Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

European Biophysics Journal

https://doi.org/10.1007/s00249-019-01346-7

ORIGINAL ARTICLE

NMR model structure of the antimicrobial peptide maximin 3


Silvia Benetti1,2   · Patrick Brendan Timmons1   · Chandralal M. Hewage1 

Received: 5 November 2018 / Revised: 20 December 2018 / Accepted: 2 January 2019


© European Biophysical Societies’ Association 2019

Abstract
Maximin 3 is a 27-residue-long cationic antimicrobial peptide found in the skin secretion and brain of the Chinese red-belly
toad Bombina maxima. The peptide is of biological interest as it possesses anti-HIV activity, not found in the other maximin
peptides, in addition to antimicrobial, antitumor and spermicidal activities. The three-dimensional structure of maximin
3 was obtained in a 50/50% water/2,2,2-trifluoroethanol-d3 mixture using two-dimensional NMR spectroscopy. Maximin
3 was found to adopt an α-helical structure from residue G1 to A22, and a coil structure with a helical propensity in the
C-terminal tail. The peptide is amphipathic, showing a clear separation between polar and hydrophobic residues. Interac-
tions with sodium dodecyl sulfate micelles, a widely used bacterial membrane-mimicking environment, were modeled using
molecular dynamics simulations. The peptide maintained an α-helical conformation, occasionally displaying a flexibility
around residues G9 and G16, which is likely responsible for the peptide’s low haemolytic activity. It is found to preferentially
adopt a position parallel to the micellar surface, establishing a number of hydrophobic and electrostatic interactions with it.

Keywords  Antimicrobial peptide maximin · NMR · AMP modeling

Introduction parasites and viruses (Gordon et al. 2005). Some peptides


are also important for wound healing, immune activation and
Antimicrobial peptides (AMPs), involved in the non-specific inflammatory processes (Yang et al. 2002, 2004), or show
response of the innate immune system (Xia et al. 2018), other biological activities such as anti-cancer (Deslouches
are present in all kinds of organisms, from viruses, bacte- et al. 2017), anti-HIV (Pace et al. 2017) and spermicidal
ria and plants (Maróti Gergely et al. 2011; da Silva Pereira (Tanphaichitr et al. 2016).
et al. 2018) to vertebrates (Avila 2017) and invertebrates AMPs have a primary sequence typically ranging between
(El Samak et al. 2018). The majority of these peptides are 10 and 50 residues and usually possess no secondary struc-
found in amphibians, with more than a 1000 reported in the ture in water. They adopt a secondary structure in the bacte-
antimicrobial peptide database, APD3 (https​://aps.unmc. rial membrane and in non-polar environments. Secondary
edu/AP/main.php). Bacterial resistance to antibiotics is the structure can be used to classify antimicrobial peptides.
motivation for the search of new antimicrobial compounds Most, such as magainin and LL-37, are α-helical. Other
that can be used without the development of resistance. structures include β-sheet, which include human α-defensins,
AMPs are interesting both as substitutes of or in combina- α–β structures, such as β-defensins, and extended structures,
tion with antibiotics because of their immediate response such as indolicidin. Many AMPs have two common features:
and activity against many pathogens such as bacteria, fungi, amphipathicity and a global positive charge, thanks to which
they interact with the bacterial cell membranes (Hsiao et al.
2018). Prokaryotic cell membranes are negatively charged
* Chandralal M. Hewage because of their composition; lipopolysaccharides (LPS) are
chandralal.hewage@ucd.ie found in Gram-negative bacterial membrane, and phospho-
1
lipids and acidic polysaccharides (teichoic acids) in Gram-
UCD School of Biomolecular and Biomedical Science, UCD
Centre for Synthesis and Chemical Biology, UCD Conway
positive bacterial one (Beveridge 1999; Weidenmaier and
Institute, University College Dublin, Belfield, Dublin 4, Peschel 2008). In fact, the interaction between peptide and
Ireland prokaryotic cell membrane is electrostatically driven and
2
Department of Chemical Sciences, Università Degli Studi Di increasing the former positive charge results in a higher
Padova, Via Marzolo 1, 35131 Padova, Italy

