, Rodriguez-Jimenez FJ, Kluver E, Ader-
De Smet K, Contreras R (2005) Human antimicrobial peptides: mann K et al (2001) Human beta-defensin 4: a novel inducible
defensins, cathelicidins and histatins. Biotechnol Lett peptide with a specific salt-sensitive spectrum of antimicrobial
27:1337–1347 activity. FASEB J 15:1819–1821
de Vos WM, Mulders JW, Siezen RJ, Hugenholtz J, Kuipers OP (1993) Geitani R, Ayoub Moubareck C, Touqui L, Karam Sarkis D (2019)
Properties of nisin Z and distribution of its gene, nisZ, in Lacto- Cationic antimicrobial peptides: alternatives and/or adjuvants to
coccus lactis. Appl Environ Microbiol 59:213–218 antibiotics active against methicillin-resistant Staphylococcus
Dhople V, Krukemeyer A, Ramamoorthy A (2006) The human beta- aureus and multidrug-resistant Pseudomonas aeruginosa. BMC
defensin-3, an antibacterial peptide with multiple biological Microbiol 19:54
functions. Biochim Biophys Acta 1758:1499–1512 Gennaro R, Scocchi M, Merluzzi L, Zanetti M (1998) Biological char-
Dimarcq JL, Bulet P, Hetru C, Hoffmann J (1998) Cysteine-rich anti- acterization of a novel mammalian antimicrobial peptide. Bio-
microbial peptides in invertebrates. Biopolymers 47:465–477 chim Biophys Acta 1425:361–368
Dolis D, Moreau C, Zachowski A, Devaux PF (1997) Aminophos- Giangaspero A, Sandri L, Tossi A (2001) Amphipathic alpha helical
pholipid translocase and proteins involved in transmembrane antimicrobial peptides. Eur J Biochem 268:5589–5600
phospholipid traffic. Biophys Chem 68:221–231 Gibbs AC, Kondejewski LH, Gronwald W, Nip AM, Hodges RS, Sykes
Durnas B, Wnorowska U, Pogoda K, Deptula P, Watek M, Piktel E et al BD et al (1998) Unusual beta-sheet periodicity in small cyclic
(2016) Candidacidal activity of selected ceragenins and human peptides. Nat Struct Biol 5:284–288
cathelicidin LL-37 in experimental settings mimicking infection Gong Y, Leroux JC, Gauthier MA (2015) Releasable conjugation of
sites. PLoS ONE 11:e0157242 polymers to proteins. Bioconjug Chem 26:1172–1181
Dürr UH, Sudheendra U, Ramamoorthy A (2006) LL-37, the only Gonzalez-Valdez J, Rito-Palomares M, Benavides J (2012) Advances
human member of the cathelicidin family of antimicrobial pep- and trends in the design, analysis, and characterization of pol-
tides. Biochem Biophys Acta 1758:1408–1425 ymer-protein conjugates for “PEGylaided” bioprocesses. Anal
Edgerton M, Koshlukova SE (2000) Salivary histatin 5 and its simi- Bioanal Chem 403:2225–2235
larities to the other antimicrobial proteins in human saliva. Adv Grassi L, Maisetta G, Esin S, Batoni G (2017) Combination strategies
Dent Res 14:16–21 to enhance the efficacy of antimicrobial peptides against bacterial
Edwards IA, Elliott AG, Kavanagh AM, Blaskovich MAT, Cooper biofilms. Front Microbiol 8:2409
MA (2017) Structure-activity and -toxicity relationships of the Gudmundsson GH, Agerberth B, Odeberg J, Bergman T, Olsson B,
antimicrobial peptide tachyplesin-1. ACS Infect Dis 3:917–926 Salcedo R (1996) The human gene FALL39 and processing
Ericksen B, Wu Z, Lu W, Lehrer RI (2005) Antibacterial activity and of the cathelin precursor to the antibacterial peptide LL-37 in
specificity of the six human {alpha}-defensins. Antimicrob granulocytes. Eur J Biochem 238:325–332
Agents Chemother 49:269–275 Gusman H, Travis J, Helmerhorst EJ, Potempa J, Troxler RF, Oppen-
Falla TJ, Karunaratne DN, Hancock RE (1996) Mode of action of the heim FG (2001) Salivary histatin 5 is an inhibitor of both host
