You are on page 1of 28

ANRV322-MI61-23 ARI 6 August 2007 18:7

Lantibiotics: Peptides
of Diverse Structure
and Function
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Joanne M. Willey1 and Wilfred A. van der Donk2


by Pennsylvania State University on 05/22/12. For personal use only.

1
Department of Biology, Hofstra University, Hempstead, New York 11549;
email: biojmw@hofstra.edu
2
Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana,
Illinois 61801; email: vddonk@uiuc.edu

Annu. Rev. Microbiol. 2007. 61:477–501 Key Words


First published online as a Review in Advance on bacteriocin, peptide antibiotic, peptide engineering,
June 18, 2007
posttranslational modification
The Annual Review of Microbiology is online at
micro.annualreviews.org Abstract
This article’s doi: The current need for antibiotics with novel target molecules has co-
10.1146/annurev.micro.61.080706.093501
incided with advances in technical approaches for the structural and
Copyright  c 2007 by Annual Reviews. functional analysis of the lantibiotics, which are ribosomally synthe-
All rights reserved
sized peptides produced by gram-positive bacteria. These peptides
0066-4227/07/1013-0477$20.00 have antibiotic or morphogenetic activity and are structurally defined
by the presence of unusual amino acids introduced by posttransla-
tional modification. Lantibiotics are complex polycyclic molecules
formed by the dehydration of select Ser and Thr residues and the
intramolecular addition of Cys thiols to the resulting unsaturated
amino acids to form lanthionine and methyllanthionine bridges, re-
spectively. Importantly, the structural and functional diversity of the
lantibiotics is much broader than previously imagined. Here we dis-
cuss this growing collection of molecules and introduce some re-
cently discovered peptides, review advances in enzymology and pro-
tein engineering, and discuss the regulatory networks that govern
the synthesis of the lantibiotics by the producing organisms.

477
ANRV322-MI61-23 ARI 6 August 2007 18:7

protein structures. In this review, we discuss


Contents the lantibiotics, with an emphasis on recent
findings. The reader is directed to a number
INTRODUCTION . . . . . . . . . . . . . . . . . 478
of other reviews that offer a more historical
AN OVERVIEW OF
perspective (12, 21, 94, 117).
LANTIBIOTIC
STRUCTURES . . . . . . . . . . . . . . . . . 478
BIOLOGICAL ACTIVITIES AND AN OVERVIEW OF
APPLICATIONS . . . . . . . . . . . . . . . . 479 LANTIBIOTIC STRUCTURES
CLASSIFICATION SCHEMES . . . . . 481
Lantibiotics are small (19–38 amino acids)
SELF-IMMUNITY . . . . . . . . . . . . . . . . . 481
peptides that undergo extensive posttrans-
LANTIBIOTIC STRUCTURE
lational modification. The least modified
AND FUNCTION . . . . . . . . . . . . . . 484
peptide to date is lactocin S (24%), and
Class I Lantibiotics: Nisin,
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

the recently described compound 107891


Subtilin, and Related Peptides . . 484
is the most heavily modified (58%) (65).
Class II Lantibiotics . . . . . . . . . . . . . . 485
by Pennsylvania State University on 05/22/12. For personal use only.

The posttranslational modifications common


Class III Lantibiotics: SapB and
to all lantibiotics involve the dehydration
SapT . . . . . . . . . . . . . . . . . . . . . . . . . 487
of Ser and Thr residues in the propep-
Posttranslational Modification
tide to yield 2,3-didehydroalanine (Dha) and
Enzymes and Engineering
(Z )-2,3-didehydrobutyrine (Dhb), respec-
Studies . . . . . . . . . . . . . . . . . . . . . . . . 487
tively (Figure 1a). This is followed by the
LANTIBIOTIC GENETICS . . . . . . . 488
stereospecific intramolecular addition of a
Biosynthetic Gene Clusters . . . . . . . 488
Cys residue onto Dha or Dhb to form a lan-
Regulation of Lantibiotic Synthesis 489
thionine (Lan) or methyllanthionine (MeLan)
CONCLUSIONS AND
bridge, respectively. A representative example
OUTLOOK . . . . . . . . . . . . . . . . . . . . . 492
is shown in Figure 1b for nisin.
The modified peptide is then exported
and a leader sequence (23- to 59-amino-acid
residues) is proteolytically removed, although
INTRODUCTION not always in that order. Lantibiotic leader se-
Lantibiotic: The lantibiotic nisin (Figure 1) produced by quences are conserved among groups of lan-
gene-encoded, Lactococcus lactis was first discovered in 1927 tibiotics (12) but bear no sequence homology
posttranslationally
(92). This peptide has become the best under- to the Sec-dependent or twin-arginine (TAT)
modified peptide
containing one or stood of over 50 lantibiotics produced by both exported proteins, with the exception of the
more Lan and/or low and high G+C gram-positive bacteria. leader peptide of cinnamycin (123). In cases
MeLan residues Lantibiotics are defined by posttranslational in which the function of the leader has been
Propeptide: modifications that introduce the thioether specifically addressed, it is required for secre-
C-terminal segment amino acids lanthionine and methyllanthio- tion (15, 58, 90, 110, 115) and ensures com-
of prepropeptide that nine; in fact the term lantibiotic is derived plete maturation (14, 29, 66, 130). For those
is posttranslationally
from lanthionine-containing antibiotics (96). lantibiotics that are proteolytically processed
modified to
ultimately become Although the lantibiotics were once thought extracellularly, the leader may prevent activa-
the mature to be a relatively small group with limited bio- tion of the lantibiotic until it is safely outside
lantibiotic logical activity, a larger collection of peptides the cell (114), as lantibiotics with their leader
Dha: 2,3- is emerging with a widening array of struc- sequence attached have no antimicrobial ac-
didehydroalanine tures and functions. These molecules are in- tivity (62, 66, 114, 130).
Dhb: (Z )-2,3- triguing because of their unique biochemistry, At present, no fewer than 15 differ-
didehydrobutyrine genetic regulation, range of biological func- ent posttranslational modifications have been
tions, and potential for engineering unique documented in lantibiotics. This structural

478 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

a H O H
O b NisA
N N Leader-ITSISLCTPGCKTGALMGCNMKTATCHCSIHVSK
Xn
R OH SH - 8 H2O NisB
(Dehydratase)
Dehydration

O H O
H Leader-IDhbDhaIDhaLCDhbPGCKDhbGALMGCNMKDhbADhbCHCSIHVDhaK
N N
Xn
R S NisC
H (Cyclase)
R=H Dha
R = CH3 Dhb
A C E
S S S
Cyclization
Leader-IDhbAlaIDhaLAlaAbuPGAlaKAbuGALMGAlaNMKAbuAAbuAlaHAlaSIHVDhaK
Xn S S
B D
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Proteolysis NisP
O S NH (Protease)
D L
by Pennsylvania State University on 05/22/12. For personal use only.

HN R O
O Ala Leu
HN Met
R=H (2S,6R)-lanthionine
R = Me (2S,3S,6R)-3-methyllanthionine Gly
Ile Leu O Gly
H
O NH N O
S NH NH S
Lys His
S NH Lys
HN O S O Me O NH
O Me NH
O O Asn Met
Me NH Ala O O
HN Pro Gly N
H
Ile Me S Ser
Me Ile
Nisin A Val His
Lys
N
O H

Figure 1
(a) Installation of lanthionine (Lan) or methyllanthionine (MeLan) residues into lantibiotic prepeptides.
(b) The posttranslational maturation process of nisin. For Lan and MeLan structures, the segments
derived from Ser/Thr are in red and those derived from Cys are in blue.

diversity releases the peptides from the con- isoleucine (76). The antibiotic 107891 con-
straints imposed by the use of only 20 amino tains two previously unknown modifications:
acids (Figure 2). For instance C-terminal a chlorinated Trp and a mono- or dihydroxy-
Lan: lanthionine
Cys residues may form S-aminovinyl-d- lated Pro (65).
MeLan:
cysteine (AviCys) or S-aminovinyl-3-methyl-
3-methyllanthionine
d-cysteine (AviMeCys) structures, and Dha
and Dhb residues that are N-terminally ex-
BIOLOGICAL ACTIVITIES AviCys: S-[(Z)-2-

posed after leader processing spontaneously


AND APPLICATIONS aminovinyl]-d-
cysteine
hydrolyze to yield 2-oxopropionyl (OPr) and At a time when there is an urgent need
AviMeCys: S-[(Z)-
2-oxobutyryl (OBu) groups (50). OPr may for new antibiotics, lantibiotics demon- 2-aminovinyl]-d-3-
be further modified by reduction to a 2- strate promising chemotherapeutic poten- methylcysteine
hydroxypropionyl (Hop) residue (30, 111). tial. Many lantibiotics are bacteriocidal OPr:
Moreover, l-Ser can be converted to d-Ala against a variety of gram-positive bacteria 2-oxopropionyl
with Dha as an intermediate (20). Cinnamycin at nanomolar levels and some are active OBu: 2-oxobutyryl
and the closely related duramycins have a lysi- against methicillin-resistant Staphylococcus au-
Hop:
noalanine bridge, as well as a hydroxylated reus (MRSA), vancomycin-resistant entero- 2-hydroxypropionyl
Asp (48, 135), and cypemycin has an allo- cocci, and oxacillin-resistant gram-positives

www.annualreviews.org • Lantibiotic Function and Diversity 479


ANRV322-MI61-23 ARI 6 August 2007 18:7

O NH O Me NH O
O H
(D) (L) (D) (L) H N
N
HN S O HN S O
H3C
meso-lanthionine (2S,3S,6R)-3-methyllanthionine 2,3-Didehydroalanine (Z)-2,3-didehydrobutyrine

Ala Ala Abu Ala Dha Dhb


S S

O
O O
H
N CH3
O NH O H
(L)
(CH2)3 (L)
(D)
HO
O 2-Oxopropionyl OPr
HN N O HN S NH
H OH
O
(2S,9S)-lysinoalanine AviCys erythro-3-hydroxy-
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

H3C
L-aspartic acid
O
Ala
Ala Lys S Asp OH 2-Oxobutyryl OBu
by Pennsylvania State University on 05/22/12. For personal use only.