13
Vol.:(0123456789)
European Biophysics Journal

antibacterial activity (Xie et al. 2017). Charges higher than Materials and methods
a certain value, dependent on the peptide considered, result
in a decrease in antimicrobial activity (Zelezetsky and Tossi Materials and NMR sample preparation
2006).
There are not many known cases of bacterial resistance 3-Trimethylsilyl propionic acid (TSP) and 2,2,2-trifluoro-
(Yeaman and Yount 2003; Perron et al. 2006) because of ethanol (TFE-d3) of analytical grade were obtained from
the non-specific mechanisms for killing bacteria that usually Sigma-Aldrich (Ireland).
involve an interaction with the membrane to disrupt it. There The peptide maximin 3 (MW =2698  g ­ m ol −1 ,
are three main mechanisms of membrane permeation: car- purity>98%) was purchased from ProteoGenix (Paris).
pet-like (or detergent-like), barrel-stave and toroidal. They Peptide (5 mg) was dissolved in 0.6 mL of 50% (v/v) TFE-
all lead to leaking of cellular components and dissipation d3–H2O solution, resulting in a peptide concentration of
of the electrical potential. The first step is always the bind- 3.09 mM.
ing with the membrane surface by electrostatic interaction.
After a threshold concentration is reached, the peptides,
respectively, disrupt the bilayer curvature and disintegrate
the membrane; or they form a pore, with the hydrophobic NMR spectroscopy
residues facing the lipid core region of the bilayer and the
polar ones making up the interior region of the lumen; or The NMR measurements were performed at 298  K on
they bend the membrane from the outer towards the inner a Bruker Avance 600 NMR spectrometer with a 5-mm
leaflet, forming a toroidal pore made up of the lipid head inverse probe head at a 1 H resonance frequency of
groups and the peptides (Shai 2002; Brogden 2005). The 600.13 MHz.
threshold concentration required to disrupt the bilayer may One-dimensional (1D) and two-dimensional (2D)
also be dependent on the bacterial target’s ability to repair NMR spectra were acquired with a relaxation delay of
damaged cell membranes (Sani et al. 2015). Another impor- 4.0 s and 1.5 s, respectively. The 2D phase-sensitive total
tant feature to consider is their haemolytic activity; some correlation spectroscopy (TOCSY) (Bax and Davis 1985),
AMPs are also lytic to mammalian cells. Human erythro- nuclear Overhauser effect spectroscopy (NOESY) (Kumar
cytes are considered as representatives of eukaryotic cells et al. 1980) and natural abundance 1H–13C heteronuclear
and their membrane is composed of neutral lipids such as single-quantum coherence spectroscopy (1H–13C-HSQC)
phosphatidylcholine/lecithin and sphingomyelin (Verkleij (John et al. 1992) experiments were performed with an
et al. 1973; Nouri-Sorkhabi et al. 1996). Some peptides are acquisition time of 0.28 s, 0.57 s and 0.14 s, respectively,
toxic to human cells and the interaction with human cells and mixing time of 80 ms for TOCSY and 200  ms for
was suggested to be hydrophobic instead of electrostatic. NOESY. The 1H spectral width was 7.2 kHz for all the 2D
Hydrophobicity is, therefore, a key feature for toxicity and data, and the 13C spectral width was 21.1 kHz. The spectra
it can be changed to modulate the selectivity, alongside the were acquired with 8, 16 and 32 transients for each of the
net charge (Yin et al. 2012; Son et al. 2013). 2048, 1024 and 1024 ­t1 increments into 512 complex data
Structural studies of AMPs have been carried out in our points for the TOCSY, NOESY and HSQC, respectively.
laboratory for various amphibian peptides, including XT-7 The sine-squared window function was applied for pro-
(Subasinghage et al. 2010), ranatuerin-2CSa (Subasinghage cessing in all the 2D spectra and the 1H signal of TSP was
et al. 2008) and alyteserin-1c (Subasinghage et al. 2011). In used as chemical shift reference. Spectra were processed
search of a peptide with potential to target many diseases, with the Bruker TopSpin program, version 2.1 (Bruker
our attention was focused on maximin family peptides. BioSpin, Germany).
Maximin 3 (GIGGKILSGLKTALKGAAKELASTYLH) is a
27-residue AMP isolated from the skin secretions (Lai et al.
2002) and the brain of the Chinese red-belly toad Bombina
maxima (Liu et al. 2011). Three families of peptides were Structure calculation
isolated from this amphibian: maximins, maximins H (Lai
et al. 2002) and maximins S (Wang et al. 2005). Maximin 3 Spectral analysis and NOESY peak integration were
shows little haemolytic activity and possesses antimicrobial, accomplished with NMRFAM-SPARKY software, ver-
antitumor and spermicidal activities like the other maximins, sion 3.131 (Lee et al. 2015). Through the calibration of
but a higher anti-HIV activity, making it an interesting anti- nuclear Overhauser effect intensity (nOe) versus distance
microbial peptide for further study (Lai et al. 2002). In this bond routine of backbone, methyl proton and flexible side
report, structural properties of maximin 3 have been studied chain classes, distance constraints were obtained from
using NMR spectroscopy and molecular modeling.

