ArticlePDF Available

Abstract and Figures

The glucagon-like peptide 1 (GLP-1) analog, liraglutide, is a GLP-1 agonist and is used in the treatment of type-2 diabetes mellitus and obesity. From a pharmaceutical perspective, it is important to know the oligomerization state of liraglutide with respect to stability. Compared to GLP-1, liraglutide has an added fatty acid (FA) moiety that causes oligomerization of liraglutide as suggested by small-angle x-ray scattering (SAXS) and multiangle static light scattering (MALS) results. SAXS data suggested a global shape of a hollow elliptical cylinder of size hexa-, hepta-, or octamer, whereas MALS data indicate a hexamer. To elaborate further on the stability of these oligomers and the role of the FA chains, a series of molecular-dynamics simulations were carried out on 11 different hexa-, hepta-, and octameric systems. Our results indicate that interactions of the fatty acid chains contribute noticeably to the stabilization. The simulation results indicate that the heptamer with paired FA chains is the most stable oligomer when compared to the 10 other investigated structures. Theoretical SAXS curves extracted from the simulations qualitatively agree with the experimentally determined SAXS curves supporting the view that liraglutide forms heptamers in solution. In agreement with the SAXS data, the heptamer forms a water-filled oligomer of elliptical cylindrical shape. Copyright © 2015 Biophysical Society. Published by Elsevier Inc. All rights reserved.
Content may be subject to copyright.
Article
Oligomerization of a Glucagon-like Peptide 1 Analog: Bridging Experiment
and Simulations
Tine M. Frederiksen,
1
Pernille Sønderby,
1
Line A. Ryberg,
1
Pernille Harris,
1,
*Jens T. Bukrinski,
2
Anne M. Scharff-Poulsen,
2
Maria N. Elf-Lind,
1
and Gu
¨nther H. Peters
1,
*
1
Department of Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark; and
2
Novozymes A/S, Bagsværd, Denmark
ABSTRACT The glucagon-like peptide 1 (GLP-1) analog, liraglutide, is a GLP-1 agonist and is used in the treatment of type-2
diabetes mellitus and obesity. From a pharmaceutical perspective, it is important to know the oligomerization state of liraglutide
with respect to stability. Compared to GLP-1, liraglutide has an added fatty acid (FA) moiety that causes oligomerization of lir-
aglutide as suggested by small-angle x-ray scattering (SAXS) and multiangle static light scattering (MALS) results. SAXS data
suggested a global shape of a hollow elliptical cylinder of size hexa-, hepta-, or octamer, whereas MALS data indicate a hex-
amer. To elaborate further on the stability of these oligomers and the role of the FA chains, a series of molecular-dynamics sim-
ulations were carried out on 11 different hexa-, hepta-, and octameric systems. Our results indicate that interactions of the fatty
acid chains contribute noticeably to the stabilization. The simulation results indicate that the heptamer with paired FA chains is
the most stable oligomer when compared to the 10 other investigated structures. Theoretical SAXS curves extracted from the
simulations qualitatively agree with the experimentally determined SAXS curves supporting the view that liraglutide forms hep-
tamers in solution. In agreement with the SAXS data, the heptamer forms a water-filled oligomer of elliptical cylindrical shape.
INTRODUCTION
The glucagon-like peptide 1 (GLP-1) receptor is a well-es-
tablished therapeutic target for the treatment of type-2
diabetes mellitus (1–3), and extensive research has estab-
lished the physiologic roles of GLP-1 and its endogenous
receptor in regulating glucose homeostasis and energy
metabolism (4,5). GLP-1-(7-37) is a 31-amino-acid incretin
hormone secreted by the endocrine L cells in the gut wall
upon glucose intake (6), and the peptide is secreted in
response to the nutrient content of the gastrointestinal tract
and thus potentiates insulin exocytosis from pancreatic b-
cells in a glucose-dependent manner (6,7). Additionally,
GLP-1 suppresses appetite, glucagon secretion, and gastric
emptying, all of which contribute to inhibition of the post-
prandial rise in plasma glucose concentrations (8). GLP-1
is responsible for up to 60% of the postprandial insulin
response (9). Recent studies show that GLP-1 is not only
a key factor in type-2 diabetes mellitus treatment, but also
has potential in the treatment of obesity (1,10) and has
shown positive effects on neuroprotection in animal models
(11), which can potentially be used for the treatment of Par-
kinson’s and Alzheimer’s disease (12–15).
While GLP-1 is interesting for a pharmaceutical applica-
tion, it cannot be used for routine treatment, because its
biological half-life is only a few minutes (16,17). The short
insulinotropic action of GLP-1 results from the degradation
of the peptide by dipeptidyl-peptidase IV (DPP IV) and
rapid renal clearance due to its relatively small size (18–
20). DPP IV degrades GLP-1 at the N-terminus by cleaving
off the first two amino acids, generating the biologically
inactive fragment, GLP-1-(9-37) (9,21). To overcome this
shortcoming, different strategies have been used. Those
include: 1) incorporation of the peptides in injectable micro-
spheres; 2) fusion with larger carrier molecules like albumin
or fragment crystallizable region of immunoglobulin G
or polyethylene glycol; and 3) attachment of a fatty acid
(FA) directing oligomerization and promoting reversible
binding to endogenous human serum albumin (20). All three
approaches result in an increased half-life partly due to the
increased size of the drug minimizing the renal clearance
mechanism. The latter approach, for instance, has been uti-
lized in designing the GLP-1 analog: liraglutide (Victoza;
Novo Nordisk A/S, Bagsværd, Denmark) (22,23). An in-
depth understanding of the oligomerization state of liraglu-
tide is not only instrumental in the understanding of the
increased half-life observed for this molecule but also to
ensure a stable oligomeric state, because uncontrolled and
extensive oligomerization can drive fibrillation (24). The
amino acid sequence of liraglutide is shown in Fig. 1.
Compared to GLP-1, two structural modifications have
been introduced. A C-16 acyl chain (palmitoyl), is linked
to Lys
26
via a g-glutamic acid spacer, and the lysine in po-
sition 34 of the native GLP-1 sequence is exchanged with
arginine to ensure homogenous palmitoylation at position
26 (25,26). The general understanding is that the acyl chain
allows a noncovalent binding to albumin, which delays both
proteolytic inactivation by DPP IV and renal clearance,
Submitted March 3, 2015, and accepted for publication July 30, 2015.
*Correspondence: ghp@kemi.dtu.dk or ph@kemi.dtu.dk
Editor: Ivet Bahar.
Ó2015 by the Biophysical Society
0006-3495/15/09/1202/12 http://dx.doi.org/10.1016/j.bpj.2015.07.051
1202 Biophysical Journal Volume 109 September 2015 1202–1213
resulting in a biological half-life of ~13–14 h and allowing
once-daily administration (2,23). A further prolongation
may also be caused by the fatty acid chain that may steri-
cally hinder DPP IV from degrading liraglutide (5). Further-
more, studies have shown that one way to stabilize GLP-1 is
to add a clustering agent that causes the peptide to oligomer-
ize (2), thus, the FA chain in liraglutide could act as a clus-
tering agent.
Although the pharmacological efficiency of liraglutide
has been established (23,27), there is a lack of a molecular
understanding of the solution structure of liraglutide. Using
analytical ultracentrifugation, Steensgaard et al. (28) could
show that liraglutide oligomerizes in a concentration-inde-
pendent manner forming, predominately, heptamers in the
concentration range of 0.004–4.501 mg/mL. Recently,
Wang et al. (29) studied the pH dependence of the size
and secondary structure of liraglutide oligomers using
light-scattering and circular dichroism, respectively. The
authors report a transformation from an octamer to a
dodecamer at pH 6.4 and 6.9 with subsequent partial loss
of the a-helical structure of liraglutide. Furthermore, it
has been shown that the oligomerization of GLP-1 and
similar peptide analogs is dependent on the pH and ionic
strength (30), and thus different solution structures may
exist (3,31).
To get further insight in the solution structure of liraglu-
tide, we have performed a series of molecular-dynamics
(MD) simulations, and the simulation results are compared
with results from small-angle x-ray scattering (SAXS) and
multiangle static light-scattering (MALS) experiments.
MATERIALS AND METHODS
SAXS
A quantity of 3 mL commercial Victoza (contains 18 mg liraglutide (free-
base, anhydrous); Novo Nordisk A/S)) and the inactive ingredients diso-
dium phosphate dihydrate, 1.42 mg; propylene glycol, 14 mg; and phenol,
5.5 mg in aqueous solution (32) were dialyzed against 3 1 L buffer con-
taining ~0.47 mg/mL (0.376 mM) Na
2
HPO
4
2H
2
O, pH 8.1, over 3 days.
Concentration determinations were performed with the NanoDrop 1000
Spectrophotometer (Thermo Fisher Scientific, Waltham, MA) at 280 nm.
The extinction coefficient was calculated to be 6990 cm
1
M
1
with the
PROTPARAM (33) tool from ExPASy.org (34) using the primary sequence
of the protein.