antimicrobial peptide indolicidin. J Biol Chem 271:19298–19303 and bacterial enzymes implicated in periodontal disease. Infect
Farhadi F, Khameneh B, Iranshahi M, Iranshahy M (2019) Antibacte- Immun 69:1402–1408
rial activity of flavonoids and their structure-activity relationship: Gyurko C, Lendenmann U, Troxler RF, Oppenheim FG (2000) Can-
an update review. Phytother Res 33:13–40 dida albicans mutants deficient in respiration are resistant to the
Feng X, Sambanthamoorthy K, Palys T, Paranavitana C (2013) The small cationic salivary antimicrobial peptide histatin 5. Antimi-
human antimicrobial peptide LL-37 and its fragments possess crob Agents Chemother 44:348–354
both antimicrobial and antibiofilm activities against multidrug- Hallock KJ, Lee DK, Ramamoorthy A (2003) MSI-78, an analogue
resistant Acinetobacter baumannii. Peptides 49:131–137 of the magainin antimicrobial peptides, disrupts lipid bilayer
Fox JL (2013) Antimicrobial peptides stage a comeback. Nat Biotech- structure via positive curvature strain. Biophys J 84:3052–3060
nol 31:379–382 Hampe IAI, Friedman J, Edgerton M, Morschhäuser J (2017) An
Fregeau Gallagher NL, Sailer M, Niemczura WP, Nakashima TT, Stiles acquired mechanism of antifungal drug resistance simultaneously
ME, Vederas JC (1997) Three-dimensional structure of leucocin enables Candida albicans to escape from intrinsic host defenses.
A in trifluoroethanol and dodecylphosphocholine micelles: PLoS Pathog 13:e1006655
spatial location of residues critical for biological activity in Hancock RE (2001) Cationic peptides: effectors in innate immunity and
type IIa bacteriocins from lactic acid bacteria. Biochemistry novel antimicrobials. Lancet Infect Dis 1:156–164
36:15062–15072 Harder J, Bartels J, Christophers E, Schroder JM (2001) Isolation and
Friedrich CL, Rozek A, Patrzykat A, Hancock RE (2001) Structure and characterization of human beta-defensin-3, a novel human induc-
mechanism of action of an indolicidin peptide derivative with ible peptide antibiotic. J Biol Chem 276:5707–5713
improved activity against gram-positive bacteria. J Biol Chem Harder J, Glaser R, Schroder JM (2007) Human antimicrobial proteins
276:24015–24022 effectors of innate immunity. J Endotoxin Res 13:317–338
Frohm Nilsson M, Sandstedt B, Sorensen O, Weber G, Borregaard Helmerhorst EJ, Van’t Hof W, Veerman EC, Simoons-Smit I, Nieuw
N, Stahle-Backdahl M (1999) The human cationic antimicrobial Amerongen AV (1997) Synthetic histatin analogues with

13
International Journal of Peptide Research and Therapeutics

broad-spectrum antimicrobial activity. Biochem J 326(Pt and molecular modeling of PEGylated human growth hormone
1):39–45 with agonist activity. Int J Biol Macromol 80:400–409
Helmerhorst EJ, Venuleo C, Sanglard D, Oppenheim FG (2006) Roles Khameneh B, Diab R, Ghazvini K, Fazly Bazzaz BS (2016) Break-
of cellular respiration, CgCDR1, and CgCDR2 in Candida throughs in bacterial resistance mechanisms and the potential
glabrata resistance to histaten 5. Antimicrob Agents Chemother ways to combat them. Microb Pathog 95:32–42
50:1100–1103 Khameneh B, Iranshahy M, Soheili V, Fazly Bazzaz BS (2019) Review
Helmerhorst EJ, Breeuwer P, van’t Hof W, Walgreen-Weterings E, on plant antimicrobials: a mechanistic viewpoint. Antimicrob
Oomen LC, Veerman EC et al (1999) The cellular target of hista- Resist Infect Control 8:118
