ArticlePDF Available

Effect of helix-promoting strategies on the biological activity of novel analogues of the B-chain of INSL3

Authors:

Abstract and Figures

Insulin-like 3 (INSL3) is a novel circulating peptide hormone that is produced by testicular Leydig cells and ovarian thecal and luteal cells. In males, INSL3 is responsible for testicular descent during foetal life and suppresses germ cell apoptosis in adult males, whereas in females, it causes oocyte maturation. Antagonists of INSL3 thus have significant potential clinical application as contraceptives in both males and females. Previous work has shown that the INSL3 receptor binding region is largely confined to the B-chain central alpha-helix of the hormone and a conformationally constrained analogue of this has modest receptor binding and INSL3 antagonist activity. In the present study, we have employed and evaluated several approaches for increasing the alpha-helicity of this peptide in order to better present the key receptor binding residues and increase its affinity for the receptor. Analogues of INSL3 with higher alpha-helicity generally had higher receptor binding affinity although other structural considerations limit their effectiveness.
Content may be subject to copyright.
ORIGINAL ARTICLE
Effect of helix-promoting strategies on the biological activity
of novel analogues of the B-chain of INSL3
Fazel Shabanpoor ÆRichard A. Hughes ÆSuode Zhang ÆRoss A. D. Bathgate Æ
Sharon Layfield ÆMohammed Akhter Hossain ÆGeoffrey W. Tregear Æ
Frances Separovic ÆJohn D. Wade
Received: 23 October 2008 / Accepted: 17 November 2008 / Published online: 7 December 2008
ÓSpringer-Verlag 2008
Abstract Insulin-like 3 (INSL3) is a novel circulating
peptide hormone that is produced by testicular Leydig cells
and ovarian thecal and luteal cells. In males, INSL3 is
responsible for testicular descent during foetal life and
suppresses germ cell apoptosis in adult males, whereas in
females, it causes oocyte maturation. Antagonists of INSL3
thus have significant potential clinical application as con-
traceptives in both males and females. Previous work has
shown that the INSL3 receptor binding region is largely
confined to the B-chain central a-helix of the hormone and
a conformationally constrained analogue of this has modest
receptor binding and INSL3 antagonist activity. In the
present study, we have employed and evaluated several
approaches for increasing the a-helicity of this peptide in
order to better present the key receptor binding residues
and increase its affinity for the receptor. Analogues of
INSL3 with higher a-helicity generally had higher receptor
binding affinity although other structural considerations
limit their effectiveness.
Keywords INSL3 RXFP2 Lactam-constraint
Disulfide-constraint Helicity Peptide
Introduction
Insulin-like peptide 3 (INSL3) was discovered in the early
1990s (Adham et al. 1993) and shown to belong to the
insulin–relaxin superfamily of polypeptide hormones. It was
originally named Leydig cell insulin-like peptide (Ley-IL)
because it was found in the Leydig cells of the testis
(Burkhardt et al. 1994) and has also been referred to as RLF
(relaxin-like factor) due to its relaxin-like activity in a mouse
interpubic ligament bioassay (Bu
¨llesbach and Schwabe
1995). In the male, INSL3 acts as a marker for fully differ-
entiated adult-type Leydig cells (Ivell and Einspanier 2002)
and is also expressed by ovarian follicles and in the corpus
luteum in the female but at lower levels compared to the male
(Roche et al. 1996; Tashima et al. 1995).
INSL3 is a circulating hormone which has important
reproductive and non-reproductive roles. During foetal life
it is principally involved in mediation of the transabdom-
inal phase of testicular decent as INSL3 or its receptor,
RXFP2, knockout male mice have been shown to have a
similar phenotype in which both are cryptorchid, i.e. they
retain their testes in the abdominal cavity, which leads to
impaired spermatogenesis and infertility (Bachelot et al.
2000; Bogatcheva et al. 2003; Feng et al. 2004; Foresta and
Ferlin 2004; Nef and Parada 1999; Spiess et al. 1999;
Zimmermann et al. 1999). In adults, the INSL3 and RXFP2
system acts as a paracrine factor in mediating gonadotropin
actions (Kawamura et al. 2004). Luteinizing hormone
(LH), which is released by the anterior pituitary gland,
stimulates INSL3 transcripts in ovarian theca and testicular
Leydig cells. INSL3 successively binds RXFP2 expressed
F. Shabanpoor S. Zhang R. A. D. Bathgate S. Layfield
M. A. Hossain G. W. Tregear J. D. Wade (&)
Howard Florey Institute, University of Melbourne,
Melbourne, VIC 3010, Australia
e-mail: john.wade@florey.edu.au
F. Shabanpoor F. Separovic J. D. Wade
School of Chemistry, University of Melbourne,
Melbourne, VIC 3010, Australia
R. A. Hughes
Department of Pharmacology, University of Melbourne,
Melbourne, VIC 3010, Australia
R. A. D. Bathgate
Department of Biochemistry and Molecular Biology,
University of Melbourne, Melbourne, VIC 3010, Australia
123
Amino Acids (2010) 38:121–131
DOI 10.1007/s00726-008-0219-2
in germ cells to activate the inhibitory G protein, thus
leading to decreases in cAMP production. This, in turn,
leads to the initiation of meiotic progression of arrested
oocytes in preovulatory follicles in vitro and in vivo and
suppresses male germ cell apoptosis in vivo (Kawamura
et al. 2004).
A recent study has shown that in males the INSL3/
RXFP2 signalling system is also involved in bone metab-
olism as RXFP2
-/-
knockout mice showed a considerable
reduction in their bone mass, mineralizing surface and bone
formation compared to wild type mice (Ferlin et al. 2008).
This study also showed that 64% of young men with
RXFP2 mutations had significant reduction in bone mass
density, a sign of osteoporosis (Ferlin et al. 2008). INSL3
may also play a role in the pathobiology of some forms of
human cancers, such as thyroid carcinoma, as its expres-
sion is upregulated in hyperplastic and neoplastic human
thyrocytes (Klonisch et al. 2005).