N
H NHR
O
O HO
O Me H O
H
H2N (D) N CH3
HN S NH
2-Hydroxypropionyl Hop
NH
Figure 2
allo-isoleucine AviMeCys ClTrp O
Posttranslational
N
modifications that O Abu Cl
H HPro
have been reported N S
D-Ala HO OH
to date for the NHR
lantibiotic family. Dihydroxyproline

(22). To date only nisin has been used com- of phospholipase A2 (69). Duramycin in-
mercially, with a 40-year history as a preser- creases chloride secretion in lung epithelium
vative in food products. There is interest in (16) and has been in clinical trials to evalu-
expanding its use to food packaging, as well as ate its efficacy in clearing mucus secretions
introducing it to clinical applications in both from the lungs associated with cystic fibro-
human and animal health products (22, 117). sis and other airway diseases (77). The ente-
Some interesting clinical applications have rococcal lantibiotic cytolysin not only targets
been proposed and/or are in clinical trials. other gram-positives, but functions as a vir-
The lantibiotic mutacin 1140 is produced ulence factor, lysing erythrocytes and poly-
by Streptococcus mutans, the major causative morphonuclear leukocytes (24). Perhaps the
agent of dental caries. The compound is effec- most novel activity is found in the peptides
tive against many strains of the same species, SapB and SapT, produced by the filamen-
prompting interest in this peptide as an agent tous soil microbes Streptomyces coelicolor and
to prevent dental caries (36, 120). Along sim- S. tendae, respectively (54, 55). Streptomyces
ilar lines, lozenges seeded with a salivaricin spp. undergo a complex, fungal-like life cycle
A–producing strain of S. salivarius have been that involves the differentiation of substrate-
introduced as a probiotic to combat S. pyogenes associated hyphae into upwardly growing
in the oral cavity and prevent halitosis (9, 122). aerial hyphae that then form reproductive
Not all lantibiotics function strictly as an- spores. SapB (54) and SapT (55) are both hy-
timicrobial agents, however. Cinnamycin and drophobic and surface active; they are thought
the related duramycins are potent inhibitors to function as biosurfactants, releasing the

480 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

surface tension at the colony-air interface protein with a conserved N-terminal cys-
to facilitate the emergence of nascent aerial teine protease domain (84). Despite the func-
hyphae (105). tional difference between class I and class II
Bacteriocin:
Thus, while lantibiotics were initially dis- exporters, they share the designation LanT. bacterial proteins
covered, further isolated, and explored as Unique within the class II molecules are the that demonstrate
bacteriocins, the variety of lantibiotic activi- two-component lantibiotics, in which two antibiotic activity
ties now realized suggests that their functional peptides assemble to form the mature antibi- against other strains
of the same species
diversity may be underestimated. By screen- otic. Each peptide is encoded by its own struc-
or closely related
ing only for antibiotic activity, lantibiotics that tural gene and modified by separate LanM species
demonstrate other, unexpected biological ac- enzymes (71, 73). However, a single LanT
tivities may have been missed. protein removes the leader peptide and se-
cretes both products.
We propose that a new, third class be es-
CLASSIFICATION SCHEMES
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

tablished for lanthionine-containing peptides


In this review we adopt a classification scheme that lack significant antibiotic activity but in-
by Pennsylvania State University on 05/22/12. For personal use only.

that places all lantibiotics in one of three stead perform another function for the pro-
classes (Class I, II, or III) on the basis of ducing cell. To date three such peptides have
the pathway by which maturation of the pep- been described: S. coelicolor secretes SapB (54,
tide occurs and the presence or absence of 55, 128), AmfS is produced by S. griseus (109),
antibiotic activity. This designation incorpo- and SapT is the product of S. tendae (55,
rates differences in leader peptide sequence, 128). Of these morphogenetic peptides, only
biosynthetic operon structure, peptide func- the structure of SapT has been confirmed by
tion, and to a lesser extent, the structure of the NMR spectroscopy; the structure of SapB is
mature lantibiotic. This approach is based on inferred from its amino acid sequence and
a scheme first suggested by Pag & Sahl (84) mass spectrometry (54). AmfS has been puri-
and is simple, is inclusive, and may be suffi- fied, but its structure, like that of three other
ciently flexible to accommodate lanthionine- SapB orthologues (two produced by S. scabies
containing peptides that have not yet been and one by S. avermitilis), has not been rigor-
discovered. ously explored (128).
Class I lantibiotic prepeptides are mod- Apart from their unusual function, other
ified by two distinct enzymes: the gener- factors further distinguish SapB and its known
ically termed LanB enzyme, which dehy- orthologues from antimicrobial lantibiotics.
drates Thr and Ser residues, and LanC, which Although the putative modification enzymes
mediates cyclization. The peptides are ex- for SapB and AmfS (RamC and AmfT, respec-
ported by LanT, a dedicated ABC transporter. tively) bear homology to the C-terminal cy-
Class I leader peptides are cleaved by LanP, clization domain of LanM enzymes, they lack
subtilisin-like serine proteases. These lantibi- the zinc ligands that are important for cycliza-
otics are more linear than class II (Figure 3). tion by LanM. Thus, the mechanism of mat-
Class II lantibiotics are modified by the uration of these class III lantibiotics may be
LanM-type modification enzymes, which are distinct.
large (900–1000 amino acids) proteins that ex-
hibit both dehydratase and cyclase activities.
Whereas LanM enzymes bear no homology SELF-IMMUNITY
to LanB proteins, they exhibit low sequence Antibiotic synthesis requires that producer or-
identity to the LanC enzymes (97), includ- ganisms have a mechanism conferring resis-
ing three zinc binding ligands (83). Class II tance to their product. In the case of lan-
lantibiotic secretion and leader processing is tibiotics, immunity can be provided by a
also performed by a single, multifunctional specific immunity protein (LanI) and/or by

www.annualreviews.org • Lantibiotic Function and Diversity 481


ANRV322-MI61-23 ARI 6 August 2007 18:7

specialized ABC transporter proteins encoded for self-immunity exists for the Bacillus subtilis
by the lanFEG genes. The mechanism of Pep5 product subtilin (102), but despite the struc-
self-immunity is perhaps one of the simplest; tural similarity between the two lantibiotics,
a single PepI peptide appears to bind at the NisI cannot protect L. lactis from subtilin
membrane–cell wall interface, thereby mask- (103). Another variation of the LanI/LanFEG
ing the Pep5 target molecule (38). In many theme is represented by NukH, which helps
cases, however, LanI cannot confer full resis- confer immunity to nukacin ISK-1. NukH
tance and it functions cooperatively with the differs from NisI-like proteins in that it is
ABC transporters. NisI, which has a lipopro- smaller and has three transmembrane do-
tein signal sequence attaching it to the outer mains. NukH functions synergistically with
face of the membrane, intercepts nisin to re- the NukFEG transporter to provide self-
duce its local concentration in the immedi- protection as well as immunity to the struc-
ate environment (103). A similar mechanism turally similar lantibiotic lacticin 481 (2).
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Class I
by Pennsylvania State University on 05/22/12. For personal use only.

Nisin A Ala Leu


Gly Met S Ser Ile His Val Dha Lys OH
E Ala
Abu C Abu
Gly Ala
Dha Ala His
H Ile Ile Leu Lys Ala D
S S Abu S
Dhb Ala A Ala Abu B Ala Asn
Lys
Pro Gly Met
S

Ala Leu
Figure 3 Subtilin Gly Gln S Lys Ile Dha Lys OH
Ala
Abu
Representative Dha
Abu Dhb Leu Ala Asn
examples of the H Trp Glu Leu
Val S Ala
S Abu
three classes of Lys Ala Ala S
Abu Ala Phe
lantibiotics. The Gln
S Pro Gly Leu
same color-coding
and shorthand
notation is used as Epidermin Asn Ala S
Phe
defined in H Lys Ile Phe Tyr
Ile S
Figure 1. In most Ala
NH
Ala Ala Ala Abu Ala Lys Ala Ala
cases the Ser/Thr S
S
is located Pro Gly Dhb Gly

N-terminal of the
cysteine that forms
107891
a Lan/MeLan with Dha
ClTrp Leu
it. Exceptions are S Thr Ala
S
Ala
S
Ala
found in H Val Dhb Ala Ala Abu Ala Phe
cinnamycin S HPro Asn
Pro Gly Ala NH
(Cys1 + Thr18, Gly Ala
S
Cys5 + Thr11) Gly Gly

(40, 135),
mersacidin Val
Lys
Gln
(Cys1 + Thr2) Pep5 Lys OH
Ala Ala Ala Gln Lys Dhb Leu Phe Abu Val
(94), and the S S Ala
α-peptide of Arg Leu Arg Ala
Gly
S
lacticin 3147 OBu Ala Gly Pro Ala Ile Lys Ala Dhb Ala
Asn
(Cys1 + Thr2) Lys
(70). Gly Lys

482 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

Class II

Cinnamycin Lacticin 481 Mersacidin


S
OH
11 Ser Gly Dhb Gly
Phe Abu Phe HO Phe H Gly
Gly Asp Ala Gly
Val Pro Gly
Asn B Val Ala Ser Ala Val Ala Ile
Ala14 Ala Pro Gly
S S C Gly Gly Ile His C B S
S Ile S Glu
Abu S Phe Abu Leu Val HN
A His Abu A Glu B
Ala1 4 Phe Lys
Gln Ala Ala Trp Gln C Dha D
Lys D Arg Ala5 H Ala Phe Abu Ala
Ala6 S Abu S
Asn Abu
HO Asn Met S Leu
N
H

Lactocin S Sublancin
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Trp Leu
Ala
Leu Leu
Val Ala His His Gly Gly
Dha Ala Gln Ala
Abu Gln Lys
Pro Lys Ala Cys
Pro D-Ala Thr Ala Ala OH Ile Cys
by Pennsylvania State University on 05/22/12. For personal use only.

Leu Val Gly


Gly Ala S S
Dhb Val Ala Asp Gly S
Leu S S Leu
OPr Ala Ala Ala S
D-Ala Ala Cys
H Glu Ala Gly
S Cys
Val Val Phe Tyr Gly Phe Arg
Met Tyr Ala Gln
D-Ala Leu Lys Gly Val Asn Gln OH H
Gly Ala
Tyr Arg

Lacticin 3147 A1 (Ltn α) Lacticin 3147 A2 (Ltn β)


Asn
Gly Asn
Dhb Asn Pro Ala
Trp Gly Ile Asn S
Dhb Dhb Thr
Ala Thr OH
S A B Ala Met Ala C Ala
Phe Tyr Ala Ala D-Ala A Abu
Ala S Glu Ala Arg Ala
Ala Ile Ile S S
Asp Trp Pro Ala
D-Ala
H C His D Trp Leu Tyr Pro B
Leu Ala Ala OBu Dhb Abu Thr Lys
Abu Ala
Abu Ala D-Ala
S Leu
S Lys OH

Haloduracin α Haloduracin β
Gly Asn Lys
Leu
Arg Gly Ala Dhb
Val Ala
A Ala Met Ala Val S
Ala Gly OH
H2N O Val Ala Leu D Ala
S Glu Pro Pro B Abu
NH Tyr
A S S Ala Dha Gln
O S S B Val C Ser Trp S Ala
Ala Ala C
Abu Pro
Ala Abu Ala Dhb
Ile S Leu Abu Thr Lys
Trp Tyr Asn S Asn OH Abu H

Class III
SapB SapT
S S
H Thr Gly Ala Ala Gly Ala Ala Ala Asn OH
S S S S
Arg Leu Ser Thr H Tyr Abu Ala Ala Ala Abu Ala Ala Abu Ala Gly OH
Gln Gly Gly Leu
Ala Leu Leu Thr Ile Ile Val Ile
Val Val
Dha Leu Dha Ile

Figure 3
(Continued )

www.annualreviews.org • Lantibiotic Function and Diversity 483


ANRV322-MI61-23 ARI 6 August 2007 18:7

LANTIBIOTIC STRUCTURE a flexible hinge region (112). The MeLan and


AND FUNCTION Lan bridging patterns found in subtilin and
ericin S produced by B. subtilis ATCC6633
Prepropeptide: Our understanding of the various modes of
precursor peptide for and B. subtilis A1/3, respectively (99), are iden-
action of lantibiotics has greatly increased
posttranslational tical to that of nisin. B. subtilis A1/3 also pro-
in recent years, as advances in peptide engi-
modification. Also duces ericin A, in which the three N-terminal
neering have facilitated the investigation of
called prepeptide bridges are conserved, but differs from sub-
structure-activity relationships. A short sum-
Lipid II: tilin by 13 residues and the position of the
mary of these developments is provided in the
undecaprenyl- two C-terminal MeLan residues. The gene
pyrophosphoryl- following sections on select members of the
clusters encoding subtilin and ericin A and S
MurNAc- three classes of lantibiotics defined above.
(pentapeptide)- are strikingly similar, with the exception that
GlcNAc, B. subtilis A1/3 possesses two prepropeptide
membrane- Class I Lantibiotics: Nisin, Subtilin, structural genes. It has been speculated that
and Related Peptides
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

associated this cluster underwent two duplications of the


peptidoglycan
L. lactis strains produce at least three struc- subtilin structural gene, spaS, to give rise to
monomer that is
by Pennsylvania State University on 05/22/12. For personal use only.

polymerized and tural variants of nisin. Nisin A is the proto- the two ericin paralogues (99).
cross-linked to type (Figure 1), nisin Z differs by one amino The pore-forming activity of nisin is
generate the acid, and nisin Q differs at four positions; unique in that the cell wall precursor lipid II is
bacterial cell wall all are 34 residues in length (27). Streptococ- used as a docking molecule (6, 8) (Figure 4).
cus uberis produces an additional variant, nisin The N-terminal amides of the A and B rings
U, which has 78% sequence identity to nisin of nisin form a cage that binds the pyrophos-
A and lacks the C-terminal three residues phate moiety of lipid II (33, 43, 44, 127).
(129). Nisin is a flexible molecule with two This binding motif is also found in other lan-
amphiphilic domains consisting of three N- tibiotics that interact with lipid II, including
terminal rings (labeled A, B, and C) and the subtilin, epidermin, gallidermin, and mutacin
C-terminal D and E rings, which are joined by 1140 (4, 34). Epidermin and gallidermin lack

Figure 4
The biosynthesis
of the bacterial cell
wall with the
binding sites for
the glycopeptide
antibiotic
vancomycin and
the lantibiotic nisin
indicated. By using
lipid II as a docking
molecule, nisin
inhibits
polymerization of
murein subunits in
addition to forming
pores in the plasma
membrane.