13
European Biophysics Journal

NOESY peak volume integrals using CALIBA (Güntert, steps, followed by another 24,000 steps without restraints.
Braun and Wüthrich 1991) labeling protons that could not Finally, the system was simulated for 81 ns.
be stereospecifically assigned as pseudoatoms.
Using the distance constraints obtained that do not
account for fixed and limit-violating distances and weight-
ing them using 1 kJ m ­ ol−1 Å−2 as force constants (­ kNOE), Results and discussion
100 structures were randomly generated using the CYANA
program (Güntert and Buchner 2015). These model struc- Conformational analysis by NMR
tures were further subjected to 20,000 steps of simulated
annealing and 20,000 of conjugated gradient minimization. The structure of maximin 3 was determined using 2D
The final step involved 2000 cycles of conjugated gradi- NMR spectroscopy in TFE-d3–water, which is known to
ent energy minimization on the 20 peptide structures with induce formation of secondary structure by enhancing the
lowest target function values, while restraining backbone α-helical character (Shiraki et al. 1995). The collected
atoms with CHARMM22 force field (MacKerell et al. 2004) spectra showed well-dispersed peaks, indicating the pres-
in NAMD, version 2.12 (Phillips et al. 2005). The struc- ence of an ordered secondary structure. The TOCSY and
1
tural analysis of the energy-minimized structures was con- H–13C HSQC spectra were used to identify the individual
ducted using VMD (visual molecular dynamics), version residue spin systems. Beginning with the peptide’s three
1.9.3 (Humphrey et al. 1996). PROCHECK (Laskowski et al. unique residues, E20, Y25 and H27, sequence-specific
1993) and wwPDB (Berman et al. 2003) were used to deter- resonance assignments were carried out using the ­HN–HN
mine the stereochemical quality and the structural statistics and the ­H N–H α regions (Figs. 1 and 2, respectively) of
of the 20 final structural models. the NOESY spectrum, resulting in the characterization of
all the individual spin systems. The chemical shift of the
amide proton could not be found for G1 because of chemi-
cal exchange.
Molecular dynamics simulation Table 1 lists all the 1H chemical shifts of maximin 3.
The intermolecular nOe connectivities are clearly identi-
A peptide–SDS micelle system was constructed using VMD fied in Fig. 3, with line thickness proportional to their
(Humphrey et al. 1996) by aligning the centers of mass of intensity. There are many d­ αN(i, i + 3), ­dαβ(i, i + 3), ­dαN(i,
the medoid maximin 3 structure and the SDS micelle. Coor- i + 4) and ­d αβ(i, i + 4) connectivities throughout, which
dinates of the SDS micelle were obtained from Jakobtorwei- are typical of an α-helical secondary structure, and some
hen et al. (Jakobtorweihen et al. 2013). The system was then ­dαN(i, i + 2) connectivities in the N-terminal tail region,
solvated with TIP3P water (Jorgensen et al. 1983). Finally, which may be suggestive of a tighter helix. There is a
chloride ions were added to neutralize the system. notable lack of d­ αN(i, i + 3) and d­ αN(i, i + 4) connectivities
The system was minimized and simulated using the spanning G16, which indicates the presence of a kink at
CHARMM36 all-atom force field (MacKerell et al. 1998, that position (Fig. 4).
2004) within NAMD version 2.12 (Phillips et al. 2005). All
the calculations took place in the NPT ensemble, utilizing
the Langevin piston Nose–Hoover method (Martyna et al.
1994; Feller et al. 1995) with periodic boundary conditions. Molecular modeling
Bonds to hydrogen atoms were constrained to a fixed value
with the SHAKE algorithm (Ryckaert et al. 1977). The inte- NOE cross-peak intensities were converted into distance
gration time step was set to 2 fs. Long-range non-bonded restraints, from which 100 random structures were obtained,
interactions were calculated up to a switching distance of 8.5 of which the 20 with the lowest target function values were
Å, beyond which a smooth switching function truncated the further energy-minimized. The structural statistics of the cal-
energy to a cutoff of 11 Å. The computation of long-range culated models are summarized in Table 2. As illustrated in
electrostatic interactions was performed for every step by the Figs. 5 and 6, maximin 3 possesses two α-helical segments,
PME method (Darden et al. 1993), while the non-bonded list separated by a kink at G16. Helix I extends from G1–K15,
was updated every step. and helix II extends from G16–A22, with S23 incorporated
The system was minimized for 2000 conjugate gradi- into the helix in some of the structures. Helix I displays
ent steps with peptide backbone atoms fixed, and 2000 some curvature, centerd on G9, and is more ordered than
steps with the α-carbons restrained. The system was helix II (G16–A22), with a smaller RMSD for both the back-
heated to 310  K over 6000 steps and then the volume bone and the heavy atoms. The C-terminal tail region shows
equilibrated with the Langevin piston at 1 atm for 24,000 disordered character.

13
European Biophysics Journal

Fig. 1  Amide region of the


200 ms NOESY spectrum
of maximin 3 in 50% TFE-
d3–H2O mixed solvent system
with ­dNN(i,i + 1) connectivities
labeled (TOPSPIN)

Fig. 2  Fingerprint region of the


200 ms NOESY spectrum of
maximin 3 in 50% TFE-d3–H2O
mixed solvent system with
backbone connectivities labeled
(TOPSPIN)

13
European Biophysics Journal

Table 1  1H chemical shifts Amino acid NH Hα Hβ Other protons


(ppm) identified for every
residue of maximin 3 in 50% Gly1 3.979, 4.006
TFE-d3/H2O
Ile2 8.505 4.187 1.920 γ1 1.285, 1.518; γ2 0.974; δ 0.917
Gly3 8.461 3.864, 3.919
Gly4 8.163 3.899
Lys5 7.967 4.159 1.977 γ 1.476, 1.554; δ 1.705
Ile6 7.955 3.859 2.000 γ1 1.215; γ2 0.934; δ 0.873
Leu7 8.170 4.169 1.739, 1.787 γ 1.644; δ 0.903
Ser8 8.046 4.198 3.998, 4.037
Gly9 8.110 3.927
Leu10 8.297 4.195 1.746, 1.817 γ 1.554; δ 0.877
Lys11 8.326 3.967 1.912, 1.971 γ 1.451; δ 1.705; ε 2.931
Thr12 7.856 4.396 3.942
Ala13 8.145 4.172 1.554
Leu14 8.470 4.164 1.891 γ 1.573; δ 0.871
Lys15 7.993 4.089 1.946; 1.999 γ 1.495; δ 1.675, 1.732; ε 2.976
Gly16 8.122 3.910
Ala17 8.138 4.211 1.525
Ala18 8.198 4.082 1.529
Lys19 7.894 4.069 1.976 γ 1.470, 1.577; δ 1.721; ε 2.991
Glu20 8.062 4.124 2.212, 2.249 γ 2.491
Leu21 8.462 4.166 1.863 γ 1.663, 1.770; δ 0.914
Ala22 8.308 4.149 1.524
Ser23 7.945 4.315 4.013, 4.052
Thr24 7.741 4.168 4.175 γ 1.108
Tyr25 7.788 4.562 2.975, 3.130 δ1 7.164; δ2 6.792; ε1 7.149; ε2 6.777
Leu26 7.817 4.324 1.537, 1.696 δ 1.619; γ 0.866, 0.904
His27 7.703 4.547 3.202, 3.316 δ2 7.225; ε1 8.499