INSL3 is expressed as a preprohormone with an
N-terminal signal peptide for secretion, a B-chain, a
C-peptide, and a C-terminal A-chain. The preprohormone
is subsequently processed into a mature peptide through
cleavage of the signal peptide and formation of two inter-
chain and an intra-A-chain disulfide bond followed by
proteolytic removal of the C-peptide (Adham et al. 1993;
Hsu 2003). Mature human INSL3 consists of an A- and
B-chain of 26 and 31 amino acids, respectively, and its
tertiary structure has recently been solved using solution
NMR spectroscopy (Rosengren et al. 2006) (Fig. 1a).
INSL3 adopts a core structure similar to that found in
insulin and relaxin, especially in the region confined by
the disulfide bonds.
To determine the residues involved in receptor binding,
recent structure–activity studies by our group using single
Ala substitution have shown that substituting Arg
B16
and
Val
B19
significantly reduced receptor binding affinity
(Rosengren et al. 2006). On the other hand, multi-Ala
substitution showed that His
B12
and Arg
B20
have a strong
synergistic effect with Arg
B16
, suggesting that His
B12
and
Arg
B20
may play a role in the initial step of receptor rec-
ognition, involving electrostatic interactions between basic
residues of the peptide and acidic residues on the receptor
(Rosengren et al. 2006). In addition to these residues,
Trp
B27
toward the C-terminus of the B-chain has also been
shown to be crucial for binding of INSL3 as the mutation
or deletion of Trp
B27
leads to loss of receptor binding
affinity (Bu
¨llesbach and Schwabe 1999; Rosengren et al.
2006). These B-chain residues collectively form a receptor
binding motif (H
B12
,R
B16
,V
19
,R
20
and W
27
). A-chain
N-terminal truncation studies of INSL3 have shown that
truncation of the INSL3 peptide to Cys
A10
results in a
peptide with high receptor binding affinity but which is
devoid of signalling activity, i.e. an antagonist (Bu
¨llesbach
and Schwabe 2005; Hossain et al. 2008).
Despite knowledge of the region of the peptide that is
involved in receptor signalling, there is no clear under-
standing of the mechanism of receptor activation. A
recent study has shown that the mechanism of receptor
activation by INSL3 is independent of the amino acid side
chains and is a function of certain peptide bonds at the
N-terminus of the A-chain (Bu
¨llesbach and Schwabe
2007). These authors proposed the backbone amide bond
around Arg
A8
and Tyr
A9
to be crucial for receptor acti-
vation, as the replacement of these residues with alanine
does not affect signalling whereas their deletion or
replacement with D-Pro has no impact on receptor binding
but severely retards receptor activation (Bu
¨llesbach and
Schwabe 2007). In contrast, a more recent study on a
relaxin-2, which also binds to INSL3 receptor (RXFP2),
has shown that there are other residues in the A-chain
which are involved in receptor activation. These authors
have shown that K
A17
is an important residue for receptor
activation as its mutation to alanine enhances RXFP2-
activation activity of relaxin-2 as a result of inducing
active conformational transformation. On the other hand,
the replacement of this residue with a polar or negatively
charged residue reduces the receptor activation activity of
relaxin-2 (Park et al. 2008).
W27
V19 R20
R16
H12
A
W27
V19 R20
R
16
H12
B
B-chain
A-chain
Fig. 1 a Solution NMR
structure of native human
INSL3 showing the important
receptor binding residues
(H
12
,R
16
,V
19
,R
20
and W
27
).
bAnalogue 30 (Table 1)in
which a truncated INSL3
A-chain (from residue Cys
A15
to
Cys
A24
) is linked via a disulfide
bond to the truncated B-chain
122 F. Shabanpoor et al.
123
INSL3, due to its role in germ cell maturation in adults,
has enormous potential as a clinical agent in the area of
fertility management; in particular, antagonists of this
peptide may have significant clinical promise for use as
both a male and female contraceptive. As discussed above,
INSL3 has been shown to bind to its receptor using the
residues primarily located on the a-helical region of the
B-chain. In an attempt to develop mimetics of INSL3
B-chain with high receptor binding affinity and antago-
nistic activity, our group recently designed and synthesized
shortened analogues of the INSL3 B-chain that had
antagonistic activity in vitro (Del Borgo et al. 2006;
Shabanpoor et al. 2007). In vivo administration of one of
these antagonists into the testes of rats resulted in a sub-
stantial decrease in testis weight probably due to the
inhibition of germ cell survival (Del Borgo et al. 2006).
However, these peptides have receptor binding affinities
within the micromolar range compared to the nanomolar
affinity of the native INSL3. This is due, in part, to the lack
of INSL3-like native a-helical structure in these peptides,
which is thought to be important for the presentation of
binding residues in the correct orientation to the binding
pocket of the receptor. Therefore, the aim of this study is
to systematically examine known methods, including
introduction of disulfide and lactam constraints or a-helix-
inducing residues and N-caps, to induce additional
a-helicity in the B-chain mimetics of INSL3 and to eval-
uate their effectiveness as INSL3 antagonists.
Materials and methods
9-Fluroenylmethoxycarbonyl (Fmoc) protected L-a-amino
acids, 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate (HBTU), N,N-dimethylformamide
(DMF), piperidine and trifluoroacetic acid (TFA) were
obtained from Auspep (West Melbourne, Australia). Fmoc-
Aib-OH, Fmoc-Asp(O-2-PhiPr)-OH, Fmoc-Dab(Mtt)-OH,
Fmoc-Glu(O-2-PhiPr)-OH, Fmoc-Lys(Mtt)-OH and PyBOP
were obtained from Novabiochem (Melbourne, Australia).
