ArticlePDF Available

Enhancement of antibacterial and lipopolysacchride binding activities of a human lactoferrin peptide fragment by the addition of acyl chain

Authors:

Abstract and Figures

Cationic antibacterial peptides are potentially therapeutic in the treatment of sepsis, because of their amalgamated antibacterial and lipopolysaccharide-binding activities. We prepared acyl analogues of the peptide fragment of human lactoferrin, which originally had weak antibacterial activity. It was found that 12 carbon units constitute the optimal acyl chain length, enhancing the antibacterial activity and binding of lipopolysaccharide by up to two orders of magnitude. Lactoferrin-based lipopeptides approached the activity of polymyxin B, a lipopeptide of natural origin, but were also active against Gram-positive bacteria.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
Journal of Antimicrobial Chemotherapy (2003) 51, 1159–1165
DOI: 10.1093/jac/dkg219
Advance Access publication 14 April 2003
1159
...................................................................................................................................................................................................................................................................
Published by Oxford University Press
Enhancement of antibacterial and lipopolysaccharide binding activities
of a human lactoferrin peptide fragment by the addition of acyl chain
Andreja Majerle, Jurka KidriQ and Roman Jerala*
Laboratory of Biotechnology, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
Received 22 October 2002; returned 13 December 2002; revised 17 February 2003; accepted 18 February 2003
Cationic antibacterial peptides are potentially therapeutic in the treatment of sepsis, because of
their amalgamated antibacterial and lipopolysaccharide-binding activities. We prepared acyl
analogues of the peptide fragment of human lactoferrin, which originally had weak antibacterial
activity. It was found that 12 carbon units constitute the optimal acyl chain length, enhancing
the antibacterial activity and binding of lipopolysaccharide by up to two orders of magnitude.
Lactoferrin-based lipopeptides approached the activity of polymyxin B, a lipopeptide of natural
origin, but were also active against Gram-positive bacteria.
Keywords: antibacterial peptide, endotoxin, human lactoferrin, lipopeptide
Introduction
Animals and plants produce cationic antibacterial peptides as
a first line of defence against invading pathogenic micro-
organisms.
1,2
The structures of these peptides are highly
diverse and can form different types of secondary structure,
but they are all amphipathic and have a net positive charge
under physiological conditions.
3
The primary target of their
action is bacterial membrane.
4
Its leaflet contains, in contrast
to multicellular animals, a large excess of anionic phospho-
lipids. Development of resistance against such peptides, by
modification of the membrane composition, is unlikely with-
out compromising the bacterial viability. Antibacterial pep-
tides may thus provide an alternative to conventional
antibiotics, which are becoming increasingly ineffective due
to the rapid emergence of resistant bacterial strains.
A major constituent of the cell wall of Gram-negative bac-
teria, lipopolysaccharide (LPS), is one of the most potent
stimulants of the immune response and can be released from
bacteria on administration of antibiotics.
5,6
Following cellular
recognition of LPS, inflammatory mediators such as cyto-
kines, adhesion molecules and others are produced
7
and may
lead to septic shock.
6,8
LPS comprises a lipid A moiety, which
is the minimal structural element necessary for endotoxic
activity.
6,9
Attempts to develop molecules that prevent LPS
binding to cellular receptors have often focused on the
lipid A-binding region from endogenous LPS-binding pep-
tides and proteins. The positive charge and hydrophobicity of
peptides seem to be important in determining their ability to
bind LPS. One of the most studied antimicrobial peptides is
cyclic lipopeptide polymyxin B, which is effective against
Gram-negative bacteria.
10
Its medical use, however, is limited
by its toxic side effects.
11
Lactoferrin is an 80 kDa iron-binding glycoprotein found
in exocrine secretions of mammals and in granules of neutro-
phils during inflammatory responses.
12
It has antibacterial
activity against a broad range of Gram-positive and Gram-
negative bacteria and fungi.
1315
Human lactoferrin binds to
lipid A with high affinity
16
and induces LPS release from the
cell wall of Gram-negative bacteria.
17
Proteolytic digestion of
human lactoferrin yields a peptide fragment called lacto-
ferricin H, which has enhanced antibacterial activity compared
to intact lactoferrin.
18
Lactoferricin contains a region that
forms an amphipathic α-helix (residues 2131), distinct from
the site of iron binding
19
and includes the LPS-binding region
(residues 2834),
20
but when isolated this peptide fragment
folds into a β-hairpin structure.
21
Peptides corresponding to
this region exhibit antibacterial activity against Gram-
positive and Gram-negative bacteria
19
and bind LPS.
19,22
Polymyxin B is one of the most potent neutralizers of LPS.
Removal of the 6-heptanoyl/octanoyl diaminobutyryl moiety
..................................................................................................................................................................................................................................................................
*Corresponding author. Tel: +386-1-476-0372; Fax: +386-1-476-0300; E-mail: roman.jerala@ki.si
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
A. Majerle et al.
1160
results in loss of antibacterial activity.
23
This led us to explore
the influence of lipophilic modification of a peptide based on
residues 2131 of human lactoferrin (LF12) on antibacterial
activity and on LPS-binding and neutralizing activity. The
method we used for obtaining recombinant antibacterial
peptide allows straightforward preparation of lipopeptide
conjugates
24
and potentially cost-effective large-scale pro-
duction. By modification with acyl chains, we have enhanced
its antibacterial activity against Gram-negative, and to a
greater extent, Gram-positive bacteria. Endotoxin binding
and in vitro neutralization were enhanced >10-fold.
Materials and methods
Reagents
The chemicals used were of the highest quality commercially
available and obtained mostly from the Sigma-Aldrich Cor-
poration (St Louis, MO, USA). A stock solution of lipid A
(hexa-acyl lipid A from F515 Escherichia coli, provided by
Dr Ulrich Zaehringer), was prepared (1 mg/mL) in endotoxin-
free water, sonicated for 10 min and stored in small aliquots at
20°C. On the day of use it was thawed and sonicated for
3 min. Laboratory glassware used in the chromogenic Limulus
amoebocyte lysate (LAL) assay was thoroughly cleaned and
baked dry for 4 h at 180°C to render it free of contaminating
LPS. Pipette tips from their original packing were wrapped in
aluminium foil piece by piece and autoclaved for 45 min at
131°C and 1.2 × 10
5
Pa.
Bacterial strains and growth conditions
E. coli DC2 (CGSC 7139) was obtained from the E. coli
Genetic Stock Centre (Yale University, New Haven, CT,
USA). Staphylococcus aureus (ATCC 25923) was obtained
from the American Type Culture Collection (Manassas, VA,
USA). Bacterial cultures were stored at 70°C and grown on
LuriaBertani (LB) medium at 37°C.
Preparation and purification of lipopeptides
Procedures for cloning, production and purification of the
recombinant dodecapeptide LF12 (FQWQRNIRKVR-
homoserine lactone) were as described for the production of
15
N-enriched peptide [
15
N]LF12.