484 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

the C-terminal tail and are shorter (∼30 Å) and three overlapping thioether bridges ren-
compared with nisin (50 Å). This difference der the C terminus globular (Figure 3). Site-
may explain why these two compounds do not directed mutagenesis of mutacin II shows that
form pores in bacteria that are susceptible to its hinge is essential for antibiotic activity and
pore formation by nisin (4). while all three thioether linkages are impor-
The C terminus of nisin inserts into the tant, if not required, for full activity (14), sub-
membrane in a perpendicular orientation with stitution of the MeLan bridge with Lan or
respect to the bilayer surface (44, 116), re- vice versa has no effect on activity in mutacin
sulting in stable pores 2–2.5 nm in diameter II and lacticin 481 (11, 14, 29).
(124) consisting of eight nisin and four lipid II Lacticin 481 and its relatives are believed
molecules (33). This stoichiometry differs to form pores in target microbes (29), al-
from the NMR structure of the 1:1 nisin– though the means by which this is accom-
lipid II complex, and it is possible that nisin- plished has not been fully explored. The A
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

nisin interactions account for the difference. ring in this subfamily shows similarity with the
Attaining a transmembrane structure requires C ring of mersacidin, which is critical for its
by Pennsylvania State University on 05/22/12. For personal use only.

the presence of the flexible hinge region; nisin activity (104). The observation that microbes
Z peptides mutated in this region are unable producing these lantibiotics have similar im-
to form pores but retain the ability to bind munity proteins that prevent and/or remove
lipid II (127, 133). Breukink and coworkers the peptide from the membrane further sug-
(34) recently demonstrated that nisin and mu- gests that the membrane is targeted (29).
tacin 1140 disrupt the functional localization
of lipid II. This cell wall precursor is nor- Mersacidin. The discovery that mersacidin
mally found at sites of nascent cell wall biosyn- is active against methicillin-resistant S. au-
thesis such as the septum and along helical reus in a murine model (57) has prompted ef-
bands encircling the cylindrical axis of rod- forts to determine the key structural compo-
shaped bacteria (25, 106, 113). Nisin-treated nents of this peptide. Unlike nisin and other
cells sequester lipid II in patches on the bac- cationic peptides, mersacidin does not form
terial membrane, removing it from the site of pores in plasma membranes. Instead, it ap-
cell wall biosynthesis. The extreme sensitiv- pears to inhibit the transglycosylation step in
ity observed in susceptible bacteria and the peptidoglycan biosynthesis (7). Although the
absence of nisin-resistant mutants reflect the molecular details remain to be resolved, its
dual modes of nisin activity: pore formation conformation changes in the presence of lipid
coupled with the inhibition of cell wall biosyn- II (42), and the C ring (particularly the Glu
thesis (33, 34). therein) is believed to be essential for recog-
nition (41, 104). In fact, this ring is conserved
in the lacticin 481 subgroup of lantibiotics,
Class II Lantibiotics the two-component lantibiotics (108), and in
The class II lantibiotics are structurally di- plantaricin C (125).
verse but are similar in that they use a single
enzyme to catalyze dehydration and cycliza-
Cinnamycin and the duramycins. Cin-
tion. It currently represents the largest of the
namycin and three duramycin variants are
three classes of lantibiotics.
streptomycete products (123) and are remark-
able for the presence of unusual modifications
Lacticin 481 and its relatives. Lacticin 481 including a lysinoalanine ring (Figures 2 and
is one of about 20 related lantibiotics (29). All 3). They bind to phosphatidylethanolamine
are hydrophobic with no net charge at neu- (PE) with a 1:1 stoichiometry (119), inducing
tral pH. In all cases, the N terminus is linear transbilayer lipid movement (68).

www.annualreviews.org • Lantibiotic Function and Diversity 485


ANRV322-MI61-23 ARI 6 August 2007 18:7

Two-component lantibiotics. Lantibiotics presumed to be important in lipid II bind-


that consist of two modified peptides in- ing. The N-terminal MeLan ring identified
clude plantaricin W, staphylococcin C55, cy- in Halβ (73) appears to be conserved in all
tolysin L, Smb, BHT-A, haloduracin, and β-peptides except lacticin 3147 and staphylo-
lacticin 3147. Both the mature peptides coccin C55. This ring is followed by a stretch
and prepeptides are often called LanA1 and of hydrophobic amino acids, where Ser to d-
LanA2; here we differentiate the two by us- Ala conversions occur in LtnA2 of lacticin
ing LanA1/LanA2 to refer to the unmodi- 3147. The B ring is also relatively conserved
fied prepeptides and Lanα/Lanβ to designate among currently known two-component lan-
the mature peptide subunits. In all cases ex- tibiotics, absent only in BHT and Smb. Fi-
cept cytolysin, the α-peptide resembles mer- nally, the two most C-terminal Lan/MeLan
sacidin and the β-peptide is relatively long and rings are conserved in all β-peptides except
flexible (70) (Figure 5). cytolysin.
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

To date only the structure of lacticin 3147 Each peptide of the two-component
has been fully elucidated by NMR spec- lantibiotics displays weak or no antimicrobial
by Pennsylvania State University on 05/22/12. For personal use only.

troscopy (70), with the structure proposed activity, but synergistic interaction of the
for haloduracin strongly supported by tan- molecules results in strong antibiotic activity.
dem mass spectrometry (73) (Figure 3). This A nisin-like model for lacticin 3147 has been
structural information combined with se- proposed (126), in which Ltnα is thought
quence homology suggests that the three C- to bind lipid II and recruit Ltnβ to form
terminal rings in all α-peptides have the same pores in target cytoplasmic membranes
topology, which in analogy to mersacidin is (78, 126). A recent study in which every

a
A B C
HalA1 MTNLLKEWKMPLERTHNNSNPAGDIFQELEDQDILAGVNGA ––CAWYNI–SCRLGNKGAYCTLTVECMPSCN 69
PlwAα MK––ISKIEAQAR––KDFFKKIDTNSNLLNVNGA –KCKWWNI–SCDLGNNGHVCTLSHECQVSCN 59
SacAα MKSSFLEKDIEEQ––VTWFEEVSEQEFDDDIFGA CSTNTFSL–SDYWGNKGNWCTATHECMSWCK 62
LtnA1 MN––––KNEIETQP–VTWLEEVSDQNFDEDVFGA CSTNTFSL–SDYWGNNGAWCTLTHECMAWCK 59
BhtA1 MK–EIQKAGLQEEL–SILMDDAN––NLEQLTAGI GTTVVNSTFSIVLGNKGYICTVTVECMRNCQ 61
SmbA1 MK–EIQKAGLQEEL–SILMDDAN––NLEQLTAGI GTTVVNSTFSIVLGNKGYICTVTVECMRNCSK 61

b A B C D
HalA2 MVNSKDLRNPEFRKAQGLQFVDEVNEKELSSLAGSGDVHAQ-TTWPC––ATVGV––––SVALC–PTTKCTSQC 65
PlwAβ MTKTSRRKNAIANYLEPVDEKSINESFGAGDPEAR–SGIPC–TIGAAVAA––SIAVC–PTTKCSKRCGKRKK 67
BhtA2 MKSNLLKINNVTEVEKDMVTLIKDEDMELAGG–––––––STPAC––AIGVVGI––TVAVTGISTACTSRCINK 62
SmbA2 MKSNLLKINNVTEMEKNMVTLIKDEDMLAGG–––––––STPAC––AIGVVGI––TVAVTGISTACTSRCINK 61
LtnA2 MKEKNMKKNDTIELQLGKYLEDD–MIELAEGDESHGG–––––––TTPATP–AISILSAYISTNTC–PTTKCTRAC 65
SacAβ MKNELGKFLEENELELGKFSESDMLEITDDEVYAAGTPLALLGGAAT––GVIGYI–––SNQTCP–TTACTRAC 67
CylS-A MLNKENQENYYSNKLELVGPSFEE––LSLEEMEAIQGSGDVQAE–TTPACFTIGLGVGALFSAKFC 63
CytL-A MENLSVVPSFEELSVEEMEAIQGSGDVQAE–TTPVC–AVAATAAA––SSAACGWVGGGIFTGVTVVVSLKHC 68

Figure 5
Sequence alignment of (a) the LanA1 and (b) the LanA2 prepeptides of haloduracin (HalA) (73),
plantaracin W (PlwA) (39), staphylococcin C55 (SacA) (80), lacticin 3147 (LtnA) (28), BHT (BhtA) (45),
Smb (SmbA) (131), and the two peptides of cytolysin (CylSA and CytLA) (24). The gene names used
here are as found in the databases, with the exception of SmbA1 (annotated as SmbB) and SmbA2
(annotated as SmbA). Ser/Thr residues that are not dehydrated are underlined. The LanT protease
cleavage sequences are shown in red; for cases in which additional proteolysis takes place (plantaricin W,
cytolysin, and haloduracin), the N terminus of the mature products is denoted with a red arrow.