Δδ values are found for shorter hydrogen bonds and vice


versa. The amide proton chemical shifts have the expected
periodicity of 3–4 (Kuntz et al. 1991) and, as expected,
the most negative Δδ values are those of hydrophilic resi-
dues, K5, S8, T12, K15 and K19, and the most positive
Δδ values are the ones of hydrophobic residues, I6, L7,
L10, L14, A18, L21 and A22 (Zhou et al. 1992). K11,
which has a higher than expected chemical shift, is an
exception to this trend, meaning that the amide proton of
K11 has a shorter-than-expected hydrogen bond with the
Fig. 3  Short and medium range connectivities for maximin 3 in 50% carbonyl group of S7. This finding is consistent with there
TFE-d3–H2O (CYANA) being a region of curvature centerd on G9, thereby bring-
ing the S7 and K11 residues closer together. The hydrogen
bond lengths in the C-terminal tail region (T24-H27) are
Chemical shift analysis outside those that would be typical of an α-helix. This is
confirmed by the investigation of the torsion angles found
The chemical shift deviation of ­HN protons was calculated in the calculated structural models; the (φ,ψ) angles do
(Fig. 4a) as the difference between the observed chemical not conform to those expected of residues in an α-helix.
shift and the random coil chemical shift (Δδ=δ obs-δ rc). The data presented in this study demonstrate that maxi-
Amide proton chemical shifts are connected with hydrogen min 3 possesses an α-helical structure between residues
bond length (Wagner et al. 1983) shown in Fig. 4b: higher G1–A22/S23, while the C-terminal tail residues are in
a coil conformation. Similar patterns were observed for

13
European Biophysics Journal

Table 2  Average structural statistics of the 20 structural models of other natural peptides, including GO 1–1 (Suh et al. 1996),
maximin 3 maximin 4 (Toke et al. 2011) and the lasioglossin series of
NOEs peptides (Čeřovský et al. 2009) (Fig. 5).

Intraresidue 165
Interresidue, sequential 140
Molecular dynamics
Interresidue, non-sequential 111
Ensemble RMSD values, Å, SA ± SDa
The NMR-determined structural model of maximin 3 was
Backbone
used for a molecular dynamics simulation in SDS micelles
 All residues 0.465 ± 0.140
to model its orientation in and interactions with the bacterial
 Helix I (G1–K15) 0.356 ± 0.162
membrane-mimicking environment. During the simulation,
 Helix II (G16–A22) 0.474 ± 0.191
the peptide was found to adopt a completely α-helical con-
 Tail (T24–H27) 0.690 ± 0.205
formation, from G1 to H27. The adoption of a more ordered
Heavy atoms
secondary structure, especially in the C-terminal tail region,
 All residues 1.030 ± 0.192
can be accounted for by the electrostatic interactions present
 Helix I (G1–K15) 0.781 ± 0.208
in SDS complementing the hydrophobic forces that are also
 Helix II (G16–A22) 0.935 ± 0.341
present in TFE (Roland Montserret et al. 2000).
 Tail (T24–H27) 1.573 ± 0.266
Interestingly, in the micellar environment, the peptide
Ramachandran plot a­ nalysisb
exhibits marked flexibility around G9 and G16, at times dis-
 Residues in most favored regions 85.49%
playing a curvature there. The glycine-induced flexibility is
 Residues in additionally allowed regions 14.51%
likely responsible for the peptide’s low haemolytic activity;
 Residues in generously allowed regions 0%
Gly to Ala substitutions which reduce conformational flex-
 Residues in disallowed regions 0%
ibility and promote α-helical character have previously been
Average energies (kcal/mol)
found to increase peptides’ non-specific interactions with
 Ebond 15.811
zwitterionic cell membranes and potentially increase toxicity
 Eangle 64.693
against eukaryotic cells (Idiong et al. 2011).
 Eimprp 4.096
During the simulation, the peptide moved from its initial
 EVdW − 49.921
position at the center of the micelle to a pose parallel to
 Edihed 69.485
the micelle surface, as illustrated in Fig. 6. The amphip-
 Etotal 4.692
athic nature of the peptide allows the side chains of all the
a
 RMSD values from VMD non-polar residues to be involved in hydrophobic interac-
b
 Based on PROCHECK tions with the micelle interior, while the hydrophilic amino
acids face the solvent, and the positively charged lysine and

Fig. 4  a Amide chemical shift deviation plot of maximin 3 in 50% bond distance as calculated from the chemical shift deviations (Δδ =
TFE-d3–H2O. The observed H ­ N chemical shift was compared with 19.2dN−3–2.3) (Wagner et al. 1983) (ORIGIN) 
the random coil chemical shift standards (Δδ=δobs-δrc). b Hydrogen

13
European Biophysics Journal

histidine residues interact with the negatively charged sulfate


headgroups.

Structural comparison between maximin 3


and maximin 4

Maximin 4 is the only other maximin whose structure has


been determined (Toke et al. 2011), and the only maximin
which has been investigated for potential clinical applica-
tions, namely by incorporation into antibacterial hydrogels
(Laverty et al. 2012). It has a helix–kink–helix configuration,
both in methanol and in negatively charged SDS micelles,
with an interhelical angle of 70–90°. While maximin 3 also
possesses a kink, the interhelical angle is comparatively
minimal at 6°. Toke and co-workers attribute part of the
stabilization of the maximin 4 kink to hydrophobic contacts
between the residues A12, A13 on helix I and L17 on helix
II. This stabilization is not present in maximin 3 as it pos-
sesses a polar residue, threonine, in position 12 and a residue
with a shorter side chain, alanine, in position 17.
It was also suggested that if maximin 4 was a linear, rigid
α-helix, there would not be a well-defined polar face. The
presence of the hinge allows the formation of two units:
one more hydrophobic and the other more polar. Maximin
3, on the other hand, possesses a well-defined distinction
Fig. 5  a Solution structure of maximin 3 in 50% TFE-d3–H2O show- between the polar and hydrophobic faces, which allows it
ing the best fit superposition of 20 backbone conformations. b Rib- to lie parallel to the negatively charged SDS micelle surface
bon representation of the medoid solution conformation showing the with a comparatively minimal interhelical angle. Meanwhile,
amphipathic helical motif, with the hydrophobic and polar faces on
the left and right, respectively (VMD)  the dynamic N-terminal helix of maximin 4 was proposed
to insert into the hydrophobic core of the membrane, while
the more polar C-terminal helix was found to assume a tilted
orientation and be in contact with the lipid headgroups. The
mechanism of membrane disruption was, thus, proposed to
be funnel-like, with pores formed by peptides aligned this
way (Toke et al. 2011). The peptides’ different orientation in
prokaryotic cell membranes may be explained by the helix
I of maximin 3 having a larger polar angle, 116°, compared
to 45° in maximin 4—a difference caused by maximin 3
having the polar residues lysine and threonine at positions 5
and 12, respectively.