Fmoc-PAL-PEG-PS and Fmoc-L-Ala-PEG-PS resins
with substitution of 0.20 mmol/g were purchased from
Applied Biosystems (Melbourne, Australia). Methanol,
diethylether, dichloromethane (Merck, Melbourne, Austra-
lia); 3,6-dioxa-1,8-octanedithiol (DODT), triisopropylsilane
(TIPS), diisopropylethylamine (DIPEA), 1,2,4,5-benzene-
tetracarboxylic dianhydride (Sigma-Aldrich, Sydney,
Australia); 2,20-dipyridyl disulfide (DPDS), (Fluka-
Switzerland); acetonitrile and NH
4
HCO
3
, (NH
4
)
2
CO
3
(BDH
Laboratory Supplies, Poole, UK); and trifluoromethanesul-
fonic acid (TFMSA) (MP Biomedicals, Sydney, Australia).
Dulbecco’s modified Eagles’ medium (DMEM), RPMI 1640
medium, 2 mM L-glutamine, foetal calf serum and penicil-
lin/streptomycin were all obtained from Trace Biosciences
(Sydney, Australia). All other reagents were obtained from
Sigma-Aldrich (Sydney, Australia).
Molecular modelling
All molecular modelling was performed using SYBYL
molecular modelling software (Tripos, version 7.0, St
Louis, MO, USA) on a Silicon Graphics O
2
workstation.
Design of disulfide constrained mimetics
All the single chain disulfide constrained mimetics were
designed as described previously (Shabanpoor et al. 2007).
Briefly, using the NMR structure of native human INSL3
as a template, the A-chain was deleted and a disulfide bond
was inserted between b-carbon atoms of residues less than
10 A
˚apart on the strand and a-helical segments of the
B-chain. The two chain disulfide constrained analogue 30
(Fig. 1b) was designed by truncating the B-chain strand
from N-terminus up to Leu
B9
and from the C-terminus until
Trp
B27
, and the A-chain was truncated from N-terminus
until Cys
A15
and from C-terminus until Cys
A24
. In native
INSL3, Cys
A24
forms a disulfide bond with Cys
B22
so,
hence, there was no need for creation of a disulfide bond.
Cys
B10
, which pairs with Cys
A11
in an inter-chain disulfide
bond, was mutated to serine (Ser
B10
). On the other hand,
Cys
A15
forms an intra-A-chain disulfide bond with Cys
A10
and in order to from the second disulfide bond, Leu
B9
,
which points toward Cys
A15
, was mutated to Cys
B9
and
then a disulfide bond was created between this pair of
cysteines. Finally, the two-chain disulfide constrained
analogue was energy minimized in vacuo as described
previously (Shabanpoor et al. 2007) using the Powell
method with the Tripos force field, Gasteiger-Marsili
charges and termination at a root mean square (RMS)
gradient of less than 0.05 kcal/mol per A
˚.
Design of i to i ?4 lactam constrained mimetics
In designing lactam constrained mimetics, we first
inspected the NMR structure of the INSL3 B-chain for an
optimum place to introduce the lactam. Phe
B14
and Leu
B18
,
spaced iand i?4 on one face of the helix opposite to the
side where the key receptor binding residues were located,
was observed to be a suitable place to introduce a lactam
constraint. We designed a series of lactam constrained
analogues of INSL3 B-chain where we truncated the
B-chain from the C-terminus until Trp
B27
and to Pro
B1
at
the N-terminus. Some of the lactam constrained analogues
were truncated further from the N-terminus to Gly
B11
.
Effect of helix-promoting strategies on the biological activity 123
123
Following this truncation, Gly
B11
was mutated to Ala
B11
,
then Phe
B14
was mutated to Lys
B14
or Dab
B14
, and Leu
B18
was mutated to Glu
B18
or Asp
B18
(Fig. 3). Finally, an amide
bond was created between the e-amino group of the Lys or
Dab side-chain and carbonyl group of either the Glu or Asp
side-chain, and the resultant analogues were energy mini-
mized as described earlier.
Incorporation of a-helix-inducing residues and N-caps
The INSL3 B-chain was truncated from the N-terminus up
to Gly
B11
, which was then mutated to a more helix-
favouring residue, Ala
B11
. Ala
B17
was mutated to a more
helix-inducing residue, a-aminoisobutyric acid (Aib).
Valine residues along B-chain helix were mutated to either
Ala or Aib. The N-cap, 2,4,5 benzenecarboxylate, which is
known to stabilize helices by acting as a surrogate H-bond
acceptor (Mimna et al. 2007), was coupled to the N-ter-
minus of the INSL3 B-chain helix.
Solid-phase peptide synthesis
In order to increase the enzymatic stability of the ana-
logues, all linear precursor peptides were synthesized as
C-terminal amides (Werle and Bernkop-Schnu
¨rch 2006)on
PAL-PEG-PS resin with 0.19–0.22 mmol/g loadings using
Fmoc chemistry. The side chain protected amino acids used
were: Arg(Pbf), Asp(OPip), Cys(Trt), Cys(Acm), Cys(tBu),
Glu(OPip), Glu(OtBu), His(Trt), Lys(Boc), Lys(Mtt) and
Trp(Boc). Peptides were synthesized on either a Pioneer
peptide synthesizer (PerSeptive Biosystems, MA, USA)
using continuous flow methodology or a microwave
peptide synthesizer (CEM, Liberty, Matthews, USA). In
continuous flow syntheses, the coupling of Fmoc protected
L-a-amino acids was accomplished using HBTU
(0.3 mmol) and DIPEA in DMF (5 ml) for 30 min and
Fmoc protecting groups were removed by treating the
resin-attached peptide with piperidine (20% v/v) in DMF
for 20 min. For microwave-assisted syntheses, a fivefold
excess of amino acid and HBTU and a tenfold excess of
DIEA were used, and the coupling and deprotection were
carried out at 75°C using 25 W microwave power for
5 min and 60 W microwave power for 3 min, respectively.
The single chain disulfide-constrained peptides were
synthesized as described previously (Shabanpoor et al.