486 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

residue of both peptides was mutated to Ala ing with the surrounding water molecules,
provides a blueprint to better understand and the hydrophobic side chains project out of
these molecular interactions (19). the water layer, contributing to the hydropho-
Enterococcal cytolysin is unusual in a bicity that characterizes streptomycete aerial
number of ways: Not only does it possess bac- structures and spores (55).
teriocin function, but it is cytolytic against Among the four orthologues of the SapB
erythrocytes and other eukaryotic cells (24). structural gene that have been identified, the
Although its structure has not been rigor- Cys and Ser residues that form both Lan
ously established, sequence comparison with bridges and the Ser residues that remain as
lacticin 3147 and haloduracin (Figure 5) Dha in mature SapB are conserved, suggesting
suggests that both of its peptides contain that these elements are important for activity.
a MeLan at the N terminus and an addi- The hydrophobicity of the two rings is also
tional internal Lan ring. The topology of conserved (128), so although the nature of the
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

the putative C-terminal ring of the CylLS interaction between peptides and the cell wall
peptide remains to be resolved. The CylLL remains obscure, it appears that amphiphilic-
by Pennsylvania State University on 05/22/12. For personal use only.

and CylLS prepropeptides are modified by ity and hydrophobicity are important.
CylM and exported by a dual-function trans-
porter/protease to generate the CylLL  and
CylLS  peptides (24). A second round of pro- Posttranslational Modification
teolysis by an extracellular serine protease, Enzymes and Engineering Studies
CylA, is required for complete peptide matu- The enzyme reaction mechanisms responsi-
ration, removing an identical six-residue se- ble for the dehydration and cyclization reac-
quence from the N terminus of each pep- tions common to all lantibiotics are intriguing
tide to yield CylLL  and CylLS  (24). The because of their biochemistry and potential
β-peptides of the two-component lantibiotics applications to peptide engineering. The de-
plantaricin W and haloduracin appear to also hydration enzymes involved in class I (LanB)
require a second round of proteolytic process- and class II (LanM) lantibiotics demonstrate
ing (39, 73); however, the responsible pro- low substrate specificity. For instance, NisB
teases have not yet been identified. dehydrates nonlantibiotic peptides fused to
the leader peptide of NisA expressed in L.
lactis (53, 91) and LctM can do the same in
Class III Lantibiotics: SapB and SapT vitro with peptides containing both proteino-
Both SapB and SapT are 21 residues genic and nonproteinogenic amino acids (11).
long, hydrophobic, and highly surface ac- LctM dehydration involves initial phospho-
tive (Figure 3). These peptides are capa- rylation of the targeted Ser/Thr, followed by
ble of self-assembly at air-water interfaces; elimination of the phosphate (10). The en-
this is thought to provide a film of surfac- zyme is processive with respect to the dehy-
tant through which nascent aerial filaments dration reaction, completing all four dehy-
emerge (105). Both peptides appear to be drations without releasing the substrate (75).
well suited for residence at an air-water in- Although the leader peptide is required for the
terface, as modeling predicts that the macro- correct action of LanB and LanM proteins, it
cyclic structures would limit internal hydro- can be appended at its N terminus with pep-
gen bonding within polar side chains and force tide tags (101, 130), including the export pep-
the nonpolar side chains to the surface of the tides from the Sec and TAT secretion path-
molecule, thereby generating hydrophilic and ways (59), without affecting posttranslational
hydrophobic patches. These peptides are dis- modification.
tinctly amphiphilic; the polar backbone atoms The LanC cyclization enzymes show
may participate in extensive hydrogen bond- weak but clear sequence homology to the

www.annualreviews.org • Lantibiotic Function and Diversity 487


ANRV322-MI61-23 ARI 6 August 2007 18:7

C-terminal domains of LanM proteins (97), 2003 we refer the reader to several compre-
including one His and two Cys residues that hensive reviews (12, 21, 61). These investiga-
function as zinc ligands in the subtilin cyclase tions show the importance of Glu17 for mer-
SpaC (83). Recent elucidation of the crystal sacidin (104) and the Lan and MeLan rings
structure of NisC shows the zinc on top of in lacticin 481 for antimicrobial activity. In-
an α,α-barrel that resembles the β-subunit of terconversion of Lan and MeLan is tolerated
protein farnesyl transferase (66). In addition by the biosynthetic machinery with relatively
to the three protein ligands, the coordination little influence on the antimicrobial activities
sphere is completed by a water molecule be- of lacticin 481 (11) and mutacin II (14). How-
lieved to be displaced by a Cys in the sub- ever, such substitution results in a large de-
strate (66). The thiolate is then electrostati- crease in activity for nisin (61). Removal of
cally activated by the electron-rich metal site the N-terminal five residues from lacticin 481
toward attack onto the dehydro amino acid. (110) and the three N-terminal lysines from
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

The active site consists of a shallow groove the related compound nukacin ISK-1 (1) re-
that accommodates the formation of rings of sults in a 10-fold and 32-fold reduction of
by Pennsylvania State University on 05/22/12. For personal use only.

different sizes, but how NisC controls the re- bioactivity, respectively, indicating the impor-
gioselectivity of cyclization remains unclear. tance of the positively charged N terminus. In
Biomimetic studies have demonstrated that addition to these investigations, several recent
the enzyme must overcome the higher re- engineering studies have focused on increas-
activity of Dha compared with that of Dhb ing production of wild-type lantibiotics (13,
in this process (134). Like the dehydration 20, 35).
carried out by NisB and LctM, the cycliza-
tion reaction catalyzed by NisC and LctM LANTIBIOTIC GENETICS
has low substrate specificity, tolerating both
The recent increase in the number of cloned
nonlantibiotic peptides (11, 53) and cysteine
lantibiotic biosynthetic genes has revealed
derivatives (11).
several trends, including the presence of reg-
Among other modification enzymes, the
ulatory and immunity genes in addition to
oxidative decarboxylases that incorporate the
biosynthetic operons and the regulated ex-
AviCys and MeAviCys structures at the C ter-
pression of these genes in a growth-phase-
mini of epidermin and mersacidin (EpiD and
dependent fashion. As more gene clusters are
MrsD, respectively) have recently been crys-
sequenced, the potential for identifying new
tallized. They catalyze the oxidation of the C-
lantibiotics through genomic approaches is
terminal Cys to the corresponding thioalde-
becoming realized (29, 64, 73).
hyde followed by decarboxylation (3, 63).
EpiD also has low substrate specificity, as it
can oxidize a variety of synthetic peptides (95). Biosynthetic Gene Clusters
The mechanism by which l-Ser is converted The genes for lantibiotic biosynthesis are gen-
to d-Ala residues in lacticin 3147 was recently erally found in clusters, which can be chro-
reported (23). As predicted (98), this is a two- mosomal, on transposable elements, or on
step process involving dehydration of l-Ser plasmids, raising questions about the impor-
to Dha by peptide-specific LtnM enzymes tance of horizontal gene transfer. In many
followed by stereospecific hydrogenation by cases the lanA structural gene is the first gene
a single dehydrogenase, LtnJ, to yield d-Ala in an operon that includes genes encoding
(23). the modification enzymes (lanB and lanC or
New studies on lantibiotic engineering for lanM ) and exporter (lanT ) (Figure 6). For
mersacidin (26, 104), lacticin 3147 (19, 20, those lantibiotics that undergo other, more
23), nukacin ISK-1 (1, 79), and lacticin 481 unusual posttranslational modifications, the
(11) have been reported; for results prior to genes encoding the required enzymes are

488 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

Nisin
L. lactis
A B T C I P R K F E G

Subtilin
B. subtilis B T C S I F E G R K

Epidermin
S. epidermis A
G E F H T B C D Q P

Pep 5 I A
S. epidermis T P B C

Lacticin 481
L. lactis A
M T F E G
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Cinnamycin
by Pennsylvania State University on 05/22/12. For personal use only.

S. cinnamoneus
3 4 7 A M X T H Y Z 8 9 R K 10 11 12 14

13
Mersacidin R1
B. subtilis K2 R2 F G E A R1 D M T

Lacticin 3147 E F I R A1 A2 M1 T M2 J
L. lactis

SapB ramC ramS ramA ramB ramR


S. coelicolor
SapB

Figure 6
Representative biosynthetic gene clusters of selected lantibiotics. Transcriptional units, where known, are
indicated by red arrows. Dashed arrow represents a putative promoter, the exact location of which has not
yet been determined (37). Note that the gene nomenclature differs for SapB for historical reasons (128).

usually found in this cluster. Recent exam- Regulation of Lantibiotic Synthesis


ples include ltnJ and cinX, which encodes the
In almost all cases, lantibiotic production
hydroxylase that oxidizes the Asp residue of
is coordinately regulated with other cellu-
cinnamycin both in vivo (M.J. Bibb, personal
lar events. In addition, biosynthesis of a
communication) and in vitro (E. Fogle & W.A.
number of lantibiotics is autoregulatory with
van der Donk, unpublished results). How-
transcription of biosynthetic and immunity
ever, this is not always the case; mrsD is con-
operons governed by two-component regu-
vergently transcribed relative to other mer-
latory systems. Many of these autoregulatory
sacidin biosynthetic enzymes. Other genes
mechanisms have been described as quorum-
involved in regulation and/or immunity may
sensing systems (32, 60, 100).
be within the biosynthetic operon or, more
Autoregulation of nisin and subtilin in-
commonly, found in closely linked operons.
volves subinhibitory concentrations of extra-
For instance, the biosynthetic gene cluster re-
cellular lantibiotics, which trigger the activity
sponsible for cinnamycin production includes
of a signal kinase/response regulator signal
17 genes under the control of nine promoters
transduction system. This results in the tran-
(123; S. O’Rourke, personal communication)
scription of biosynthetic and immunity genes,
(Figure 6).

www.annualreviews.org • Lantibiotic Function and Diversity 489


ANRV322-MI61-23 ARI 6 August 2007 18:7

with peptide production beginning during otic production, and sporulation. Subtilin
mid-exponential growth and peaking at the biosynthesis depends on transcription of the
log-phase to stationary-phase transition (60, spaRK operon, which encodes the response
100). The nisin and subtilin autoregula- regulator and signal kinase. This operon is
tory systems have inspired the construction in turn dependent on the alternative sigma
of lantibiotic-specific tools for heterologous factor, σH , which is regulated at both the
gene expression: the so-called NICE (nisin- transcriptional and translational levels. For
controlled gene expression) (61) and SURE instance, sigH transcription is inhibited by
(subtilin-regulated gene expression) systems the transition state regulator AbrB (100)
(5). (Figure 7).
Advances in B. subtilis genetics have re- Although Bacillus sp. HILY-85 also seems
vealed a more complex regulatory network to coordinately regulate mersacidin biosyn-
regulating subtilin biosynthesis. The cell in- thesis with other stationary-phase events,
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

tegrates multiple environmental signals prior it is not produced until stationary phase
to initiating a suite of late-log- and stationary- commences, and it is σH independent (31),
by Pennsylvania State University on 05/22/12. For personal use only.

phase behaviors such as competence, antibi- as recently demonstrated in an engineered

Figure 7
Presubtilin is the product of the spaS gene; it is modified and transported by the membrane-associated
subtilin synthetase complex (52), which consists of SpaB, SpaC, and SpaT. After export, the leader is
cleaved by any one of three exoproteases (subtilisin, WprA, Vpr) (18). Mature subtilin serves as ligand for
the sensor kinase SpaK, which when activated phosphorylates SpaR, a positive regulator of spaS and the
biosynthetic and immunity operons. Stationary-phase control of spaS transcription is regulated indirectly
through σH control of spaRK; other genes in the cluster are dependent on σA for transcription. σH itself
is subject to transcriptional control by the transition state regulator AbrB (100).