24
Peptide LF12 was derived
as follows: 170460 nmol of purified recombinant LF12 was
dried in a centrifuge evaporator, completely lactonized by
the addition of 100% trifluoroacetic acid (TFA) 20 µL and
dried in a rotary evaporator. The dried pellet was dissolved in
anhydrous N, N-dimethylformamide (DMF) 50 µL, delivered
by a gas-tight syringe, and triethylamine (Et
3
N) 8 µL was
added. Alkylamine (hexylamine, n-octylamine, dodecylamine,
tetradecylamine, hexadecylamine, oleylamine) at 100-fold
molar excess over peptide was added to the lactonized peptide
solution and incubated overnight at 45°C. Lipopeptides were
isolated by reverse-phase (RP)-HPLC and eluted at room
temperature with a linear gradient from 30% to 70% of buffer
B (80% acetonitrile, 0.05% TFA in deionized and degassed
water) over 20 min and from 70% to 100% of buffer B over
10 min, at a flow rate of 0.5 mL/min. The identity of the
lipopeptides was confirmed by fast atom bombardment mass
spectrometry (FAB-MS) using a mass spectrometer, AutoSpec
(MicroMass, Manchester, UK).
Spectroscopic characterization of lipopeptides and their
interaction with lipid A
A PTI (Photon Technology International, Lawrenceville, NJ,
USA) spectrofluorimeter was used to measure the intrinsic
tryptophan fluorescence of the lipopeptides. Emission spectra
were recorded from 320 to 370 nm at 25°C in a 10 mm quartz
cuvette with excitation at 280 nm. Slit widths were set at 1 nm.
Fluorescence measurements were similarly used to character-
ize peptide binding to lipid A. Binding of lipopeptides to
lipid A in 20 mM K-phosphate buffer pH 7.0 was monitored
by observing the change in the intrinsic tryptophan fluores-
cence of each lipopeptide. Lipid A was gradually added to a
fixed amount of peptide (1 µM) to a final concentration from
0.5 to 6 µM. From the fit of the fluorescence versus lipid A
concentration curve to the equation: F = F
max
{K
d
+ P
0
+ L
0
[(K
d
P
0
L
0
)
2
4
P
0
L
0
]}/2 where F is fluorescence
intensity, F
max
maximal fluorescence intensity, K
d
dissocia-
tion constant, P
0
total peptide concentration and L
0
total lipid
A concentration, taking into account ligand (lipid A)
depletion,
25
the dissociation constant was determined for
each peptide using a non-linear curve fit implemented in the
program Origin (Microcal).
In vitro LPS binding
The ability of lipopeptides to neutralize LPS in vitro was
assayed by the chromogenic LAL test
26
according to the
manufacturers instructions (Cape Cod Associates, Fal-
mouth, MA, USA). LPS [3.2 endotoxin units (EU)/mL] was
mixed with various concentrations of peptides in endotoxin-
free water. A total of 50 µL of each mixture was added to an
equal volume of the pyrochrome reagent in endotoxin-free
water and incubated for 22 min at 37°C in a 96-well
endotoxin-free microtitre plate pre-equilibrated at 37°C. The
reaction was terminated by adding acetic acid to 10%.
The absorbance at 405 nm was read with a microplate reader
3550-UV (Bio-Rad, Hercules, CA, USA).
Determination of antibacterial activity
Two methods were used to assay the antibacterial activity of
the lipopeptides. In an agar plate assay,
27
overnight cultures of
E. coli and S. aureus in LB medium (tryptone 10 g/L and yeast
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
Addition of acyl chain to enhance antibacterial peptides
1161
extract 5 g/L) were diluted 1:10 into a top agar (6% bacterio-
logical agar in LB medium). Two millilitres of the top agar
lawned with bacteria was poured over LB agar plates warmed
at 37°C. Ten microlitres of different concentrations of pep-
tides were spotted on to solidified top agar. Peptides were
serially diluted in sterile deionized water in a final volume of
10 µL. The plates were incubated at 37°C for 3 h.
Additionally, the antibacterial activity of lipopeptides was
assayed on E. coli and S. aureus using a cfu assay:
28
cells were
incubated with various concentrations of peptides at 37°C for
4 h in 10 mM sodium phosphate buffer (pH 7.4), or at 37°C for
2 h in LB medium, and the number of cfu was determined by
plating the diluted cell suspension on to LB agar plates.
Results
Preparation and purification of LF12 lipopeptides
Production of recombinant peptides in bacteria may be a cost-
efficient alternative to chemical synthesis, particularly for
isotope-labelled peptides. The bacterial production of pep-
tides requires a special approach because of their sensitivity to
proteolytic degradation. An additional problem is the toxicity
of the peptides to bacteria that produce them. Both problems
were solved by prod ucing the peptides in the form of insoluble
fusion proteins.
29
We have used peptide fusion with keto-
steroid isomerase (KSI), a protein that is highly insoluble in
water as well as in the cytoplasm of bacteria.
24
We prepared
peptide LF12, with Met-27 in the original sequence of human
lactoferrin replaced by isoleucine, which is found at position
27 in the porcine variant of lactoferrin.
19
This modification
was necessary because of the use of cyanogen bromide
(CNBr) for cleavage of the fusion protein between the peptide
and carrier protein. Additionally, potential oxidation of the
methionine residue was avoided. This modification did not
change the antibacterial activity of the peptide.
19
Yields of
inclusion bodies of KSILF12His
6
fusion protein
30
exceeded 400 mg/L of the bacterial culture in LB medium.
Cleavage of KSILF12His
6
fusion protein with CNBr
released dodecapeptide LF12, containing, owing to the
CNBr cleavage, a reactive homoserine-lactone group at its
C-terminus. KSI peptide carrier protein, which is hydro-
phobic, precipitated, while the LF12 peptide was present in
the solution and was separated from other peptides (i.e. ter-
minal His
6
-containing C-terminal peptide) by RP-HPLC. A
range of LF12 conjugates was prepared by reaction with
alkylamines with substituent hydrocarbon chains ranging
from six to 18 carbon units. Reaction products were separ-
ated by RP-HPLC and their identity confirmed by mass
spectra (Table 1). The maximum in emission fluorescence
wavelength (λ
max
) decreased with increasing acyl chain
length up to 12 carbon units (LF12-C12) and then increased
again for longer chain lengths (data not shown). This may be
because the addition of acyl chains up to 12 carbon units
increased the hydrophobicity of the environment of trypto-
phan, whereas the lipopeptides with longer acyl chains may
have formed micelles or other aggregates with aliphatic
domains segregated from the tryptophan residue, as recently
suggested for lipophilic acid-modified magainin.
31
Lipid A binding and in vitro neutralization of LPS
Dissociation constants of lipopeptides to lipid A were deter-
mined by fluorescence titration. Binding of lipid A to each
peptide resulted in a blue-shift and an intensity increase of the
fluorescence emission spectra. From the fit of the fluores-
cence versus concentration curves, the dissociation constant
for each peptide was determined (Figure 1, Table 2). Derivatiz-
ation of LF12 with the C
12
chain (LF12-C12) enhanced its
binding to li pid A 14-fold. Its K
d
at 1.5 µM was only three-fold
higher in comparison with polymyxin B.