2007). Analogues 30 and 31 (Table 1) with two inter-chain
disulfide bonds were synthesized with two Cys(Trt)s and
two Cys(Acm)s, one of each in either chain. The formation
of a disulfide bond between the two Cys(Trt) was carried
out by dissolving the A and B-chains in an equimolar ratio
in 0.1 M NH
4
CO
3
, adding 300 ll of 100 mM DPDS and
stirring the reaction mixture for 30 min. The second inter-
chain disulfide bond was formed by first dissolving the
peptide in acetic acid (2 mg/ml) followed by the addition
of 60 mM HCl (0.1 ml/mg) and 20 mM I
2
(42 eq/Acm).
The reaction mixture was stirred at room temperature for
1 h and the progression of the reaction was monitored by
HPLC.
The all-linear form of the lactam-constrained peptides
were synthesized at the 0.1-mmol scale on PAL-PEG-PS
resin (substitution 0.20 mmol/g) using a microwave-assis-
ted peptide synthesizer and the conditions described above.
The formation of an amide bond between the side chains of
two residues, Lys or Dab and Glu or Asp, was carried out
on-resin. The phenylisopropyl ester (OPip) of aspartic and
glutamic acids and methyltrityl (Mtt) group of lysine and
Dab were removed by treating the peptide resin with 3%
TFA/5% TIPS in DCM (2 930 min) (Shepherd et al.
2006). The on-resin cyclization was carried out in three
different ways. In the first instance, we attempted to cyclize
the peptide on-resin using a standard protocol of coupling
with 3 equivalents of HBTU and 3.5 equivalents of DIPEA
in 3 ml of DMF overnight. Second, the resin-bound peptide
was treated with PyBOP/HOAt/DIPEA (3:3:3.5) in 3 ml of
DMF/DMSO/NMP (1:1:1) overnight. Finally, the cycliza-
tion was carried out in a microwave-assisted peptide
synthesizer using HBTU (3 eq) DIPEA (3.5 eq) for 10 min
at 75°C, 25 W.
The syntheses of peptides with helicogenic residues and
N-caps were carried out in the same way as for the disulfide
constrained mimetics. The N-terminus was either capped
with acetic anhydride (10 eq) or 1,2,4,5-benzene-tetra-
carboxylic dianhydride (10 eq) in DMF in the presence of
DIEA (10 eq).
The cleavage of peptides was carried out using a TFA:
H
2
O:DODT:TIS (94:2.5:2.5:1, 20 ml) mixture for 90 min.
Cleaved peptides were precipitated in ice-cold diethyl ether,
centrifuged at 3,000 rpm for 3 min; the pellet was washed by
resuspending it in ice-cold diethylether and centrifuging it
again for three times. Peptides were analysed and purified by
RP-HPLC on Waters XBridge
TM
columns (4.6 9250 mm,
C18, 5 lm) and (19 9150 mm, C18, 5 lm), respectively,
using H
2
O with 0.1% TFA as solvent A and acetonitrile with
0.1% TFA as solvent B, with a gradient of 1% change in
buffer B per min over 30 min. Peptide 24 (Table 1) N-cap-
ped with 1,2,4,5-benzene-tetracarboxylic dianhydride was
dissolved in 1 M (NH
4
)
2
CO
3
and lyophilized before HPLC
analysis and purification.
Matrix-assisted laser desorption ionization time-of-flight
mass spectrometry (MALDI-TOF/TOF MS, Bruker Dal-
tonics, Germany) was used to characterize the peptides at
each intermediate step using sinapinic acid, a-cyano-4-
hydroxy-cinnamic acid and 2,5-dihydroxy benzoic acid
(Bruker Daltonics, Germany) as matrices, based on the
molecular size of a peptide. The matrices were made up in
50% acetonitrile containing 0.05% TFA. The peptide
124 F. Shabanpoor et al.
123
Table 1 Primary amino acid sequence, monoisotopic mass, calculated and theoretical a-helicity in PBS and 20% TFE, and binding affinity (pK
i
,
n=3) of INSL3 analogues
Peptide No Sequence pKi
Calcul Exper PBS 20% TFE Theo Mean ± SEM (n=3)
H-PTPEMREKLCGHHFVRALVRVCGGPRWSTEA-OH
H-AAATNPARYCCLSGCTQQDLLTLCPY-OH
1 Ac-TPEMREKLSGHHFVRALVRVSGGPRW-NH23044.5 3045 10 55 42 5.31 ± 0.24
2H2N-CPEMREKLSGHHFVRALVRCSGGPRW-NH23009.6 3009.7 8 37 42 6.09 ± 0.05
3Ac-
CPEMREKLSGHHFVRALVRCSGGPRW-NH2
3050.5 3050.9 8 30 42 6.41 ± 0.11
4Ac-
CPEMREKLSGAHFVRALVRCSGGPRW-NH2
2982.5 2982.8 10 32 42 5.1 ± 0.09
5Ac-CPEMREKLSGHHFVAALVRCSGGPRW-NH2
2965.4 2965.6 11 35 42 <4
6Ac-
CPEMREKLSGHHFVRALARCSGGPRW-NH2
3018.5 3019 8 28 42 5.07 ± 0.06
7Ac-CPEMREKLSGHHFVRALVACSGGPRW-NH2
2965.4 2965.6 9 35 42 5.93 ± 0.06
8Ac-CPEMREKLSGHHFVRALVRCSGGPRA-NH22934 2934.6 8 34 42 NB
9Ac-CPEMREKLSGHHFVAALVRCSGGPRBAl-NH23068 3068.3 - - - 5.65 ± 0.08
10 Ac-CPEMREKLSGHHFVRALARCSGGPR(1NAL)-NH23061 3061 - - - 5.63 ± 0.03
11 Ac-CPEMREKLSGHHFVRALVRCSAAARW-NH23052.6 3052.6 8 41 42 5.74 ± 0.20
12
Ac-CPEMREKLSGHHCVRACVRCSGGPRW-NH22989.5 2989.4 5 12 42 < 4
13 Ac-TPEMREKLSGHHCVRACVRVSGGPRW -NH22986.4 2987 8 25 42 NB
14 Ac-TPEMREKLSGHHDVRAKVRVSGGPRW-NH23010.5 3010.9 17 47 42 5.82 ± 0.08
15 Ac-TPEMREKLSGHHEVRAKVRVSGGPRW -NH23023.5 3023.8 18 58 42 6.09 ± 0.08