SAXS measurements were performed at the MAX IV laboratories at
beamline I911-SAXS, MAX IV Laboratory, Lund, Sweden (35). The sam-
ple detector distance and the direct beam position were calibrated
using AgBe (silver behenate). Parameters are shown in Table S2 in the
Supporting Material. Measurements on pure water were used to get the
data on an absolute scale. Buffers were measured both before and
after each sample and averaged before subtraction. The sample size was
~50 mL injected manually in a flow cell. Measurements were performed
on a series of liraglutide samples at the approximate concentrations of
1, 2, and 4.7 mg/mL.
All calibrations and corrections of the SAXS data were done using the
in-house software BLI911-4 (35). Buffer averaging and subsequent subtrac-
tion before data analysis was done in the program PRIMUS (36). The
software package ATSAS, Ver. 2.4 (37), was used for further data analysis.
Evaluation of the Guinier region was performed within PRIMUS. The pair
distribution function, p(r), was evaluated using the interactive program
GNOM (38).
Asymmetric flow field flow
fractionation-UV-MALS
Asymmetric flow field flow fractionation (AF4) separation was performed
using a Dionex UltiMate 3000 autosampler and pump (Thermo Fisher
Scientific, Waltham, MA) connected to an Eclipse AF4 separation system
(Wyatt Technology Europe, Dernbach, Germany) followed by a Dionex
UltiMate 3000 RS variable wavelength UV detector (Thermo Fisher Scien-
tific) set at 280 nm, and a Dawn Heleos-II 18-angle MALS detector (Wyatt
Technology Europe). Separations were performed using a 10-kDa molecu-
lar-mass cutoff PES (polyethersulfone) membrane in a 17.5 cm separation
channel with an S-350 mm spacer. Samples were introduced to the channel
at 0.2 mL/min and subsequently focused at the head of the channel at a
focus flow rate of 1.5 mL/min. Samples were eluted over 15 min with
a channel flow rate of 1 mL/min and a cross flow gradient of 4.0–
2.5 mL/min. Undiluted Victoza (6 mg/mL liraglutide) and 10diluted Vic-
toza (diluted with eluent) were injected and eluted with 20 mM phosphate,
100 mM NaCl, and 0.05% NaN
3
, pH 8.1, filtrated through a 0.1 mm filter.
Different injection volumes of undiluted and diluted Victoza were used, and
the resulting mass loads were 6, 12, and 18 mg liraglutide. The molecular
mass of liraglutide was calculated using the software ASTRA, Ver. 6.1.2
(Wyatt Technology Europe) with dn/dc¼0.185 mL/g and UV extinction
coefficient (280 nm) ¼6990 cm
1
M
1
.
MD simulations
Several orientations and oligomers of liraglutide have been investigated.
All are based on the solution NMR structure of liraglutide (PDB: 4APD)
obtained from the Protein Data Bank (39). The coordinates from the
PDB file were copied, translated, and rotated in a circle with a radius
of 20 A
˚, corresponding to the results from SAXS experiments. This re-
sulted in several oligomers containing six, seven, or eight monomers,
respectively. A set of oligomers was created where the monomers were
oriented so that two FA chains were paired in the direction of the ellip-
tical cylinder arrangement, which hereafter will be referred to as hexa-,
hepta-, and octamer systems (Fig. 2). In the heptamer, one monomer
was oriented with the FA chain pointing outward of the elliptical
arrangement.
Furthermore, another set of oligomers were created where some of the
monomers were flipped upside down to see if interactions between the C-
and N-terminal charges would stabilize the structures. For this set of olig-
omers, the hexa- and one octamer were made with every second monomer
FIGURE 1 Amino-acid sequence of liraglutide. To see this figure in
color, go online.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1203
flipped upside down. In the case of the heptamer, only the monomer
with the FA chain pointing outward was flipped upside down. Also,
another octameric structure was prepared where every second monomer
pair was flipped upside down. These configurations are hereafter referred
to as the AA6_3ud, AA7_1ud, AA8_4udp, and AA8_4uds systems where
ud,p, and sare short for upside-down, pair, and single, respectively
(Fig. 2). To clarify the extent of the stabilizing effect of the FA chain
on the structures, another set of oligomers were created. One oligomer
is constructed according to the rotation and translation of the first
heptamer, but it does not contain any FA chains (hereafter referred to as
‘AA7_glp1’’ and represented in Fig. 2). Three other oligomers were
also created, namely, a hexa-, hepta-, and octamer where all the monomers
are rotated so that the FA chains are pointing outward of the elliptical
arrangement. These will hereafter be referred to as the AA6_FAout,
AA7_FAout, and AA8_FAout systems (Fig. 2). This gives a total of 11
oligomeric structures.
The structures were solvated using the program SOLVATE from
Grubmu
¨ller and Groll (40). Water molecules were described by the TIP3
water model (41). Next, the systems were neutralized by adding 3 Na
þ
ions per monomer. Simulations were performed at an ionic strength of
0.1 M NaCl (see details in Table 1). All simulations were performed using
the computer program NAMD (42) with the CHARMM36 force field (43).
The same simulation parameters were used as described by Madsen et al.
(44). (See the Supporting Material for a detailed description.) Analyses
of the trajectories were carried out in the software VMD (45).
Theoretical SAXS curves
The program CRYSOL (46), which is part of the program package ATSAS,
Ver. 2.6 (37), was used to compare the SAXS curves of the structures ex-
tracted from the MD simulations with the experimental measured SAXS
curve of the oligomer. CRYSOL calculates the scattering intensity based
on the atomic coordinates of the protein and adds a hydration layer simpli-
fied as a continuous outer envelope (37).
FIGURE 2 Representation of the start structures
for the 11 oligomeric structures. All structures are
shown from a top view. (Red line) Orientation of
the FA chain on each monomer; (ellipses) size
and shape of the oligomers. To see this figure in
color, go online.
TABLE 1 System and simulation details of the 11 oligomeric
systems
Structure
No. of
Atoms
No. of
Waters
No. of
NaCl
Initial Box
Size (A
˚)
Simulation
Time (ns)
Hexamer 140,203 45,615 86 113 118 115 69
Heptamer 53,774 16,665 31 83 88 89 129
Octamer 136,414 44,000 83 113 115 115 71
AA6_3ud 57,419 18,055 34 91 88 85 34
AA7_1ud 57,090 17,769 33 91 87 87 39
AA8_4udp 54,814 16,834 32 91 85 85 41
AA8_4uds 54,928 16,872 32 91 85 85 43
AA6_FAout 52,289 16,247 31 83 90 85 21
AA7_FAout 53,114 16,445 31 83 88 89 51
AA8_FAout 54,937 16,875 32 83 90 90 21
AA7_glp1 51,729 16,131 30 83 87 87 69
Biophysical Journal 109(6) 1202–1213
1204 Frederiksen et al.
RESULTS
In this section, experimental results from SAXS and AF4-
UV-MALS are presented, followed by the computational
results.
SAXS
SAXS intensity curves of liraglutide measured at different
concentrations are shown in Fig. 3. Repulsion is observed
already at 1 mg/mL, while the shape of the curve is consis-
tent over the concentration range, reflecting a similar shape
of molecule. This is also reflected in the Kratky plots shown
in Fig. S1. Corresponding pair distribution functions are
provided in Fig. S2.
Table 2 summarizes the parameters extracted from the
SAXS measurements. The radius of gyration (R
g
), maximum
particle diameter (D
max
) and I(0)/cshow a slight decrease
with concentration, as expected from the repulsive behavior.
The partial specific volume, n, used for calculating the
molecular mass is chosen to match pure protein and is set
to the average value of 0.73 cm
3
/g. To compare the experi-
mental results with the model structures extracted from the
simulations, the experimental data were extrapolated to q¼
0 to avoid interparticle repulsion.
AF4-UV-MALS
Analysis of liraglutide showed a single peak and a uniform
molecular weight across the peak in all analyses (Fig. 4,left)
indicating that repulsive behavior is negligible at the con-
centration range found in the detector. The average molecu-
lar mass across the peak was 22 kDa, which corresponds to a
hexamer assuming a monomer molecular mass of 3.7 kDa.
Undiluted Victoza (6 mg/mL liraglutide) and 10diluted
Victoza were analyzed and different injection volumes
were used, which resulted in mass loads of 6, 12, and
18 mg liraglutide. Liraglutide is diluted during analysis by
the eluate, and the resulting liraglutide concentration was
quantified in the eluate passing the UV detector (Fig. 4,
right). Peak liraglutide concentrations of 0.012, 0.024, and
0.036 mg/mL were observed.
MD simulations
Simulations were performed on several sets consisting of
hexa-, hepta-, and octamer oligomers, to study the structural
arrangement and stability of the oligomers including the
role of the FA in promoting the stability of the oligomers.
The last structures taken from the simulations are shown
in Figs. S3–S5.
Figs. S3–S5 show that although the internal structures for
all 11 oligomers are highly distorted compared to the start
structures, all of them but AA6_3ud (Fig. S4 a) maintain
a tunnel-like structure which is, however, more or less flat-
tened and resembling an elliptical shape.
The solvent-accessible surface area (SASA) of the oligo-
mers seen in Figs. 5,S6, and S7 relates to the packing of the
monomers.