32
In vitro, the LPS
neutralization potency of lipopeptides was determined using
the LAL assay. The most potent inhibitor of the LAL reaction
was again LF12-C12, where the neutralizing concentration
at 2.4 µM was only two-fold higher in comparison with
polymyxin B (Table 1)
33
and was 12-fold lower than the 50%
endotoxin-neutralizing concentration (ENC
50
) of the parent
peptide LF12.
Antibacterial activity
A cfu assay was used for determination of MICs. The anti-
bacterial activities of peptides were also determined by agar
plate assay. MIC values in the cfu assay were lower by up to
two orders of magnitude in the solution assay, in comparison
with the agar plate assay, which is probably due to the inter-
actions of peptides with agar or high inocula of bacteria. All
lipopeptides were more effective than the parent peptide
LF12 against both Gram-negative and Gram-positive bacteria
(Table 2) and showed higher antibacterial activity against
E. coli than against S. aureus (Figure 2). The optimal acyl
Tabl e 1 . Properties of synthesized (lipo)peptides
a
Isolation by RP-HPLC (linear gradient from 30% to 70% of buffer B over
20 min and from 70% to 100% of buffer B over 10 min, at a flow rate of
0.5 mL/min and room temperature).
b
Determined by FAB-MS.
Peptide
Elution at %
of buffer B
a
Molecular mass experimental
b
(calculated) (Da)
LF12 45 1613.6 (1613.8)
LF12-C6 52 1715.0 (1714.9)
LF12-C8 54 1743.2 (1743.2)
LF12-C12 58 1798.7 (1798.7)
LF12-C14 72 1828.1 (1827.2)
LF12-C16 76 1854.3 (1854.3)
LF12-C18 80 1881.6 (1881.3)
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
A. Majerle et al.
1162
chain length was 12 carbon units, with the highest enhance-
ment of antibacterial activity of 50-fold against E. coli, and
78- or 75-fold enhancement of activity against S. aureus when
compared with the parent peptide. The results of the assay
in sodium phosphate buffer showed similar antibacterial
activity of peptides against both types of bacteria (Table 2,
Figure 2).
Discussion
In the present study, we have improved the antibacterial activ-
ity of a peptide based on a human protein by the addition of
acyl chain. Polymyxin B nonapeptide, a derivative of poly-
myxin B lacking the 6-heptanoyl/octanoyl diaminobutyryl
group of the parent compound, has no antibacterial activity
and poor anti-endotoxic activity.
23
NMR experiments have
shown that the interaction between polymyxin B and LPS
involves electrostatic interactions between the polar head
group of lipid A and the charged residues of polymyxin B, and
hydrophobic interactions between the lipid chains of LPS and
the acyl chain, as well as of a cluster of hydrophobic residues
on polymyxin B.
34
We have selected LF12, a peptide based on human lacto-
ferricin, as a host peptide in the design of novel antibacterial
compounds. MIC values of peptides based on human, mouse
and goat lactoferricins are in the range 63240 µM for
E. coli and S. aureus, whereas the bovine variant is more
potent at 9 µM.
35
This higher antibacterial activity is believed
to be the result of the additional tryptophan residue. The
added hydrophobic chain in our lipopeptides probably fulfils
the same function as the tryptophan residue, which was found
to penetrate into the membrane as a hydrophobic anchor.
36
The dissociation constants of lipopeptides for their binding
to lipid A were similar to the neutralizing concentrations in
LAL, confirming that lipopeptides bound specifically to the
lipid A moiety. Our results suggest that LF12 lipopeptides
bind to LPS in a similar manner to polymyxin B:
34,3739
both electrostatic interactions, particularly between cationic
residues of the lipopeptide and phosphate groups of LPS, and
Figure 1. Binding of LF11-C12 and LF12 peptides to lipid A as moni-
tored by intrinsic tryptophan fluorescence. Upper curve: LF12-C12;
lower curve: LF12. Lipid A was added to 1 µM solution of peptide in 20
mM K-phosphate buffer pH 7 .0. Solid lines represent th e best non-linea
r
fits as described in the Materials and methods section. Fraction of the
complex was determined from (F F
0
)/F
max
(F
0
, fluorescence intensity
at 330 nm without ligand; F
max
, fluorescence intensity at ligand satura-
tion). Only a low ligand concentration range is shown for the LF12
experiment (measured to 50 µM).
Tabl e 2 . Antibacterial and endotoxin-binding activities of (lipo)peptides
K
d
, dissociation constant; ND, not determined.
a
ENC
50
of peptide in the chromogenic LAL test using 3.2 EU/mL of LPS. ENC
50
values (means ± S.E.M.) were
calculated by sigmoidal curve fitting.
b
Cfu assay, incubation in 10 mM sodium phosphate buffer (pH 7.4).
c
Cfu assay, incubation in LB medium.
Lipid A-binding
activity
In vitro LPS
neutralization Antibacterial activity (µM)
Peptide K
d
(µM) ENC
50
(µM)
a
E. coli
b
E. coli
c
S. aureus
b
S. aureus
c
LF12 20.7 30.0 ± 2.4 40 200 200 >600
LF12-C6 6.41 11.2 ± 2.2 1.5 25 15 25
LF12-C8 3.21 8.84 ± 3.3 2 19 10 19
LF12-C12 1.50 2.43 ± 1.6 0.3 4 8 8
LF12-C14 3.35 19.7 ± 1.0 0.6 7 15 15
LF12-C16 7.93 12.1 ± 1.4 3 7 14 14
LF12-C18 8.95 ND 5 8 ND ND
Polymyxin B 0.50
32
1.4 ± 0.5 0.2 0.2 2 2
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
Addition of acyl chain to enhance antibacterial peptides
1163
hydrophobic interactions between the acyl chain of the
lipopeptide and the lipid chains of LPS contribute to the
interaction in solution. The acyl moiety of the lipopeptide is
probably important for disorganizing the bacterial membrane
by disrupting the lipid packing and the supramolecular struc-
ture of LPS, which is important for its endotoxic activity.
Results on LF12 lipopeptides are similar to the data on
modification of peptide antibiotic octapeptin, where the
best antibacterial activity was achieved at chain lengths of
C
8
for E. coli and C
12
and C
14
for Bacillus subtilis.
40
We
have observed that the distribution of antimicrobial activity
as a function of acyl chain length was skewed towards
longer chains for Gram-positive bacteria. Improvement of
antibacterial activity by peptide acylation was higher for
S. aureus than for E. coli. This indicates that antibacterial
specificity as well as efficiency can be altered by the nature of
the hydrophobic substituent.
The efficiency of (lipo)peptides depended on the com-
position of the medium with complex medium (LB) increas-
ing the MIC by up to15-fold, in comparison with the buffer at
low ionic strength (Table 2). The presence of sodium chloride
(171 mM) in the medium did not affect the antibacterial activ-
ity against Gram-negative and Gram-positive bacteria (data
not shown). This is important for their potential therapeutic
use, because many antibacterial peptides have reduced
activity under physiological or increased salt conditions
(e.g. chronic inflammation of lungs by patients with cystic
fibrosis).
41,42
The modification of antibacterial peptide with acyl chains
has the potential for further improvement. Human peptides
and fragments of proteins are an attractive source of host
peptides, since they are less likely to cause antigenic reaction.