490 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

mersacidin expression system (26). Two dif-


ferent response regulators separately govern
self-immunity and mersacidin biosynthesis:
MrsR2 and its cognate sensor kinase, MrsK2,
regulate immunity, and MrsR1 governs
biosynthesis. The kinase that phosphory-
lates MrsR1 has not yet been identified, and
as mersacidin does not appear to be au-
toregulatory, the signal that triggers its pro-
duction remains unknown (31). Mersacidin
is not the only lantibiotic whose synthe-
sis is controlled, at least in part, by a so-
called orphan response regulator; lacticin
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

3147 (72), mutacin II (87), epidermin (85),


and SapB (49, 81, 82) are also subject to such
by Pennsylvania State University on 05/22/12. For personal use only.

regulation.
S. mutans is a principal component of oral
biofilms, where interspecies competition is
presumed to be intense owing to limited re-
sources and high biodiversity. This would
make antibiotic production an adaptive ad-
vantage, and indeed mutacin I production is
stimulated when S. mutans is grown on solid
substrates (88). Its production is regulated in
concert with other cellular activities includ-
ing competence, biofilm formation, and acid
tolerance (74, 121). A complex regulatory net-
work is emerging that involves the autoinduc-
ing signal LuxS, which is needed for efficient
transcription of the mutA structural gene and,
to a lesser extent, of mutR; a transcriptional
repressor, irvA, which itself is negatively reg-
ulated by LuxS (74, 107); and a sensor kinase
(CiaH) (89).
Simultaneous bacteriocin production and
competence development might provide a
mechanism to ensure a pool of homologous
DNA for uptake by selectively targeting non-
competent cells of the same strain (56). Smb
production by S. mutans GS5 is coregulated
with competence, as Smb production depends Figure 8
on the quorum-sensing signal competence- (a) In the absence of a target microbe or erythrocyte, CylR2 binds to the
stimulating peptide (CSP) encoded by the promoter that governs cytolysin biosynthetic genes. However, basal
transcription enables a small amount of mature CylL  and CylS  to
comC gene. A com box-like element is present accumulate. Under these circumstances, CylL  titrates CylS  . (b) In the
upstream of the smbA structural gene (131), presence of a target cell, CylL  preferentially binds the lipid bilayer. CylS 
and an S. mutans GS5 comC mutant that lacks is thus free to (i) interact with the membrane-bound CylR1, which leads to
antibiotic production activity is rescued by derepression of the cytolysin biosynthetic operon, and (ii) bind to CylL
the addition of synthetic CSP to the medium localized to the target cell. Both peptides are needed for cytolytic activity.

www.annualreviews.org • Lantibiotic Function and Diversity 491


ANRV322-MI61-23 ARI 6 August 2007 18:7

(132). An 18-residue CSP was recently puri- brane binding protein of unknown function,
fied from S. mutans GS5; apparently cleavage CylR2, is also involved (24). This economy of
of the three C-terminal amino acids by an un- regulation allows Enterococcus faecalis to use a
known extracellular protease potentiates CSP single peptide to probe the environment for
activity (86). cytolysin targets and induce its production
The biosynthesis of epidermin by S. epi- only when appropriate.
dermidis is, at least in part, also controlled by Extracellular pH regulates the production
global regulators of cellular stress response of lacticin 481 by strains of L. lactis. The lac-
and biofilm production (51). Transcriptional ticin 481 biosynthetic operon (lctAMTFEG)
regulation of epiA is governed by the response is governed by two promoters, P1 and P3,
regulator EpiQ (85); however, peptide matu- which map upstream of lctA. Acidification of
ration is under the control of the global reg- the medium by lactic acid stimulates expres-
ulatory system, agr (accessory gene regula- sion from both promoters, and while the P3
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

tor) (46, 47). When the agr quorum system is promoter appears to be the stronger of the
active, many surface proteins are downregu- two, P1 is more tightly regulated (37). The
by Pennsylvania State University on 05/22/12. For personal use only.

lated while many exoproteins are upregulated P1 promoter features a novel 14-bp regulatory
(118). Epidermin activity is thus controlled region to which the positive regulatory pro-
via the agr-regulated protease EpiP, which tein RcfB binds. RcfB is a member of the Crp-
cleaves the epidermin leader peptide (51). Fnr family of transcriptional activators and is
The cell-density-dependent mechanism cotranscribed with a universal stress-like pro-
that regulates the production of the two- tein and a multidrug transporter. RcfB null
component lantibiotic cytolysin reflects its mutants are significantly diminished in their
role as an Enterococcus faecalis virulence fac- ability to survive acidic conditions, suggesting
tor (Figure 8). In the absence of target cells, that RcfB is involved in regulating multiple
cytolysin production is repressed by CylR1, genes whose expression is required for acid
which dimerizes and binds specifically to an tolerance (67).
inverted repeat that overlaps the –35 re-
gion of the cytolysin operon promoter (93).
However, basal transcription of the biosyn- CONCLUSIONS AND OUTLOOK
thetic operon results in low-level produc- The past decade has seen great leaps in our un-
tion of CylLS  and CylLL  , which form an derstanding of the mode of action and biosyn-
insoluble complex that has neither cytolytic thesis of lantibiotics, and it has revealed that
nor regulatory activity. When target cells are this class of molecules is not limited to antimi-
present, CylLL  preferentially binds to phos- crobial peptides. The list of new lantibiotics
phatidylcholine:cholesterol lipid bilayers and and potential applications continues to expand
can no longer titrate free CylLS  , whose ac- and the immediate future is likely to see new
cumulation results in high-level cytolysin ex- breakthroughs in the discovery, engineering,
pression (17). Although it is not known how and mode of action of lantibiotics (see Future
CylLS  causes derepression, a second, mem- Issues, below).

SUMMARY POINTS
1. Lantibiotics are translated as prepropeptides that are first modified by the dehydration
of Ser and Thr residues in the C-terminal half of the peptides. Lan or MeLan bridges
are then introduced by the intramolecular addition of Cys thiols to the dehydrated
amino acids. The peptides do not gain full biological function until the leader sequence
is cleaved.

492 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

2. A new classification scheme is proposed: Class I peptides are modified by separate


dehydratases (LanB) and cyclases (LanC). Class II lantibiotics are dehydrated and
cyclized by a single LanM-type enzyme. Class III peptides have little or no antibiotic
activity; rather, they provide alternative physiological functions.
3. Most lantibiotics exert their antibiotic effect by either forming pores in the target
cell membrane or inhibiting cell wall synthesis. Importantly, there is little evidence of
stable resistance mutants arising in food products that have been treated with nisin,
which has been used for almost 50 years.
4. In vitro reconstitution of LanM-type enzymes reveals that the Ser and Thr residues
are first phosphorylated prior to elimination, and both in vitro and in vivo studies
show that the biosynthetic enzymes have remarkable substrate promiscuity. The latter
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

findings are promising for lantibiotic engineering.


5. The genes for lantibiotic biosynthesis, regulation, and self-immunity are found in
by Pennsylvania State University on 05/22/12. For personal use only.

clusters. Lantibiotic biosynthesis is frequently coregulated as part of a stress response


when cells enter late-log or stationary phase.

FUTURE ISSUES
1. As new lantibiotic peptides are discovered, it is most likely that new structural elements
and functionalities will be revealed.
2. New clinical applications of a variety of lantibiotics are currently being pursued,
underscoring their potential as important agents in disease control.
3. Protein engineering is readily performed in these ribosomally synthesized peptides,
and thus lantibiotics are especially amenable to rational drug design.
4. Database mining and PCR analysis of genomic DNA could greatly expand the number
and diversity of currently known lantibiotics.
5. The mode of action of lantibiotics such as Pep5, lacticin 481, and epilancin that are
structurally unrelated to nisin, mersacidin, or cinnamycin will be the subject of further
investigation.

DISCLOSURE STATEMENT
The authors are not aware of any biases that might be perceived as affecting the objectivity of
this review.

ACKNOWLEDGMENTS
The authors would like to thank Mr. Alex Smiros for his assistance in preparing the figures.
The authors also acknowledge support by the National Institutes of Health; WAD is supported
by NIH R01 GM58822, and JMW by NIH R15 GM069398.

www.annualreviews.org • Lantibiotic Function and Diversity 493


ANRV322-MI61-23 ARI 6 August 2007 18:7

LITERATURE CITED
1. Asaduzzaman SM, Nagao J, Aso Y, Nakayama J, Sonomoto K. 2006. Lysine-oriented
charges trigger the membrane binding and activity of nukacin ISK-1. Appl. Environ.
Microbiol. 72:6012–17
2. Aso Y, Okuda K, Nagao J, Kanemasa Y, Thi Bich Phuong N, et al. 2005. A novel type of
immunity protein, NukH, for the lantibiotic nukacin ISK-1 produced by Staphylococcus
warneri ISK-1. Biosci. Biotechnol. Biochem. 69:1403–10
3. Blaesse M, Kupke T, Huber R, Steinbacher S. 2000. Crystal structure of the peptidyl-
cysteine decarboxylase EpiD complexed with a pentapeptide substrate. EMBO J. 19:6299–
310
4. Bonelli RR, Schneider T, Sahl HG, Wiedemann I. 2006. Insights into in vivo activities of
lantibiotics from gallidermin and epidermin mode-of-action studies. Antimicrob. Agents
Chemother. 50:1449–57
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

5. Bongers RS, Veening JW, Van Wieringen M, Kuipers OP, Kleerebezem M. 2005. Devel-
opment and characterization of a subtilin-regulated expression system in Bacillus subtilis:
by Pennsylvania State University on 05/22/12. For personal use only.

strict control of gene expression by addition of subtilin. Appl. Environ. Microbiol. 71:8818–
24
6. Breukink E, Wiedemann I, van Kraaij C, Kuipers OP, Sahl H, de Kruijff B. 1999. Use of
the cell wall precursor lipid II by a pore-forming peptide antibiotic. Science 286:2361–64
7. Brötz H, Bierbaum G, Reynolds PE, Sahl HG. 1997. The lantibiotic mersacidin inhibits
peptidoglycan biosynthesis at the level of transglycosylation. Eur. J. Biochem. 246:193–99
8. Brötz H, Josten M, Wiedemann I, Schneider U, Götz F, et al. 1998. Role of lipid-
bound peptidoglycan precursors in the formation of pores by nisin, epidermin and other
lantibiotics. Mol. Microbiol. 30:317–27
9. Burton JP, Chilcott CN, Moore CJ, Speiser G, Tagg JR. 2006. A preliminary study of
the effect of probiotic Streptococcus salivarius K12 on oral malodour parameters. J. Appl.
Microbiol. 100:754–64
10. Chatterjee C, Miller LM, Leung YL, Xie L, Yi M, et al. 2005. Lacticin 481 synthetase
phosphorylates its substrate during lantibiotic production. J. Am. Chem. Soc. 127:15332–
33
11. Chatterjee C, Patton GC, Cooper L, Paul M, van der Donk WA. 2006. Engineering
dehydro amino acids and thioethers into peptides using lacticin 481 synthetase. Chem.
Biol. 13:1109–17
12. Chatterjee C, Paul M, Xie L, van der Donk WA. 2005. Biosynthesis and mode of action
of lantibiotics. Chem. Rev. 105:633–84
13. Cheigh CI, Park H, Choi HJ, Pyun YR. 2005. Enhanced nisin production by increasing
genes involved in nisin Z biosynthesis in Lactococcus lactis subsp. lactis A164. Biotechnol.
Lett. 27:155–60
14. Chen P, Novak J, Kirk M, Barnes S, Qi F, Caufield PW. 1998. Structure-activity study
of the lantibiotic mutacin II from Streptococcus mutans T8 by a gene replacement strategy.
Appl. Environ. Microbiol. 64:2335–40
15. Chen P, Qi FX, Novak J, Krull RE, Caufield PW. 2001. Effect of amino acid substitutions
in conserved residues in the leader peptide on biosynthesis of the lantibiotic mutacin II.
FEMS Microbiol. Lett. 195:139–44
16. Cloutier MM, Guernsey L, Mattes P, Koeppen B. 1990. Duramycin enhances chloride
secretion in airway epithelium. Am. J. Physiol. 259:C450–54
17. Coburn PS, Pillar CM, Jett BD, Haas W, Gilmore MS. 2004. Enterococcus faecalis senses
target cells and in response expresses cytolysin. Science 306:2270–72