Biology

An array of other peptides in the maximin family has been


characterized, although none have had their three-dimen-
Fig. 6  Final pose of maximin 3 in a simulated SDS micelle. Lysine sional structures determined. There are two features that dif-
and histidine side residues are shown interacting with the negatively ferentiate maximin 3 from most other peptides of the maxi-
charged sulfate headgroups. Positively charged residues are colored min family: it terminates with a histidine amino acid and
in blue, negatively charged residues colored in red, polar residues not with an asparagine or a glutamine, and is not amidated
colored in purple, glycine residues colored in orange and hydrophobic
residues colored in gray (VMD)  in the C-terminal.

13
European Biophysics Journal

The histidine termination of maximin 3 has been pro-


posed to be correlated to its anti-HIV activity, found only
for this maximin (Lai et al. 2002). Other maximin peptides
terminating with a histidine have been discovered since then
in the brain of the same amphibian (Liu et al. 2011), but
they have not been tested for anti-HIV activity. While no
direct correlation had been found between amidation and
efficiency/selectivity of AMPs, it was demonstrated that
peptides with a low hydrophobicity gradient, that lay paral- Fig. 7  Multiple sequence alignment of selected maximin peptides and
magainin 2 (CLUSTAL)
lel to the membrane surface and disrupt it by the carpet-like
mechanism, increase their toxicity when their C termini are
amidated (Dennison et al. 2009). positions in both peptides, E19 for magainin 2 and E23
for maximin 3. As for the secondary structure, they both
are α-helices apart from the termini (Gesell et al. 1997).
Maximin family residue substitution analysis Magainin 2 is known for dimerizing in an antiparallel ori-
entation on binding to phospholipid bilayers (Wakamatsu
Maximin 32 differs from maximin 3 by only one amino acid; et al. 2002). The dimers are assumed to be stabilized by
it has a glycine in position 8 instead of serine. This substitu- dipole–dipole interactions, aromatic–aromatic interactions
tion results in a decrease in activity against Gram-positive between the side chains of F5 and F12/F16 of the other helix,
and Gram-negative bacteria, and yeast (Lai et al. 2002; Liu and electrostatic interactions between E19 and positively
et al. 2011). The change can be attributed to either glycine’s charged residues found near the N terminus (Wakamatsu
property of being a helix breaker, or a reduction in the polar- et al. 2002). Considering the similarities found between
ity of the polar face. Two substitutions in maximin 32, K11R these peptides, maximin 3 could also dimerize, aided by
and S23A, make maximin 45 as active against bacteria as ionic or π-cation interactions between Glu/Tyr and positively
maximin 3. The restored activity of maximin 45 could come charged residues either at or near the N or C terminus.
either from R11 or A23. Arginine and lysine side chains are In conclusion, this study is the first to determine the
similarly long and charged, while alanine and serine have three-dimensional structure of maximin 3 and underline the
different hydrophobicity. There are two cases in the maximin importance of its α-helical structure and amphipathicity for
family in which there is a single substitution at position 23, the bioactivity of this peptide. It was found that the peptide
S23F; this leads from maximin 5 to maximin 42 and from adopts a helix–kink–helix conformation, followed by a dis-
maximin 28 to maximin 39. In both cases, the substitution ordered C-terminal tail. Elucidation of the solution structure
of serine with phenylalanine results in reduced antibacterial of maximin 3 will facilitate further studies to determine its
activity, which indicates the importance of having a polar mechanism of action, while the residue substitution analysis
residue at this position. may lead to design of novel analogs containing additional
amino acid substitutions with improved biological activity
and therapeutic indices.
Comparison of maximin 3 and magainin 2
Acknowledgements  CH is grateful to John O’Brien and Manuel
Ruether at Trinity College Dublin for NMR facilities, University Col-
One of the most studied antimicrobial peptides is magainin lege Dublin for a Research Scholarship to PBT, and ICHEC for access
2. It can be found in the skin secretion of the African clawed to supercomputer facilities. SB and PBT are joint first authors of this
frog Xenopus laevis, and like maximin 3, it has a net charge work. The solution structure of maximin 3 was deposited to the PDB
of +3. It has antibacterial, antiviral, antifungal, antipara- at the RSCB with deposition code 6HZ2.
sitic, spermicidal, antimalarial, and antitumor activities and
a selective toxicity (Zasloff 1987; Wojcik et al. 2000; Albiol
Matanic and Castilla 2004; Lehmann et al. 2006; Mor 2009;
Sinha et al. 2016). Comparing the antimicrobial activity References
against the same microorganisms as maximin 3, the latter
Albiol Matanic VC, Castilla V (2004) Antiviral activity of antimi-
has lower MICs, while both show little haemolytic activity. crobial cationic peptides against Junin virus and herpes sim-
Comparing the peptides’ primary structures, the first plex virus. Int J Antimicrob Agents 23:382–389. https​://doi.
apparent difference is the length: magainin 2 has 23 amino org/10.1016/j.ijant​imica​g.2003.07.022
acids, making it 4 residues shorter than maximin 3. Align- Avila EE (2017) Functions of antimicrobial peptides in vertebrates.
Curr Protein Pept Sci. https​://doi.org/10.2174/13892​03717​66616​
ing the two primary structures (Fig. 7), the similarity is low, 08131​62629​
but it is pertinent to note that glutamate occurs at equivalent