16 Ac-TPEMREKLSGHHEVRADabVRVSGGPRW -NH22996.5 2966.8 11 14 42 <5
17 Ac-AHHKVRADVRVSGGPRW -NH21952.2 1952.4 17 36 65 <5
18 Ac-AHHDVRAKVRVSGGPRW -NH21952.2 1952 8 27 65 <5
19 Ac-AHHDVRAKVRVSGGPRW -NH21970.1 1970.1 7 22 65 <5
20 Ac-AHHKVRAEVRVSGGPRW -NH21965.2 1965.7 4 17 65 <5
21 Ac-AHHEVRAKVRVSGGPRW -NH21965.2 1965.7 17 48 65 <5
22 Ac-AHHEVRAKVRVSGGPRW -NH21983.2 1983.7 7 23 65 <5
23 H
2
N-AHHFVRALVRVSGGPRW-NH
2
1944.1 1944.04 7 49 65 <5
24 Ac-AHHFVRALVRVSGGPRW -NH
2
1986.1 1986.8 8 46 65 <5
25 Da-AHHFVRALVRVSGGPRW -NH
2
2181.1 2181.3 6 41 65 <5
26 Ac-AHHFVRAibLVRVSGGPRW -NH
2
2001.5 2001.1 7 46 65 <5
27 Ac-GHHFAibRALVRAib SGGPRW-NH
2
1944 1944.9 7 21 65 <5
28 Ac-GHHFAibRAibLVRAibSGGPRW-NH
2
1958 1959.2 7 18 65 5.5 ± 0.12
29 Ac-AHHFV(A)RALVRV(A)SGGPRW-NH
2
1930.2 1930.7 7 43 65 <5
Ac-CTQQDLLTLC-NH2
CSAHHFVRALVRVCGGPRW
H-PTPEMREKLCGHHFVRALVRVCGGPRWSTEA-OH
H-PTPEMREKLCGHHFVRALVRVCGGPRW STEA-OH
020.0 ± 41.713328.0733073303 58
9.27 ± 0.06
-
%α-helix [M+H]
7029.52 32 53 42 8.43 ± 0.06
3339268.29263LSNIn -
5.920713
NB No binding
Effect of helix-promoting strategies on the biological activity 125
123
content was determined using vapour-phase acid hydrolysis
in 6 M HCl containing 2% phenol at 110°C for 24 h. The
individual amino acids were converted to stable, fluores-
cent derivatives using Waters AccQ.Tag kit (Waters,
Sydney, Australia). The derivatized amino acids were
separated using a Shim-Pak XR-ODS (3 975 mm,
2.2 lm) column on a Shimadzu RP-HPLC system (Shi-
madzu, VIC, Australia). The concentrations of individual
amino acids were standardized against an internal standard
(norvaline) at a concentration of 100 pmol/ll sample
injected.
Circular dichroism spectroscopy
The peptides were made up to a concentration of 0.1 lMin
phosphate buffered saline (PBS: 10 mM potassium phos-
phate buffer containing 137 mM NaCl pH 7.4). The far UV
circular dichroism (CD) spectra of peptides were acquired
using a JASCO model J815 spectropolarimeter between the
wavelengths of 195–250 nm at room temperature with a
resolution of 0.1 nm, bandwidth of 0.1 nm and a cell of
0.1 cm path length (P). The recorded spectra in millidegrees
of ellipticity (h) were converted to mean residue ellipticity
(MRE) in degcm
2
dmol
-1
. The CD spectra data were first
transformed from machine unit hto delta epsilon (De) using
GraphPad PRISM 4 (GraphPad Inc., San Diego, USA) with
a user defined formula (De=h9(0.1 9MRW)/
(P 9C)93,298) (C: Peptide concentration, MRW: Pep-
tide molecular weight/number of residues). The converted
values were then submitted to the DichroWeb server (Lees
et al. 2006; Lobley and Wallace 2001; Lobley et al. 2002;
Whitmore and Wallace 2004) for the calculation of sec-
ondary structure using the CDSSTR (Compton and Johnson
1986) and K2D (Andrade et al. 1993) analysis algorithms.
Ligand binding assay
Human embryonic kidney (HEK)-293T cells stably trans-
fected with RXFP2 and europium-labelled INSL3 were
used in whole cell binding assays. Cells were plated out at
a density of 80,000 cells per 200 ll per well in 96 well
Isoplate with white wall and clear bottom precoated with
poly-L-lysine. Competition binding experiments were car-
ried out as described previously (Shabanpoor et al. 2008)
with 300 pM of Eu-DTPA-INSL3 (K
d
: 0.892 nM) in
presence of increasing concentrations of peptide analogues.
Non-specific binding was determining with an excess
(500 nM) of unlabelled INSL3. Each concentration point
was performed in triplicate and the data expressed as the
mean ±SEM (standard error of mean) of the percentage of
total specific binding of triplicates from at least three
independent experiments. Curves were fitted using a one-
site binding model in GraphPad PRISM 4 (GraphPad Inc.,
San Diego, USA). The inhibition constants (K
i
) were
determined from IC
50
values using the Cheng-Prusoff
equation, and the statistical differences in pK
i
values were
calculated using one-way ANOVA followed by Bonfer-
roni’s multiple comparison test for multiple group
comparisons.
Functional cAMP assay
A cAMP reporter gene assay was used to assess the receptor
signalling of INSL3 and analogue 31 in HEK-293T cell line
co-transfected with RXFP2 (LGR8) and a pCRE-b-galac-
tosidase reporter plasmid. The assay was carried out as
described previously (Scott et al. 2006). Briefly, co-trans-
fected cells were incubated with increasing concentrations
of INSL3 and analogue 31 for 6 h after which the medium
was removed and cells frozen at -80°C overnight. The
amount of cAMP-driven b-galactosidase expression was
measured by lysing the cells. Each concentration point was
performed in triplicate and the data expressed as the
mean ±SEM of three independent experiments.