Human proteins that interact with LPS more effectively than
lactoferrin (e.g. LPS-binding protein, bactericidal/permeability-
increasing protein),
4345
or which are involved in activation of
immune cells caused by LPS (CD14, TLR4, MD-2),
4648
are
particularly interesting as future donors of host peptides.
Acknowledgements
We thank Dr Ulrich Zaehringer from the Forschungscentrum,
Borstel, Germany for lipid A, Robert Bremšak for his excel-
lent technical help, Professor Rober H. Pain for his comments
on the manuscript, and Bogdan Kralj and Dušan higon (The
National Mass Spectrometry Center at the Joief Stefan Insti-
tute in Ljubljana, Slovenia) for measuring the mass spectra of
lipopeptides. This research was supported by the Ministry of
Education, Science and Sport of the Republic of Slovenia.
References
1. Hancock, R. E., Falla, T. & Brown, M. (1995). Cationic bacteri-
cidal peptides.
Advances in Microbial Physiology
37, 135–75.
2. Zasloff, M. (2002). Antimicrobial peptides of multicellular organ-
isms.
Nature
415, 389–95.
3. Hancock, R. E. (1997). The bacterial outer membrane as a drug
barrier.
Trends in Microbiology
5, 37–42.
4. Hancock, R. E. & Chapple, D. S. (1999). Peptide antibiotics.
Antimicrobial Agents and Chemotherapy
43, 1317–23.
5. Morrison, D. C. (1998). Antibiotic-mediated release of endo-
toxin and the pathogenesis of Gram-negative sepsis.
Progress in
Clinical and Biological Research
397, 199–207.
6. Rietschel, E. T., Kirikae, T., Schade, F. U., Mamat, U., Schmidt,
G., Loppnow, H.
et al
. (1994). Bacterial endotoxin: molecular rela-
tionships of structure to activity and function.
FASEB Journal
8,
217–25.
7. Ulevitch, R. J. & Tobias, P. S. (1995). Receptor-dependent
mechanisms of cell stimulation by bacterial endotoxin.
Annual
Review of Immunology
13, 437–57.
8. Morrison, D. C. & Ryan, J. L. (1987). Endotoxins and disease
mechanisms.
Annual Review of Medicine
38, 417–32.
9. Galanos, C., Luderitz, O., Rietschel, E. T., Westphal, O., Brade,
H., Brade, L.
et al
. (1985). Synthetic and natural
Escherichia coli
free lipid A express identical endotoxic activities.
European Journal
of Biochemistry
148, 1–5.
10. Stokes, D. C., Shenep, J. L., Fishman, M., Hildner, W. K.,
Bysani, G. K. & Rufus, K. (1989). Polymyxin B prevents lipopolysac-
Figure 2. Enhancement of antibacterial activity by modification o
f
LF12 peptides with acyl chains. Ratios of MIC were calculated fo
r
E
. coli and S. aureus as [MIC(LF12)]/[MIC(lipopeptide)] in buffer (a)
and in LB medium (b).
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
A. Majerle et al.
1164
charide-induced release of tumor necrosis factor-alpha from alve-
olar macrophages.
Journal of Infectious Diseases
160, 527.
11. Cohen, J. & Glauser, M. P. (1991). Septic shock: treatment.
Lancet
338, 7369.
12. Masson, P. L., Heremans, J. F. & Schonne, E. (1969). Lacto-
ferrin, an iron-binding protein in neutrophilic leukocytes.
Journal of
Experimental Medicine
130, 64358.
13. Arnold, R. R., Cole, M. F. & McGhee, J. R. (1977). A bacteri-
cidal effect for human lactoferrin.
Science
197, 2635.
14. Bortner, C. A., Miller, R. D. & Arnold, R. R. (1986). Bactericidal
effect of lactoferrin on
Legionella pneumophila
.
Infection and
Immunity
51, 3737.
15. Kalmar, J. R. & Arnold, R. R. (1988). Killing of
Actinobacillus
actinomycetemcomitans
by human lactoferrin.
Infection and
Immunity
56, 25527.
16. Appelmelk, B. J., An, Y. Q., Geerts, M., Thijs, B. G., de Boer, H.
A., MacLaren, D. M.
et al
. (1994). Lactoferrin is a lipid A-binding pro-
tein.
Infection and Immunity
62, 262832.
17. Ellison, R. T., Giehl, T. J. & LaForce, F. M. (1988). Damage of
the outer membrane of enteric Gram-negative bacteria by lacto-
ferrin and transferrin.
Infection and Immunity
56, 277481.
18. Bellamy, W., Takase, M., Wakabayashi, H., Kawase, K. &
Tomita, M. (1992). Antibacterial spectrum of lactoferricin B, a potent
bactericidal peptide derived from the N-terminal region of bovine
lactoferrin.
Journal of Applied Bacteriology
73, 4729.
19. Odell, E. W., Sarra, R., Foxworthy, M., Chapple, D. S. & Evans,
R. W. (1996). Antibacterial activity of peptides homologous to a loop
region in human lactoferrin.
FEBS Letters
382, 1758.
20. Elass-Rochard, E., Roseanu, A., Legrand, D., Trif, M., Salmon,
V., Motas, C.
et al
. (1995). Lactoferrinlipopolysaccharide inter-
action: involvement of the 2834 loop region of human lactoferrin
in the high-affinity binding to
Escherichia coli
055B5 lipopoly-
saccharide.
Biochemical Journal
312, 83945.
21. Hwang, P. M., Zhou, N., Shan, X., Arrowsmith, C. H. & Vogel,
H. J. (1998). Three-dimensional solution structure of lactoferricin B,
an antimicrobial peptide derived from bovine lactoferrin.
Biochem-
istry
37, 428898.
22. Chapple, D. S., Mason, D. J., Joannou, C. L., Odell, E. W.,
Gant, V. & Evans, R. W. (1998). Structurefunction relationship of
antibacterial synthetic peptides homologous to a helical surface
region on human lactoferrin against
Escherichia coli
serotype O111.
Infection and Immunity
66, 243440.
23. Duwe, A. K., Rupar, C. A., Horsman, G. B. & Vas, S. I. (1986).
In vitro
cytotoxicity and antibiotic activity of polymyxin B nona-
peptide.
Antimicrobial Agents and Chemotherapy
30, 3401.
24. Majerle, A., Kidric, J. & Jerala, R. (2000). Production of stable
isotope enriched antimicrobial peptides in
Escherichia coli
: an appli-
cation to the production of
15
N-enriched fragment of lactoferrin.
Journal of Biomolecular NMR
18, 14551.
25. Swillens, S. (1995). Interpretation of binding curves obtained
with high receptor concentrations: practical aid for computer
analysis.
Molecular Pharmacology
47, 1197203.
26. Lindsay, G. K., Roslansky, P. F. & Novitsky, T. J. (1989).
Single-step, chromogenic
Limulus
amebocyte lysate assay for
endotoxin.
Journal of Clinical Microbiology
27, 94751.
27. Zasloff, M. (1987). Magainins, a class of antimicrobial peptides
from
Xenopus
skin: isolation, characterization of two active forms,
and partial cDNA sequence of a precursor.
Proceedings of the
National Academy of Sciences, USA
84, 544953.