13
European Biophysics Journal

Bax A, Davis DG (1985) MLEV-17-based two-dimensional homonu- John BK, Plant D, Webb P, Hurd RE (1992) Effective combina-
clear magnetization transfer spectroscopy. J Magn Reson 65:355– tion of gradients and crafted RF pulses for water suppression
360. https​://doi.org/10.1016/0022-2364(85)90018​-6 in biological samples. J Magn Reson 98:200–206. https​://doi.
Berman H, Henrick K, Nakamura H (2003) Announcing the worldwide org/10.1016/0022-2364(92)90125​-Q
Protein Data Bank. Nat Struct Mol Biol 10:980–980. https​://doi. Jorgensen WL, Chandrasekhar J, Madura JD et al (1983) Comparison
org/10.1038/nsb12​03-980 of simple potential functions for simulating liquid water. J Chem
Beveridge TJ (1999) Structures of gram-negative cell walls and their Phys 79:926–935. https​://doi.org/10.1063/1.44586​9
derived membrane vesicles. J Bacteriol 181:4725–4733 Kumar A, Ernst RR, Wüthrich K (1980) A two-dimensional nuclear
Brogden KA (2005) Antimicrobial peptides: pore formers or metabolic overhauser enhancement (2D NOE) experiment for the elucidation
inhibitors in bacteria? Nat. Rev. Microbiol. 3:238–250 of complete proton-proton cross-relaxation networks in biological
Čeřovský V, Buděšínsky M, Hovorka O et al (2009) Lasioglossins: macromolecules. Biochem Biophys Res Commun 95:1–6. https:​ //
three novel antimicrobial peptides from the venom of the eusocial doi.org/10.1016/0006-291X(80)90695​-6
bee Lasioglossum laticeps (Hymenoptera: Halictidae). ChemBio- Kuntz ID, Kosen PA, Craig EC (1991) Amide chemical shifts in many
Chem 10:2089–2099. https​://doi.org/10.1002/cbic.20090​0133 helices in peptides and proteins are periodic. J Am Chem Soc
da Silva Pereira L, do Nascimento VV, de Fátima Ferreira Ribeiro S, 113:1406–1408. https​://doi.org/10.1021/ja000​04a05​0
et al (2018) Characterization of Capsicum annuum L. leaf and Lai R, Zheng YT, Shen JH et al (2002) Antimicrobial peptides from
root antimicrobial peptides: antimicrobial activity against phy- skin secretions of Chinese red belly toad Bombina maxima. Pep-
topathogenic microorganisms. Acta Physiol Plant 40:107. https​ tides 23:427–435. https​://doi.org/10.1016/S0196​-9781(01)00641​
://doi.org/10.1007/s1173​8-018-2685-9 -6
Darden T, York D, Pedersen L (1993) Particle mesh Ewald: An N Laskowski RA, MacArthur MW, Moss DS et al (1993) PROCHECK:
⋅log( N ) method for Ewald sums in large systems. J Chem Phys a program to check the stereochemical quality of protein struc-
98:10089–10092. https​://doi.org/10.1063/1.46439​7 tures. J Appl Crystallogr 26:283–291. https​://doi.org/10.1107/
Dennison SR, Harris F, Bhatt T et al (2009) The effect of C-terminal S0021​88989​20099​44
amidation on the efficacy and selectivity of antimicrobial and Laverty G, Gorman SP, Gilmore BF (2012) Antimicrobial peptide
anticancer peptides. Mol Cell Biochem 332:43–50. https​://doi. incorporated poly(2-hydroxyethyl methacrylate) hydrogels for the
org/10.1007/s1101​0-009-0172-8 prevention of Staphylococcus epidermidis-associated biomaterial
Deslouches B, Di YP, Deslouches B et al (2017) Antimicrobial pep- infections. J Biomed Mater Res Part A 100A:1803–1814. https​://
tides with selective antitumor mechanisms: prospect for anticancer doi.org/10.1002/jbm.a.34132​
applications. Oncotarget 8:46635–46651. https:​ //doi.org/10.18632​ Lee W, Tonelli M, Markley JL (2015) NMRFAM-SPARKY: Enhanced
/oncot​arget​.16743​ software for biomolecular NMR spectroscopy. Bioinformatics
Feller SE, Zhang Y, Pastor RW, Brooks BR (1995) Constant pressure 31:1325–1327. https​://doi.org/10.1093/bioin​forma​tics/btu83​0
molecular dynamics simulation: the Langevin piston method. J Lehmann J, Retz M, Sidhu SS et al (2006) Antitumor activity of the