Overall, the packing of the heptamers appears to be more
prominent than the octamers throughout the simulations,
indicated by the lower SASA. The AA6_3ud structure has
a significant lower packing than the AA7_1ud, AA8_4udp,
and AA8_4uds structures, which most likely is a result of a
complete opening of the elliptical structure as seen in Fig
S4 a. The relatively high SASA for the AA6_FAout olig-
omer could be due to the elongation of some of the mono-
mers, which appear to unfold from the helix structure
(Fig. S5 a).
The time evolution of the root-mean-square deviation
(RMSD) is shown for the 11 structures in Figs. 6,S8, and S9.
From the initial steep increase in RMSD, it is evident that
the oligomers rearrange to some extent within the first 6 ns.
RMSD converges for the hexamer, AA6_3ud and heptamer,
AA7_1ud, AA8_4udp, AA8_4uds, AA8_FAout, and
AA7_glp1 conformations. These structures appear to be sta-
ble when it comes to the overall movement of the systems.
The octamer, AA6_FAout and AA7_FAout, however, do not
converge. Furthermore, the three hexameric systems and
AA7_FAout present a significantly higher RMSD value
compared to the other systems.
To further monitor the movement of the monomers, the
two-dimensional positions of the a-carbon in the FA-Lys
linker (Fig. S11) for all 11 oligomer conformations, pro-
jected onto the yz plane (the monomers are translated and
rotated around the xaxis), are shown as a function of simu-
lation time in Figs. 7,8, and 9.
From Fig. 7, we can conclude that the spread of the
hexamer is larger than for the heptamer and the octamer
throughout the simulation. Furthermore, the hexamer is
squeezed to give a more flattened shape. For the octamer,
it appears that the elliptical structure is unstable because
one monomer seems to migrate from the oligomeric
FIGURE 3 Scattering curves normalized for concentration. Plots at con-
centrations 1, 2, and 4.7 mg/mL. (Inset) Scattering over entire measured
scattering range. (Lir is liraglutide.) To see this figure in color, go online.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1205
structure. In the case of the heptamer, Fig. 7 b,itis
evident that the position of the a-carbon is rather dense
throughout the simulation and that the structure resembles
an ellipse.
Fig. 8 ashows that the AA6_3ud structure flattens drasti-
cally, which corresponds to the more open structure seen in
Fig. S4 a. The AA8_4udp structure maintains a more ellip-
tical arrangement, even though one monomer seems to be
leaving the structure (indicated by the circumference). The
AA8_4uds structure is rather mobile and gains a squared
shape. The AA7_1ud structure is, like the hexamer in
Fig. 7 a, squeezed so that the elliptical structure is
destroyed.
In AA7_glp1, Fig. 9 d, monomers are more mobile than
compared with the other structures resulting in a disordered
(unstable) structure. In Fig. 9 b, the AA7_FAout structure
appears to maintain an elliptical structure, but the move-
ment of the a-carbons is very spread out. Fig. 9 cshows
that the a-carbon movement of the AA8_FAout system is
rather centered on the starting position throughout the
simulation, which indicates a stable system. However, as
seen for the octamer and AA8_4udp systems, one mono-
mer escapes from the elliptical arrangement. The AA6_FA-
out system (Fig. 9 a) moves significantly throughout
the simulation, and this movement results in a flattened
structure.
Mean energies based on the structures taken at every
50 ps throughout the simulations are given in Table 3.
The energies calculated for the oligomeric system (pep-
tide-peptide, P-P) and the oligomer-water interactions
(P-W) are normalized to the number of monomers in
each oligomer to make comparison of the different systems
possible. From these, it can be seen that the total P-P van
der Waals (vdW) energy for the heptamer is lower than
for any of the other oligomers. Considering the P-W inter-
actions, the energy for the heptamer system is less nega-
tive than found for the other systems. In general, all
the hexameric systems of liraglutide (hexamer, AA6_3ud,
AA6_FAout) are higher in P-P vdW energy, which might
indicate that these structures are less stable than the hepta-
and octameric liraglutide oligomers. The energies show
relatively large fluctuations (data not shown) which is
also reflected by the relatively large standard deviations.
This indicates that internally the 11 oligomeric systems
are flexible structures.
The analyses were done for segment-segment interac-
tions where FA chains are facing each other (S-S FA), no
FA chains are between them (S-S), one FA chain pointing
outward (S-S FA out), and all FA chains pointing outward
(S-S all FA out). See Figs. 10 and 2for illustration of S-S,
S-S FA, S-S FA out, and S-S all FA out (AA6/7/8_FAout
structures). The two last analyses were only done for the
heptamer, AA7_1ud, and for the oligomer, AA6_FAout,
AA7_FAout, and AA8_FAout because these were the
only conformations relevant for such investigation. Results
for the AA7_glp1 structure (with no Fa chains attached)
are also reported since the monomers in the AA7_glp1
structure are rotated the same way as those in the heptamer
with paired FA chains. It can be seen that for the structures
with FA pairs, the energies are significantly lower for the
TABLE 2 Parameter overview extracted from SAXS measurements
Concentration (mg/mL) R
g
Guinier (A
˚)R
g
p(r)(A
˚)D
max
(A
˚)I(0) Guinier I(0) p(r)I(0)/cMolecular Mass (kDa)
1 23.1 23.8 82.0 0.021 0.021 0.021 26
2 21.7 22.3 75.9 0.040 0.041 0.020 25
4.7 22.2 22.2 74.2 0.096 0.096 0.020 25
FIGURE 4 AF4-UV-MALS analysis of undiluted Victoza (6 mg/mL lir-
aglutide) and 10diluted Victoza (F10). Different injection volumes were
tested. All analyses showed a single peak in the UV chromatogram and a
uniform molar mass of ~22 kDa across the peak (left). No other peaks
were observed in the chromatogram. The liraglutide concentration in the
eluate passing the UV detector is shown in the right graph. To see this figure
in color, go online.
FIGURE 5 SASA as a function of time for the hexamer, heptamer, and
octamer. The area has been normalized according to the number of mono-
mers. SASA was calculated every 50 ps along the trajectory using a van der
Waals radius of 1.4 A
˚. To see this figure in color, go online.
Biophysical Journal 109(6) 1202–1213
1206 Frederiksen et al.
monomer pairs that have FA chains facing each other than
for those where no FA chains are between them. This sup-
ports the view that interactions of the FA chains contribute
to the stabilization of the oligomers of liraglutide (2,5).
DISCUSSION
The experimental SAXS curves (Fig. 3) show that even
at the lowest measured concentration of 1 mg/mL, repul-
sive interactions between liraglutide oligomers are pre-
sent, which increases with increasing concentration.
Repulsive interactions can lead to an underestimation
of the molecular weight. The SAXS data suggest that
the solution structure of liraglutide is a hexa-, hepta-,
or octamer. The uncertainty of the structure arises from
uncertainties related to the measured concentration and
the estimated partial specific volume (v) (and hence
the number of monomers in the oligomer). To our
knowledge, there is no value of nfor liraglutide avail-
able in the literature. In this study, v¼0.73 cm
3
/g (cor-
responding to an average value for pure proteins) was
used. The FA chain could contribute to an increase in
the partial specific volume, but to which extent is diffi-
cult to estimate. Using v¼0.74 cm
3
/g as also reported
by Mylonas and Svergun (47), the molecular weight in-
creases to that resembling an octamer. From the SAXS
measurements, it can only be concluded that the solution
structure of liraglutide is an oligomer of approximately
heptameric molecular size, with a consistent shape of
an elliptical cylinder, and that this oligomerization is
concentration-independent within the measured concen-
tration range.
The MALS results indicate a hexameric solution structure
of liraglutide. In contrast to the SAXS data, no repulsion in-
teractions between oligomers were observed in the concen-
tration range of 0.012–0.036 mg/mL. Note that the SAXS
data were measured in the concentration range of 1.0–
4.7 mg/mL, where 1 mg/mL corresponds to the lowest con-
centration that can be measured.
Simulation results suggest that the hexamer is an unfa-
vorable arrangement for the monomers as seen from the
structural deviation of the oligomers, given by the RMSD
(Fig. 6) and the two-dimensional plot of the a-carbon in
the FA-Lys linker (Fig. 7). However, the packing of the
structure is relatively tight with a low SASA (Fig. 5), which
indicates that more interactions are obtainable. This is also
FIGURE 7 Position of a-carbon in the FA-Lys
linker of the hexamer (a), heptamer (b), and oc-
tamer (c), as a function of simulation time. One
monomer is highlighted (circle) in the octamer
(c), to indicate the possible disintegration of this
structure. Coordinates taken from the first 15 ns,
5 ns in the middle of the simulation, and then the
last ~5 ns, are shown. (Circles and dashed lines)
Average structure within the 5-ns intervals (dots).
The yz a-carbon atom coordinates for the FA-Lys
linker were plotted in intervals of 50 ps. To see
this figure in color, go online.