494 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

18. Corvey C, Stein T, Dusterhus S, Karas M, Entian KD. 2003. Activation of subtilin
precursors by Bacillus subtilis extracellular serine proteases subtilisin (AprE), WprA, and
Vpr. Biochem. Biophys. Res. Commun. 304:48–54
19. Cotter PD, Deegan LH, Lawton EM, Draper LA, O’Connor PM, et al. 2006. Com-
19. Complete
plete alanine scanning of the two-component lantibiotic lacticin 3147: generating scanning
a blueprint for rational drug design. Mol. Microbiol. 62:735–47 mutagenesis was
20. Cotter PD, Draper LA, Lawton EM, McAuliffe O, Hill C, Ross RP. 2006. Overpro- performed on each
duction of wild-type and bioengineered derivatives of the lantibiotic lacticin 3147. Appl. of the 59 amino
Environ. Microbiol. 72:4492–96 acids in lacticin
3147, and a core set
21. Cotter PD, Hill C, Ross RP. 2005. Bacterial lantibiotics: strategies to improve therapeutic
of invariable
potential. Curr. Protein Pept. Sci. 6:61–75 residues were
22. Cotter PD, Hill C, Ross RP. 2005. Bacteriocins: developing innate immunity for food. identified.
Nat. Rev. Microbiol. 3:777–88
23. Cotter PD, O’Connor PM, Draper LA, Lawton EM, Deegan LH, et al. 2005. Post-
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

23. Confirms that


translational conversion of L-serines to D-alanines is vital for optimal production conversion of Ser
and activity of the lantibiotic lacticin 3147. Proc. Natl. Acad. Sci. USA 102:18584–89
by Pennsylvania State University on 05/22/12. For personal use only.

to D-Ala is a
24. Cox CR, Coburn PS, Gilmore MS. 2005. Enterococcal cytolysin: a novel two component two-step process
peptide system that serves as a bacterial defense against eukaryotic and prokaryotic cells. involving
Curr. Protein Pept. Sci. 6:77–84 dehydration of Ser
to Dha followed by
25. Daniel RA, Errington J. 2003. Control of cell morphogenesis in bacteria: two distinct
isomerization to
ways to make a rod-shaped cell. Cell 113:767–76 D-Ala.
26. Dawson MJ, Cortes Bargallo J, Rudd BAM, Boakes S, Bierbaum G, et al. 2005. U.S.
Patent No. 5716931
27. Delves-Broughton J, Blackburn P, Evans RJ, Hugenholtz J. 1996. Applications of the
bacteriocin, nisin. Antonie van Leeuwenhoek 69:193–202
28. Dougherty BA, Hill C, Weidman JF, Richardson DR, Venter JC, Ross RP. 1998. Sequence
and analysis of the 60 kb conjugative, bacteriocin-producing plasmid pMRC01 from
Lactococcus lactis DPC3147. Mol. Microbiol. 29:1029–38
29. Dufour A, Hindré T, Haras D, Le Pennec JP. 2007. The biology of the lantibiotics of
the lacticin 481 subgroup is coming of age. FEMS Microbiol. Rev. 31:134–67
30. Ekkelenkamp MB, Hanssen M, Danny Hsu ST, de Jong A, Milatovic D, et al. 2005.
Isolation and structural characterization of epilancin 15X, a novel lantibiotic from a
clinical strain of Staphylococcus epidermidis. FEBS Lett. 579:1917–22
31. Guder A, Schmitter T, Wiedemann I, Sahl HG, Bierbaum G. 2002. Role of the single reg-
ulator MrsR1 and the two-component system MrsR2/K2 in the regulation of mersacidin
production and immunity. Appl. Environ. Microbiol. 68:106–13
32. Haas W, Shepard BD, Gilmore MS. 2002. Two-component regulator of Enterococcus
faecalis cytolysin responds to quorum-sensing autoinduction. Nature 415:84–87
33. Hasper HE, de Kruijff B, Breukink E. 2004. Assembly and stability of nisin-lipid II pores.
Biochemistry 43:11567–75
34. Hasper HE, Kramer NE, Smith JL, Hillman JD, Zachariah C, et al. 2006. An
34. Shows that
alternative bacteriocidal mechanism of action for lantibiotic peptides that target nisin and mutacin
lipid II. Science 313:1636–37 1140 inhibit cell
35. Heinzmann S, Entian KD, Stein T. 2006. Engineering Bacillus subtilis ATCC 6633 for wall biosynthesis
improved production of the lantibiotic subtilin. Appl. Microbiol. Biotechnol. 69:532–36 by sequestering
36. Hillman JD. 2002. Genetically modified Streptococcus mutans for the prevention of dental lipid II to
nonphysiological
caries. Antonie Van Leeuwenhoek 82:361–66
patches along the
37. Hindré T, Le Pennec JP, Haras D, Dufour A. 2004. Regulation of lantibiotic lacticin 481 nascent cell wall.
production at the transcriptional level by acid pH. FEMS Microbiol. Lett. 231:291–98

www.annualreviews.org • Lantibiotic Function and Diversity 495


ANRV322-MI61-23 ARI 6 August 2007 18:7

38. Hoffmann A, Schneider T, Pag U, Sahl HG. 2004. Localization and functional analysis
of PepI, the immunity peptide of Pep5-producing Staphylococcus epidermidis strain 5. Appl.
Environ. Microbiol. 70:3263–71
39. Holo H, Jeknic Z, Daeschel M, Stevanovic S, Nes IF. 2001. Plantaricin W from Lactobacil-
lus plantarum belongs to a new family of two-peptide lantibiotics. Microbiology 147:643–51
40. Hosoda K, Ohya M, Kohno T, Maeda T, Endo S, Wakamatsu K. 1996. Structure de-
termination of an immunopotentiator peptide, cinnamycin, complexed with lysophos-
phatidylethanolamine by 1H-NMR. J. Biochem. 119:226–30
41. Hsu ST. 2006. Structural motifs of lipid II-binding lantibiotics as a blueprint for novel
antibiotics. Anti-Infect. Agents Med. Chem. 5:245–54
42. Hsu ST, Breukink E, Bierbaum G, Sahl HG, de Kruijff B, et al. 2003. NMR study of
mersacidin and lipid II interaction in dodecylphosphocholine micelles. Conformational
changes are a key to antimicrobial activity. J. Biol. Chem. 278:13110–17
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

43. Hsu ST, Breukink E, de Kruijff B, Kaptein R, Bonvin AM, van Nuland NA. 2002. Map-
ping the targeted membrane pore formation mechanism by solution NMR: the nisin Z
by Pennsylvania State University on 05/22/12. For personal use only.

and lipid II interaction in SDS micelles. Biochemistry 41:7670–76


44. Hsu ST, Breukink E, Tischenko E, Lutters MA, De Kruijff B, et al. 2004. The nisin-lipid
II complex reveals a pyrophosphate cage that provides a blueprint for novel antibiotics.
Nat. Struct. Mol. Biol. 11:963–67
45. Hyink O, Balakrishnan M, Tagg JR. 2005. Streptococcus rattus strain BHT produces both
a class I two-component lantibiotic and a class II bacteriocin. FEMS Microbiol. Lett.
252:235–41
46. Ji G, Beavis R, Novick RP. 1997. Bacterial interference caused by autoinducing peptide
variants. Science 276:2027–30
47. Ji G, Beavis RC, Novick RP. 1995. Cell density control of staphylococcal virulence me-
diated by an octapeptide pheromone. Proc. Natl. Acad. Sci. USA 92:12055–59
48. Kaletta C, Entian KD, Jung G. 1991. Prepeptide sequence of cinnamycin (Ro 09-0198):
the first structural gene of a duramycin-type lantibiotic. Eur. J. Biochem. 199:411–15
49. Keijser BJ, van Wezel GP, Canters GW, Vijgenboom E. 2002. Developmental regulation
of the Streptomyces lividans ram genes: involvement of RamR in regulation of the ramCSAB
operon. J. Bacteriol. 184:4420–29
50. Kellner R, Jung G, Josten M, Kaletta C, Entian KD, Sahl HG. 1989. Pep5: structure
elucidation of a large lantibiotic. Angew. Chem. 101:618–21
51. Kies S, Vuong C, Hille M, Peschel A, Meyer C, et al. 2003. Control of antimicrobial
peptide synthesis by the agr quorum sensing system in Staphylococcus epidermidis: Activity
of the lantibiotic epidermin is regulated at the level of precursor peptide processing.
Peptides 24:329–38
52. Kiesau P, Eikmanns U, Gutowski-Eckel Z, Weber S, Hammelmann M, Entian KD. 1997.
Evidence for a multimeric subtilin synthetase complex. J. Bacteriol. 179:1475–81
53. Kluskens LD, Kuipers A, Rink R, de Boef E, Fekken S, et al. 2005. Post-translational
modification of therapeutic peptides by NisB, the dehydratase of the lantibiotic nisin.
55. Reports
Biochemistry 44:12827–34
structure and
activity of SapT, 54. Kodani S, Hudson ME, Durrant MC, Buttner MJ, Nodwell JR, Willey JM. 2004. The
which is capable of SapB morphogen is a lantibiotic-like peptide derived from the product of the develop-
restoring mental gene ramS in Streptomyces coelicolor. Proc. Natl. Acad. Sci. USA 101:11448–53
morphogenesis to a 55. Kodani S, Lodato MA, Durrant MC, Picart F, Willey JM. 2005. SapT, a lanthionine-
SapB deficient S.
containing peptide involved in aerial hyphae formation in the streptomycetes. Mol.
coelicolor mutant.
Microbiol. 58:1368–80

496 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

56. Kreth J, Merritt J, Shi W, Qi F. 2005. Co-ordinated bacteriocin production and compe-
tence development: a possible mechanism for taking up DNA from neighbouring species.
Mol. Microbiol. 57:392–404
57. Kruszewska D, Sahl HG, Bierbaum G, Pag U, Hynes SO, Ljungh A. 2004. Mersacidin
eradicates methicillin-resistant Staphylococcus aureus (MRSA) in a mouse rhinitis model.
J. Antimicrob. Chemother. 54:648–53
58. Kuipers A, De Boef E, Rink R, Fekken S, Kluskens LD, et al. 2004. NisT, the transporter
of the lantibiotic nisin, can transport fully modified, dehydrated and unmodified prenisin
and fusions of the leader peptide with non-lantibiotic peptides. J. Biol. Chem. 279:22176–
82
59. Kuipers A, Wierenga J, Rink R, Kluskens LD, Driessen AJM, et al. 2006. Sec-
59. Reports the
mediated transport of post-translationally dehydrated peptides in Lactococcus lactis. modification and
Appl. Environ. Microbiol. 72:7626–33 export of
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

60. Kuipers OP, Beerthuyzen MM, de Ruyter PG, Luesink EJ, de Vos WM. 1995. Autoreg- prelantibiotics
engineered with
ulation of nisin biosynthesis in Lactococcus lactis by signal transduction. J. Biol. Chem.
Sec or Tat signal
by Pennsylvania State University on 05/22/12. For personal use only.