Chem Phys 103:4613–4621. https​://doi.org/10.1063/1.47064​8 antimicrobial peptide magainin II against bladder cancer cell
Gesell J, Zasloff M, Opella SJ (1997) Two-dimensional 1H NMR lines. Eur Urol 50:141–147. https​://doi.org/10.1016/J.EURUR​
experiments show that the 23-residue magainin antibiotic peptide O.2005.12.043
is an α-helix in dodecylphosphocholine micelles, sodium dode- Liu R, Liu H, Ma Y et al (2011) There are abundant antimicrobial
cylsulfate micelles, and trifluoroethanol/water solution. J Biomol peptides in brains of two kinds of Bombina Toads. J Proteome Res
NMR 9:127–135. https​://doi.org/10.1023/A:10186​98002​314 10:1806–1815. https​://doi.org/10.1021/pr101​285n
Gordon YJ, Romanowski EG, McDermott AM (2005) A review of anti- MacKerell AD, Bashford D, Bellott M et al (1998) All-atom empirical
microbial peptides and their therapeutic potential as anti-infective potential for molecular modeling and dynamics studies of pro-
drugs. Curr Eye Res 30:505–515. https​://doi.org/10.1080/02713​ teins. J Phys Chem B 102:3586–3616. https​://doi.org/10.1021/
68059​09686​37 jp973​084f
Güntert P, Buchner L (2015) Combined automated NOE assignment MacKerell AD, Feig M, Brooks CL (2004) Extending the treatment
and structure calculation with CYANA. J Biomol NMR 62:453– of backbone energetics in protein force fields: limitations of gas-
471. https​://doi.org/10.1007/s1085​8-015-9924-9 phase quantum mechanics in reproducing protein conformational
Güntert P, Braun W, Wüthrich K (1991) Efficient computation of three- distributions in molecular dynamics simulation. J Comput Chem
dimensional protein structures in solution from nuclear magnetic 25:1400–1415. https​://doi.org/10.1002/jcc.20065​
resonance data using the program DIANA and the supporting Maróti Gergely G, Kereszt A, Kondorosi É, Mergaert P (2011) Natural
programs CALIBA, HABAS and GLOMSA. J Mol Biol 217:517– roles of antimicrobial peptides in microbes, plants and animals.
530. https​://doi.org/10.1016/0022-2836(91)90754​-T Res Microbiol 162:363–374. https​://doi.org/10.1016/j.resmi​
Hsiao Y-W, Hedström M, Losasso V et al (2018) Cooperative modes of c.2011.02.005
action of antimicrobial peptides characterized with atomistic sim- Martyna GJ, Tobias DJ, Klein ML (1994) Constant pressure molecular
ulations: a study on cecropin B. J Phys Chem B 122:5908–5921. dynamics algorithms. J Chem Phys 101:4177–4189. https​://doi.
https​://doi.org/10.1021/acs.jpcb.8b019​57 org/10.1063/1.46746​8
Humphrey W, Dalke A, Schulten K (1996) VMD: Visual molecular Montserret R, McLeish MJ, Böckmann A, et al (2000) Involvement
dynamics. J Mol Graph 14:33–38. https​://doi.org/10.1016/0263- of electrostatic interactions in the mechanism of peptide folding
7855(96)00018​-5 induced by sodium dodecyl sulfate binding. Biochemistry. https​
Idiong G, Won A, Ruscito A et al (2011) Investigating the effect of a ://doi.org/10.1021/BI000​208X
single glycine to alanine substitution on interactions of antimi- Mor A (2009) Multifunctional host defense peptides: antipara-
crobial peptide latarcin 2a with a lipid membrane. Eur Biophys J sitic activities. FEBS J 276:6474–6482. https​://doi.org/10.111
40:1087–1100. https​://doi.org/10.1007/s0024​9-011-0726-z 1/j.1742-4658.2009.07358​.x
Jakobtorweihen S, Ingram T, Smirnova I (2013) Combination of COS- Nouri-Sorkhabi MH, Wright LC, Sullivan DR, Kuchel PW (1996)
MOmic and molecular dynamics simulations for the calculation of Quantitative31P nuclear magnetic resonance analysis of the
membrane-water partition coefficients. J Comput Chem 34:1332– phospholipids of erythrocyte membranes using detergent. Lipids
1340. https​://doi.org/10.1002/jcc.23262​ 31:765–770. https​://doi.org/10.1007/BF025​22893​