FIGURE 6 RMSD of the entire oligomeric structure for the hexa-, hepta-,
and octamer. Structures were aligned to the first frame (t¼0), and devia-
tions are determined for the backbone chains. To see this figure in color,
go online.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1207
supported by the relatively low P-P energy of the hexameric
system compared to the others (Table 3). The heptamer
seems to present a very favorable arrangement with a tight
packing as indicated by the low SASA (Fig. 5), little
displacement given by the low and converging RMSD
(Fig. 6), and the overall elliptical shape seen in the two-
dimensional plot of the a-carbon in the FA-Lys linker
(Fig. 7). This is also the structure with the overall lowest
P-P energy of ~200 kcal/mol. The octamer, like the hex-
amer, also appears to present an unfavorable arrangement
with a slight opening and disintegration of the elliptical
structure as seen in the end structure (Fig. S3), the two-
dimensional plot of the a-carbon (Fig. 7), and the high
SASA (Fig. 5). The AA6_3ud presents the worst monomer
arrangement out of the 11 investigated structures based on
the end structure (Fig. S4) that opens completely, resulting
in an unstable conformation as also seen from the relatively
large SASA (Fig. S6), increasing RMSD (Fig. S8), and the
scattered a-carbon position (Fig. 8). The AA7_1ud,
AA8_4udp, and AA8_4uds systems all maintain a relatively
well-arranged end structure (Fig. S4), even though one
monomer appears to be leaving the general elliptical struc-
ture of AA8_4up. In contrast to this, the overall packing of
the three systems is relatively tight (Fig. S6), but the overall
movement is scattered and very spread out as seen in the
two-dimensional plot of the a-carbon (Fig. 8). Thus, it
seems like interactions between the C- and N-termini are
not contributing to the energy, hence, liraglutide is most
likely not to arrange like the flipped structures (AA6_3ud,
AA7_1ud, AA8_4udp, and AA8_4uds). The AA6/7/8_FA-
out structures also present unfavorable arrangements of
the monomers, as can be seen by the large scattering
of the individual monomers in the two-dimensional plot
of the a-carbon (Fig. 9) and large overall displacement rep-
resented by the RMSD (Fig. S9).
The most important analysis result for these structures is,
however, that the S-S all FAout energies for all three
systems are less negative than those of the segment inter-
actions in systems where the FA chains are pointing toward
each other (Table 3). This amplifies the hypothesis of
liraglutide oligomer structures that give rise to FA inter-
actions. However, when the FA chains are pointing out-
ward they could, in theory, wrap around the elliptical
structure in such a manner that they interact. This appears
not to be the case, based on the energy calculations (Table
3) and the fact that all the FA chains in all the three
FIGURE 8 Position of a-carbon in the FA-Lys linker of the AA6_3ud (a), AA7_1ud (b), AA8_4udp (c), and AA8_4uds (d) as a function of simulation
time. One monomer is highlighted in a circumference in AA8_4udp (c), to indicate the possible disintegration of the AA8_4udpsystem. See Fig. 7 legend for
details. To see this figure in color, go online.
Biophysical Journal 109(6) 1202–1213
1208 Frederiksen et al.
structures seem to be randomly laying on the surface of
the oligomers (Fig. S5). This arrangement could promote
clustering of oligomers. However, this is not the case
because SAXS and MALS data indicate the presence of
one defined oligomeric species. The AA7_glp1 was made
as a reference structure when considering the role of the
FA chains. The results show that the movement of the indi-
vidual monomers of this system (Fig. 9) is rather large. This
FIGURE 9 Position of a-carbon in the FA-Lys linker of the AA6_FAout (a), AA7_FAout (b), AA8_FAout (c), and AA7_glp1 (d), as a function of simu-
lation time. See Fig. 7 legend for details. To see this figure in color, go online.
TABLE 3 Mean energies and corresponding standard error of the mean (for P-P and P-W) and standard deviation (for S-S) for the 11
systems are calculated from the simulations
Structure P-P vdW (kcal/mol) P-W vdW (kcal/mol) S-S vdW (kcal/mol) S-S FA vdW (kcal/mol) S-S FA out vdW (kcal/mol)
Hexamer 188 518.9 84.7 521.1 20 54.5 44 56.1 —
Heptamer 200 514.8 68.1 519.3 37 55.6 56 54.7 23 53.8
Octamer 187 513.7 84.4 518.0 21 53.8 62 54.1 —
AA6_3ud 185 516.4 88.1 522.1 4.6 52.2 52 57.1 —
AA7_1ud 191 519.8 83.0 522.4 12 54.6 50 54.7 38 54.8
AA8_4udp 189 515.1 86.8 518.9 18 53.1 55 53.5 —
AA8_4uds 190 516.5 85.3 521.1 24 53.6 54 54.4 —
AA7_glp1 160 515.1 84.9 519.0 23 55.1 26 55.0 29 55.2
Structure P-P vdW (kcal/mol) P-W vdW (kcal/mol) S-S all FA out vdW (kcal/mol)
AA6_FAout 171 513.4 104 521.5 23 53.0
AA7_FAout 187 518.9 86.5 525.3 33 53.0
AA8_FAout 176 513.1 98.6 522.8 35 53.0
The vdW energies for the peptide-peptide (P-P), peptide-water (P-W), segment-segment (seg-seg) with FA pairs (S-S FA), seg-seg without FA pairs inter-
acting (S-S), and—for the heptamer—seg-seg interactions for the monomer pairs including the monomer with the FA pointing outward (S-S FA out), are
calculated. Energies are also calculated for the systems where all FA chains are pointing outward (S-S all FA out). The energies for the P-P and P-W in-
teractions are normalized according to the number of monomers in the structure. MDENERGY from the program NAMD (http://www.ks.uiuc.edu/
Research/namd/) was used to calculate the energies in intervals of 50 ps.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1209
emphasizes the stabilization effect of the FA chains. The
system seems to have a tight packing (Fig. S7); however,
this fact is more likely to be a result of the missing FA
chains in the structure, and hence, less surface area. The
S-S energies bear witness to a great lack in possible interac-
tions because these energies are significantly less negative
(~29 kcal/mol) than for those systems with FA chains pre-
sent (S-S FA average energy ~54 kcal/mol). This corre-
sponds well with the hydrophobic/hydrophilic areas of the
monomer shown in Fig. 10, where it is evident that there
is a difference in the hydrophobicity around the monomeric
structure. All in all, it shows that the FA chains are impor-
tant in stabilizing the liraglutide oligomer.
It thus appears that the heptamer is the most favorable
arrangement, which is further supported by the comparison
between the experimentally determined SAXS (SAXS
exp
)
curve and curves extracted from the simulations. The theo-
retical SAXS (SAXS
comp
) curves along with the experimen-
tally determined curve are shown in Fig. S10. SAXS
comp
curves were calculated from the last structure of the 11
simulations.
The discrepancies (c
2
values) for the 11 curves compared
to the experimentally obtained SAXS curve are, with the
radii of gyration, given in Table S1. Besides the radius of gy-
ration for the octamer, the radii of the systems lie very close
to those found from the experimental data seen in Table 2.
Also, the discrepancies given in Table S1 do not present
one specific candidate with the best fit, as several of the sys-
tems have low discrepancies between their end structures
and the experimentally obtained data.
To clarify the best fit further, we show the time evolution
of the discrepancies between SAXS
comp
and SAXS
exp
for
the 11 oligomers in Fig. 11.
The discrepancy of the heptamer is rather stable and low
throughout the simulation. So is c
2
of the hexamer,
AA8_4udp, AA8_4uds, and the AA7_glp1 systems. On
the contrary, the discrepancy of the octamer and AA6_3ud
is very high and increases with simulation time. That of
the AA6_FAout, AA7_FAout, AA8_FAout, and AA7_1ud
systems fluctuate significantly throughout the simulations.
The results show that the global structure and size seems
to be correct, especially for the heptamer and hexamer,
but none of our simulated structures captures the precise
shape of SAXS
exp
.
Summarizing and combining all the results (see Table 4
for a combined scoring chart), it appears that the most likely
solution structure of liraglutide is a heptamer where the
monomers are oriented in such a way that the attached FA
chains can interact in three pairs in the direction of the ellip-
tical arrangement, and with the remaining monomer ori-
ented so that the FA chain is pointing out.
The heptamer, with the above-mentioned conformation,
presents the best model with a total score of 1.9 being the
best scoring structure in four out of seven categories and
landing either a second, third, or fourth place in the remain-
ing categories. It presents the best energy interactions as
well as the least structural deviation of the monomers and
highest packing. The hexamer presents the second-best
solution, but does not score best in any category. The
FIGURE 10 A surface plot of the liraglutide monomer showing the seg-
seg orientations. (Green) Hydrophobic; (orange) hydrophilic; (gray)FA
chain. (ac) Different hydrophobicities on either side of the monomer. To
see this figure in color, go online.
FIGURE 11 Discrepancies calculated between the SAXS
comp
curves and
the experimentally determined SAXS curve of liraglutide as a function of
simulation time, calculated every 5 ns. (Arrows)c
2
values continuously in-
crease with simulation time. To see this figure in color, go online.
Biophysical Journal 109(6) 1202–1213
1210 Frederiksen et al.
AA6_3ud structure presents the worst arrangement of the
monomers, with a total score of 10.