Results and discussion
INSL3 binds to its receptor principally using residues
confined to the B-chain. Deletion of the INSL3 A-chain
leads to the loss of B-chain a-helical structure and, there-
fore, loss of receptor binding affinity of the B-chain in
isolation. The a-helix is the most abundant secondary
structure which accounts for 30% of all protein residues
(Barlow and Thornton 1988). The helix plays a crucial role
in many protein-mediated biological processes such as
receptor binding (Beck-Sickinger and Jung 1995) and thus
is an attractive target for the design of mimetics. However,
peptides derived from these regions may not be biologi-
cally active when in isolation. This is usually due to the
loss of a-helicity where the peptides exhibit little or no
secondary structure as a result of loss of stabilizing inter-
actions within the parent protein. As part of our ongoing
effort to design short mimetics of INSL3 B-chain with
antagonistic action, we have utilized and evaluated various
possible ways of increasing the a-helicity within the
B-chain of INSL3. Towards this aim, we have designed
three series of analogues that include disulfide (primarily
helix to strand) constraints, ito i?4 lactam constraints,
and helix-inducing N-caps and residues (Table 1).
All analogues were synthesized on the solid-phase and
subjected to analysis by RP-HPLC and mass spectrometry.
The on-resin cyclization of lactam analogues was difficult.
In the first instance, using standard coupling procedures,
the reaction did not give the desired cyclic compound. A
second attempt using different coupling reagents, PyBOP
126 F. Shabanpoor et al.
123
and HOAt, gave the lactam but also resulted in the for-
mation of various major side products. Best results were
obtained using HBTU as coupling reagent and heating to
75°C using microwave power, in that the reaction went to
completion with minor side-products formation only and
an overall yield of 10–15%.
The introduction of a disulfide constraint between the
strand and the helix of the B-chain had little impact on the
level of helicity but improved the receptor binding affinity
in the analogue 3(pK
i
=6.41 ±0.11) compared to its
linear counterpart analogue 1(pK
i
=5.31 ±0.24). In
order to determine the mode of receptor interaction of this
analogue, the residues H
B12
,R
B16
,V
B19
,R
B20
and W
B27
,
which have been shown previously to be important for
binding of native INSL3 to RXFP2, were each mutated in
analogue 3. Alanine mutation of Arg
B16
significantly
reduced the receptor binding affinity of analogue 3(Fig. 2).
The replacement of His
B12
and Val
B19
with Ala also caused
40–50 times reduction in the binding affinity of analogue 3
whereas replacement of Arg
B20
caused only a slight drop in
receptor binding affinity. Finally, replacement of Trp
B27
in
this analogue with Ala led to almost complete loss of
receptor binding affinity. This trend of single replacement
of binding residues in the B-chain disulfide constrained
analogue and its loss of receptor binding affinity resembles
that of native INSL3 (Rosengren et al. 2006), suggesting
that, despite their lower affinity for RXFP2, the B-chain
mimetics bind to the receptor in the same fashion as native
INSL3.
The small improvement in receptor binding of the linear
B-chain with the disulfide constraint between the strand
and the a-helix was probably not as a result of induction of
further a-helical structure as there was little difference in
the level of a-helicity between the linear compound 1and
its cyclic counterpart analogue 3. It is likely that the
disulfide constraint between the strand and the helix of the
B-chain pulls the helix and the strand—very flexible in
native INSL3—closer together and this is possibly causing
a conformational change in the spatial orientation of the
Trp
B27
at the C-terminus of the B-chain which might be
better placed for interaction with the receptor. To further
investigate this possibility, three residues (G
B23,24
and
P
B25
) toward the C-terminus of the B-chain were replaced
with alanine. This change resulted in an analogue (11) with
binding affinity (pK
i
=5.74 ±0.2, n=3) similar to that
of linear B-chain (analogue 1). The GGP residues appear to
act as a hinge that provides flexibility for the B-chain C-
terminal region and this flexibility may be required for
proper interaction of Trp
B27
with the receptor. Since Trp
B27
is a crucial binding residue in the B-chain analogues,
we further investigated the role of both the individual
indole and benzene rings by replacing Trp
B27
in analogue 3
with b-(benzothien-3-yl)alanine (Bal, analogue 9) and
b-(naphtha-1-yl)alanine (1-Nal, analogue 10). Bal is an
analogue of Trp that has sulfur instead of nitrogen in the
indole ring and 1-Nal is another analogue in which the
indole ring is replaced with a benzene ring. The drop in
the level of receptor binding affinity of these two analogues
was similar which shows both rings of Trp
B27
are equally
important in receptor interaction.
Further constraining analogue 3by incorporating an ito
i?4 disulfide constraint along the helix at Phe
B14
and
Leu
B18
gave analogue 12 which exhibited low level of
helicity even in the presence of 20% TFE. This peptide had
a very low level of receptor affinity compared to analogue
3. The ito i?4 disulfide constraint along the helix would
cause analogue 12 to lose its flexibility around the helical
region and prevent the peptide from adopting an active
conformation for high affinity receptor binding. This was
investigated by inserting only the ito i?4 disulfide
constraint at Phe
B14
and Leu
B18
in analogue 13.This
analogue did not show any affinity for receptor binding,
which is likely due to the loss of flexibility and inability to
adopt a conformation with the key receptor binding resi-
dues in a correct orientation to the binding pocket on the
receptor.
The introduction of a lactam bridge in the B-chain was
the second approach that was investigated for the restora-
tion of helical structure in the isolated B-chain of INSL3.
Side chain–side chain lactam bridges have long been used
as a convenient and flexible method for introducing con-
formational constraints into a peptide structure. The points
to consider in the design of peptides with a lactam bridge
are the spacing of the two residues to be linked, the side-
chain orientation and the position of the bridge. A Glu–Lys
Fig. 2 Competition binding studies of native INSL3, disulfide
constrained analogue 3and its Ala-substituted analogues (Table 1).