270:27299–304 peptides.
61. Kuipers OP, Bierbaum G, Ottenwälder B, Dodd HM, Horn N, et al. 1996. Protein
engineering of lantibiotics. Antonie Van Leeuwenhoek 69:161–69
62. Kuipers OP, Rollema HS, de Vos WM, Siezen RJ. 1993. Biosynthesis and secretion of a
precursor of nisin Z by Lactococcus lactis, directed by the leader peptide of the homologous
lantibiotic subtilin from Bacillus subtilis. FEBS Lett. 330:23–27
63. Kupke T, Götz F. 1997. The enethiolate anion reaction products of EpiD. pKa value of
the enethiol side chain is lower than that of the thiol side chain of peptides. J. Biol. Chem.
272:4759–62
64. Lawton EM, Cotter PD, Hill C, Ross RP. 2007. Identification of a novel two-peptide
lantibiotic, haloduracin, produced by the alkaliphile Bacillus halodurans C-125. FEMS
Microbiol. Lett. 267:64–71
65. Lazzarini A, Gastaldo L, Candiani G, Ciciliato I, Losi D, et al. 2005. International Patent
Publication No. WO 2005/014628 A1
66. Li B, Yu JPJ, Brunzelle JS, Moll GN, van der Donk WA, Nair SK. 2006. Structure 66. Demonstrates
and mechanism of the lantibiotic cyclase involved in nisin biosynthesis. Science that NisC has
311:1464–67 broad substrate
67. Madsen SM, Hindré T, Le Pennec JP, Israelsen H, Dufour A. 2005. Two acid-inducible specificity and is
structurally similar
promoters from Lactococcus lactis require the cis-acting ACiD-box and the transcription
in fold and
regulator RcfB. Mol. Microbiol. 56:735–46 substrate activation
68. Makino A, Baba T, Fujimoto K, Iwamoto K, Yano Y, et al. 2003. Cinnamycin (Ro 09- to mammalian
0198) promotes cell binding and toxicity by inducing transbilayer lipid movement. J. Biol. farnesyl
Chem. 278:3204–9 transferases.
69. Märki F, Hanni E, Fredenhagen A, van Oostrum J. 1991. Mode of action of the
lanthionine-containing peptide antibiotics duramycin, duramycin B and C, and cin-
namycin as indirect inhibitors of phospholipase A2. Biochem. Pharmacol. 42:2027–35
70. Martin NI, Sprules T, Carpenter MR, Cotter PD, Hill C, et al. 2004. Structural charac-
terization of lacticin 3147, a two-peptide lantibiotic with synergistic activity. Biochemistry
43:3049–56
71. McAuliffe O, Hill C, Ross RP. 2000. Each peptide of the two-component lantibiotic
lacticin 3147 requires a separate modification enzyme for activity. Microbiology 146:2147–
54

www.annualreviews.org • Lantibiotic Function and Diversity 497


ANRV322-MI61-23 ARI 6 August 2007 18:7

72. McAuliffe O, O’Keeffe T, Hill C, Ross RP. 2001. Regulation of immunity to the two-
component lantibiotic, lacticin 3147, by the transcriptional repressor LtnR. Mol. Microbiol.
39:982–93
73. McClerren AL, Cooper LE, Quan C, Thomas PM, Kelleher NL, van der Donk WA.
73. A genetic
approach resulted 2006. Discovery and in vitro biosynthesis of haloduracin, a new two-component
in the identification lantibiotic. Proc. Natl. Acad. Sci. USA 103:17243–48
of the 74. Merritt J, Kreth J, Shi W, Qi F. 2005. LuxS controls bacteriocin production in Streptococcus
two-component mutans through a novel regulatory component. Mol. Microbiol. 57:960–69
lantibiotic 75. Miller LM, Chatterjee C, van der Donk WA, Kelleher NL. 2006. The dehydration activity
haloduracin, the
of lacticin 481 synthetase is highly processive. J. Am. Chem. Soc. 128:1420–21
first to be produced
by a Bacillus sp. 76. Minami Y, Yoshida K, Azuma R, Urakawa A, Kawauchi T, et al. 1994. Structure of
cypemycin, a new peptide antibiotic. Tetrahedron Lett. 35:8001–4
77. Molina y Vedia LM, Stutts MJ, Boucher JRC, Henke DC. 1995. U.S. Patent No. 5716931
78. Morgan SM, O’Connor PM, Cotter PD, Ross RP, Hill C. 2005. Sequential actions of the
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

two component peptides of the lantibiotic lacticin 3147 explain its antimicrobial activity
at nanomolar concentrations. Antimicrob. Agents Chemother. 49:2606–11
by Pennsylvania State University on 05/22/12. For personal use only.

79. Nagao J, Harada Y, Shioya K, Aso Y, Zendo T, et al. 2005. Lanthionine introduction
into nukacin ISK-1 prepeptide by co-expression with modification enzyme NukM in
Escherichia coli. Biochem. Biophys. Res. Commun. 336:507–13
80. Navaratna MA, Sahl HG, Tagg JR. 1999. Identification of genes encoding two-
component lantibiotic production in Staphylococcus aureus C55 and other phage group
II S. aureus strains and demonstration of an association with the exfoliative toxin B gene.
Infect. Immun. 67:4268–71
81. Nguyen KT, Willey JM, Nguyen LD, Nguyen LT, Viollier PH, Thompson CJ. 2002. A
central regulator of morphological differentiation in the multicellular bacterium Strepto-
myces coelicolor. Mol. Microbiol. 46:1223–38
82. O’Connor TJ, Nodwell JR. 2005. Pivotal roles for the receiver domain in the mechanism
of action of the response regulator RamR of Streptomyces coelicolor. J. Mol. Biol. 351:1030–
47
83. Okeley NM, Paul M, Stasser JP, Blackburn N, van der Donk WA. 2003. SpaC and NisC,
the cyclases involved in subtilin and nisin biosynthesis, are zinc proteins. Biochemistry
42:13613–24
84. Pag U, Sahl HG. 2002. Multiple activities in lantibiotics—models for the design of novel
antibiotics? Curr. Pharm. Des. 8:815–33
85. Peschel A, Augustin J, Kupke T, Stevanovic S, Götz F. 1993. Regulation of epidermin
biosynthetic genes by EpiQ. Mol. Microbiol. 9:31–39
86. Petersen FC, Fimland G, Scheie AA. 2006. Purification and functional studies
86. The
two-component of a potent modified quorum-sensing peptide and a two-peptide bacteriocin in
lantibiotic Smb is Streptococcus mutans. Mol. Microbiol. 61:1322–34
shown to be 87. Qi F, Chen P, Caufield PW. 1999. Functional analyses of the promoters in the lantibiotic
regulated by CSP, mutacin II biosynthetic locus in Streptococcus mutans. Appl. Environ. Microbiol. 65:652–58
which is most 88. Qi F, Chen P, Caufield PW. 2000. Purification and biochemical characterization of
potent after
mutacin I from the group I strain of Streptococcus mutans CH43, and genetic analysis of
proteolysis.
mutacin I biosynthesis genes. Appl. Environ. Microbiol. 66:3221–29
89. Qi F, Merritt J, Lux R, Shi W. 2004. Inactivation of the ciaH gene in Streptococcus mutans
diminishes mutacin production and competence development, alters sucrose-dependent
biofilm formation, and reduces stress tolerance. Infect. Immun. 72:4895–99
90. Qiao M, Saris PE. 1996. Evidence for a role of NisT in transport of the lantibiotic nisin
produced by Lactococcus lactis N8. FEMS Microbiol. Lett. 144:89–93

498 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

91. Rink R, Kuipers A, de Boef E, Leenhouts KJ, Driessen AJ, et al. 2005. Lantibiotic struc-
tures as guidelines for the design of peptides that can be modified by lantibiotic enzymes.
Biochemistry 44:8873–82
92. Rogers LA. 1928. The inhibiting effect of Streptococcus lactis on Lactobacillus bulgaricus. J.
Bacteriol. 16:321–25
93. Rumpel S, Razeto A, Pillar CM, Vijayan V, Taylor A, et al. 2004. Structure and DNA-
binding properties of the cytolysin regulator CylR2 from Enterococcus faecalis. EMBO J.
23:3632–42
94. Sahl HG, Bierbaum G. 1998. Lantibiotics: biosynthesis and biological activities of
uniquely modified peptides from gram-positive bacteria. Annu. Rev. Microbiol. 52:41–79
95. Schmid DG, Majer F, Kupke T, Jung G. 2002. Electrospray ionization Fourier trans-
form ion cyclotron resonance mass spectrometry to reveal the substrate specificity of the
peptidyl-cysteine decarboxylase EpiD. Rapid Commun. Mass Spectrom. 16:1779–84
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

96. Schnell N, Entian KD, Schneider U, Götz F, Zahner H, et al. 1988. Prepeptide se-
quence of epidermin, a ribosomally synthesized antibiotic with four sulphide-rings. Na-
by Pennsylvania State University on 05/22/12. For personal use only.

ture 333:276–78
97. Siezen RJ, Kuipers OP, de Vos WM. 1996. Comparison of lantibiotic gene clusters and
encoded proteins. Antonie van Leeuwenhoek 69:171–84
98. Skaugen M, Nissenmeyer J, Jung G, Stevanovic S, Sletten K, et al. 1994. In vivo con-
version of L-serine to D-alanine in a ribosomally synthesized polypeptide. J. Biol. Chem.
269:27183–85
99. Stein T, Borchert S, Conrad B, Feesche J, Hofemeister B, et al. 2002. Two different
lantibiotic-like peptides originate from the ericin gene cluster of Bacillus subtilis A1/3. J.
Bacteriol. 184:1703–11
100. Stein T, Borchert S, Kiesau P, Heinzmann S, Kloss S, et al. 2002. Dual control of subtilin
biosynthesis and immunity in Bacillus subtilis. Mol. Microbiol. 44:403–16
101. Stein T, Entian KD. 2002. Maturation of the lantibiotic subtilin: matrix-assisted laser
desorption/ionization time-of-flight mass spectrometry to monitor precursors and their
proteolytic processing in crude bacterial cultures. Rapid Commun. Mass Spectrom. 16:103–
10
102. Stein T, Heinzmann S, Dusterhus S, Borchert S, Entian KD. 2005. Expression and
functional analysis of the subtilin immunity genes spaIFEG in the subtilin-sensitive host
Bacillus subtilis MO1099. J. Bacteriol. 187:822–28
103. Stein T, Heinzmann S, Solovieva I, Entian KD. 2003. Function of Lactococcus lactis nisin
immunity genes nisI and nisFEG after coordinated expression in the surrogate host Bacillus
subtilis. J. Biol. Chem. 278:89–94
104. Szekat C, Jack RW, Skutlarek D, Farber H, Bierbaum G. 2003. Construction of an ex-
pression system for site-directed mutagenesis of the lantibiotic mersacidin. Appl. Environ.
Microbiol. 69:3777–83
105. Tillotson RD, Wosten HA, Richter M, Willey JM. 1998. A surface active protein involved
in aerial hyphae formation in the filamentous fungus Schizophillum commune restores the
capacity of a bald mutant of the filamentous bacterium Streptomyces coelicolor to erect aerial
structures. Mol. Microbiol. 30:595–602
106. Tiyanont K, Doan T, Lazarus MB, Fang X, Rudner DZ, Walker S. 2006. Imaging pepti-
doglycan biosynthesis in Bacillus subtilis with fluorescent antibiotics. Proc. Natl. Acad. Sci.
USA 103:11033–38
107. Tsang P, Merritt J, Shi W, Qi F. 2006. IrvA-dependent and IrvA-independent pathways
for mutacin gene regulation in Streptococcus mutans. FEMS Microbiol. Lett. 261:231–34

www.annualreviews.org • Lantibiotic Function and Diversity 499


ANRV322-MI61-23 ARI 6 August 2007 18:7

108. Turner DL, Brennan L, Meyer HE, Lohaus C, Siethoff C, et al. 1999. Solution structure
of plantaricin C, a novel lantibiotic. Eur. J. Biochem. 264:833–39
109. Ueda K, Oinuma K, Ikeda G, Hosono K, Ohnishi Y, et al. 2002. AmfS, an extracellular
peptidic morphogen in Streptomyces griseus. J. Bacteriol. 184:1488–92
110. Uguen P, Hindré T, Didelot S, Marty C, Haras D, et al. 2005. Maturation by LctT is
required for biosynthesis of full-length lantibiotic lacticin 481. Appl. Environ. Microbiol.
71:562–65
111. van de Kamp M, Horstink LM, Van den Hooven HW, Konings RNH, Hilbers CW,
et al. 1995. Sequence analysis by NMR spectroscopy of the peptide lantibiotic epilancin
K7 from Staphylococcus epidermidis K7. Eur. J. Biochem. 227:757–71
112. van den Hooven HW, Doeland CC, Van de Kamp M, Konings RN, Hilbers CW, Van de
Ven FJM. 1996. Three-dimensional structure of the lantibiotic nisin in the presence of
membrane-mimetic micelles of dodecylphosphocholine and of sodium dodecylsulphate.
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Eur. J. Biochem. 235:382–93


113. van der Donk WA. 2006. Lighting up the nascent cell wall. ACS Chem. Biol. 1:425–28
114. van der Meer JR, Polman J, Beerthuyzen MM, Siezen RJ, Kuipers OP, de Vos WM. 1993.
by Pennsylvania State University on 05/22/12. For personal use only.