CONCLUSIONS
From a pharmaceutical perspective, it is important to know
the oligomerization state of liraglutide with respect to sta-
bility, because uncontrolled and extensive oligomerization
can drive fibrillation. Hence, an important criterion for a sta-
ble formulation and, in turn, long shelf-life in the liquid
form is the solution structure. To gain further insight in
the solution structure of liraglutide, we performed a series
of MD simulations accompanied by MALS and SAXS ex-
periments. MALS provides information about the mass,
and SAXS provides information on mass, radius of gyra-
tion, and shape. The SAXS curves indicate that liraglutide
undergoes concentration-independent oligomerization. De-
pending on the partial specific volume used for deducing
the molecular weight and in turn the number of monomers
in an oligomer, liraglutide may form hexa-, hepta-, or oc-
tamers in solution. In contrast, the MALS results suggest
that liraglutide forms hexamers in solution. The experi-
mental results are not conclusive with respect to the size
of the oligomers. No information can be deduced from these
measurements regarding the orientation and stabilizing role
of the acyl chains in the oligomers.
We, therefore, performed MD simulations on several
oligomeric sets consisting of hexa-, hepta-, and octamers.
Our simulation results indicate that interactions between
the FA chains contribute to the stabilization of the structure
and that the heptamer presents the best representation of
the investigated liraglutide oligomers. Furthermore, com-
paring the experimentally determined SAXS curve with
the SAXS curves determined from the structures extracted
from the simulations shows qualitative agreement for
the overall size and shape. This indicates that liraglutide
in solution is most likely to form heptamers in a hollow,
water-filled, elliptical cylindrical-shaped structure where
the monomers are oriented in such a way that the FA chains
can interact pairwise. From the simulations, as of this
writing, we are not able to identify the absolute position
of the FA chains in the heptamer, but it is clear that interac-
tions between them are significant.
SUPPORTING MATERIAL
Supporting Materials and Methods, eleven figures, and two tables are avail-
able at http://www.biophysj.org/biophysj/supplemental/S0006-3495(15)
00815-2.
AUTHOR CONTRIBUTIONS
T.M.F. wrote the article and performed and analyzed simulations; P.S.
conducted and analyzed SAXS experiments, and contributed in writing
the article; L.A.R. performed preliminary simulations and contributed
to the article; P.H. conducted SAXS experiments and contributed in writing
the article; J.T.B. conducted SAXS experiments and contributed in writing
the article; A.M.S.-P. conducted and analyzed AF4-MALS experiments,
and contributed in writing the article; M.N.E.-L. performed preliminary
simulations; and G.H.P. performed and analyzed simulations, and contrib-
uted in writing the article.
ACKNOWLEDGMENTS
Simulations were performed at the Danish Center for Scientific Computing
at the Technical University of Denmark. MAXIV Synchrotron is acknowl-
edged for providing beamtime and for support during the experiments.
VMD (45) was used for all graphical representations of liraglutide.
The research leading to these results has received funding from the Euro-
pean Community’s Seventh Framework Program (grant No. FP7/2007-
2013) BioStruct-X, under grant agreement No. 283570. DANSCATT (the
Danish Agency for Science, Technology and Innovation) is acknowledged
for financial support.
SUPPORTING CITATIONS
References (48–50) appear in the Supporting Material.
TABLE 4 Scoring chart of the 11 oligomeric systems
Structure SASA RMSD 2D Projection
Energy SAXS
Total ScoreP-P vdW P-W vdW c
2
Value c
2
Value Fluctuation
Hexamer 2 8 3 5 5 7 4 4.9
Heptamer 1 4 1 1 1 3 2 1.9
Octamer 7 10 6 6 3 11 10 7.6
AA6_3ud 10 11 11 8 9 10 11 10
AA7_1ud 4 5 10 2 2 6 7 5.1
AA8_4udp 6 3 5 3 8 5 6 5.1
AA8_4uds 5 2 8 4 7 4 3 4.7
AA6_FAout 11 9 2 10 11 2 8 7.6
AA7_FAout 8 7 9 7 4 9 9 7.6
AA8_FAout 9 1 4 9 10 1 5 5.6
AA7_glp1 3 6 7 11 6 8 1 6.0
Each system is evaluated and compared to each of the others in these categories: lowest and most stable SASA, lowest and most stable RMSD, most stable
two-dimensional (2D) projection and elliptical shape, lowest P-P vdW energy, highest P-W vdW energy, lowest discrepancy from measured SAXS curve
taken for the end structure, and lowest mean and standard deviation of the discrepancy throughout the simulation (data taken from Fig. 11). The SASA,
RMSD, and 2D projection plots are inspected visually; 1 is the best score and 11 is the worst. The total score is normalized.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1211
REFERENCES
1. Kaspar, A. A., and J. M. Reichert. 2013. Future directions for peptide
therapeutics development. Drug Discov. Today. 18:807–817.
2. Li, Y., M. Shao, ., M. Gong. 2013. Self-assembling peptides improve
the stability of glucagon-like peptide-1 by forming a stable and sus-
tained complex. Mol. Pharm. 10:3356–3365.
3. Chang, X., D. Keller, ., J. J. Led. 2002. NMR studies of the aggrega-
tion of glucagon-like peptide-1: formation of a symmetric helical
dimer. FEBS Lett. 515:165–170.
4. Willard, F. S., and K. W. Sloop. 2012. Physiology and emerging
biochemistry of the glucagon-like peptide-1 receptor. Exp. Diabetes
Res. 2012:470851.
5. Nauck, M. A. 2008. Liraglutide, a once-daily human GLP-1 analogue.
Br. J. Diabetes Vasc. Dis. 8:S26–S33.
6. Jang, H.-J., Z. Kokrashvili, ., J. M. Egan. 2007. Gut-expressed gust-
ducin and taste receptors regulate secretion of glucagon-like peptide-1.
Proc. Natl. Acad. Sci. USA. 104:15069–15074.
7. Moran-Ramos, S., A. R. Tovar, and N. Torres. 2012. Diet, friend or foe
of enteroendocrine cells: how it interacts with enteroendocrine cells.
Adv. Nutr. Int. Rev. J. 3:8–20.
8. Sakurai, K., E. Y. Lee, ., T. Miki. 2012. Glucagon-like peptide-1
secretion by direct stimulation of L cells with luminal sugar vs non-
nutritive sweetener. J. Diabetes Investig. 3:156–163.
9. Doyle, M. E., and J. M. Egan. 2007. Mechanisms of action of glucagon-
like peptide 1 in the pancreas. Pharmacol. Ther. 113:546–593.
10. Madsbad, S. 2014. The role of glucagon-like peptide-1 impairment in
obesity and potential therapeutic implications. Diabetes Obes. Metab.
16:9–21.
11. Briyal, S., S. Shah, and A. Gulati. 2014. Neuroprotective and anti-
apoptotic effects of liraglutide in the rat brain following focal cerebral
ischemia. Neuroscience. 281C:269–281.
12. Talbot, K., and H. Y. Wang. 2014. The nature, significance, and
glucagon-like peptide-1 analog treatment of brain insulin resistance
in Alzheimer’s disease. Alzheimers Dement. 10 (Suppl 1):S12–S25.
13. Velmurugan, K., R. Bouchard, ., S. Pugazhenthi. 2012. Neuroprotec-
tive actions of glucagon-like peptide-1 in differentiated human neuro-
progenitor cells. J. Neurochem. 123:919–931.
14. Zhao, L., J. Xu, ., Y. Fang. 2015. Protective effect of rhGLP-1 (7-36)
on brain ischemia/reperfusion damage in diabetic rats. Brain Res.
1602:153–159.
15. McClean, P. L., and C. Ho
¨lscher. 2014. Liraglutide can reverse memory
impairment, synaptic loss and reduce plaque load in aged APP/PS1
mice, a model of Alzheimer’s disease. Neuropharmacology. 76 (Pt
A):57–67.
16. Deacon, C. F. 2004. Therapeutic strategies based on glucagon-like pep-
tide 1. Diabetes. 53:2181–2189.
17. Tasyurek, H. M., H. A. Altunbas, ., S. Sanlioglu. 2014. Incretins: their
physiology and application in the treatment of diabetes mellitus. Dia-
betes Metab. Res. Rev. 30:354–371.
18. Drucker, D. J., and M. A. Nauck. 2006. The incretin system: glucagon-
like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in
type 2 diabetes. Lancet. 368:1696–1705.
19. Orskov, C., A. Wettergren, and J. J. Holst. 1993. Biological effects and
metabolic rates of glucagonlike peptide-1 7-36 amide and glucagonlike
peptide-1 7-37 in healthy subjects are indistinguishable. Diabetes.
42:658–661.
20. Lund, A., F. K. Knop, and T. Vilsbøll. 2014. Glucagon-like peptide-1
receptor agonists for the treatment of type 2 diabetes: differences and
similarities. Eur. J. Intern. Med. 25:407–414.
21. Gallwitz, B., T. Ropeter, ., W. E. Schmidt. 2000. GLP-1-analogues
resistant to degradation by dipeptidyl-peptidase IV in vitro. Regul.
Pept. 86:103–111.
22. Ahre
´n, B. 2011. GLP-1 for type 2 diabetes. Exp. Cell Res. 317:1239–
1245.
23. Ladenheim, E. E. 2015. Liraglutide and obesity: a review of the data so
far. Drug Des. Devel. Ther. 9:1867–1875.
24. Bemporad, F., and F. Chiti. 2012. Protein misfolded oligomers: exper-
imental approaches, mechanism of formation, and structure-toxicity re-
lationships. Chem. Biol. 19:315–327.
25. Russell-Jones, D. 2009. Molecular, pharmacological and clinical as-
pects of liraglutide, a once-daily human GLP-1 analogue. Mol. Cell.