28. Valore, E. V., Park, C. H., Quayle, A. J., Wiles, K. R., McCray,
P. B., Jr & Ganz, T. (1998). Human β-defensin-1: an antimicrobial
peptide of urogenital tissues.
Journal of Clinical Investigation
101,
163342.
29. Piers, K. L., Brown, M. H. & Hancock, R. E. (1993). Recom-
binant DNA procedures for producing small antimicrobial cationic
peptides in bacteria.
Gene
134, 713.
30. Kuliopulos, A. & Walsh, C. T. (1994). Production, purification
and cleavage of tandem repeats of recombinant peptides. Journal
of the American Chemistry Society 116, 4599607.
31. Avrahami, D. & Shai, Y. (2002). Conjugation of a magainin
analogue with lipophilic acids controls hydrophobicity, solution
assembly, and cell selectivity.
Biochemistry
41, 225463.
32. David, S. A., Balasubramanian, K. A., Mathan, V. I. & Balaram,
P. (1992). Analysis of the binding of polymyxin B to endotoxic
lipid A and core glycolipid using a fluorescent displacement probe.
Biochimica et Biophysica Acta
1165, 14752.
33. David, S. A., Silverstein, R., Amura, C. R., Kielian, T. &
Morrison, D. C. (1999). Lipopolyamines: novel antiendotoxin com-
pounds that reduce mortality in experimental sepsis caused by
Gram-negative bacteria.
Antimicrobial Agents and Chemotherapy
43, 91219.
34. Pristovsek, P. & Kidric, J. (1999). Solution structure of poly-
myxins B and E and effect of binding to lipopolysaccharide: an NMR
and molecular modeling study.
Journal of Medical Chemistry
42,
460413.
35. Rekdal, O., Andresen, J., Vorland, L. H. & Svendsen, J. S.
(1999). Construction and synthesis of lactoferricin derivatives with
enhanced antimicrobial activity.
Journal of Peptide Science
5,
3245.
36. Schibli, D. J., Hwang, P. M. & Vogel, H. J. (1999). The structure
of the antimicrobial active center of lactoferricin B bound to sodium
dodecyl sulfate micelles.
FEBS Letters
446, 2137.
37. Wang, H. Y., Tummler, B. & Boggs, J. M. (1989). Use of spin
labels to determine the percentage of interdigitated lipid in com-
plexes with polymyxin B and polymyxin B nonapeptide.
Biochimica
et Biophysica Acta
985, 18298.
38. Thomas, C. J., Gangadhar, B. P., Surolia, N. & Surolia, A.
(1998). Kinetics and mechanism of the recognition of endotoxin by
polymyxin B.
Journal of the American Chemistry Society
120,
1242834.
39. Thomas, C. J. & Surolia, A. (1999). Kinetics of the interaction of
endotoxin with polymyxin B and its analogs: a surface plasmon
resonance analysis.
FEBS Letters
445, 4204.
40. Swanson, P. E., Paddy, M. R., Dahlquist, F. W. & Storm, D. R.
(1980). Characterization of octapeptinmembrane interactions
using spin-labeled octapeptin.
Biochemistry
19, 330714.
41. Bals, R., Wang, X., Wu, Z., Freeman, T., Bafna, V., Zasloff, M.
et al
. (1998). Human β-defensin 2 is a salt-sensitive peptide anti-
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
Addition of acyl chain to enhance antibacterial peptides
1165
biotic expressed in human lung.
Journal of Clinical Investigation
102, 87480.
42. Beringer, P. M. (1999). New approaches to optimizing anti-
microbial therapy in patients with cystic fibrosis.
Current Opinion in
Pulmonary Medicine
5, 3717.
43. Marra, M. N., Wilde, C. G., Griffith, J. E., Snable, J. L. & Scott,
R. W. (1990). Bactericidal/permeability-increasing protein has
endotoxin-neutralizing activity.
Journal of Immunology
144, 6626.
44. Su, G. L., Simmons, R. L. & Wang, S. C. (1995). Lipopoly-
saccharide binding protein participation in cellular activation by
LPS.
Critical Reviews in Immunology
15, 20114.
45. Tobias, P. S., Soldau, K., Iovine, N. M., Elsbach, P. & Weiss, J.
(1997). Lipopolysaccharide (LPS)-binding proteins BPI and LBP
form different types of complexes with LPS.
Journal of Biological
Chemistry
272, 186825.
46. Wright, S. D., Ramos, R. A., Tobias, P. S., Ulevitch, R. J. &
Mathison, J. C. (1990). CD14, a receptor for complexes of lipopoly-
saccharide (LPS) and LPS binding protein.
Science
249, 14313.
47. Beutler, B. (2000). Tlr4: central component of the sole mam-
malian LPS sensor.
Current Opinion in Immunology
12, 206.
48. Aderem, A. & Ulevitch, R. J. (2000). Toll-like receptors in the
induction of the innate immune response.
Nature
406, 7827.
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
by guest on June 8, 2013http://jac.oxfordjournals.org/Downloaded from
... In-58 modified by unsaturated fatty acid exhibited more potent bactericidal, resistant to proteinase K, and less toxic to mammalian cells (Vasilchenko et al. 2017). What's more, previous studies have demonstrated that a human Lfcin was conjugated with a twelve-carbon fatty acid, which is the optimal length of tail to improve activity (Majerle et al. 2003). ...
... Recently, researchers have paid attention to design fatty acid conjugation with peptides in order to improve bactericidal activity, and the strategy borrows from the naturally occurring polymyxins and daptomycin . It was investigated that Nterminal fatty acid conjugation increased bactericidal activity of lactoferrin peptides (Majerle et al. 2003). In this study, Lf4NH2 and Lf5NH2 were firstly conjugated with the unsaturated fatty acid linoleic acid (18-C) at the N-terminal to detect their antibacterial activity against S. hyicus in vitro and topically therapeutic efficacy in vivo. ...
Article
Full-text available
There is an urgent need to explore new antimicrobial agents due to the looming threat of bacteria resistance. Bovine lactoferricin (LfcinB), as a multifunctional peptide, has the potential to be a new active drug in the future. In this study, it aims to investigate the effect of fatty acid conjugation on antimicrobial peptide activity and topical therapeutic efficacy in a mouse model infected with Staphylococcus hyicus. Both Lfcin4 and Lfcin5 were conjugated with the unsaturated fatty acid linoleic acid (18-C) at their N-terminus and modified by acylation at the C-terminus. The derived peptides of Lin-Lf4NH2 and Lin-Lf5NH2 showed better antibacterial activity (MICs of 3.27 to 6.64 μM) than their parent peptides (MICs of 1.83 to 59.57 μM). Lin-Lf4NH2 (63.2%, 5 min) and Lin-Lf5NH2 (35.8%, 5 min) could more rapidly penetrate bacterial membrane than Lf4NH2 (2.34%, 5 min) and Lf5NH2 (1.94%, 5 min), which further confirmed by the laser scanning confocal microscopy (LSCM). Electron microscopy observations showed Lin-Lf4NH2 and Lin-Lf5NH2 disrupted S. hyicus cell membranes and led to the leakage of contents. Furthermore, after treatment with Lin-Lf4NH2 and Lin-Lf5NH2, the abscess symptoms of mice were significantly alleviated; the recovery rate of abscesses scope of Lin-Lf4NH2 (73.25%) and Lin-Lf5NH2 (71.71%) were 38.8 and 37.9-fold higher than that of untreated group (1.89%), respectively, and superior to Lf4NH2 (46.87%) and Lf5NH2 (58.75%). They significantly reduced the bacterial load and the levels of the pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and chemokine (MCP-1) in S. hyicus skin lesions. This study provides evidence that conjugation of a fatty acid to antimicrobial peptides can improve the activity and have potential for topical therapeutic of S. hyicus skin infections. Key points • Lin-Lfcin4NH2/Lfcin5NH2 showed stronger antimicrobial activity than parent peptides. • Lin-Lfcin4NH2/Lfcin5NH2 had a more effective ability to destroy bacterial membranes. • Lin-Lfcin4NH2/Lfcin5NH2 showed a topically higher efficacy than parent peptides.