Europium-labelled-INSL3 (0.3 nM) was used as a labelled ligand in
the competition binding assay in the presence of increasing concen-
tration of INSL3 and analogues. Singly substituting R
B12
(5) and
W
B27
(8) with Ala caused these peptides to almost completely lose
their receptor binding affinity. Replacing H
B12
(4) and V
19
(6) with
alanine also led to a major loss of receptor binding affinity. Mutation
of R
B20
had minor impact in the level of binding affinity
Effect of helix-promoting strategies on the biological activity 127
123
(E-K) lactam at the spacing of iand i?4 in the middle of
the peptide has been shown to be more effective at stabi-
lizing helical structure than two Glu-Lys lactams
positioned one at each end of the peptide (Houston et al.
1995). In the B-chain of INSL3, we introduced a lactam
bridge (i,i?4) in the mid-region of the B-chain a-helix
by mutating Phe
B14
and Leu
B18
to Glu, Asp, Lys or Dab.
Various orientations of the lactam bridge were evaluated in
the long form (analogues 1416) (Fig. 3a) and truncated
B-chain of INSL3 (analogues 1722).
Analogues 14 and 15 had higher a-helicity in PBS
(17 and 18%, respectively) compared to the linear analogue
1(10%); this small improvement in a-helicity was
accompanied by a small increase in receptor binding
affinity. In analogue 15, Lys
B18
was replaced with diam-
inobutyric acid (Dab) (analogue 16). This modification was
observed to not only reduce the level of a-helicity but also
cause a drop in receptor binding affinity (Table 1). This
may be due to the shorter side-chain of Dab which, on
cyclization with the Glu side-chain, results in an unfa-
vourable conformational change in the B-chain. The
introduction of a lactam bond in the truncated INSL3
B-chain analogues had little effect on either a-helicity or
receptor binding. Analogues 17 and 21with K-D and E-K
lactam orientation, respectively, were more effective in
inducing helical structure compared to D-K and K-E lac-
tams in analogues 18 and 20. Analogue 18 with D-K lactam
orientation did not have higher a-helicity compared to its
linear counterpart analogue 19. On the other hand, ana-
logue 21with E-K lactam orientation exhibited higher
a-helicity compared to its linear analogue 22. Both the
short and long forms of E-K orientated lactams (15 and 21)
had the highest helical content both in PBS and 20% TFE
compared to any other lactam orientation and this is in
accordance with previous literature reports (Houston et al.
1995). Despite some of the lactam constrained analogues
being able to induce a-helicity in the B-chain, this did not
result in high affinity binding. The introduction of a lactam
bridge in the mid-region of the B-chain backbone likely led
to the loss of B-chain flexibility and a more rigid structure,
which is no longer capable of adjusting binding residues to
align with the binding pocket on the receptor.
Analogues 2325 show the effect of ‘capping’ the
N-terminal of the truncated B-chain with acetyl and 2,4,5
benzenecarboxylate (Fig. 3B), the latter which has recently
been reported to increase the a-helicity of a model peptide
from 17 to 70% by providing carbonyl groups that act as
surrogate H-bond acceptors (Mimna et al. 2007). The
N-terminus capping of the B-chain a-helix was not effec-
tive in inducing a-helicity in PBS but in 20% TFE the level
of a-helicity increased significantly, which was likely a
reflection of the tendency of these peptides to adopt a
helical structure.
The a-helix-inducing effect of residues such as Ala and
Aib on the INSL3 B-chain helix was also investigated. The
use of a-helicogenic residues in enhancing the activity of
peptides has been demonstrated by substituting these at
specific positions in the helical N-terminal fragment of
parathyroid hormone PTH(1–11), which made the resulting
peptide 3,500 times more potent (Barazza et al. 2005). The
a-helix stabilization of alanine has been related to the
hydrophobic interaction, steric effects and solvation of
the polar groups of the a-helix backbone (Avbelj 2000;
Avbelj and Fele 1998; Bai and Englander 1994; Bai et al.
1993; Blaber et al. 1993,1994; Connelly et al. 1993).
Aib-based peptides have a remarkable tendency to form
a-helical conformations in solution due to the restricted
rotation about (N-Ca) and (Ca-C
1
) bonds, which is caused
by the presence of two methyl groups on the Caatom
(Ma et al. 2007; Marshall et al. 1990). Alanine and Aib
residues were incorporated along the a-helical segment of
INSL3 B-chain in place of valine in analogues 2629.
None of these analogues had high helicity in PBS, although
the helicity increased upon addition of 20% TFE. These
peptides also showed very low receptor binding affinity due
to the lack of helical structure.
W27
L18 R20
R16
F14
V19
H12
O
OO
O
OH
OH
OH
N-term inus
C-terminus
BA
W27
R20
R16
V19
H12
K18
E14
E18
K14
K18
D14
D18
K14
1, 2, 4, 5-Benzene-tetraca rbox
y
lic d ia nh
y
dride
Fig. 3 a An INSL3 B-chain
analogue in which a lactam
constraint was incorporated in
the middle of the a-helix on the
side opposite to where the
binding residue is located.
F
14
and L
18
were replaced with
K, E, D or Dab in different
orientations. bTruncated INSL3
B-chain until G
B11
and the
N-terminus capped with either
acetic anhydride or 1,2,4,5-
benzene-tetracarboxylic
dianhydride
128 F. Shabanpoor et al.
123
The disulfide constrained analogue 30, where the
B-chain helix was stabilized using the C-terminal a-helical
region of the A-chain and analogue 31, which is a B-chain
dimer, exhibited 23 and 32% a-helicity in PBS, which was
higher than any other single B-chain analogues. The higher
level of a-helicity was accompanied by an increase in
receptor binding affinity of these two analogues (Fig. 4a).
To determine if 31 was able to activate the receptor and
induce intracellular cAMP production, the analogue was
tested in HEK-293T cells expressing RXFP2 (LGR8).