Endocrinol. 297:137–140.
26. Knudsen, L. B., P. F. Nielsen, ., H. Agersø. 2000. Potent derivatives of
glucagon-like peptide-1 with pharmacokinetic properties suitable for
once daily administration. J. Med. Chem. 43:1664–1669.
27. Pabreja, K., M. A. Mohd, ., S. G. B. Furness. 2014. Molecular mech-
anisms underlying physiological and receptor pleiotropic effects medi-
ated by GLP-1R activation. Br. J. Pharmacol. 171:1114–1128.
28. Steensgaard, D. B., J. K. Thomsen, ., L. B. Knudsen. 2008. The mo-
lecular basis for the delayed absorption of the once-daily Human GLP-
1 analogue, liraglutide. Diabetes. 57 (Suppl 1):A164.
29. Wang, Y., A. Lomakin, ., G. B. Benedek. 2015. Transformation of
oligomers of lipidated peptide induced by change in pH. Mol. Pharm.
12:411–419.
30. Trier, S., L. Linderoth, ., U. L. Rahbek. 2014. Acylation of Glucagon-
like peptide-2: interaction with lipid membranes and in vitro intestinal
permeability. PLoS One. 9:e109939.
31. Chang, X., D. Keller, ., J. J. Led. 2001. Structure and folding of
glucagon-like peptide-1-(7-36)-amide in aqueous trifluoroethanol stud-
ied by NMR spectroscopy. Magn. Reson. Chem. 39:477–483.
32. RxList, The Internet Drug Index. 2015. Victoza. http://www.rxlist.com/
victoza-drug.htm. Accessed July 5, 2015.
33. Gasteiger, E., C. Hoogland, ., A. Bairoch. 2005. Protein identification
and analysis tools in the ExPASy server. In The Proteomics Protocols
Handbook. Humana Press, New York, pp. 571–607.
34. Gasteiger, E., A. Gattiker, ., A. Bairoch. 2003. ExPASy: the prote-
omics server for in-depth protein knowledge and analysis. Nucleic
Acids Res. 31:3784–3788.
35. Labrador, A., Y. Cerenius, ., T. Plivelic. 2013. The yellow mini-hutch
for SAXS experiments at MAX IV Laboratory. J. Phys. Conf. Ser.
425:72019.
36. Konarev, P. V., V. V. Volkov, ., D. I. Svergun. 2003. PRIMUS: a Win-
dows PC-based system for small-angle scattering data analysis. J. Appl.
Cryst. 36:1277–1282.
37. Petoukhov, M. V., D. Franke, ., D. I. Svergun. 2012. New develop-
ments in the ATSAS program package for small-angle scattering data
analysis. J. Appl. Cryst. 45:342–350.
38. Semenyuk, A. V., and D. I. Svergun. 1991. GNOM. A program package
for small-angle scattering data processing. J. Appl. Cryst. 24:537–540.
39. Bernstein, F. C., T. F. Koetzle, ., M. Tasumi. 1977. The Protein Data
Bank. A computer-based archival file for macromolecular structures.
Eur. J. Biochem. 80:319–324.
40. Grubmu
¨ller, H., and V. Groll. 2015. SOLVATE. Biophysical Chemistry
Department, Max Planck Institute of Biophysics, Frankfurt, Germany.
http://www.mpibpc.mpg.de/grubmueller/solvate. Accessed February
28, 2015.
41. Jorgensen, W. L., J. Chandrasekhar, ., M. L. Klein. 1983. Comparison
of simple potential functions for simulating liquid water. J. Chem.
Phys. 79:926.
42. Nelson, M. T., W. Humphrey, ., K. Schulten. 1996. NAMD: a parallel,
object-oriented molecular dynamics program. Int. J. High Perform.
Comput. Appl. 10:251–268.
43. MacKerell, A. D., D. Bashford, ., M. Karplus. 1998. All-atom empir-
ical potential for molecular modeling and dynamics studies of proteins.
J. Phys. Chem. B. 102:3586–3616.
44. Madsen, J. J., L. Linderoth, ., G. H. Peters. 2011. Secretory phospho-
lipase A2 activity toward diverse substrates. J. Phys. Chem. B.
115:6853–6861.
Biophysical Journal 109(6) 1202–1213
1212 Frederiksen et al.
45. Humphrey, W., A. Dalke, and K. Schulten. 1996. VMD: visual molec-
ular dynamics. J. Mol. Graph. 14:33–38, 27–28.
46. Svergun, D., C. Barberato, and M. H. Koch. 1995. CRYSOL—a pro-
gram to evaluate x-ray solution scattering of biological macromole-
cules from atomic coordinates. J. Appl. Cryst. 28:768–773.
47. Mylonas, E., and D. I. Svergun. 2007. Accuracy of molecular mass
determination of proteins in solution by small-angle x-ray scattering.
J. Appl. Cryst. 40:s245–s249.
48. Darden, T., D. York, and L. Pedersen. 1993. Particle mesh Ewald: an N
log(N) method for Ewald sums in large systems. J. Chem. Phys.
98:10089–10092.
49. Essmann, U., L. Perera, ., L. G. Pedersen. 1995. A smooth particle
mesh Ewald method. J. Chem. Phys. 103:8577–8593.
50. Feller, S. E., Y. Zhang, ., B. R. Brooks. 1995. Constant-pressure mo-
lecular-dynamics simulation—the Langevin piston method. J. Chem.
Phys. 103:4613–4621.
Biophysical Journal 109(6) 1202–1213
Oligomerization of a GLP1 Analog 1213
... GLP-1 is a peptide used to treat several disorders, most notably obesity and type-2 diabetes [78][79][80] . It reduces appetite, glucagon secretion and slows down gastric emptying 80 , and has a low risk of inducing hypoglycemia, a common side effect for diabetes drugs 78 . ...
... GLP-1 is a peptide used to treat several disorders, most notably obesity and type-2 diabetes [78][79][80] . It reduces appetite, glucagon secretion and slows down gastric emptying 80 , and has a low risk of inducing hypoglycemia, a common side effect for diabetes drugs 78 . GLP-1 is a 36 amino acid long peptide that when cleaved at the N-terminus produces its active form: GLP-1 7-36 amide 78 . ...
... GLP-1 is a 36 amino acid long peptide that when cleaved at the N-terminus produces its active form: GLP-1 7-36 amide 78 . The drawback of GLP-1 in its native form is that, like most peptides, it has a short half-life and fast clearance rate 80 . The GLP-1 derivatives liraglutatide and semaglutide were developed to overcome this issue 80,81 . ...
Article
Full-text available
Non-natural amino acids are increasingly used as building blocks in the development of peptide-based drugs as they expand the available chemical space to tailor function, half-life and other key properties. However, while the chemical space of modified amino acids (mAAs) such as residues containing post-translational modifications (PTMs) is potentially vast, experimental methods for measuring the developability properties of mAA-containing peptides are expensive and time consuming. To facilitate developability programs through computational methods, we present CamSol-PTM, a method that enables the fast and reliable sequence-based prediction of the intrinsic solubility of mAA-containing peptides in aqueous solution at room temperature. From a computational screening of 50,000 mAA-containing variants of three peptides, we selected five different small-size mAAs for a total number of 37 peptide variants for experimental validation. We demonstrate the accuracy of the predictions by comparing the calculated and experimental solubility values. Our results indicate that the computational screening of mAA-containing peptides can extend by over four orders of magnitude the ability to explore the solubility chemical space of peptides and confirm that our method can accurately assess the solubility of peptides containing mAAs. This method is available as a web server at https://www-cohsoftware.ch.cam.ac.uk/index.php/camsolptm.
... A comprehensive example involves liraglutide, an Active Pharmaceutical Ingredient (API) peptide used in the treatment of Type 2 diabetes and chronic obesity, with the brand names Victoza and Saxenda. A first study, exploiting an AF4-UV-MALS platform, published by Frederiksen et al., measured the aggregation state of liraglutide as hexameric ( Figure 5) and compared the result with the ones stemming from theoretical calculations [89]. These evaluations were, however, performed using a mobile phase different from the formulation environment, which could greatly affect the aggregation. . ...
... Different colors refer to different injection amounts. Image adapted from [89]. ...
Article
Full-text available
Field-flow fractionation (FFF) is a family of single-phase separative techniques exploited to gently separate and characterize nano- and microsystems in suspension. These techniques cover an extremely wide dynamic range and are able to separate analytes in an interval between a few nm to 100 µm size-wise (over 15 orders of magnitude mass-wise). They are flexible in terms of mobile phase and can separate the analytes in native conditions, preserving their original structures/properties as much as possible. Molecular biology is the branch of biology that studies the molecular basis of biological activity, while biotechnology deals with the technological applications of biology. The areas where biotechnologies are required include industrial, agri-food, environmental, and pharmaceutical. Many species of biological interest belong to the operational range of FFF techniques, and their application to the analysis of such samples has steadily grown in the last 30 years. This work aims to summarize the main features, milestones, and results provided by the application of FFF in the field of molecular biology and biotechnology, with a focus on the years from 2000 to 2022. After a theoretical background overview of FFF and its methodologies, the results are reported based on the nature of the samples analyzed.