tin 5 on Candida albicans is the energized mitochondrion. J Biol Khameneh B, Iranshahy M, Vahdati-Mashhadian N, Sahebkar A, Fazly
Chem 274:7286–7291 Bazzaz BS (2019) Non-antibiotic adjunctive therapy: a promis-
Helmerhorst EJ, Reijnders IM, van’t Hof W, Simoons-Smit I, Veerman ing approach to fight tuberculosis. Pharmacol Res 146:104289
EC, Amerongen AV (1999) Amphotericin B- and fluconazole- Khurshid Z, Naseem M, Sheikh Z, Najeeb S, Shahab S, Zafar MS
resistant Candida spp., Aspergillus fumigatus, and other newly (2016) Oral antimicrobial peptides: types and role in the oral
emerging pathogenic fungi are susceptible to basic antifungal cavity. Saudi Pharm J 24:515–524
peptides. Antimicrob Agents Chemother 43:702–704 Khurshid Z, Najeeb S, Mali M, Moin SF, Raza SQ, Zohaib S et al
Hill CP, Yee J, Selsted ME, Eisenberg D (1991) Crystal structure of (2017a) Histatin peptides: pharmacological functions and their
defensin HNP-3, an amphiphilic dimer: mechanisms of mem- applications in dentistry. Saudi Pharm J 25:25–31
brane permeabilization. Science 251:1481–1485 Khurshid Z, Najeeb S, Mali M, Moin SF, Raza SQ, Zohaib S et al
Huang Y, Huang J, Chen Y (2010) Alpha-helical cationic antimicrobial (2017b) Histatin peptides: Pharmacological functions and their
peptides: relationships of structure and function. Protein Cell applications in dentistry 25:25–31
1:143–152 Koehbach J, Craik DJ (2019) The vast structural diversity of antimicro-
Hunter AC, Elsom J, Wibroe PP, Moghimi SM (2012) Polymeric bial peptides. Trends Pharmacol Sci 40:517–528
particulate technologies for oral drug delivery and targeting: a Komatsu T, Salih E, Helmerhorst EJ, Offner GD, Oppenheim FG
pathophysiological perspective. Maturitas 73:5–18 (2011) Influence of histatin 5 on Candida albicans mitochondrial
Hwang PM, Vogel HJ (1998) Structure-function relationships of anti- protein expression assessed by quantitative mass spectrometry. J
microbial peptides. Biochem Cell Biol 76:235–246 Proteome Res 10:646–655
Hwang PM, Zhou N, Shan X, Arrowsmith CH, Vogel HJ (1998) Kondejewski LH, Jelokhani-Niaraki M, Farmer SW, Lix B, Kay CM,
Three-dimensional solution structure of lactoferricin B, an anti- Sykes BD et al (1999) Dissociation of antimicrobial and hemo-
microbial peptide derived from bovine lactoferrin. Biochemistry lytic activities in cyclic peptide diastereomers by systematic
37:4288–4298 alterations in amphipathicity. J Biol Chem 274:13181–13192
Hwang B, Hwang JS, Lee J, Kim JK, Kim SR, Kim Y et al (2011) Koshlukova SE, Lloyd TL, Araujo MW, Edgerton M (1999) Salivary
Induction of yeast apoptosis by an antimicrobial peptide, Papili- histatin 5 induces non-lytic release of ATP from Candida albi-
ocin. Biochem Biophys Res Commun 408:89–93 cans leading to cell death. J Biol Chem 274:18872–18879
Inthanachai T, Thammahong A, Edwards SW, Virakul S, Kiatsurayanon Kragol G, Lovas S, Varadi G, Condie BA, Hoffmann R, Otvos L Jr
C, Chiewchengchol D (2020) The inhibitory effect of human (2001) The antibacterial peptide pyrrhocoricin inhibits the
beta-defensin-3 on Candida Glabrata isolated from patients ATPase actions of DnaK and prevents chaperone-assisted protein
with Candidiasis. Immunol Invest. https:​ //doi.org/10.1080/08820​ folding. Biochemistry 40:3016–3026
139.2020.17553​07 Lee MK, Cha L, Lee SH, Hahm KS (2002) Role of amino acid residues
Isola R, Isola M, Conti G, Lantini MS, Riva A (2007) Histatin-induced within the disulfide loop of thanatin, a potent antibiotic peptide.
alterations in Candida albicans: a microscopic and submicro- J Biochem Mol Biol 35:291–296
scopic comparison. Microsc Res Tech 70:607–616 Lee C-C, Sun Y, Qian S, Huang HW (2011) Transmembrane pores
Javia A, Amrutiya J, Lalani R, Patel V, Bhatt P, Misra A (2018) Anti- formed by human antimicrobial peptide LL-37. Biophys J
microbial peptide delivery: an emerging therapeutic for the treat- 100:1688–1696
ment of burn and wounds. Ther Deliv 9:375–386 Lehrer RI, Lichtenstein AK, Ganz T (1993) Defensins: antimicrobial
Kalmodia S, Son K-N, Cao D, Lee B-S, Surenkhuu B, Shah D et al and cytotoxic peptides of mammalian cells. Annu Rev Immunol
(2019) Presence of histatin-1 in human tears and association 11:105–128
with aqueous deficient dry eye diagnosis: a preliminary study. Leszczynska K, Namiot A, Fein DE, Wen Q, Namiot Z, Savage PB
Sci Rep 9:1–10 et al (2009) Bactericidal activities of the cationic steroid CSA-13
Kang W, Liu H, Ma L, Wang M, Wei S, Sun P et al (2017) Effec- and the cathelicidin peptide LL-37 against Helicobacter pylori
tive antimicrobial activity of a peptide mutant Cbf-14-2 against in simulated gastric juice. BMC Microbiol 9:187
penicillin-resistant bacteria based on its unnatural amino acids. Li R, Kumar R, Tati S, Puri S, Edgerton M (2013) Candida albicans
Eur J Pharm Sci 105:169–177 Flu1-mediated efflux of salivary histatin 5 reduces its cytosolic
Kavanagh K, Dowd S (2004) Histatins: antimicrobial peptides with concentration and fungicidal activity. Antimicrob Agents Chem-
therapeutic potential. J Pharm Pharmacol 56:285–289 other 57:1832–1839
Kawano K, Yoneya T, Miyata T, Yoshikawa K, Tokunaga F, Terada Liu W, Hansen JN (1993) The antimicrobial effect of a structural vari-
Y et al (1990) Antimicrobial peptide, tachyplesin I, isolated ant of subtilin against outgrowing Bacillus cereus T spores and
from hemocytes of the horseshoe crab (Tachypleus tridentatus). vegetative cells occurs by different mechanisms. Appl Environ
NMR determination of the beta-sheet structure. J Biol Chem Microbiol 59:648–651
265:15365–15367 Long T, Li H, Wang X, Yue B (2017) A comparative analysis of anti-
Khameneh B, Iranshahy M, Ghandadi M, Ghoochi Atashbeyk D, Fazly bacterial properties and inflammatory responses for the KR-12
Bazzaz BS, Iranshahi M (2015) Investigation of the antibacterial peptide on titanium and PEGylated titanium surfaces. RSC Adv
activity and efflux pump inhibitory effect of co-loaded piperine 7:34321–34330
and gentamicin nanoliposomes in methicillin-resistant Staphy- Lysenko ES, Gould J, Bals R, Wilson JM, Weiser JN (2000) Bacterial
lococcus aureus. Drug Dev Ind Pharm 41:989–994 phosphorylcholine decreases susceptibility to the antimicrobial
Khameneh B, Jaafari MR, Hassanzadeh-Khayyat M, Varasteh A, peptide LL-37/hCAP18 expressed in the upper respiratory tract.
Chamani J, Iranshahi M et al (2015) Preparation, characterization Infect Immun 68:1664–1671

13
International Journal of Peptide Research and Therapeutics

MacKay BJ, Pollock JJ, Iacono VJ, Baum BJ (1984) Isolation of mil- peptides inhibit macromolecular synthesis in Escherichia coli.
ligram quantities of a group of histidine-rich polypeptides from Antimicrob Agents Chemother 46:605–614
human parotid saliva. Infect Immun 44:688–694 Pazgier M, Hoover D, Yang D, Lu W, Lubkowski JJC (2006a) & CMLS
Maisetta G, Batoni G, Esin S, Luperini F, Pardini M, Bottai D et al MLS. Human β-defensins 63:1294–1313
(2003) Activity of human beta-defensin 3 alone or combined Pazgier M, Hoover DM, Yang D, Lu W, Lubkowski J (2006b) Human
with other antimicrobial agents against oral bacteria. Antimicrob beta-defensins. Cell Mol Life Sci 63:1294–1313
Agents Chemother 47:3349–3351 Peters BM, Shirtliff ME, Jabra-Rizk MA (2010) Antimicrobial
Makowski M, Silva IC, Pais do Amaral C, Goncalves S, Santos NC peptides: primeval molecules or future drugs? PLoS Pathog
(2019) Advances in Lipid And Metal Nanoparticles For Antimi- 6:e1001067
crobial Peptide Delivery. Pharmaceutics 11:588 Porter EM, van Dam E, Valore EV, Ganz T (1997) Broad-spectrum
Malaekeh-Nikouei B, Fazly Bazzaz BS, Mirhadi E, Tajani AS, antimicrobial activity of human intestinal defensin 5. Infect
Khameneh B (2020) The role of nanotechnology in combating Immun 65:2396–2401
biofilm-based antibiotic resistance. J Drug Delivery Sci Technol Puri S, Edgerton M (2014) How does it kill?: understanding the
60:101880 candidacidal mechanism of salivary histatin 5. Eukaryot Cell
Malanovic N, Lohner K (2016) Antimicrobial peptides targeting Gram- 13:958–964
positive bacteria. Pharmaceuticals (Basel) 9:59 Pushpanathan M, Gunasekaran P, Rajendhran J (2013) Antimicrobial
Manteghi R, Pallagi E, Olajos G, Csoka I (2020) Pegylation and for- peptides: versatile biological properties. Int J Pept 2013:675391
mulation strategy of anti-microbial peptide (AMP) according to Rahnamaeian M (2011) Antimicrobial peptides: modes of mecha-
the quality by design approach. Eur J Pharm Sci 144:105197 nism, modulation of defense responses. Plant Signal Behav
Miyasaki KT, Bodeau AL, Ganz T, Selsted ME, Lehrer RI (1990) 6:1325–1332
In vitro sensitivity of oral, gram-negative, facultative bacteria Raj PA, Edgerton M, Levine MJ (1990) Salivary histatin 5: dependence
to the bactericidal activity of human neutrophil defensins. Infect of sequence, chain length, and helical conformation for candi-
Immun 58:3934–3940 dacidal activity. J Biol Chem 265:3898–3905
Moffa EB, Mussi MC, Xiao Y, Garrido SS, Machado MA, Giampaolo Raj PA, Marcus E, Sukumaran DK (1998) Structure of human salivary
ET et al (2015) Histatin 5 inhibits adhesion of C. albicans to histatin 5 in aqueous and nonaqueous solutions. Biopolymers
reconstructed human oral epithelium. Front Microbiol 6:885 45:51–67
Mookherjee N, Hancock RE (2007) Cationic host defence peptides: Ramanathan B, Davis EG, Ross CR, Blecha F (2002) Cathelicidins:
innate immune regulatory peptides as a novel approach for treat- microbicidal activity, mechanisms of action, and roles in innate
ing infections. Cell Mol Life Sci 64:922–933 immunity. Microbes Infect 4:361–372
Nagaoka I, Hirota S, Yomogida S, Ohwada A, Hirata M (2000) Syner- Reddy KV, Yedery RD, Aranha C (2004) Antimicrobial peptides:
gistic actions of antibacterial neutrophil defensins and cathelici- premises and promises. Int J Antimicrob Agents 24:536–547
dins. Inflamm Res 49:73–79 Ryley HC (2001) Human antimicrobial peptides. Rev Med Microbiol
Neville F, Cahuzac M, Konovalov O, Ishitsuka Y, Lee KYC, Kuzmenko 12:177–186
I et al (2006) Lipid headgroup discrimination by antimicrobial Sadat SM, Jahan ST, Haddadi A (2016) Effects of size and surface
peptide LL-37: insight into mechanism of action. Biophys J charge of polymeric nanoparticles on in vitro and in vivo applica-
90:1275–1287 tions. J Biomater Nanobiotechnol 7:91
Oppenheim FG, Xu T, McMillian FM, Levitz SM, Diamond RD, Sajjan US, Tran LT, Sole N, Rovaldi C, Akiyama A, Friden PM et al
Offner GD et al (1988) Histatins, a novel family of histidine-rich (2001) P-113D, an antimicrobial peptide active against Pseu-
proteins in human parotid secretion. Isolation, characterization, domonas aeruginosa, retains activity in the presence of sputum
primary structure, and fungistatic effects on Candida albicans. from cystic fibrosis patients. Antimicrob Agents Chemother
J Biol Chem 263:7472–7477 45:3437–3444
Oren Z, Lerman JC, Gudmundsson GH, Agerberth B, Shai Y (1999) Sansom MS (1998) Peptides and lipid bilayers: dynamic interactions.
Structure and organization of the human antimicrobial peptide Curr Opin Colloid Interface Sci 3:518–524
LL-37 in phospholipid membranes: relevance to the molecu- Santos R, Costa C, Mil-Homens D, Romão D, de Carvalho CCCR, Pais
lar basis for its non-cell-selective activity. Biochem J 341(Pt P et al (2017) The multidrug resistance transporters CgTpo1_1
3):501–513 and CgTpo1_2 play a role in virulence and biofilm forma-
Otvos L Jr, Wade JD (2014) Current challenges in peptide-based drug tion in the human pathogen Candida glabrata. Cell Microbiol
discovery. Front Chem 2:62 19:e12686
Pardi A, Zhang XL, Selsted ME, Skalicky JJ, Yip PF (1992) NMR Schneider JJ, Unholzer A, Schaller M, Schafer-Korting M, Korting HC
studies of defensin antimicrobial peptides. 2. Three-dimensional (2005) Human defensins. J Mol Med (Berl) 83:587–595
structures of rabbit NP-2 and human HNP-1. Biochemistry Shah D, Ali M, Pasha Z, Jaboori AJ, Jassim SH, Jain S et al (2016)
31:11357–11364 Histatin-1 expression in human lacrimal epithelium. PLoS ONE
Park C, Lee DG (2010) Melittin induces apoptotic features in Candida 11:e0148018
albicans. Biochem Biophys Res Commun 394:170–172 Sharma S, Verma I, Khuller GK (2000) Antibacterial activity of
Park CB, Yi KS, Matsuzaki K, Kim MS, Kim SC (2000) Structure- human neutrophil peptide-1 against Mycobacterium tuberculo-
activity analysis of buforin II, a histone H2A-derived antimicro- sis H37Rv: in vitro and ex vivo study. Eur Respir J 16:112–117
bial peptide: the proline hinge is responsible for the cell-penetrat- Sharma S, Verma I, Khuller GK (2001) Therapeutic potential of human
ing ability of buforin II. Proc Natl Acad Sci USA 97:8245–8250 neutrophil peptide 1 against experimental tuberculosis. Antimi-
Park SC, Park Y, Hahm KS (2011) The role of antimicrobial peptides crob Agents Chemother 45:639–640
in preventing multidrug-resistant bacterial infections and biofilm Soheili V, Tajani AS, Ghodsi R, Bazzaz BSF (2019) Anti-PqsR com-
formation. Int J Mol Sci 12:5971–5992 pounds as next-generation antibacterial agents against Pseu-
Park MS, Kim JI, Lee I, Park S, Bae J-Y, Park M-S (2018) Towards the domonas aeruginosa: a review. Eur J Med Chem 172:26–35
application of human defensins as antivirals. Biomol Ther 26:242 Sonawane A, Santos JC, Mishra BB, Jena P, Progida C, Sorensen OE
Patrzykat A, Friedrich CL, Zhang L, Mendoza V, Hancock RE (2002) et al (2011) Cathelicidin is involved in the intracellular killing
Sublethal concentrations of pleurocidin-derived antimicrobial of mycobacteria in macrophages. Cell Microbiol 13:1601–1617

13
International Journal of Peptide Research and Therapeutics

Swidergall M, Ernst JF (2014) Interplay between Candida albicans White SH, Wimley WC, Selsted ME (1995) Structure, function,
and the antimicrobial peptide armory. Eukaryot Cell 13:950–957 and membrane integration of defensins. Curr Opin Struct Biol
Tamamura H, Kuroda M, Masuda M, Otaka A, Funakoshi S, 5:521–527
Nakashima H et al (1993) A comparative study of the solution Wimley WC, Hristova K (2011) Antimicrobial peptides: successes,
structures of tachyplesin I and a novel anti-HIV synthetic peptide, challenges and unanswered questions. J Membr Biol 239:27–34
T22 ([Tyr5,12, Lys7]-polyphemusin II), determined by nuclear Winter J, Wenghoefer M (2012) Human defensins: potential tools for
magnetic resonance. Biochim Biophys Acta 1163:209–216 clinical applications. Polymers 4:691–709
Taneja NK, Ganguly T, Bakaletz LO, Nelson KJ, Dubey P, Poole LB Worthington RJ, Melander C (2013) Combination approaches to com-
et al (2013) d-alanine modification of a protease-susceptible bat multidrug-resistant bacteria. Trends Biotechnol 31:177–184
outer membrane component by the Bordetella pertussis dra locus Xhindoli D, Pacor S, Benincasa M, Scocchi M, Gennaro R, Tossi A
promotes resistance to antimicrobial peptides and polymorpho- (2016) The human cathelicidin LL-37—a pore-forming antibac-
nuclear leukocyte-mediated killing. J Bacteriol 195:5102–5111 terial peptide and host-cell modulator. Biochim Biophys Acta
Thennarasu S, Tan A, Penumatchu R, Shelburne CE, Heyl DL, Rama- 1858:546–566
moorthy A (2010) Antimicrobial and membrane disrupting Xiong YQ, Yeaman MR, Bayer AS (1999) In vitro antibacterial activi-
activities of a peptide derived from the human cathelicidin anti- ties of platelet microbicidal protein and neutrophil defensin
microbial peptide LL37. Biophys J 98:248–257 against Staphylococcus aureus are influenced by antibiotics dif-
Theuretzbacher U, Outterson K, Engel A, Karlen A (2020) The global fering in mechanism of action. Antimicrob Agents Chemother
preclinical antibacterial pipeline. Nat Rev Microbiol 18:275–285 43:1111–1117
Toke O (2005) Antimicrobial peptides: new candidates in the fight Xu T, Levitz SM, Diamond RD, Oppenheim FG (1991) Antican-
against bacterial infections. Biopolymers 80:717–735 didal activity of major human salivary histatins. Infect Immun
Tomasinsig L, Zanetti M (2005) The cathelicidins–structure, function 59:2549–2554
and evolution. Curr Protein Pept Sci 6:23–34 Yang L, Harroun TA, Weiss TM, Ding L, Huang HW (2001) Barrel-
Turner J, Cho Y, Dinh NN, Waring AJ, Lehrer RI (1998) Activities stave model or toroidal model? A case study on melittin pores.
of LL-37, a cathelin-associated antimicrobial peptide of human Biophys J 81:1475–1485
neutrophils. Antimicrob Agents Chemother 42:2206–2214 Yeaman MR, Yount NY (2003) Mechanisms of antimicrobial peptide
Umerska A, Cassisa V, Bastiat G, Matougui N, Nehme H, Manero F action and resistance. Pharmacol Rev 55:27–55
et al (2017) Synergistic interactions between antimicrobial pep- Zasloff M (2002) Antimicrobial peptides of multicellular organisms.
tides derived from plectasin and lipid nanocapsules containing Nature 415:389–395
monolaurin as a cosurfactant against Staphylococcus aureus. Int Zhai Y, Wang Y, Rao N, Li J, Li X, Fang T et al (2019) Activation
J Nanomed 12:5687–5699 and biological properties of human beta defensin 4 in stem cells
Vandamme D, Landuyt B, Luyten W, Schoofs L (2012) A compre- derived from human exfoliated deciduous teeth. Front Physiol
hensive summary of LL-37, the factotum human cathelicidin 10:1304
peptide. Cell Immunol 280:22–35 Zhang L, Falla TJ (2006) Antimicrobial peptides: therapeutic potential.
Verma C, Seebah S, Low SM, Zhou L, Liu SP, Li J et  al (2007) Expert Opin Pharmacother 7:653–663
Defensins: antimicrobial peptides for therapeutic development. Zhang J, Feng Y, Teng K, Lin Y, Gao Y, Wang J et al (2014) Type AII
Biotechnol J 2:1353–1359 lantibiotic bovicin HJ50 with a rare disulfide bond: structure,
Wade D, Boman A, Wahlin B, Drain CM, Andreu D, Boman HG et al structure-activity relationships and mode of action. Biochem J
(1990) All-D amino acid-containing channel-forming antibiotic 461:497–508
peptides. Proc Natl Acad Sci USA 87:4761–4765
Wang G (2008) Structures of human host defense cathelicidin LL-37 Publisher’s Note Springer Nature remains neutral with regard to
and its smallest antimicrobial peptide KR-12 in lipid micelles. J jurisdictional claims in published maps and institutional affiliations.
Biol Chem 283:32637–32643
Wang G (2014) Human antimicrobial peptides and proteins. Pharma-
ceuticals (Basel) 7:545–594
Wang L, Hu C, Shao L (2017) The antimicrobial activity of nanoparti-
cles: present situation and prospects for the future. Int J Nanomed
12:1227–1249

13

You might also like