... Fibrous bodies are visible in almost every captured picture, except for the effect of SHP-3.3. This could be explained by the ability of lipopeptides to aggregate (Shai 2002;Majerle et al. 2003;Zanetti et al. 1995). Enhanced binding activities and aggregation of lipopeptides could cause the packaging of cells and their slower destruction. ...
Article
Full-text available
The aim of this article is to introduce the topic of newly designed peptides as well as their biological activity. We designed nine encoded peptides composed of six amino acids. All these peptides were synthesized with C-terminal amidation. To investigate the importance of increased hydrophobicity at the amino end of the peptides, all of them were subsequently synthesized with palmitic or lithocholic acid at the N-terminus. Antimicrobial activity was tested on Gram-positive and Gram-negative bacteria and fungi. Cytotoxicity was measured on HepG2 and HEK 293 T cell cultures. Peptides bearing a hydrophobic group exhibited the best antimicrobial activity. Lipopeptides with palmitic or lithocholic acid (PAL or LCA peptides) at the N-terminus and with C-terminal amidation were highly active against Gram-positive bacteria, especially against strains of Staphylococcus aureus and Candida tropicalis. The LCA peptide SHP 1.3 with the sequence LCA-LVKRAG-NH2, had high efficiency on HepG2 human liver hepatocellular carcinoma cells (97%).
... Chu et al. found that three Nal tagged at the C-terminus of peptide (S1-Nal-Nal-Nal) resulted in > 20 % hemolysis at 6.25 g/mL [30] . An increasing body of evidence suggests that the hemolysis of AMPs is significantly increased when the fatty acid chain length exceeds 12 carbons [31][32][33] . Hence, the length of the hydrophobic tags in our design was less than or equal to 3 amino acids or 12 carbons (fatty acids). ...
Article
Full-text available
Recently, much emphasis has been placed on solving the intrinsic defects of antimicrobial peptides (AMPs), especially their susceptibility to protease digestion for the systemic application of antibacterial biomaterials. Although many strategies have increased the protease stability of AMPs, antimicrobial activity was severely compromised, thereby substantially weakening their therapeutic effect. To address this issue, we introduced hydrophobic group modifications at the N-terminus of proteolysis-resistant AMPs D1 (AArIIlrWrFR) through end-tagging with stretches of natural amino acids (W and I), unnatural amino acid (Nal) and fatty acids. Of these peptides, N1 tagged with a Nal at N-terminus showed the highest selectivity index (GMSI = 19.59), with a 6.73-fold improvement over D1. In addition to potent broad-spectrum antimicrobial activity, N1 also exhibited high antimicrobial stability toward salts, serum and proteases in vitro and ideal biocompatibility and therapeutic efficacy in vivo. Furthermore, N1 killed bacteria through multiple mechanisms, involving disruption of bacterial membranes and inhibition of bacterial energy metabolism. Indeed, appropriate terminal hydrophobicity modification opens up new avenues for developing and applying high-stability peptide-based antibacterial biomaterials. STATEMENT OF SIGNIFICANCE: To improve the potency and stability of proteolysis-resistant antimicrobial peptides (AMPs) without increasing toxicity, we constructed a convenient and tunable platform based on different compositions and lengths of hydrophobic end modifications. By tagging an Nal at the N-terminal, the obtained target compound N1 exhibited strong antimicrobial activity and desirable stability under multifarious environments in vitro (protease, salts and serum), and also showed favorable biocompatibility and therapeutic efficacy in vivo. Notably, N1exerted its bactericidal effect by damaging bacterial cell membranes and inhibiting bacterial energy metabolism in a dual mode. The findings provide a potential method for designing or optimizing proteolysis-resistant AMPs thus promoting the development and application of peptide-based antibacterial biomaterial.
Article
Full-text available
This study investigates short cationic antimicrobial lipopeptides composed of 2–4 amino acid residues and C12-C18 fatty acids attached to the N-terminal part of the peptides. The findings were discussed in the context of the relationship among biological activity, self-assembly, stability, and membrane interactions. All the lipopeptides showed the ability to self-assemble in PBS solution. In most cases, the critical aggregation concentration (CAC) much surpassed the minimal inhibitory concentration (MIC) values, suggesting that monomers are the main active form of lipopeptides. The introduction of β-alanine into the peptide sequence resulted in a compound with a high propensity to fibrillate, which increased the peptide stability and activity against S. epidermidis and C. albicans and reduced the cytotoxicity against human keratinocytes. The results of our study indicated that the target of action of lipopeptides is the bacterial membrane. Interestingly, the type of peptide counterion may affect the degree of penetration of the lipid bilayer. In addition, the binding of the lipopeptide to the membrane of Gram-negative bacteria may lead to the release of calcium ions necessary for stabilization of the lipopolysaccharide layer.
Article
The increasingly severe bacterial resistance worldwide pushes people to discover and design potential antibacterial drugs unavoidably. In this work, a series of short, mirror-symmetric peptides were designed and successfully synthesized, centered on “RRR” and labeled with hydrophobic amino acids at both ends. Based on the structure-activity relationship analysis, LWWR (LWWRRRWWL-NH2) was screened as a desirable mirror-symmetric peptide for further study. As expected, LWWR displayed broad-spectrum antibacterial activity against the standard bacteria and antibiotic-resistant strains. Undoubtedly, the high stability of LWWR in a complex physiological environment was an essential guarantee to maximizing its antibacterial activity. Indeed, LWWR also exhibited a rapid bactericidal speed and a low tendency to develop bacterial resistance, based on the multiple actions of non-receptor-mediated membrane actions and intra-cellular mechanisms. Surprisingly, although LWWR showed similar in vivo antibacterial activity compared with Polymyxin B and Melittin, the in vivo safety of LWWR was far higher than that of them, so LWWR had better therapeutic potential. In summary, the desirable mirror-symmetric peptide LWWR was promised as a potential antibacterial agent to confront the antibiotics resistance crisis. Statement of Significance Witnessing the growing problem of antibiotic resistance, a series of short, mirror-symmetric peptides based on the symmetric center “RRR” and hydrophobic terminals were designed and synthesized in this study. Amongst, LWWR (LWWRRRWWL-NH2) presented broad-spectrum antibacterial activity both in vitro and in vivo due to its multiple mechanisms and good stability. Meanwhile, the low drug resistance and toxicity of LWWR also suggested its potential for clinical application. The findings of this study will provide some inspiration for the design and development of potential antibacterial agents, and contribute to the elimination of bacterial infections worldwide as soon as possible.