... GLP-1 is a peptide used to treat several disorders, most notably obesity and type-2 diabetes [77][78][79] . It reduces appetite, glucagon secretion and slows down gastric emptying 79 , and has a low risk of inducing hypoglycemia, a common side effect for diabetes drugs 77 . ...
... The drawback of GLP-1 in its native form is that, like most peptides, it has a short half-life and fast clearance rate 79 . The GLP-1 derivatives liraglutatide and semaglutide were developed to overcome this issue 79,80 . The half-life of these drugs is significantly extended compared to its native form by introducing long fatty acid chains that improves drug half-life primarily by enabling albumin binding [81][82][83][84][85][86] . ...
Preprint
Full-text available
Non-natural amino acids are increasingly used as building blocks in the development of peptide-based drugs, as they expand the available chemical space to tailor function, half-life and other key properties. However, while the chemical space of modified amino acids (mAAs) is potentially vast, experimental methods for measuring the developability properties of mAA-containing peptides are expensive and time consuming. To facilitate developability programs through computational methods, we present CamSol-PTM, a method that enables the fast and reliable sequence-based prediction of the solubility of mAA-containing peptides. From a computational screening of 50,000 mAA-containing variants of three peptides, we selected five different mAAs for a total number of 30 peptide variants for experimental validation. We demonstrate the accuracy of the predictions by comparing the calculated and experimental solubility values. Our results indicate that the computational screening of mAA-containing peptides can extend by over four orders of magnitude the ability to explore the chemical space of to increase the solubility of peptides. This method is available as a web server at https://www-cohsoftware.ch.cam.ac.uk/index.php/camsolptm.
... Based on static light scattering, another group reported Liraglutide forms octamers at pH 7.4, which are reversibly converted to dodecamers below pH 6.7 (Wang et al. 2015). Liraglutide was also found to form hexamers to octamers using small-angle X-ray scattering (Frederiksen et al. 2015). ...
Article
Full-text available
The drug Liraglutide is an acylated analogue of glucagon-like peptide-1 (GLP-1) which is indicated for the treatment of type 2 diabetes mellitus and chronic obesity. Stability testing of biopharmaceuticals is an essential part of their quality control, aimed at revealing possible degradation pathways and identification of potential degradation products. Up to now, no information on the Liraglutide behaviour in formulation medium under stress conditions has been provided. In the present study, the original innovator and an analogue product of Liraglutide were subjected to stress by exposing to various temperature, pH and oxidation conditions to generate possible degradation products and aggregation. Liquid chromatography-mass spectrometry was used to determine impurity profiles and identify degradation products of stressed drug samples from different suppliers. Results showed that Liraglutide products exhibit high stability at room temperature. Exposure to high pH resulted in the formation of aggregates and chemical modifications, including oxidation, while exposure to low pH caused peptide precipitation. Liraglutide formulations showed slight susceptibility to oxidation on tryptophan residue. Overall, no significant differences were observed between the originator and the analogue of Liraglutide. Both exhibited similar behaviour in their impurity profiles during forced degradation. The approach used in this study offers potential benefits for industrial purposes in formulation development and product characterization. Graphical Abstract
... It has also been claimed that LP aggregation could contribute significantly to its prolonged blood circulation [17]. In this context, it has been shown that the dimensions of LG aggregates are strongly dependent on the pH, with LG aggregates being formed by eight LP units at pH 8 [18] and twelve LP units at pH 6.4 [19]. Innovative procedures have also been applied to the synthesis of LG using total chemical routes [20]. ...
Article
Full-text available
The pharmacokinetics of peptide drugs are strongly affected by their aggregation prop- erties and the morphology of the nanostructures they form in their native state as well as in their therapeutic formulation. In this contribution, we analyze the aggregation properties of a Liraglutide analogue (LG18), a leading drug against diabetes type 2. LG18 is a lipopeptide characterized by the functionalization of a lysine residue (K26) with an 18C lipid chain. To this end, spectroscopic experiments, dynamic light scattering measurements, and molecular dynamics simulations were carried out, following the evolution of the aggregation process from the small LG18 clusters formed at sub-micromolar concentrations to the mesoscopic aggregates formed by aged micromolar solutions. The critical aggregation concentration of LG18 in water (pH = 8) was found to amount to 4.3 μM, as assessed by the pyrene fluorescence assay. MD simulations showed that the LG18 nanostructures are formed by tetramer building blocks that, at longer times, self-assemble to form micrometric supramolecular architectures.
... Lipidation is another common strategy to improve the stability of peptide drugs in vivo [12][13][14] . Lipidation is also known to extend the systemic circulation half-life of the peptide molecule by allowing reversible binding to serum albumin 15 . This strategy has been applied to GLP-1 to produce liraglutide and semaglutide which are commercially available lipidated forms of GLP-1 analogues 16 , as well as other lipidated GLP-1 analogues that are currently under clinical evaluation 17,18 . ...
Thesis
Full-text available
Aggregation and physical instability of peptide-based drugs poses a great challenge to the pharmaceutical industry. Glucagon-like peptide 1 (GLP-1) is a hormone that is used in the treatment of type-2 diabetes. However, GLP-1 has a short half-life in vivo and it is prone to aggregate which complicates its pharmaceutical usage. Strategies to overcome the short half-life in vivo include numerous chemical modifications of the native peptide. The focus of this Thesis is on the effect of two sets of chemical modification strategies, lipidation and C-terminal amidation, on the physical stability of the peptide. The first part of this work combines experimental and computational approaches to better understand the molecular basis of the aggregation of GLP-1 and its C-terminally amidated variant, GLP-1-Am. In particular, the off-pathway aggregation of GLP-1 and GLP-1-Am into disordered low-molecular weight oligomers is described. This process competes with the amyloid formation pathway and the addition of pre-formed off-pathway oligomers slightly slows down the fibrillation rate. Energy Landscape Theory was employed to investigate and rationalize the conformational behaviour and aggregation propensity of GLP-1 in different protonation states. Under all conditions studied, the GLP-1 energy landscape possesses a multi-funnel character with a variety of structurally different ensembles with low energy, which is a typical feature of intrinsically disordered proteins and aggregating systems. It is also shown that β-structure-containing conformations are more energetically favoured at acidic pH compared to neutral pH conditions, which agrees with a greater propensity of GLP-1 for aggregation at acidic pH which was observed experimentally. The second part of this Thesis focuses on the self-assembly and aggregation of lipidated analogues of GLP-1. Four lipidated GLP-1 analogues, which varied in the position of lipidation, and one additional analogue differing by the nature of the lipid moiety, were studied to establish the effect of the lipidation site and the lipid moiety on the physical stability of the peptide. The lipidation was shown to induce formation of large stable oligomers (i.e. > 7 monomeric units). The aggregation mechanism and kinetics were shown to be highly dependent on the lipidation position and the nature of the lipid moiety. Moreover, the aggregation kinetics of lipidated analogues were rarely observed to follow a classical nucleation-elongation mechanism but were rather likely to consist of more complex processes. Aggregates with a high content of β-sheet were formed by all analogues studied, however, they were distinct in their tertiary structure and aggregate morphology.
Article
Full-text available
The prevalence of obesity worldwide has nearly doubled since 1980 with current estimates of 2.1 billion in 2013. Overweight and obesity lead to numerous adverse conditions including type 2 diabetes, cardiovascular disease, stroke, and certain cancers. The worldwide spread of obesity and associated comorbidities not only threatens quality of life but also presents a significant economic burden. While bariatric surgery has proven to be a viable treatment option for the morbidly obese, there is clearly a need for less invasive alternatives. Recent research has suggested that long-acting analogs of the gut hormone, glucagon-like peptide 1 (GLP-1), may have potential as an antiobesity treatment. The GLP-1 receptor agonist, liraglutide (trade name Saxenda), was recently approved by the US Food and Drug Administration as an obesity treatment option and shown in clinical trials to be effective in reducing and sustaining body weight loss. This review presents the basis for GLP-1-based therapies with a specific focus on animal and human studies examining liraglutide's effects on food intake and body weight.
Article
Full-text available
Background: Acylation of peptide drugs with fatty acid chains has proven beneficial for prolonging systemic circulation as well as increasing enzymatic stability without disrupting biological potency. Acylation has furthermore been shown to increase interactions with the lipid membranes of mammalian cells. The extent to which such interactions hinder or benefit delivery of acylated peptide drugs across cellular barriers such as the intestinal epithelia is currently unknown. The present study investigates the effect of acylating peptide drugs from a drug delivery perspective. Purpose: We hypothesize that the membrane interaction is an important parameter for intestinal translocation, which may be used to optimize the acylation chain length for intestinal permeation. This work aims to characterize acylated analogues of the intestinotrophic Glucagon-like peptide-2 by systematically increasing acyl chain length, in order to elucidate its influence on membrane interaction and intestinal cell translocation in vitro. Results: Peptide self-association and binding to both model lipid and cell membranes was found to increase gradually with acyl chain length, whereas translocation across Caco-2 cells depended non-linearly on chain length. Short and medium acyl chains increased translocation compared to the native peptide, but long chain acylation displayed no improvement in translocation. Co-administration of a paracellular absorption enhancer was found to increase translocation irrespective of acyl chain length, whereas a transcellular enhancer displayed increased synergy with the long chain acylation. Conclusions: These results show that membrane interactions play a prominent role during intestinal translocation of an acylated peptide. Acylation benefits permeation for shorter and medium chains due to increased membrane interactions, however, for longer chains insertion in the membrane becomes dominant and hinders translocation, i.e. the peptides get 'stuck' in the cell membrane. Applying a transcellular absorption enhancer increases the dynamics of membrane insertion and detachment by fluidizing the membrane, thus facilitating its effects primarily on membrane associated peptides.