Characterization of the Lactococcus lactis nisin A operon genes nisP, encoding a subtilisin-
like serine protease involved in precursor processing, and nisR, encoding a regulatory
protein involved in nisin biosynthesis. J. Bacteriol. 175:2578–88
115. van der Meer JR, Rollema HS, Siezen RJ, Beerthuyzen MM, Kuipers OP, de Vos WM.
1994. Influence of amino acid substitutions in the nisin leader peptide on biosynthesis
and secretion of nisin by Lactococcus lactis. J. Biol. Chem. 269:3555–62
116. van Heusden HE, de Kruijff B, Breukink E. 2002. Lipid II induces a transmembrane
orientation of the pore-forming peptide lantibiotic nisin. Biochemistry 41:12171–78
117. van Kraaij C, de Vos WM, Siezen RJ, Kuipers OP. 1999. Lantibiotics: biosynthesis, mode
of action and applications. Nat. Prod. Rep. 16:575–87
118. Vuong C, Gotz F, Otto M. 2000. Construction and characterization of an agr deletion
mutant of Staphylococcus epidermidis. Infect. Immun. 68:1048–53
119. Wakamatsu K, Choung SY, Kobayashi T, Inoue K, Higashijima T, Miyazawa T. 1990.
Complex formation of peptide antibiotic Ro09-0198 with lysophosphatidylethanolamine:
1H NMR analyses in dimethyl sulfoxide solution. Biochemistry 29:113–18
120. Wang BY, Kuramitsu HK. 2005. Interactions between oral bacteria: inhibition of Strep-
tococcus mutans bacteriocin production by Streptococcus gordonii. Appl. Environ. Microbiol.
71:354–62
121. Wen ZT, Burne RA. 2004. LuxS-mediated signaling in Streptococcus mutans is involved
in regulation of acid and oxidative stress tolerance and biofilm formation. J. Bacteriol.
186:2682–91
122. Wescombe PA, Upton M, Dierksen KP, Ragland NL, Sivabalan S, et al. 2006. Production
of the lantibiotic salivaricin A and its variants by oral streptococci and use of a specific
induction assay to detect their presence in human saliva. Appl. Environ. Microbiol. 72:1459–
66
125. Demonstrates
that pore-forming 123. Widdick DA, Dodd HM, Barraille P, White J, Stein TH, et al. 2003. Cloning and engi-
activity of neering of the cinnamycin biosynthetic gene cluster from Streptomyces cinnamoneus cin-
plantaricin C is namoneus DSM 40005. Proc. Natl. Acad. Sci. USA 100:4316–21
strain specific and 124. Wiedemann I, Benz R, Sahl HG. 2004. Lipid II-mediated pore formation by the peptide
requires lipid II, antibiotic nisin: a black lipid membrane study. J. Bacteriol. 186:3259–61
but that it also
125. Wiedemann I, Bottiger T, Bonelli RR, Schneider T, Sahl HG, Martinez B. 2006.
blocks lipid II
biosynthesis. Lipid II-based antimicrobial activity of the lantibiotic plantaricin C. Appl. Environ.
Microbiol. 72:2809–14

500 Willey · van der Donk


ANRV322-MI61-23 ARI 6 August 2007 18:7

126. Wiedemann I, Bottiger T, Bonelli RR, Wiese A, Hagge SO, et al. 2006. The mode 126. Evidence
of action of the lantibiotic lacticin 3147—a complex mechanism involving specific suggests that
interaction of two peptides and the cell wall precursor lipid II. Mol. Microbiol. LtnA1 binds to
61:285–96 lipid II and then
recruits LtnA2,
127. Wiedemann I, Breukink E, van Kraaij C, Kuipers OP, Bierbaum G, et al. 2001. Specific
resulting in pore
binding of nisin to the peptidoglycan precursor lipid II combines pore formation and formation and
inhibition of cell wall biosynthesis for potent antibiotic activity. J. Biol. Chem. 276:1772– inhibition of cell
79 wall biosynthesis.
128. Willey JM, Willems A, Kodani S, Nodwell JR. 2006. Morphogenetic surfactants and their
role in the formation of aerial hyphae in Streptomyces coelicolor. Mol. Microbiol. 59:731–42
129. Wirawan RE, Klesse NA, Jack RW, Tagg JR. 2006. Molecular and genetic characteri-
zation of a novel nisin variant produced by Streptococcus uberis. Appl. Environ. Microbiol.
72:1148–56
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

130. Xie L, Miller LM, Chatterjee C, Averin O, Kelleher NL, van der Donk WA. 2004.
Lacticin 481: in vitro reconstitution of lantibiotic synthetase activity. Science 303:679–81
by Pennsylvania State University on 05/22/12. For personal use only.

131. Yonezawa H, Kuramitsu HK. 2005. Genetic analysis of a unique bacteriocin, Smb, pro-
duced by Streptococcus mutans GS5. Antimicrob. Agents Chemother. 49:541–48
132. Yoshida A, Kuramitsu HK. 2002. Multiple Streptococcus mutans genes are involved in
biofilm formation. Appl. Environ. Microbiol. 68:6283–91
133. Yuan J, Zhang ZZ, Chen XZ, Yang W, Huan LD. 2004. Site-directed mutagenesis of
the hinge region of nisin Z and properties of nisin Z mutants. Appl. Microbiol. Biotechnol.
64:806–15
134. Zhu Y, Gieselman M, Zhou H, Averin O, van der Donk WA. 2003. Biomimetic studies on
the mechanism of stereoselective lanthionine formation. Org. Biomol. Chem. 1:3304–15
135. Zimmermann N, Freund S, Fredenhagen A, Jung G. 1993. Solution structures of the
lantibiotics duramycin B and C. Eur. J. Biochem. 216:419–28

www.annualreviews.org • Lantibiotic Function and Diversity 501


AR322-FM ARI 9 July 2007 9:23

Annual Review of
Microbiology

Volume 61, 2007 Contents

Frontispiece
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Margarita Salas p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p xiv


by Pennsylvania State University on 05/22/12. For personal use only.

40 Years with Bacteriophage Ø29


Margarita Salas p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
The Last Word: Books as a Statistical Metaphor for Microbial Communities
Patrick D. Schloss and Jo Handelsman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 23
The Mechanism of Isoniazid Killing: Clarity Through the Scope
of Genetics
Catherine Vilchèze and William R. Jacobs, Jr. p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 35
Development of a Combined Biological and Chemical Process for
Production of Industrial Aromatics from Renewable Resources
F. Sima Sariaslani p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 51
The RNA Degradosome of Escherichia coli: An mRNA-Degrading
Machine Assembled on RNase E
Agamemnon J. Carpousis p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 71
Protein Secretion in Gram-Negative Bacteria via the Autotransporter
Pathway
Nathalie Dautin and Harris D. Bernstein p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 89
Chlorophyll Biosynthesis in Bacteria: The Origins of Structural and
Functional Diversity
Aline Gomez Maqueo Chew and Donald A. Bryant p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p113
Roles of Cyclic Diguanylate in the Regulation of Bacterial Pathogenesis
Rita Tamayo, Jason T. Pratt, and Andrew Camilli p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p131
Aggresomes and Pericentriolar Sites of Virus Assembly:
Cellular Defense or Viral Design?
Thomas Wileman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p149
As the Worm Turns: The Earthworm Gut as a Transient Habitat for
Soil Microbial Biomes
Harold L. Drake and Marcus A. Horn p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p169

vi
AR322-FM ARI 9 July 2007 9:23

Biogenesis of the Gram-Negative Bacterial Outer Membrane


Martine P. Bos, Viviane Robert, and Jan Tommassen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p191
SigB-Dependent General Stress Response in Bacillus subtilis and
Related Gram-Positive Bacteria
Michael Hecker, Jan Pané-Farré, and Uwe Völker p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p215
Ecology and Biotechnology of the Genus Shewanella
Heidi H. Hau and Jeffrey A. Gralnick p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p237
Nonhomologous End-Joining in Bacteria: A Microbial Perspective
Robert S. Pitcher, Nigel C. Brissett, and Aidan J. Doherty p p p p p p p p p p p p p p p p p p p p p p p p p p p p259
Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Postgenomic Adventures with Rhodobacter sphaeroides


Chris Mackenzie, Jesus M. Eraso, Madhusudan Choudhary, Jung Hyeob Roh,
by Pennsylvania State University on 05/22/12. For personal use only.

Xiaohua Zeng, Patrice Bruscella, Ágnes Puskás, and Samuel Kaplan p p p p p p p p p p p p p p p p p283
Toward a Hyperstructure Taxonomy
Vic Norris, Tanneke den Blaauwen, Roy H. Doi, Rasika M. Harshey,
Laurent Janniere, Alfonso Jiménez-Sánchez, Ding Jun Jin,
Petra Anne Levin, Eugenia Mileykovskaya, Abraham Minsky,
Gradimir Misevic, Camille Ripoll, Milton Saier, Jr., Kirsten Skarstad,
and Michel Thellier p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p309
Endolithic Microbial Ecosystems
Jeffrey J. Walker and Norman R. Pace p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p331
Nitrogen Regulation in Bacteria and Archaea
John A. Leigh and Jeremy A. Dodsworth p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p349
Microbial Metabolism of Reduced Phosphorus Compounds
Andrea K. White and William W. Metcalf p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p379
Biofilm Formation by Plant-Associated Bacteria
Thomas Danhorn and Clay Fuqua p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p401
Heterotrimeric G Protein Signaling in Filamentous Fungi
Liande Li, Sara J. Wright, Svetlana Krystofova, Gyungsoon Park,
and Katherine A. Borkovich p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p423
Comparative Genomics of Protists: New Insights into the Evolution
of Eukaryotic Signal Transduction and Gene Regulation
Vivek Anantharaman, Lakshminarayan M. Iyer, and L. Aravind p p p p p p p p p p p p p p p p p p p p453
Lantibiotics: Peptides of Diverse Structure and Function
Joanne M. Willey and Wilfred A. van der Donk p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p477
The Impact of Genome Analyses on Our Understanding of
Ammonia-Oxidizing Bacteria
Daniel J. Arp, Patrick S.G. Chain, and Martin G. Klotz p p p p p p p p p p p p p p p p p p p p p p p p p p p p p503

Contents vii
AR322-FM ARI 9 July 2007 9:23

Morphogenesis in Candida albicans


Malcolm Whiteway and Catherine Bachewich p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p529
Structure, Assembly, and Function of the Spore Surface Layers
Adriano O. Henriques and Charles P. Moran, Jr. p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p555
Cytoskeletal Elements in Bacteria
Peter L. Graumann p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p589

Indexes

Cumulative Index of Contributing Authors, Volumes 57–61 p p p p p p p p p p p p p p p p p p p p p p p p619


Annu. Rev. Microbiol. 2007.61:477-501. Downloaded from www.annualreviews.org

Cumulative Index of Chapter Titles, Volumes 57–61 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p622


by Pennsylvania State University on 05/22/12. For personal use only.

Errata

An online log of corrections to Annual Review of Microbiology articles may be found


at http://micro.annualreviews.org/

viii Contents

You might also like