13
European Biophysics Journal

Pace BT, Lackner AA, Porter E, Pahar B (2017) The role of defensins Verkleij A, Zwaal RF, Roelofsen B et al (1973) The asymmetric dis-
in HIV pathogenesis. Mediat Inflamm 2017:1–12. https​://doi. tribution of phospholipids in the human red cell membrane. A
org/10.1155/2017/51869​04 combined study using phospholipases and freeze-etch electron
Perron GG, Zasloff M, Bell G (2006) Experimental evolution of resist- microscopy. Biochim Biophys Acta Biomembr 323:178–193.
ance to an antimicrobial peptide. Proc Biol Sci 273:251–256. https​ https​://doi.org/10.1016/0005-2736(73)90143​-0
://doi.org/10.1098/rspb.2005.3301 Wagner G, Pardi A, Wuethrich K (1983) Hydrogen bond length
Phillips JC, Braun R, Wang W et al (2005) Scalable molecular dynam- and proton NMR chemical shifts in proteins. J Am Chem Soc
ics with NAMD. J Comput Chem 26:1781–1802. https​://doi. 105:5948–5949. https​://doi.org/10.1021/ja003​56a05​6
org/10.1002/jcc.20289​ Wakamatsu K, Takeda A, Tachi T, Matsuzaki K (2002) Dimer struc-
Ryckaert J-P, Ciccotti G, Berendsen HJ (1977) Numerical integration ture of magainin 2 bound to phospholipid vesicles. Biopolymers
of the cartesian equations of motion of a system with constraints: 64:314–327. https​://doi.org/10.1002/bip.10198​
molecular dynamics of n-alkanes. J Comput Phys 23:327–341. Wang T, Zhang J, Shen JH et al (2005) Maximins S, a novel group
https​://doi.org/10.1016/0021-9991(77)90098​-5 of antimicrobial peptides from toad Bombina maxima. Biochem
El Samak M, Solyman SM, Hanora A (2018) Antimicrobial activity of Biophys Res Commun 327:945–951. https​://doi.org/10.1016/j.
bacteria isolated from Red Sea marine invertebrates. Biotechnol bbrc.2004.12.094
Rep. https​://doi.org/10.1016/J.BTRE.2018.E0027​5 Weidenmaier C, Peschel A (2008) Teichoic acids and related cell-wall
Sani M-A, Henriques ST, Weber D, Separovic F (2015) Bacteria may glycopolymers in Gram-positive physiology and host interac-
cope differently from similar membrane damage caused by the tions. Nat Rev Microbiol 6:276–287. https​://doi.org/10.1038/
Australian tree frog antimicrobial peptide maculatin 1.1. J Biol nrmic​ro186​1
Chem 290:19853–19862. https:​ //doi.org/10.1074/jbc.M115.64326​ Wojcik C, Sawicki W, Marianowski P et  al (2000) Cyclodextrin
2 enhances spermicidal effects of magainin-2-amide. Contracep-
Shai Y (2002) Mode of action of membrane active antimicrobial pep- tion 62:99–103. https​://doi.org/10.1016/S0010​-7824(00)00143​-8
tides. Biopolym Pept Sci Sect 66:236–248 Xia X, Cheng L, Zhang S et al (2018) The role of natural antimicro-
Shiraki K, Nishikawa K, Goto Y (1995) Trifluoroethanol-induced sta- bial peptides during infection and chronic inflammation. Antonie
bilization of the α-helical structure of β-lactoglobulin: implica- Van Leeuwenhoek 111:5–26. https​: //doi.org/10.1007/s1048​
tion for non-hierarchical protein folding. J Mol Biol 245:180–194. 2-017-0929-0
https​://doi.org/10.1006/JMBI.1994.0015 Xie J, Zhao Q, Li S et al (2017) Novel antimicrobial peptide CPF-C1
Sinha S, Singh A, Medhi B, Sehgal R (2016) Systematic review: insight analogs with superior stabilities and activities against multidrug-
into antimalarial peptide. Int J Pept Res Ther 22:325–340. https​:// resistant bacteria. Chem Biol Drug Des 90:690–702. https​://doi.
doi.org/10.1007/s1098​9-016-9512-1 org/10.1111/cbdd.12988​
Son M, Lee Y, Hwang H et al (2013) Disruption of interactions between Yang D, Biragyn A, Kwak LW, Oppenheim JJ (2002) Mammalian
hydrophobic residues on nonpolar faces is a key determinant in defensins in immunity: more than just microbicidal. Trends Immu-
decreasing hemolysis and increasing antimicrobial activities of nol 23:291–296. https:​ //doi.org/10.1016/S1471-​ 4906(02)02246-​ 9
α-helical amphipathic peptides. ChemMedChem 8:1638–1642. Yang D, Biragyn A, Hoover DM et al (2004) Multiple roles of anti-
https​://doi.org/10.1002/cmdc.20130​0264 microbial defensins, cathelicidins, and eosinophil-derived neuro-
Subasinghage AP, Conlon JM, Hewage CM (2008) Conformational toxin in host defense. Annu Rev Immunol 22:181–215. https:​ //doi.
analysis of the broad-spectrum antibacterial peptide, ranatuerin- org/10.1146/annur​ev.immun​ol.22.01270​3.10460​3
2CSa: identification of a full length helix-turn-helix motif. Bio- Yeaman MR, Yount NY (2003) Mechanisms of antimicrobial pep-
chim Biophys Acta Proteins Proteomics 1784:924–929. https​:// tide action and resistance. Pharmacol Rev 55:27–55. https​://doi.
doi.org/10.1016/j.bbapa​p.2008.02.019 org/10.1124/pr.55.1.2
Subasinghage AP, Conlon JM, Hewage CM (2010) Development of Yin LM, Edwards MA, Li J et al (2012) Roles of hydrophobicity and
potent anti-infective agents from Silurana tropicalis: conforma- charge distribution of cationic antimicrobial peptides in peptide–
tional analysis of the amphipathic, alpha-helical antimicrobial membrane interactions. J Biol Chem 287:7738–7745. https​://doi.
peptide XT-7 and its non-haemolytic analogue [G4K]XT-7. Bio- org/10.1074/jbc.M111.30360​2
chim Biophys Acta Proteins Proteom 1804:1020–1028. https​:// Zasloff M (1987) Magainins, a class of antimicrobial peptides from
doi.org/10.1016/J.BBAPA​P.2010.01.015 Xenopus skin: isolation, characterization of two active forms, and
Subasinghage AP, O’Flynn D, Conlon JM, Hewage CM (2011) Con- partial cDNA sequence of a precursor. Proc Natl Acad Sci USA
formational and membrane interaction studies of the antimicro- 84:5449–5453. https​://doi.org/10.1073/PNAS.84.15.5449
bial peptide alyteserin-1c and its analogue [E4K]alyteserin-1c. Zelezetsky I, Tossi A (2006) Alpha-helical antimicrobial peptides-
Biochim Biophys Acta Biomembr 1808:1975–1984. https​://doi. Using a sequence template to guide structure-activity relationship
org/10.1016/j.bbame​m.2011.04.012 studies. Biochim. Biophys. Acta - Biomembr. 1758:1436–1449
Suh J-Y, Lee K-H, Chi S-W et al (1996) Unusually stable helical kink Zhou NE, Zhu BY, Sykes BD, Hodges RS (1992) Relationship between
in the antimicrobial peptide—a derivative of gaegurin. FEBS Lett amide proton chemical shifts and hydrogen bonding in amphip-
392:309–312. https​://doi.org/10.1016/0014-5793(96)00840​-X athic.alpha.-helical peptides. J Am Chem Soc 114:4320–4326.
Tanphaichitr N, Srakaew N, Alonzi R et al (2016) Potential use of https​://doi.org/10.1021/ja000​37a04​2
antimicrobial peptides as vaginal spermicides/microbicides. Phar-
maceuticals 9:13. https​://doi.org/10.3390/ph901​0013 Publisher’s Note Springer Nature remains neutral with regard to
Toke O, Bánóczi Z, Király P et al (2011) A kinked antimicrobial pep- jurisdictional claims in published maps and institutional affiliations.
tide from Bombina maxima. I. Three-dimensional structure deter-
mined by NMR in membrane-mimicking environments. Eur Bio-
phys J 40:447–462. https​://doi.org/10.1007/s0024​9-010-0657-0

13

You might also like