Article
Full-text available
Aims/Introduction: Oral ingestion of carbohydrate triggers secretion of glucagon‐like peptide (GLP)‐1, which inhibits the postprandial rise in blood glucose levels. However, the mechanism of carbohydrate‐induced GLP‐1 secretion from enteroendocrine L cells remains unclear. In the present study, GLP‐1 secretion was examined by meal tolerance tests of healthy Japanese volunteers. Materials and Methods: Twenty‐one healthy Japanese men participated in the study. The meal tolerance test was performed with modified nutrient compositions, with or without pretreatment with the α‐glucosidase inhibitor acarbose, or with substitution of sucrose with an equivalent dose of sweeteners in the meal. Blood concentrations of glucose, insulin, GLP‐1, and apolipoprotein (Apo) B‐48 were measured. Results: GLP‐1 secretion started concomitant with the increase in blood glucose levels 10 min after meal ingestion. Insulin secretion started at 5 min, before the increase in blood glucose levels, reflecting the contribution of direct nutrient stimulation on the former parameter and neural regulation in the latter. Carbohydrate retention in the gut lumen induced by acarbose pretreatment extended postprandial GLP‐1 secretion and negated the increase in serum ApoB‐48 levels. GLP‐1 secretion was markedly decreased by a reduction in the amount of sucrose in the meal and was not restored by an equivalent dose of sweeteners used to compensate for the sweet taste. Conclusions: The results indicate that direct stimulation of L cells with sugar, but not sweetener, is required for carbohydrate‐induced GLP‐1 secretion. In addition, inhibition of digestion of dietary carbohydrate by α‐glucosidase inhibitors may prevent postprandial hyperglycemia by increasing GLP‐1 secretion and by inhibiting glucose absorption. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2011.00163.x, 2011)
Article
Full-text available
Alzheimer's disease (AD) is an age-related neurodegenerative disease leading over the course of decades to the most common form of dementia. Many of its pathologic features and cognitive deficits may be due in part to brain insulin resistance recently demonstrated in the insulin receptor→insulin receptor substrate-1 (IRS-1) signaling pathway. The proximal cause of such resistance in AD dementia and amnestic mild cognitive impairment (aMCI) appears to be serine inhibition of IRS-1, a phenomenon likely due to microglial release of inflammatory cytokines triggered by oligomeric Aβ. Studies on animal models of AD and on human brain tissue from MCI cases at high risk of AD dementia have shown that brain insulin resistance and many other pathologic features and symptoms of AD may be greatly reduced or even reversed by treatment with FDA-approved glucagon-like peptide-1 (GLP-1) analogs such as liraglutide (Victoza). These findings call attention to the need for further basic, translational, and clinical studies on GLP-1 analogs as promising AD therapeutics.
Article
In recent years, GLP-1 and its analogs have been developed for the treatment of type 2 diabetes. It has been reported that stimulating the GLP-1 receptor can protect neurons against metabolic and oxidative insults, and therefore can be used in the treatment of stroke and Parkinson's disease. The present study aimed to examine the neuroprotective effects of rhGLP-1 (7-36) and its possible mechanisms against acute ischemia/reperfusion injuries induced by middle cerebral artery occlusion (MCAO) in diabetic rats. The type 2 diabetic rat model was established by a combination of a high-fat diet and low-dose streptozotocin (STZ). RhGLP-1 (7-36) (20, 40, 80μg/kg) was given intraperitioneally before reperfusion. The neuroprotective effects of rhGLP-1 (7-36) were evaluated by changes in neurological deficit scores and 2,3,5-Triphenyltetrazolium chloride (TTC) staining. Changes in blood glucose were used to assess hypoglycemic effects. The content of malondialdehyde (MDA) and the activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), inducible nitric oxide syntheses (iNOS) and endothelial nitric oxide syntheses (eNOS) after MCAO/R administration (2h and 46h) were examined to investigate the possible mechanisms of RhGLP-1 (7-36). Haematoxylin and eosin (H&E) staining was used for histopathological observation. Compared with the control group, rhGLP-1 (7-36)-treated groups decreased nerve function deficiency scores; significantly reduced infarction volume percentage, MDA, iNOS and blood glucose; and significantly increased SOD, GSH-PX and eNOS. In addition, rhGLP-1 (7-36) groups enhanced the density of surviving neurons and increased vascular proliferation. The current study suggests a neuroprotective effect of rhGLP-1 (7-36) in diabetic MCAO/R rats since anti-oxidative and anti-nitrosative stress effects can contribute to beneficial effects against ischemia/reperfusion injury. Copyright © 2015. Published by Elsevier B.V.
Article
Oligomerization of lipidated peptides is of general scientific interest and is important in biomedical and pharmaceutical applications. We investigated the solution properties of a lipidated peptide, Liraglutide, which is one of the glucagon like peptide-1 (GLP-1) agonists used for the treatment of type II diabetes. Liraglutide can serve as a model system for studying biophysical and biochemical properties of micelle-like self-assemblies of the lipidated peptides. Here, we report a transformation induced in Liraglutide oligomers by changing pH in the vicinity of pH 7. This fully reversible transformation is characterized by changes in the size and aggregation number of the oligomer and an associated change in the secondary structure of the constituent peptides. This transformation has quite slow kinetics: the equilibrium is reached in a course of several days. Interestingly, while the transformation is induced by changing pH, its kinetics is essentially independent of the final pH. We interpreted these findings using a model in which desorption of the monomer from the oligomer is the rate-limiting step in the transformation, and we determined the rate constant of the monomer desorption.
Article
Stroke is a leading cause of death and serious, long-term disability worldwide. We report that rats receiving liraglutide show markedly attenuated infarct volumes and neurological deficit following ischemic insult. We have also investigated the effect of liraglutide on apoptosis and oxidative stress pathways after ischemic injury in diabetic and non-diabetic rats. Male Sprague-Dawley rats weighing 300 to 350 g were used. Diabetes was induced by streptozotocin. Rats were pretreated with either vehicle or liraglutide (50 μg/kg, s.c.) for 14 days and thereafter subjected to middle cerebral artery occlusion (MCAO). Twenty-four hours after occlusion, rats were assessed for neurological deficit, motor function and subsequently sacrificed for estimation of infarct volume, oxidative stress and apoptotic markers. Vehicle-treated non-diabetic and diabetic rats showed significant (P<0.001) neurological deficit following cerebral ischemia. Liraglutide pretreatment resulted in significantly (P<0.001) less neurological deficit compared to vehicle-treated MCAO rats. Cerebral ischemia produced significant (P<0.0001) infarction in vehicle-treated rats; however, the infarct volume was significantly (p<0.001) less in liraglutide pretreated rats. Oxidative stress markers were increased following ischemia but were attenuated in liraglutide-treated rats. Anti-apoptotic protein Bcl-2 expression was decreased and pro-apoptotic protein Bax expression was increased in vehicle-treated MCAO rats compared to sham (p<0.0001). On the other hand liraglutide pretreatment showed significantly (p<0.01) increased expression of Bcl-2 and decreased expression of Bax in MCAO rats. In vehicle-treated group, the number of TUNEL-positive cells significantly (p<0.0001) increased in the ischemic hemisphere compared to sham-operated group. The number of TUNEL-positive cells in vehicle group was 73.5±3.3 and 85.5±5.2/750 μm(2) in non-diabetic and diabetic vehicle-treated MCAO rats, respectively. Following liraglutide treatment the number of TUNEL-positive cells was remarkably attenuated to 25.5±2.8 and 41.5±4.1/750 μm(2) (p<0.001) in non-diabetic and diabetic rats, respectively. The results demonstrate that GLP-1 agonist, liraglutide, is a neuroprotective agent and attenuates the neuronal damage following cerebral ischemia in rats by preventing apoptosis and decreasing oxidative stress.
Article
Glucagon-like peptide-1 (GLP-1) is a gastrointestinal hormone, secreted in response to ingestion of nutrients, and has important effects on several of the pathophysiological features of type 2 diabetes (T2D). The effects include potentiation of insulin secretion, suppression of glucagon secretion, slowing of gastric emptying and suppression of appetite. In circulation, GLP-1 has a half-life of approximately 2 min due to rapid degradation by the enzyme dipeptidyl peptidase 4 (DPP-4). Because of this short half-life GLP-1 receptor (GLP-1R) agonists, resistant to degradation by DPP-4 have been developed. At the moment four different compounds are available for the treatment of T2D and many more are in clinical development. These compounds, although all based on the effects of native GLP-1, differ with regards to structure, pharmacokinetics and size, which ultimately leads to different clinical effects. This review gives an overview of the clinical data on GLP-1R agonists that have been compared in head-to-head studies and focuses on relevant differences between the compounds. Highlighting these similarities and differences could be beneficial for physicians in choosing the best treatment strategy for their patients.