Native INSL3 induced cAMP production (pEC50 =
10.35 ±0.12) whereas analogue 31 was unable to activate
the receptor as intracellular cAMP accumulation was not
induced (Fig. 4b), which suggests that this peptide is an
antagonist of INSL3.
In summary, a systematic approach was taken to induce
a-helicity in the isolated INSL3 B-chain and study the
binding mode of interaction with the receptor. Isolated
INSL3 B-chain that was constrained with a disulfide bond
was found to bind to the receptor in the same way as native
INSL3. Attempting to induce an a-helical conformation by
constraining the B-chain of INSL3 did not fully compen-
sate for the stabilizing interactions of the missing A-chain
as constraining the B-chain by either a disulfide or lactam
bond did not increase a-helicity and receptor binding
affinity simultaneously. However, constraining the B-chain
with a short region of A-chain or dimerizing the B-chain
not only increased the a-helicity but also the receptor
binding affinity. On this basis, we have identified two novel
INSL3 antagonists which can be used as lead compounds
to be further minimized and optimized for development as
clinically useful INSL3 antagonists which may be used as
potential male and female contraceptives.
Acknowledgments We thank Tania Ferraro for help with binding
assays. This work was funded by National Health and Medical
Research Council of Australia Project grants #350245 and 509048 to
JDW, RADB and RAH.
References
Adham IM, Burkhardt E, Benahmed M et al (1993) Cloning of a
cDNA for a novel insulin-like peptide of the testicular Leydig
cells. J Biol Chem 268:26668–26672
Andrade MA, Chacon P, Merelo JJ et al (1993) Evaluation of
secondary structure of proteins from UV circular dichroism
spectra using an unsupervised learning neural network. Protein
Eng 6:383–390. doi:10.1093/protein/6.4.383
Avbelj F (2000) Amino acid conformational preferences and solva-
tion of polar backbone atoms in peptides and proteins. J Mol Biol
300:1335–1359. doi:10.1006/jmbi.2000.3901
Avbelj F, Fele L (1998) Role of main-chain electrostatics, hydro-
phobic effect and side-chain conformational entropy in
determining the secondary structure of proteins. J Mol Biol
279:665–684. doi:10.1006/jmbi.1998.1792
Bachelot T, Ray-Coquard I, Catimel G et al (2000) Multivariable
analysis of prognostic factors for toxicity and survival for
patients enrolled in phase I clinical trials. Ann Oncol 11:151–
4773. doi:10.1023/A:1008368319526
Bai Y, Englander SW (1994) Hydrogen bond strength and beta-sheet
propensities: the role of a side chain blocking effect. Proteins
18:262–266. doi:10.1002/prot.340180307
Bai Y, Milne JS, Mayne L et al (1993) Primary structure effects on
peptide group hydrogen exchange. Proteins 17:75–86. doi:
10.1002/prot.340170110
Barazza A, Wittelsberger A, Fiori N et al (2005) Bioactive N-terminal
undecapeptides derived from parathyroid hormone: the role of
alpha-helicity. J Pept Res 65:23–35. doi:10.1111/j.1399-3011.
2005.00207.x
Barlow DJ, Thornton JM (1988) Helix geometry in proteins. J Mol
Biol 201:601–619. doi:10.1016/0022-2836(88)90641-9
Beck-Sickinger AG, Jung G (1995) Structure-activity relationships of
neuropeptide Y analogues with respect to Y1 and Y2 receptors.
Biopolymers 37:123–142. doi:10.1002/bip.360370207
Fig. 4 a Comparison of competition binding curves of INSL3
(pK
i
=9.27 ±0.06) with analogues 30 (pK
i
=7.14 ±0.02) and
31 (pK
i
=8.43 ±0.06). As the level of a-helicity increased
(Table 1) so does the receptor binding affinity. bFunctional cAMP
studies of nINSL3 analogue 31. Native INSL3 can induce cAMP
production (pEC50 =10.35 ±0.12) in HEK-293T cells co-transfec-
ted with RXFP2 (LGR8) and pCRE-b-galactosidase whereas
treatment of these cells with analogue 31 does not induce cAMP
production, i.e. it is an antagonist. Each concentration point was
performed in triplicate and the data are expressed as the mean ±SEM
of three independent experiments
Effect of helix-promoting strategies on the biological activity 129
123
Blaber M, Zhang XJ, Matthews BW (1993) Structural basis of amino
acid alpha helix propensity. Science 260:1637–1640. doi:10.1126/
science.8503008
Blaber M, Zhang XJ, Lindstrom JD et al (1994) Determination of
alpha-helix propensity within the context of a folded protein.
Sites 44 and 131 in bacteriophage T4 lysozyme. J Mol Biol
235:600–624. doi:10.1006/jmbi.1994.1016
Bogatcheva NV, Truong A, Feng S et al (2003) GREAT/LGR8 is the
only receptor for insulin-like 3 peptide. Mol Endocrinol
17:2639–2646. doi:10.1210/me.2003-0096
Bu
¨llesbach EE, Schwabe C (1995) A novel Leydig cell cDNA-
derived protein is a relaxin-like factor. J Biol Chem 270:16011–
16015. doi:10.1074/jbc.270.27.16011
Bu
¨llesbach EE, Schwabe C (1999) Tryptophan B27 in the relaxin-like
factor (RLF) is crucial for RLF receptor-binding. Biochemistry
38:3073–3078. doi:10.1021/bi982687u
Bu
¨llesbach EE, Schwabe C (2005) LGR8 signal activation by the
relaxin-like factor. J Biol Chem 280:14586–14590. doi:10.1074/
jbc.M414443200
Bu
¨llesbach EE, Schwabe C (2007) Structure of the transmembrane
signal initiation site of the relaxin-like factor (RLF/INSL3).
Biochemistry 46:9722–9727. doi:10.1021/bi700708s
Burkhardt E, Adham IM, Hobohm U et al (1994) A human cDNA
coding for the Leydig insulin-like peptide (Ley I-L). