You are on page 1of 12

International Journal of Medical Microbiology 304 (2014) 51–62

Contents lists available at ScienceDirect

International Journal of Medical Microbiology


journal homepage: www.elsevier.com/locate/ijmm

Mini review

Lantibiotics: Promising candidates for future applications in health


care
Jasmin Dischinger, Shradha Basi Chipalu, Gabriele Bierbaum ∗
Institute of Medical Microbiology, Parasitology and Immunology (IMMIP), University of Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany

a r t i c l e i n f o a b s t r a c t

Keywords: The immense potential of bacteria for production of antimicrobials represents an inexhaustible source
Bacteriocins of new antibiotics. An emerging class of natural products is constituted by ribosomally synthesized and
Lanthipeptides posttranslationally modified peptides (RiPPs). “Lantibiotics” (lanthionine and/or methyl-lanthionine con-
Multiresistant pathogens
taining antibiotics) belong to the earliest members of this class. The characteristic thioether amino acids
Pharmaceutical applications
are introduced into the precursor peptides by enzyme-mediated posttranslational modifications. The
Alternative therapeutics
Lantibiotics encouraging antimicrobial activity of lantibiotics against multiresistant clinical pathogens, their stability
against proteases, heat and oxidation make lantibiotics interesting candidates for novel antimicrobial
applications in many areas of the healthcare sector and associated industries. In addition to applications
as alternatives to classical antibiotics, lantibiotics can be used as probiotics, prophylactics or additives.
Furthermore, the in vitro activity of the lantibiotic modification machinery opens the possibility to gen-
erate either improved synthetic lantibiotic peptides or to introduce thioether cross-links into existing
therapeutics.
© 2013 Elsevier GmbH. All rights reserved.

Introduction The group comprises peptides with an elongated or a globular


structure as shown in Fig. 1. The early compounds were all isolated
Ribosomally synthesized and post-translationally modified from gram-positive producer strains, i.e. firmicutes and acti-
peptides (RiPPs) constitute an emerging class of natural products nobacteria, and displayed antibacterial activity. Only when several
(Arnison et al., 2013). Production of such, structurally very diverse, lanthionine containing peptides without antibacterial activity had
peptides is a common feature of bacteria, fungi and higher eukary- been described (Meindl et al., 2010; Goto et al., 2010), one of which
otes. Bacterial peptides with antimicrobial activity have often was even produced by a cyanobacterium (Li et al., 2010; Tang
been referred to as “bacteriocins”. After the first bacteriocinogenic and van der Donk, 2012), it became obvious that the lantibiotics
strain had been identified in 1925, bacteriocin production was do not form a distinct family themselves, but instead represent a
described for many Gram-positive and Gram-negative bacteria as subgroup of a larger family of lanthipeptides (Arnison et al., 2013).
well as some archaea (archaeacins) (Gratia, 1925; Tagg et al., 1976;
Rodriguez-Valera et al., 1982). According to Klaenhammer (1988),
nearly 99% of all bacteria secrete at least one bacteriocin. Indeed, a Gene clusters and regulation
plethora of new substances has recently been characterized, lead-
ing to an enormous gain of knowledge in the field of ribosomally Similar to most biosynthetic pathways in bacteria, the genes
synthesized compounds (for a comprehensive overview and the for lantibiotic biosynthesis are clustered and are designated by the
new classification and nomenclature of this class see Arnison et al., generic locus symbol lan, with a more specific genotypic desig-
2013). nation for each lantibiotic member (e.g. nis for nisin). Lantibiotic
The term “lantibiotic” was introduced by Schnell et al. (1988) biosynthetic gene clusters may be found on conjugative trans-
as an abbreviation for “lanthionine containing antibiotic” after posable elements (e.g. nisin), on plasmids (e.g. lacticin 481) or
identification of the first lantibiotic structural gene. The first com- on the chromosome of the producer (e.g. subtilin) (Chatterjee
pound, nisin, had already been described in 1928 (Rogers, 1928). et al., 2005). The precursor peptide (LanA) consists of an N-
terminal leader sequence and the core peptide that is modified
after ribosomal synthesis. It is encoded in a biosynthetic gene
cluster that typically comprises genes encoding the modifica-
∗ Corresponding author. Tel.: +49 228 28719103; fax: +49 228 28714808. tion enzymes (lanB and lanC, lanM, labKC, lanL etc.), an exporter
E-mail address: bierbaum@microbiology-bonn.de (G. Bierbaum). (lanT), a protease (lanP) and immunity proteins (lanI, lanH,

1438-4221/$ – see front matter © 2013 Elsevier GmbH. All rights reserved.
http://dx.doi.org/10.1016/j.ijmm.2013.09.003
52 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

Fig. 1. Structures of different lanthipeptides. (Me)Lan and labionin rings are highlighted by color and residues contributing to the same thioether amino acids are marked in
same color. Didehydro amino acids are designated in orange.

lanFEG). The structural gene lanA is often clustered with the genes Lanthipeptide biosynthesis and modification
of the modification enzymes in an operon. Besides this, there is
no uniform gene order in the individual gene clusters (Siezen After ribosomal biosynthesis, the serines and threonines of the
et al., 1996; Willey and van der Donk, 2007; Bierbaum and Sahl, lanthipeptides are dehydrated to give didehydroalanines (Dha) and
2009). didehydrobutyrines (Dhb), respectively. In a subsequent Michael
The biosynthesis of many lantibiotics is regulated by a quorum- addition, involving the cysteine SH-groups and the double bonds
sensing system consisting of a receptor-histidine kinase (LanK) of the dehydro amino acids, the thioether cross-links of lanthion-
and its cognate transcriptional response regulator (LanR). The ine (Lan) and methyllanthionine (MeLan) are formed. In contrast
active lantibiotic peptides themselves act as triggering agents to previous observations on the stereochemistry of the thioether
and lead to a signal cascade initiated by autophosphorylation bridges, an ll- as well as a dl-configuration can occur (Tang and
of the LanK histidine residue (Kuipers et al., 1995; Schmitz van der Donk, 2012, 2013). A typical lantibiotic contains three
et al., 2006). Subsequently, the phosphate group is transferred to six (methyl-)lanthionines in addition to several Dha and Dhb.
to the LanR response regulator that often functions as a tran- Other thioether amino acids comprise the S-aminovinyl-d-cysteine
scriptional activator of biosynthesis (van Kraaij et al., 1999; (AviCys) or S-aminovinyl-3-methyl-d-cysteine (AviMeCys) or the
Yonezawa and Kuramitsu, 2005; Willey and van der Donk, labionin (Lab) ring structures (Knerr and van der Donk, 2012). In
2007). addition, structural modifications, that are not involved in ring for-
In addition to the more than 95 lanthipeptides described so far mation such as the hydroxylation of Asp in cinnamycin and the
(Table 1 ), genome mining has shown that hundreds of further gene duramycins, hydroxylation of Pro and chlorination of Trp in micro-
clusters still await characterization. Furthermore, these gene clus- bisporicin, and epimerization reactions resulting in d-Ala in lacticin
ters are not limited to the firmicutes and actinomycetes but are also 3147 and lactocin S, occur frequently (Sahl and Bierbaum, 1998).
found in proteobacteria, chlamydiae, bacteroidetes and cyanobac- Lantibiotics have been categorized on the basis of their biosyn-
teria (Marsh et al., 2010; Li et al., 2010). thetic pathways (Willey and van der Donk, 2007; Knerr and
J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62 53

Table 1
Overview of lanthipeptides (01/2013, adapted from Dischinger et al., 2013). Classification according to Knerr and van der Donk (2012), Residues involved in (Me)Lan bridges
are highlighted in colored boxes, whereas subsidiary residues of one thioether aa are marked in same colors. The positions (x) of additional modifications are marked by
black, bold letters. Ser and Thr residues that are found to be dehydrated to Dha and Dhb are designated in orange.
54 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

Table 1 (Continued)

SA-FF22)
J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62 55

Table 1 (Continued)

michiganensis subspec.

actagardine)

Lact

3436

pneumoniae

Geobacillus spec.

7aa
56 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

Table 1 (Continued)

a
Birri et al. (2012).
b
Garg et al. (2012).
c
Lohans et al. (2012).
d
Daly et al. (2010).
e
Fagundes et al. (2011).
f
Teng et al. (2012).
g
Wescombe et al. (2012).
h
Mantovanin et al. (2002) and Mantovani and Russell (2008).
i
Simone et al. (2013).
j
Tang and van der Donk (2013).
k
Sawa et al. (2012).
l
Krawczyk et al. (2012).
m
Voeller et al. (2012).
n
Wang and van der Donk (2012).

van der Donk, 2012). In class I lantibiotics (e.g. nisin), the LanB phosphorylation (Paul et al., 2007) and their cyclase domains
dehydratase converts Ser and Thr to didehydroalanine (Dha) and contain three essential zinc binding ligands (Willey and van der
didehydrobutyrine (Dhb), respectively. Then the LanC cyclase Donk, 2007). Moreover, class I peptides are transported by the
catalyzes the intramolecular addition of Cys thiols to Dha/Dhb exporter LanT and the leader is removed by the protease LanP. In
to form the lanthionine (Lan) and methyllanthionine (MeLan) contrast, in class II peptides, both, secretion and processing, are
cross-links in a zinc dependent manner (Fig. 2). In contrast, in mediated by an exporter with an N-terminal protease domain,
class II lantibiotics (e.g. mersacidin), both the steps are catalyzed LanT(P).
by the bifunctional modification enzyme LanM. The C-terminal Class III lanthipeptides (e.g. SapB, labyrinthopeptins) lack
cyclase domain of LanM has homology to the LanC enzymes, antimicrobial activity, but appear to perform morphogenetic and
but the N-terminal dehydratase domain does not share sim- signaling functions for the producer cells. Labyrinthopeptins are
ilarities with LanB enzymes (Siezen et al., 1996). The LanM characterized by a carbacyclic amino acid called labionin (Meindl
enzymes dehydrate hydroxy amino acids via an ATP dependent et al., 2010). They are posttranslationally processed by the protein
J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62 57

Fig. 2. Formation of the thioether amino acid (methyl-) lanthionine. Hydroxy amino acids are enzymatically dehydrated to the corresponding didehydro amino acid.
Subsequently, in a nucleophilic Michael addition, a Cys derived SH group is connected to the didehydro amino acid resulting in the formation of (methyl-) lanthionine.

kinase-cyclase LabKC, which catalyzes the cyclization of the Gram-negatives are generally not affected by lantibiotics due to
labionin ring structure in two steps, which include a dehydration a protective effect of the outer membrane (OM). Consequently,
via a GTP dependent phosphorylation and subsequently a 2-fold lantibiotic treatment in concert with an OM destabilizing agent
Michael-type addition (Müller et al., 2010). The C-terminal cyclase showed an antibiotic effect even against Gram-negative strains
domain bears limited homology to LanM and LanC and the con- (Kordel et al., 1988; Stevens et al., 1991). Some lantibiotics (e.g.
served zinc ligands are missing. Thus, the maturation mechanism nisin Z) can affect selected Gram-negatives such as E. coli, Heli-
of class III lantibiotics differs from that of the other classes (Kodani cobacter pylori and Neisseria at high concentrations, probably due
et al., 2005; Willey et al., 2006). In addition, the gene clusters of to either a self-promoted uptake or a lantibiotic-based destabiliza-
class III lanthipeptides possess two LanT-like transporters lacking tion of the OM by binding to lipopolysaccharides which is a typical
a dedicated protease domain (Müller et al., 2011; Knerr and van der feature of cationic amphiphilic peptides (Nagao et al., 2009).
Donk, 2012). The antibacterial effect generally relies on (a) the inhibition of
A fourth class of lanthionine synthetases termed LanL was the cell wall biosynthesis via complex formation and sequestra-
recently discovered from Streptomyces venezuelae (Goto et al., tion of the membrane-bound cell wall precursor lipid II and/or (b)
2010). Lanthipeptides (e.g. venezuelin) modified by this new the disruption of membrane integrity and pore formation (Fig. 3).
enzyme-type are classified as class IV lanthipeptides (Knerr and van Nisin, the most intensively studied lantibiotic, acts by a dual mode
der Donk, 2012). The C-terminus of LanL also contains a LanC-like of action (MoA) combining both the mechanisms (Wiedemann
cyclase domain including the conserved zinc binding sites found in et al., 2001, 2004). The N-terminal rings (A and B) form a bind-
LanM and LanC. The N-terminus harbors a lyase domain and the ing pocket, the pyrophosphate cage that allows binding to the
central part a serine/threonine kinase domain, both of which align pyrophosphate moiety of lipid I/II (Hsu et al., 2004). Complex
well with the class III synthetases (Goto et al., 2010; Knerr and van formation with lipid II prevents transglycosylation by steric hin-
der Donk, 2012). The venezuelin gene cluster possesses a LanT and drance and results in the sequestration of the precursors and,
LanH, encoding the ATP binding and membrane permease subunits, hence, in its abduction from the sites of nascent cell wall biosyn-
respectively, which may be involved in the export (Goto et al., 2010; thesis (Brötz et al., 1998; Breukink et al., 2003). Moreover, nisin
Knerr and van der Donk, 2012). The gene cluster does not contain forms membrane spanning and potential-dependent pores con-
a protease involved in leader processing or any immunity-related sisting of 4 lipid II and 8 nisin molecules (Hasper et al., 2004).
genes, however the latter function appears to be fulfilled by the Here, lipid II serves as a docking molecule and mediates a ‘tar-
exporter LanTH, which is not common for lantibiotics (Goto et al., geted’ pore formation (Brötz et al., 1998). The assembly of pores
2010). with 2–2.5 nm in diameter (Wiedemann et al., 2004) allows small
The role of the leader peptide has not yet been fully eluci- molecules to leak from the cell, resulting in disruption of the bar-
dated. The leader appears to be important for the recognition rier function and, consequently, in dissipation of the membrane
of the precursor peptide by the modification enzymes, e.g. an potential. Finally, this results in the abrupt arrest of all cellular pro-
increase of the dehydration activity, processivity and directional- cesses and in cell death (Sahl and Brandis, 1982; Wiedemann et al.,
ity was shown for the lacticin 481 modifying enzyme (Levengood 2001).
et al., 2007) – however, in vitro the peptide was also processed By dissipation of the membrane potential and inhibition of cell
when the leader was present in trans or even absent (Levengood wall biosynthesis, nisin and other lanthipeptides are able to inhibit
et al., 2007). In contrast, the labyrinthopeptin synthetase, LabKC, spore outgrowth at the stage of spore germination. This effect also
does not modify its substrate if the leader peptide is not directly depends on membrane depolarization and lipid II binding (Gut
attached to the core peptide (Müller et al., 2011). Therefore, the et al., 2011).
necessity of leader peptides for the modification reactions might Other lantibiotics only act by one of these two mechanisms.
differ between the different biosynthetic classes (Knerr and van der Due to its elongated structure Pep5 apparently inserts into the
Donk, 2012). For those lantibiotics that are processed after export membrane, most likely by binding to a so far unknown docking
from the cell e.g. nisin, the leader peptide keeps the mature lan- molecule (Sahl and Brandis, 1982; Kordel et al., 1988). In con-
tibiotic inactive inside the cell, thereby protecting the producer trast, gallidermin preserves a nisin-like lipid II binding motif, but,
cell. since the molecule is shorter than nisin, it is incapable of pore
formation in most species and solely acts by inhibition of cell
Modes of action and targets wall biosynthesis (Bonelli et al., 2006). This MoA was also shown
for mersacidin-like and lacticin 481-like peptides. These peptides
The overwhelming majority of lanthipeptides displays share a conserved TxS/TxEC motif within their essential C-ring
an antibacterial activity. With exception of the Neisseriae, (Szekat et al., 2003; Cotter et al., 2006) or A-ring (Boettiger et al.,
58 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

Fig. 3. Overview over the MoA of antimicrobially active lanthipeptides: Almost all lantibiotics act either by inhibition of cell wall biosynthesis by binding and dislocalization
of the membrane bound cell wall precursor lipid II and/or disruption of the membrane integrity by pore formation.

2009), respectively, which represents their lipid II binding site (Hsu like low molecular weights, thermal and protease stability, lack of
et al., 2004). toxicity, low tendency to generate resistance and low immuno-
Two-peptide lantibiotics may also possess a dual MoA, genicity, which make them suitable for potential applications in
depending on two peptides that act synergistically (Morgan different health care associated settings such as in human and vet-
et al., 2005). While lipid II binding is mediated by the ␣- erinary medicine and in biochemical, pharmaceutical, agricultural
peptide, which is characterized by the lipid II binding motive or food industries. In the following, an overview of possible applica-
of mersacidin, the elongated ␤-peptide acts on the mem- tions of lanthipeptides currently under investigation is presented.
brane by pore formation (Wiedemann et al., 2006). Lipid II
complex formation of the ␣-peptide enhances the affinity for
(A) Lanthipeptides in use
the ␤-peptide and allows the ␤-peptide to adopt a trans-
bilayer orientation and to form a pore (Wiedemann et al.,
The first and – so far – only lanthipeptide in commercial use
2006).
is nisin (Fig. 1). It exhibits an antimicrobial activity against Gram-
Besides, other biological effects have been discovered for mem-
positive bacteria, including agents causing food spoilage like Listeria
bers of the lanthipeptide family. The cinnamycin-like peptides
monocytogenes (Denny et al., 1961) or clostridia, thus it has been
hold antimicrobial activities, but display a different MoA and act
employed as a biological food preservative (E234, Nisaplin® ) in
by the inhibition of enzymatic functions (Märki et al., 1991).
processed dairy products, canned fruits and vegetables for more
These peptides indirectly inhibit phospholipase A2 by forming
than 50 years. Nisin is also commercially used in animal care
a complex with its lipid substrate, phosphatidylethanolamine
products (e.g. Wipe-OUT, ImmuCell) for the prevention of bovine
(PE). Subsequently, this promotes a transbilayer movement of
mastitis caused by Staphylococcus aureus or Streptococcus agalactiae
lipids and reorganization of model membranes (Hosoda et al.,
(Dawson, 2007).
1996; Makino et al., 2003). The lanthipeptides SapT and SapB
(class III) exhibit a morphogenetic rather than an antibacte-
rial effect; they act as biosurfactants during the emergence of (B) Application as antibiotics
aerial hyphae of streptomycetes (Kodani et al., 2005, 2004).
Members of the recently identified labionin-containing lan- Lantibiotics are among the most promising candidates for
thipeptides also lack antimicrobial activity (Meindl et al., 2010; future antimicrobials due to their capacity to inhibit the growth
Voeller et al., 2012), however, the labyrinthopeptins A1-3 have of clinically significant pathogens including multidrug-resistant
been demonstrated to numb neuropathic pain in a mouse staphylococci, streptococci, enterococci and clostridia. In addition,
model (Meindl et al., 2010). For other lanthipeptides e.g. the some lantibiotics are also selectively active against a few species of
prochlorosins, bioactivities have not been identified so far Gram-negative bacteria such as Neisseria and Helicobacter strains.
(Li et al., 2010). Many lantibiotics bind to the cell wall precursor lipid II, which
is also targeted by clinically used antibiotics, but in contrast to
the glycopeptides vancomycin and teicoplanin, that complex the
Lanthipeptides and their applications d-Ala-d-alanyl group of lipid II, lantibiotics bind to a different site
on the target molecule, i.e. the pyrophosphate-sugar moiety (van
Today, more than 95 structurally diverse lanthipeptides have Heel et al., 2011). Thus, they might overcome the problem of pre-
been identified and several exhibit intriguing and encouraging existing resistance mechanisms.
bioactivities which make them interesting candidates or lead The potential of lantibiotics to act as antimicrobials is combined
structures for future applications in many areas. In addition, the with a low tendency to generate resistance, and therefore these
lanthipeptides hold many characteristics and chemical properties, compounds are highly attractive for medical applications (van Heel
J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62 59

et al., 2011; Wilson-Standford and Smith, 2011). Several lantibiotics addition to dental applications has been discussed (Suda et al.,
have been investigated in vitro and in vivo regarding their potential 2011). The salivaricins have interesting effects against Streptococcus
as antimicrobials and a few are currently in preclinical and clini- pyogenes strains. Therefore, the salivaricin A producer was supple-
cal development. For example, the semisynthetic carboxy-amide mented as a probiotic to milk drinks and was demonstrated to
derivative of the globular class II lantibiotic deoxy-actagardine B persist in the oral cavity. Thus, S. pyogenes infections were pre-
(NVB302, Novacta Biosystems Limited) is currently undergoing a vented by a probiotic bacterial replacement strategy using a strain
phase I clinical trial as a drug candidate for the treatment of Clostrid- of the indigenous oral tongue microbiotia that is known for its lan-
ium difficile infections (www.novactabio.com; Boakes et al., 2010; Li tibiotic production (Dierksen et al., 2007). Moreover, chewing gums
et al., 2012). The lantibiotic derivative NVB333 (Novacta Biosystems and lozenges, that include salivaricin-producing S. salivarius strains,
Limited) was selected as an injectable drug candidate for the treat- have been developed recently and are currently marketed as an
ment of nosocomial infections caused by Gram-positive pathogens oral care product for prophylaxis of dental caries, paradontosis and
and its promising inhibition spectrum also includes staphylococcal infection of the oral cavity (BLIS Technologies).
and enterococcal strains resistant to the reserve antibiotics dapto-
mycin and linezolid (www.novactabio.com; Li et al., 2012). Therapy (D) Application as other medical therapeutics/pharmaceuticals
of nosocomial infection is also targeted by the drug candidate
microbisporicin (NAI-107, NAICON and Sentinella Pharmaceuticals Some lanthipeptides hold additional bioactivities that are
INC), the most active lantibiotic identified so far (Castiglione et al., interesting for medical applications. The lanthipeptides of the cin-
2008). In in vivo experiments, NAI-107 was highly effective in the namycin subgroup were found to influence eukaryotic metabolic
treatment of infections caused by multiresistant bacteria, such as functions. Duramycin (Moli1901) stimulates chloride secretion of
rat endocarditis caused by MRSA (Jabes et al., 2009). The synthetic bronchial epithelia through the activation of calcium-activated
lanthipeptide mutacin 1140 (Mu1140-S, Organics) is currently in chloride channels (Cloutier et al., 1990; Roberts et al., 1991). This
preclinical development for the treatment of Gram-positive infec- effect mainly relies on unspecific changes in the cell membrane
tions and showed interesting in vivo activities against MRSA, rather than on a direct effect on the ion channels (Oliynyk et al.,
vancomycin resistant enterococci, C. difficile, Mycobacterium tuber- 2010). In a phase II clinical trial, duramycin was proven to be a
culosis and Bacillus anthracis (www.organics.com). safe and effective therapeutic for the treatment of cystic fibrosis by
Nisin exhibits a high efficacy against various relevant human inhalation (Grasemann et al., 2007; Grasemann, 2012). Duramycin
and animal pathogens, making it an effective agent e.g. for the treat- additionally inhibits the eukaryotic phospholipase A2, which is
ment of peptic ulcers (Delves-Broughton et al., 1996). In clinical involved in inflammation by release of inflammation promot-
trials, nisin was highly effective in the treatment of staphylococcal ing substances e.g. the prostaglandins (Märki et al., 1991). Thus,
mastitis in lactating dairy cattle (Cao et al., 2007) and in humans duramycin might serve as anti-inflammatory or anti-allergy drug.
(Fernández et al., 2008) and currently, an intramammary infusion The related ancovenin is a natural inhibitor of the angiotensin I
product (Mast Out® , ImmuCell) is under development for the treat- converting enzyme, that is involved in the regulation of cardiac
ment of mastitis in lactating cows. and vascular functions, thereby providing an alternative strategy to
Nevertheless, particularly for medical applications, some chem- treat high blood pressure (Kido et al., 1983). A contraceptive effect
ical or pharmacokinetic hurdles often have to be overcome for some due to spermicidal activities has been reported for nisin (Clara et al.,
lantibiotics with promising biological effects. For nisin, these are its 2004).
low solubility, lack of peptide stability especially against intestinal For a few lanthipeptides, antiviral activities have been described
enzymes and its low activity at higher pH as well as its tendency to in vitro. Cinnamycin is active against retroviruses such as herpes
interact with blood components (Boakes and Wadman, 2008). simplex (Naruse et al., 1989), whereas the labyrinthopeptins addi-
tionally exhibit a promising anti-dengue-virus effect (Alen et al.,
(C) Application as probiotics, prophylactics, preservatives and 2012). For the latter a dose dependent inhibition of the early step
additives in the replication cycle of the virus was observed. Most likely this
is caused by a direct binding to the virus envelope glycoprotein
Besides the classical antibiotic use, there are further applications E, which finally blocks the viral infection of targeted cells. In addi-
for lantibiotics. Gallidermin and lacticin 3147 are active against the tion, as described above, the labyrinthopeptins hold an unexpected
acne causing bacterium Propionibacterium acnes, thus providing the bioactivity to neuropathic pain (Meindl et al., 2010).
opportunity to use these substances as additives in cosmetics and
personal-care products (Zähner et al., 1998; Lawton et al., 2007). (E) Applications of the lanthipeptide biosynthesis machinery
In an in vitro colonization experiment, Pep5 and epidermin pre-
vented adhesion of coagulase-negative staphylococci, specifically Usage of the lanthipeptide biosynthesis machinery in in vivo
of S. epidermidis, to catheters coated with silicone (Fontana et al., and in vitro bioengineering approaches is a promising technology
2006). The coating of medical devices provides an elegant strategy to design novel antimicrobial compounds. The structure–function
to reduce catheter-related infections. In line with this, a feasible relationship of several lantibiotics has been studied by genetic
application as prophylactic against implantate associated infec- engineering of variant peptides (for a recent review see Ross
tions has been evaluated for gallidermin by the incorporation of and Vederas, 2010). Furthermore it has been possible to engineer
the active peptides into prosthetic joint cement (Sandiford et al., variant peptides with increased activity, a broader antibacterial
2010). Another prophylactic strategy to combat surgical infections spectrum or improved physico-chemical properties (e.g. Appleyard
is discussed for mersacidin. In a murine rhinitis model this lantibi- et al., 2009; Field et al., 2012).
otic was shown to be potent in eradicating the nasal colonization The promiscuity of the lantibiotic modification enzymes and
by MRSA (Kruszewska et al., 2004). Due to their structures and sur- their ability to display activity in vitro, allow introduction of
face activity, amphiphilic lanthipeptides such as nisin have been lantibiotic modifications even into synthetic analogs containing
discussed to serve as emulsifiers (Bani-Jaber et al., 2000) or as additional unnatural amino acid side chain substitutions. Thus,
absorption promoters allowing a nasal administration of therapeu- novel antibiotics could be designed on the basis of the introduc-
tics across mucosal membranes (Bower et al., 2001). tion of thioether cross-links into existing therapeutics (Rink et al.,
Lacticin 3147 and the mutacins exert antimicrobial effects 2005; Kluskens et al., 2005). For example, the successful improve-
on different cariogenic Streptococcus mutants strains and their ment of therapeutically used peptide variants of enkephalin and
60 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

somatostatin by introduction of thioether bridges has already been Boakes, S., Wadman, S., 2008. The therapeutic potential of lantibiotics. Innovation in
reported (Ösapay et al., 1997; Rew et al., 2002). To this end, Bosma Pharmaceutical Technology: Drug Discovery and Development (IPT 27), 22–25.
Boettiger, T., Schneider, T., Martinez, B., Sahl, H.G., Wiedemann, I., 2009. Influence
et al. established an in vivo system that is based on the nisin biosyn- of Ca2+ ions on the activity of lantibiotics containing a mersacidin-like lipid II
thesis and transport machinery (Kuipers et al., 2005; Bosma et al., binding motif. Appl. Environ. Microbiol. 75, 4427–4434.
2011) and introduced didehydro- as well as thioether-amino acids Bonelli, R.R., Schneider, T., Sahl, H.G., Wiedemann, I., 2006. Insights into in vivo
activities of lantibiotics from gallidermin and epidermin mode-of-action studies.
into non-lantibiotic peptides. Using this system they successfully Antimicrob. Agents Chemother. 50, 1449–1457.
produced lanthionine-bridged variants of the therapeutics vaso- Bosma, T., Kuipers, A., Bulten, E., de Vries, L., Rink, R., Moll, G.N., 2011. Bacterial
pressin, enkephalin and angiotensin. display and screening of posttranslationally thioether-stabilized peptides. Appl.
Environ. Microbiol. 7, 6794–6801.
Bower, C.K., Bothwell, M.K., McGuire, J., 2001. Lantibiotics as surface active agents
Conclusions for biomedical applications. Colloids Surf. B Biointerfaces 22, 259–265.
Breukink, E., van Heusden, H.E., Vollmerhaus, P.J., Swiezewska, E., Brunner, L.,
Walker, S., Heck, A.J., de Kruijff, B., 2003. Lipid II is an intrinsic component of the
The potential of lanthipeptides as promising alternatives for tra- pore induced by nisin in bacterial membranes. Biol. Chem. 278, 19898–19903.
ditional antimicrobial therapeutics, as probiotics or preservatives Broetz, H., Josten, M., Wiedemann, I., Schneider, U., Götz, F., Bierbaum, G., Sahl, H.G.,
1998. Role of lipid-bound peptidoglycan precursors in the formation of pores by
in different areas of the healthcare sector and in associated indus- nisin epidermin and other lantibiotics. Mol. Microbiol. 30, 317–327.
tries has been discussed for many years. So far, only nisin has been Cao, L.T., Wu, J.Q., Xie, F., Hu, S.H., Mo, Y., 2007. Efficacy of nisin in treatment of
successfully introduced as a preservative into food industry or as a clinical mastitis in lactating dairy cows. J. Dairy Sci. 90, 3980–3985.
Castiglione, Lazzarini, F., Carrano, A., Corti, L., Ciciliato, E., Gastaldo, I., Candiani, L.,
prophylactic in veterinary settings. Although to our knowledge no Losi, P., Marinelli, D., Selva, F., Parenti, E., 2008. Determining the structure and
lantibiotic has reached the approval as an antibacterial therapeutic mode of action of microbisporicin a potent lantibiotic active against multiresis-
so far, currently several clinical and preclinical trials explore their tant pathogens. Chem. Biol. 15, 22–31.
Chatterjee, C., Paul, M., Xie, L., van der Donk, W.A., 2005. Biosynthesis and mode of
high potency as effective pharmaceuticals, mainly as antimicrobial
action of lantibiotics. Chem. Rev. 105, 633–684.
therapeutics or prophylactics. Additionally, the identification of Clara, A., Gupta, S.M., Reddy, K.V.R., 2004. Spermicidal activity of Nisin: in vitro and
lanthipeptides with intriguing bioactivities other than antimicro- in vitro studies. Contraception 69, 333–342.
bial effects will enlarge their possibilities for applications and it is Cloutier, M.M., Guernsey, L., Mattes, P., Koeppen, B., 1990. Duramycin enhances
chloride secretion in airway epithelium. Am. J. Physiol. 259, 450–454.
very likely that investigations in this field will result in the identifi- Cotter, P.D., Deegan, L.H., Lawton, E.M., Draper, L.A., O’Connor, P.M., Hill, C., Ross,
cation of novel MoA and biological effects. Besides, the biosynthesis R.P., 2006. Complete alanine scanning of the two-component lantibiotic lac-
apparatus itself also holds a high potential for biotechnological use. ticin 3147: generating a blueprint for rational drug design. Mol. Microbiol. 62,
735–747.
Its ability to modify peptides in vitro with low selectivity, opens a Daly, K.M., Upton, M., Sandiford, S.K., Draper, L.A., Wescombe, P.A., Jack, R.W.,
promising strategy for the generation of improved lanthipeptide O’Connor, P.M., Rossney, A., Götz, F., Hill, C., Cotter, P.D., Ross, R.P., Tagg, J.R.,
analogs or the introduction of thioether cross-links into existing 2010. Production of the Bsa lantibiotic by community-acquired Staphylococcus
aureus strains. J. Bacteriol. 192, 1131–1142.
therapeutics. For these very reasons, it is very likely that lanthipep- Delves-Broughton, J., Blackburn, P., Evans, R.J., Hugenholtz, J., 1996. Applications of
tides or their biochemically modified derivatives will reach the the bacteriocin, nisin. Antonie Van Leeuwenhoek 69, 193–202.
pharmaceutical market. Denny, C.B., Sharpe, L.E., Bohrer, C.W., 1961. Effects of tylosin and nisin on canned
food spoilage bacteria. Appl. Microbiol. 9, 108–110.
Dierksen, K.P., Moore, C.J., Inglis, M., Wescombe, P.A., Tagg, J.R., 2007. The effect
Acknowledgements of ingestion of milk supplemented with salivaricin A-producing Streptococcus
salivarius on the bacteriocin-like inhibitory activity of streptococcal populations
on the tongue. FEMS Microbiol. Ecol. 59, 584–591.
We gratefully acknowledge the German Research Foundation Dischinger, J., Wiedemann, I., Bierbaum, G., Sahl, H.G., 2013. Lantibiotics. In: Abba
(DFG, BI 504/9-2 to GB) for funding. Additional support was pro- Kastin, J. (Ed.), Handbook of Biologically Active Peptides. , second ed. Academic
Press Incorporation, Elsevier, San Diego, USA, pp. 119–128.
vided by the BONFOR program of the Medical Faculty, University of Fagundes, P., Ceotto, H., Potter, A., Vasconcelos de, P., Maria, A., Brede, D., Nes, I.F.,
Bonn. Bastos, M., 2011. Hyicin 3682, a bioactive peptide produced by Staphylococcus
hyicus 3682 with potential applications for food preservation. Res. Microbiol.
162, 1052–1059.
References Fernandez, L., Delgado, S., Herrero, H., Maldonado, A., Rodriguez, J.M., 2008. The
bacteriocin nisin, an effective agent for the treatment of staphylococcal mastitis
Alen, M.F., Neyts, J., Süssmuth, R.D., Brönstrup, M., Schols, D., 2012. during lactation. J. Hum. Lact. 24, 311–316.
Labyrinthopeptins, a new class of lantibiotics, exhibit potent anti-dengue Field, D., Begley, M., O’Connor, P.M., Daly, K.M., Hugenholtz, F., Cotter, P.D., Hill,
virus activity. In: Twenty-Fifth International Conference on Antiviral Research C., Ross, R.P., 2012. Bioengineered nisin A derivatives with enhanced activity
(ICAR), April 16–19, Sapporo, Japan. against both Grampositive and Gram- negative pathogens. PLoS ONE 7, e46884.
Appleyard, A.N., Choi, S., Read, D.M., Lightfoot, A., Boakes, S., Hoffmann, A., Chopra, I., Fontana, M.B., de Bastos, M.C., Brandelli, A., 2006. Bacteriocins Pep5 and epider-
Bierbaum, G., Rudd, B.A., Dawson, M.J., Cortes, J., 2009. Dissecting structural and min inhibit Staphylococcus epidermidis adhesion to catheters. Curr. Microbiol.
functional diversity of the lantibiotic mersacidin. Chem. Biol. 29 (16), 490–498. 52, 350–353.
Arnison, P.G., Bibb, M.J., Bierbaum, G., Bowers, A.A., Bugni, T.S., Bulaj, G., Camarero, Garg, N., Tang, W., Goto, Y., Nair, S.K., van der Donk, W.A., 2012. Lantibiotics from
J.A., Campopiano, D.J., Challis, G.L., Clardy, J., Cotter, P.D., Craik, D.J., Dawson, M., Geobacillus thermodenitrificans. PNAS 109, 5241–5246.
Dittmann, E., Donadio, S., Dorrestein, P.C., Entian, K.D., Fischbach, M.A., Garavelli, Goto, Y., Li, B., Claesen, J., Shi, Y., Bibb, M.J., van der Donk, W.A., 2010. Discovery
J.S., Göransson, U., Gruber, C.W., Haft, D.H., Hemscheidt, T.K., Hertweck, C., Hill, of unique lanthionine synthetases reveals new mechanistic and evolutionary
C., Horswill, A.R., Jaspars, M., Kelly, W.L., Klinman, J.P., Kuipers, O.P., Link, A.J., insights. PLoS Biol. 8, e1000339.
Liu, W., Marahiel, M.A., Mitchell, D.A., Moll, G.N., Moore, B.S., Müller, R., Nair, Grasemann, H., 2012. New developments in pharmaceutical treatments for cystic
S.K., Nes, I.F., Norris, G.E., Olivera, B.M., Onaka, H., Patchett, M.L., Piel, J., Reaney, fibrosis. Curr. Pharm. Des. 18, 613.
M.J., Rebuffat, S., Ross, R.P., Sahl, H.G., Schmidt, E.W., Selsted, M.E., Severinov, K., Grasemann, H., Stehling, F., Brunar, H., Widmann, R., Laliberte, T.W., Molina, L.,
Shen, B., Sivonen, K., Smith, L., Stein, T., Süssmuth, R.D., Tagg, J.R., Tang, G.L., Tru- Döring, G., Ratjen, F., 2007. Inhalation of Moli1901 in patients with cystic fibrosis.
man, A.W., Vederas, J.C., Walsh, C.T., Walton, J.D., Wenzel, S.C., Willey, J.M., van Chest 131, 1461–1466.
der Donk, W.A., 2013. Ribosomally synthesized and post-translationally modi- Gratia, A., 1925. Sur un remarquable example d’antagonisme entre deux souches de
fied peptide natural products: overview and recommendations for a universal colibacille. Comp. Rend. Soc. Biol 93, 1040–1041.
nomenclature. Nat. Prod. Rep. 30, 108–160. Gut, I.M., Blanke, S.R., van der Donk, W.A., 2011. Mechanism of inhibition of Bacillus
Bani-Jaber, A., McGuire, J., Ayres, J.W., Daeschel, M.A., 2000. Efficacy of the Antimi- anthracis spore outgrowth by the lantibiotic nisin. ACS Chem. Biol. 6, 744–752.
crobial Peptide Nisin in Emulsifying Oil in Water. J. Food Sci 65, 502–506. Hasper, H.E., Kruijff, B., de Breukink, E., 2004. Assembly and stability of nisin-lipid II
Bierbaum, G., Sahl, H.G., 2009. Lantibiotics: mode of action, biosynthesis and bio- pores. Biochemistry 43, 11567–11575.
engineering. Curr. Pharm. Biotechnol. 10, 2–18. Hosoda, K., Ohya, M., Kohno, T., Maeda, T., Endo, S., Wakamatsu, K., 1996. Structure
Birri, D.J., Brede, D.A., Nes, I.F., 2012. Salivaricin D, a novel intrinsically trypsin- determination of an immunopotentiator peptide, cinnamycin, complexed with
resistant lantibiotic from Streptococcus salivarius 5M6c isolated from a healthy lysophosphatidylethanolamine by 1H-NMR1. J. Biochem. 119, 226–230.
infant. Appl. Environ. Microbiol. 78, 402–410. Hsu, S.T., Breukink, E., Tischenko, E., Lutters, M.A., de Kruijff, B., Kaptein, R., Bonvin,
Boakes, S., Appleyard, A.N., Cortes, J., Dawson, M.J., 2010. Organization of the biosyn- A.M., van Nuland, N.A., 2004. The nisin-lipid II complex reveals a pyrophosphate
thetic genes encoding deoxyactagardine B (DAB), a new lantibiotic produced by cage that provides a blueprint for novel antibiotics. Nat. Struct. Mol. Biol. 11,
Actinoplanes liguriae NCIMB41362. J. Antibiot. 63, 351–358. 963–967.
J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62 61

Jabes, D., Brunati, C., Guglierame, S., Donadio, S., 2009. In vitro antibacterial profile of Ösapay, G., Prokai, L., Kim, H.S., Medzihradszky, K.F., Coy, D.H., Liapakis, G., Rei-
the new lantibiotic NAI-107. In: 49th Annual Meeting of Interscience Conference sine, T., Melacini, G., Zhu, Q., Wang, S.H., Mattern, R.H., Goodman, M., 1997.
on Antimicrobial Agents and Chemotherapy, September 12–15, San Francisco. Lanthionine-somatostatin analogs: synthesis, characterization, biological activ-
Kido, Y., Hamakado, T., Yoshida, T., Anno, M., Motoki, Y., Wakamiya, T., Shiba, ity, and enzymatic stability studies. J. Med. Chem. 40, 2241–2251.
T., 1983. Isolation and characterization of ancovenin, a new inhibitor of Paul, M., Patton, G.C., van der Donk, W.A., 2007. Mutants of the zinc ligands of lac-
angiotensin I converting enzyme, produced by actinomycetes. J. Antibiot. 36, ticin 481 synthetase retain dehydration activity but have impaired cyclization
1295–1299. activity. Biochemistry 46, 6268–6276.
Klaenhammer, T.R., 1988. Bacteriocins of lactic acid bacteria. Biochimie 70, Rew, Y., Malkmus, S., Svensson, C., Yaksh, T.L., Chung, N.N., Schiller, P.W., Cassel, J.A.,
337–349. DeHaven, R.N., Taulane, J.P., Goodman, M., 2002. Synthesis and biological activ-
Kluskens, L.D., Kuipers, A., Rink, R., de Boef, E., Fekken, S., Driessen, A.J., Kuipers, ities of cyclic lanthionine enkephalin analogues: delta-opioid receptor selective
O.P., Moll, G.N., 2005. Post-translational modification of therapeutic peptides by ligands. J. Med. Chem. 45, 3746–3754.
NisB, the dehydratase of the lantibiotic nisin. Biochemistry 44, 12827–12834. Rink, R., Kuipers, A., de Boef, E., Leenhouts, K.J., Driessen, A.J., Moll, G.N., Kuipers, O.P.,
Knerr, P.J., van der Donk, W.A., 2012. Discovery, biosynthesis, and engineering of 2005. Lantibiotic structures as guidelines for the design of peptides that can be
lantipeptides. Annu. Rev. Biochem. 81, 479–505. modified by lantibiotic enzymes. Biochemistry 44, 8873–8882.
Kodani, S., Hudson, M.E., Durrant, M.C., Buttner, M.J., Nodwell, J.R., Willey, J.M., 2004. Roberts, M., Hladky, S.B., Pickles, R.J., Cuthbert, A.W., 1991. Stimulation of sodium
The SapB morphogen is a lantibiotic-like peptide derived from the product of the transport by duramycin in cultured human colonic epithelia. J. Pharmacol. Exp.
developmental gene ramS in Streptomyces coelicolor. PNAS 101, 11448–11453. Ther. 259, 1050–1058.
Kodani, S., Lodato, M.A., Durrant, M.C., Picart, F., Willey, J.M., 2005. SapTa Rodríguez-Valera, F., Juez, J., Kushner, D.J., 1982. Halocins: salt dependent bacteri-
lanthionine-containing peptide involved in aerial hyphae formation in the strep- ocins produced by extremely halophilic rods. Can. J. Microbiol. 28, 151–154.
tomycetes. Mol. Microbiol. 58, 1368–1380. Rogers, L.A., 1928. The inhibiting effect of Streptococcus lactis on Lactobacillus bul-
Kordel, M., Benz, R., Sahl, H.G., 1988. Mode of action of the staphylococcin like garicus. J. Bacteriol. 16, 321–325.
peptide Pep 5: voltage-dependent depolarization of bacterial and artificial mem- Ross, A.C., Vederas, J.C., 2010. Fundamental functionality: recent developments
branes. J. Bacteriol. 170, 84–88. in understanding the structure-activity relationships of lantibiotic peptides. J.
Krawczyk, B., Voller, G.H., Voller, J., Ensle, P., Suessmuth, R.D., 2012. Curvopeptin: a Antibiot. (Tokyo) 64, 27–34.
new lanthionine-containing class III lantibiotic and its co-substrate promiscuous Sahl, H.G., Bierbaum, G., 1998. Lantibiotics: biosynthesis and biological activities of
synthetase. ChemBioChem 13, 2065–2071. uniquely modified peptides from Gram-positive bacteria. Annu. Rev. Microbiol.
Kruszewska, D., Sahl, H.-G., Bierbaum, G., Pag, U., Hynes, S.O., Ljungh, A., 2004. 52, 41–79.
Mersacidin eradicates methicillin-resistant Staphylococcus aureus (MRSA) in a Sahl, H.G., Brandis, H., 1982. Mode of action of the staphylococcin-like peptide Pep 5
mouse rhinitis model. J. Antimicrob. Chemother. 54, 648–653. and culture conditions affecting its activity. Zentralbl. Bakteriol. Mikrobiol. Hyg.
Kuipers, O.P., Beerthuyzen, M.M., Ruyter, P.G., de Luesink, E.J., de Vos, W.M., 1995. 252, 166–175.
Autoregulation of nisin biosynthesis in Lactococcus lactis by signal transduction. Sandiford, S., Kay, P., Upton, M., 2010. The use of antimicrobial peptides in the pre-
J. Biol. Chem. 270, 27299–27304. vention of biofilm forming S. epidermidis strains associated with orthopaedic
Levengood, M.R., Patton, G.C., van der Donk, W.A., 2007. The leader peptide is not infection. In: Seventh SICOT/SIROT Annual International Conference and SOF
required for posttranslational modification by lacticin 481 synthetase. J. Am. Ortopediveckan, Gothenburg.
Chem. Soc. 129, 10314–10315. Sawa, N., Wilaipun, P., Kinoshita, S., Zendo, T., Leelawatcharamas, V., Nakayama, J.,
Li, B., Sher, D., Kelly, L., Shi, Y., Huang, K., Knerr, P.J., Joewono, I., Rusch, D., Chisholm, Sonomoto, K., 2012. Isolation and characterization of enterocin W a novel two-
S.W., van der Donk, W.A., 2010. Catalytic promiscuity in the biosynthesis of cyclic peptide lantibiotic produced by Enterococcus faecalis NKR-4-1. Appl. Environ.
peptide secondary metabolites in planktonic marine cyanobacteria. PNAS 107, Microbiol. 78, 900–903.
10430–10435. Schmitz, S., Hoffmann, A., Szekat, C., Rudd, B., Bierbaum, G., 2006. The lantibiotic
Li, P., Li, X., Saravanan, R., Li, C.M., Leong, S.S.J., 2012. Antimicrobial macromolecules: mersacidin is an autoinducing peptide. Appl. Environ. Microbiol. 72, 7270–7277.
synthesis methods and future applications. RSC Adv. 2, 4031–4044. Schnell, N., Entian, K.D., Schneider, U., Götz, F., Zähner, H., Kellner, R., Jung, G., 1988.
Lohans, C.T., Huang, Z., van Belkum, M.J., Giroud, M., Sit, C.S., Steels, E.M., Zheng, J., Prepeptide sequence of epidermin, a ribosomally synthesized antibiotic with
Whittal, R.M., McMullen, L.M., Vederas, J.C., 2012. Structural characterization four sulphide-rings. Nature 333, 276–278.
of the highly cyclized lantibiotic Paenicidin A via a partial desulfuriza- Siezen, R.J., Kuipers, O.P., de Vos, W.M., 1996. Comparison of lantibiotic gene clusters
tion/reduction strategy. J. Am. Chem. Soc. 134, 19540–19543. and encoded proteins. Antonie Van Leeuwenhoek 69, 171–184.
Makino, A., Baba, T., Fujimoto, K., Iwamoto, K., Yano, Y., Terada, N., Ohno, S., Sato, S.B., Simone, M., Monciardini, P., Gaspari, E., Donadio, S., Maffioli, S.I., 2013. Isolation
Ohta, A., Umeda, M., Matsuzaki, K., Kobayashi, T., 2003. Cinnamycin (Ro 09-0198) and characterization of NAI-802, a new lantibiotic produced by two different
promotes cell binding and toxicity by inducing transbilayer lipid movement. J. Actinoplanes strains. J. Antibiot. (Tokyo) 66, 73–78.
Biol. Chem. 278, 3204–3209. Stevens, K.A., Sheldon, B.W., Klapes, N.A., Klaenhammer, T.R., 1991. Nisin treatment
Märki, F., Hänni, E., Fredenhagen, A., van Oostrum, J., 1991. Mode of action of the for inactivation of Salmonella species and other Gram-negative bacteria. Appl.
lanthionine-containing peptide antibiotics duramycin, duramycin B and C, and Environ. Microbiol. 57, 3613–3615.
cinnamycin as indirect inhibitors of phospholipase A2. Biochem. Pharmacol. 42, Suda, S., Cotter, P.D., Hill, C., Ross, R.P., 2011. Lacticin 3147 – biosynthesis, molecular
2027–2035. analysis, immunity, bioengineering and applications. Curr. Protein Pept. Sci. 13,
Mantovani, H.C., Hu, H., Worobo, R.W., Russell, J.B., 2002. Bovicin HC5, a bacteriocin 193–204.
from Streptococcus bovis HC5. Microbiology 148, 3347–3352. Szekat, C., Jack, R.W., Skutlarek, D., Färber, H., Bierbaum, G., 2003. Construction of an
Mantovani, H.C., Russell, J.B., 2008. Bovicin HC5 a lantibiotic produced by Strepto- expression system for site-directed mutagenesis of the lantibiotic mersacidin.
coccus bovis HC5, catalyzes the efflux of intracellular potassium but not ATP. Appl. Environ. Microbiol. 69, 3777–3783.
Antimicrob. Agents Chemother. 52, 2247–2249. Tagg, J.R., Dajani, A.S., Wannamaker, L.W., 1976. Bacteriocins of Gram-positive bacte-
Marsh, A., O’Sullivan, O., Ross, R.P., Cotter, P., Hill, C., 2010. In silico analysis high- ria. Bacteriol. Rev. 40, 722–756.
lights the frequency and diversity of type 1 lantibiotic gene clusters in genome Tang, W., van der Donk, W.A., 2012. Structural characterization of four prochlorosins:
sequenced bacteria. BMC Genom. 11, 679. a novel class of lantipeptides produced by planktonic marine cyanobacteria.
Meindl, K., Schmiederer, T., Schneider, K., Reicke, A., Butz, D., Keller, S., Gühring, H., Biochemistry 51, 4271–4279.
Vértesy, L., Wink, J., Hoffmann, H., Brönstrup, M., Sheldrick, G.M., Süssmuth, R.D., Tang, W., van der Donk, W.A., 2013. The sequence of the enterococcal
2010. Labyrinthopeptins: a new class of carbacyclic lantibiotics. Angew. Chem. cytolysin imparts unusual lanthionine stereochemistry. Nat. Chem. Biol.,
49, 1151–1154. http://dx.doi.org/10.1038/nchembio.1162 (2013 Jan 13 [Epub ahead of print]
Morgan, S.M., O’Connor, P.M., Cotter, P.D., Ross, R.P., Hill, C., 2005. Sequential actions PubMed PMID: 23314913).
of the two component peptides of the lantibiotic lacticin 3147 explain its antimi- Teng, Y., Zhao, W., Qian, C., Li, O., Zhu, L., Wu, X., 2012. Gene cluster analysis for the
crobial activity at nanomolar concentrations. Antimicrob. Agents Chemother. 49, biosynthesis of elgicins, novel lantibiotics produced by Paenibacillus elgii B69.
2606–2611. BMC Microbiol. 12, 45.
Müller, W.M., Ensle, P., Krawczyk, B., Suessmuth, R.D., 2011. Leader peptide-directed van Heel, A.J., Montalban-Lopez, M., Kuipers, O.P., 2011. Evaluating the feasibility
processing of labyrinthopeptin A2 precursor peptide by the modifying enzyme of lantibiotics as an alternative therapy against bacterial infections in humans.
LabKC. Biochemistry 50, 8362–8373. Expert Opin. Drug Metab. Toxicol. 7, 675–680.
Müller, W.M., Schmiederer, T., Ensle, P., Suessmuth, R.D., 2010. In vitro biosyn- van Kraaij, C., de Vos, W.M., Siezen, R., Kuipers, O.P., 1999. Lantibiotics: biosynthesis
thesis of the prepeptide of type-III lantibiotic labyrinthopeptin A2 including mode of action and applications. Nat. Prod. Rep. 16, 575–587.
formation of a C C bond as a post-translational modification. Angew. Chem. Voeller, G.H., Krawczyk, J.M., Pesic, A., Krawczyk, B., Nachtigall, J., Süssmuth,
49, 2436–2440. R.D., 2012. Characterization of new Class III Lantibiotics—Erythreapeptin,
Nagao, J., Morinaga, Y., Islam, M.R., Asaduzzaman, S.M., Aso, Y., Nakayama, J., Avermipeptin and Griseopeptin from Saccharopolyspora erythraea, Streptomyces
Sonomoto, K., 2009. Mapping and identification of the region and secondary avermitilis and Streptomyces griseus demonstrates stepwise N-Terminal leader
structure required for the maturation of the nukacin ISK-1 prepeptide. Peptides processing. ChemBioChem 13 (8), 1174–1183.
30, 1412–1420. Wang, H., van der Donk, W.A., 2012. Biosynthesis of the class III lantipeptide
Naruse, N., Tenmyo, O., Tomita, K., Konishi, M., Miyaki, T., Kawaguchi, H., Shiba, T., catenulipeptin. ACS Chem. Biol. 7, 1529–1535.
1989. Lanthiopeptin, a new peptide antibiotic. Production, isolation and prop- Wescombe, P.A., Dyet, K.H., Dierksen, K.P., Power, D.A., Jack, R.W., Burton, J.P.,
erties of lanthiopeptin. J. Antibiot. 42, 837–845. Inglis, M.A., Wescombe, A.L., Tagg, J.R., 2012. Salivaricin G32, a homolog
Oliynyk, I., Varelogianni, G., Roomans, G.M., Johannesson, M., 2010. Effect of of the prototype Streptococcus pyogenes nisin-like lantibiotic SA-FF22, pro-
duramycin on chloride transport and intracellular calcium concentration in cys- duced by the commensal species Streptococcus salivarius. Int. J. Microbiol.,
tic fibrosis and non-cystic fibrosis epithelia. A.P.M.I.S 118, 982–990. http://dx.doi.org/10.1155/2012/738503 (in press).
62 J. Dischinger et al. / International Journal of Medical Microbiology 304 (2014) 51–62

Wiedemann, I., Benz, R., Sahl, H.G., 2004. Lipid II-mediated pore formation by Willey, J.M., Willems, A., Kodani, S., Nodwell, J.R., 2006. Morphogenetic surfactants
the peptide antibiotic nisin: a black lipid membrane study. J. Bacteriol. 186, and their role in the formation of aerial hyphae in Streptomyces coelicolor. Mol.
3259–3261. Microbiol. 59, 731–742.
Wiedemann, I., Böttiger, T., Bonelli, R.R., Wiese, A., Hagge, S.O., Gutsmann, T., Sey- Wilson-Stanford, S., Smith, L., 2011. Commercial development and appli-
del, U., Deegan, L., Hill, C., Ross, P., Sahl, H.G., 2006. The mode of action of the cation of type A lantibiotics. Recent Pat. Antiinfect. Drug Discov. 6,
lantibiotic lacticin 3147 – a complex mechanism involving specific interaction 175–185.
of two peptides and the cell wall precursor lipid II. Mol. Microbiol. 61, 285–296. Yonezawa, H., Kuramitsu, H.K., 2005. Genetic analysis of a unique bacteriocin,
Wiedemann, I., Breukink, E., van Kraaij, C., Kuipers, O.P., Bierbaum, G., Kruijff, B., de Smb, produced by Streptococcus mutans GS5. Antimicrob. Agents Chemother.
Sahl, H.G., 2001. Specific binding of nisin to the peptidoglycan precursor lipid 49, 541–548.
II combines pore formation and inhibition of cell wall biosynthesis for potent Zaehner, H., Goetz, F., Hoerner, T., Werner, R.G., Allgaier, H., Jung, G., Kellner, R.,
antibiotic activity. J. Biol. Chem. 276, 1772–1779. 1998. New peptide antibiotic gallidermin is produced by Staphylococcus gal-
Willey, J.M., van der Donk, W.A., 2007. Lantibiotics: peptides of diverse structure linarum and used esp. for treating eczema, impetigo, cellulitis and acne. U.S.
and function. Ann. Rev. Microbiol. 61, 477–501. patent 5,843,709-A.

You might also like