Article
Growing multidrug bacterial resistance has become a life-threatening problem. Polymyxins are a “last resort” antibiotic against Gram-negative bacteria (Gram-ne⁻), but exhibit weak activity against Gram-positive bacteria (Gram-po⁺) caused by different bacterial membrane structures. Herein, it is proposed that the challenge can be overcome by hydrophobically modified polymyxin E for enhancing the hydrophobic interaction with the membrane. After synthesizing mono-and di-fatty acyl conjugates to Thr residues of polymyxin E, seven new polymyxin analogs were screened out with in vitro activity against Gram-ne⁻ and Gram-po⁺. Among them, di-caprylic acid-modified (comp. 2) and mono-linoleic acid-modified compounds (comp. 3) were the most potent activity against both bacteria. Moreover, the membrane research showed that polymyxin analogs obtain activity against Gram-po⁺ by higher ability to dispute cell membrane and wall than polymyxin. Additionally, the results showed a trend of increasing hydrophobicity of analogs showing no-to-no activity against Gram-po⁺ and gradually decreasing activity against Gram-ne⁻, hence, we speculated that controlling Log P at 1-3 helps to obtain broad-spectrum antibiotic. Crucially, the maximum tolerated doses (MTD) of analogs were significantly higher than polymyxin, and reduced the nephrotoxicity. As expected, hydrophobic modification polymyxin E not only enhance the activity of Gram-po⁺, but also could retain the activity of Gram-ne⁻. This study offers more insights into the synthesis of new antibiotics based on polymyxin E structure, especially broad-spectrum antibiotics.
Article
Antimicrobial resistance (AMR) is one of the greatest threats to human health that, by 2050, will lead to more deaths from bacterial infections than cancer. New antimicrobial agents, both broad-spectrum and selective, that do not induce AMR are urgently required. Antimicrobial peptides (AMPs) are a novel class of alternatives that possess potent activity against a wide range of Gram-negative and positive bacteria with little or no capacity to induce AMR. This has stimulated substantial chemical development of novel peptide-based antibiotics possessing improved therapeutic index. This review summarises recent synthetic efforts and their impact on analogue design as well as their various applications in AMP development. It includes modifications that have been reported to enhance antimicrobial activity including lipidation, glycosylation and multimerization through to the broad application of novel bio-orthogonal chemistry, as well as perspectives on the direction of future research. The subject area is primarily the development of next-generation antimicrobial agents through selective, rational chemical modification of AMPs. The review further serves as a guide toward the most promising directions in this field to stimulate broad scientific attention, and will lead to new, effective and selective solutions for the several biomedical challenges to which antimicrobial peptidomimetics are being applied.
Article
Full-text available
Multi-drug-resistant bacteria are becoming more prevalent, and treating these bacteria is becoming a global concern. One alternative approach to combat bacterial resistance is to use antimicrobial (AMPs) or host-defense peptides (HDPs) because they possess broad-spectrum activity, function in a variety of ways, and lead to minimal resistance. However, the therapeutic efficacy of HDPs is limited by a number of factors, including systemic toxicity, rapid degradation, and low bioavailability. One approach to circumvent these issues is to use lipidation, i.e., the attachment of one or more fatty acid chains to the amine groups of the N-terminus or a lysine residue of an HDP. In this review, we examined lipidated analogs of 66 different HDPs reported in the literature to determine: (i) whether there is a link between acyl chain length and antibacterial activity; (ii) whether the charge and (iii) the hydrophobicity of the HDP play a role; and (iv) whether acyl chain length and toxicity are related. Overall, the analysis suggests that lipidated HDPs with improved activity over the nonlipidated counterpart had acyl chain lengths of 8–12 carbons. Moreover, active lipidated peptides attached to short HDPs tended to have longer acyl chain lengths. Neither the charge of the parent HDP nor the percent hydrophobicity of the peptide had an apparent significant impact on the antibacterial activity. Finally, the relationship between acyl chain length and toxicity was difficult to determine due to the fact that toxicity is quantified in different ways. The impact of these trends, as well as combined strategies such as the incorporation of d- and non-natural amino acids or alternative approaches, will be discussed in light of how lipidation may play a role in the future development of antimicrobial peptide-based alternatives to current therapeutics.
Article
Full-text available
Antimicrobial peptides are widely distributed mediators of innate host defense in animals and plants. A 36 amino acid antimicrobial peptide belonging to the defensin family, and named human beta-defensin-1 (HBD-1), was purified recently from hemodialysate fluid, but its tissue sources were not identified. By Northern blotting, we found the highest concentrations of HBD-1 mRNA in the kidney and the female reproductive tract. In situ hybridization localized the HBD-1 mRNA in the epithelial layers of the loops of Henle, distal tubules, and the collecting ducts of the kidney and the epithelial layers of the vagina, ectocervix, endocervix, uterus, and fallopian tubes in the female reproductive tract. Using a novel technique designed to detect cationic peptides in urine, we recovered several forms of HBD-1 ranging in length from 36 to 47 amino acid (aa) residues and differing from each other by amino terminal truncation. The total concentration of HBD-1 forms in voided urine was estimated at 10-100 microg/liter, with individual variations in the total amount of HBD-1 peptides and the relative proportion of HBD-1 forms. Multiple forms of HBD-1 (size 36-47 aa) were also found in the blood plasma, bound to carrier macromolecules that released the peptide under acid conditions, and in vaginal mucosal secretions (39, 40, and 44 aa). By immunostaining, HBD-1 was located in the kidney within the lumen of the loops of Henle, but no intracellular storage sites were identified in renal or female reproductive tissues. Recombinant HBD-1 forms (36, 39, and 42 aa) and natural HBD-1 forms were antimicrobial to laboratory and clinical strains of Escherichia coli at micromolar concentrations. HBD-1 activity was not changed appreciably by low pH, but was inhibited by high salt conditions. Some of the HBD-1 peptides retained their activity against E. coli in unconcentrated (low conductance) urine, and the 36 aa form was microbicidal even in normal (high conductance) urine. Production of HBD-1 in the urogenital tract could contribute to local antimicrobial defense.
Article
We report here, for the first time, the elementary steps involved in the recognition of the endotoxin (lipopolysaccharide) molecule by polymyxin B (PMB), a cyclic cationic decapeptide. Miniscule amounts of lipopolysaccharide, the invariant structural component of Gram-negative bacterial outer membranes, in circulation in humans elicits “endotoxic shock” syndrome, which is fatal in almost 60% of the instances. PMB, despite its harmful side effects, is the only useful drug for combating endotoxic shock. It neutralizes the endotoxin by binding to it. The kinetics and mechanism of this important biological recognition, investigated by stopped-flow spectrofluorometry, provide considerable insight about the endotoxin neutralizing activity of this antibiotic. This process consists of a pair of kinetically distinguishable but consecutive association and dissociation reactions, with rate constants of 1.98 × 105 M-1 s-1 (k1), 0.458 s-1 (k2), 0.458 s-1 (k-1), and 0.0571 s-1 (k-2) at 20 °C. Analysis of the activation parameters for this recognition suggests that, during the first phase of the reaction, which is bimolecular in nature, PMB associates with endotoxin in a relatively less constrained manner. Subsequently, a considerable reorganization of this initial complex occurs, which entails a significant expenditure of energy. Design of analogues of PMB which are able to overcome the constraints of the rate-limiting step should serve as more effective agents for treating endotoxic shock.
Article
We describe a method to construct tandem repeats of coding sequences for polypeptides interspersed by single methionine residues and terminating in a His(6) tag that can be purified by Ni chelate chromatography and then cleaved by cyanogen bromide (CNBr) into homogeneous peptide units, Annealing and unidirectional ligation of complementary 42mer oligonucleotides encoding the 13 amino acid residue yeast alpha-mating factor (alpha F) followed by ligation of the oligomerized 42mers into a pET vector placed the alpha-factor tandem repeats downstream of the ketosteroid isomerase (KSI) gene and upstream of a His(6) cassette. A KSI-(alpha F)(5)-His(6) fusion was overproduced in Escherichia coli, purified by Ni chelate chromatography, and then cleaved with CNBr to release insoluble KSI, the His(6) tail, and the alpha-factor peptide units, each terminating with homoserine (HS) lactone. HPLC yielded pure peptide in a yield of 56 mg/L. The alpha-factor-HS(lactone) could be ammonolyzed or hydrolyzed to yield alpha-factor-HS-amide or alpha-factor-HS, respectively. The alpha-factor-HS peptide had similar biological potency as authentic alpha-factor in yeast cell arrest assays. The alpha-factor-HS(lactone) was also reacted with a number of other compounds including analogs of fluorescein, dansyl, and biotin to produce alpha-factor peptides derivatized exclusively at the C-terminus. To test the ability of the expression system to produce longer peptides, 60-67 amino acid residue peptides encoding the Gla domain of profactor IX (FIXQS, FIXQS-His(6)) were also produced. Yields of 50-55 mg/L of pure FIXQS-His(6) and FIXQS-HS(lactone) were obtained.
Article
A series of peptides derived from sequences from human, bovine, murine and caprine lactoferrin has been prepared and investigated for antibacterial effect. Among the four species investigated peptides based on the bovine sequence displayed significant activity. The bovine sequence, bovine lactoferricin, showed a MIC value of 30 μg/mL on E. coli and S. aureus, whereas the three other lactoferricins possessed MIC values above 200 μg/mL. Based on these findings, novel peptides with enhanced antibacterial activities, were prepared with sequences designed by molecular modelling and structure-activity studies. Copyright © 1999 European Peptide Society and John Wiley & Sons, Ltd.
Article
The solution structure of bovine lactoferricin (LfcinB) has been determined using 2D 1H NMR spectroscopy. LfcinB is a 25-residue antimicrobial peptide released by pepsin cleavage of lactoferrin, an 80 kDa iron-binding glycoprotein with many immunologically important functions. The NMR structure of LfcinB reveals a somewhat distorted antiparallel beta-sheet. This contrasts with the X-ray structure of bovine lactoferrin, in which residues 1-13 (of LfcinB) form an alpha-helix. Hence, this region of lactoferricin B appears able to adopt a helical or sheetlike conformation, similar to what has been proposed for the amyloidogenic prion proteins and Alzheimer's beta-peptides. LfcinB has an extended hydrophobic surface comprised of residues Phe1, Cys3, Trp6, Trp8, Pro16, Ile18, and Cys20. The side chains of these residues are well-defined in the NMR structure. Many hydrophilic and positively charged residues surround the hydrophobic surface, giving LfcinB an amphipathic character. LfcinB bears numerous similarities to a vast number of cationic peptides which exert their antimicrobial activities through membrane disruption. The structures of many of these peptides have been well characterized, and models of their membrane-permeabilizing mechanisms have been proposed. The NMR solution structure of LfcinB may be more relevant to membrane interaction than that suggested by the X-ray structure of intact lactoferrin. Based on the solution structure, it is now possible to propose potential mechanisms for the antimicrobial action of LfcinB.
Article
Streptococcus mutans and Vibrio cholerae, but not Escherichia coli, were killed by incubation with purified human apolactoferrin. Concentrations of lactoferrin below that necessary for total inhibition resulted in a marked reduction in viable colony-forming units. This bactericidal effect was contingent upon the metal-chelating properties of the lactoferrin molecule.
Article
Dansylcadaverine, a cationic fluorescent probe binds to bacterial lipopolysaccharide and lipid A, and is displaced competitively by other compounds which possess affinity toward endotoxins. The binding parameters of dansylcadaverine for lipid A were determined by Scatchard analysis to be two apparently equivalent sites with apparent dissociation constants (Kd) ranging between 16 microM to 26 microM, while that obtained for core glycolipid from Salmonella minnesota Re595 yielded a Kd of 22 microM to 28 microM with three binding sites. The Kd of polymyxin B for lipid A was computed from dansylcadaverine displacement by the method of Horovitz and Levitzki (Horovitz, A., and Levitzki, A. (1987) Proc. Natl. Acad. Sci. USA 84, 6654-6658). The applicability of this method for analyzing fluorescence data was validated by comparing the Kds of melittin for lipid A obtained by direct Scatchard analysis, and by the Horovitz-Levitzki method. The displacement of dansylcadaverine from lipid A by polymyxin B was distinctly biphasic with Kds for polymyxin B-lipid A interactions corresponding to 0.4 microM and 1.5 microM, probably resulting as a consequence of lipid A being a mixture of mono- and di-phosphoryl species. This was not observed with core glycolipid, for which the Kd for polymyxin was estimated to range from 1.1 microM to 5.8 microM. The use of dansylcadaverine as a displacement probe offers a novel and convenient method of quantitating the interactions of a wide variety of substances with lipid A.
Article
A physiologically diverse range of Gram-positive and Gram-negative bacteria was found to be susceptible to inhibition and inactivation by lactoferricin B, a peptide produced by gastric pepsin digestion of bovine lactoferrin. The list of susceptible organisms includes Escherichia coli, Salmonella enteritidis, Klebsiella pneumoniae, Proteus vulgaris, Yersinia enterocolitica, Pseudomonas aeruginosa, Campylobacter jejuni, Staphylococcus aureus, Streptococcus mutans, Corynebacterium diphtheriae, Listeria monocytogenes and Clostridium perfringens. Concentrations of lactoferricin B required to cause complete inhibition of growth varied within the range of 0.3 to 150 micrograms/ml, depending on the strain and the culture medium used. The peptide showed activity against E. coli O111 over the range of pH 5.5 to 7.5 and was most effective under slightly alkaline conditions. Its antibacterial effectiveness was reduced in the presence of Na+, K+, Mg2+ or Ca2+ ions, or in the presence of various buffer salts. Lactoferricin B was lethal, causing a rapid loss of colony-forming capability in most of the species tested. Pseudomonas fluorescens, Enterococcus faecalis and Bifidobacterium bifidum strains were highly resistant to this peptide.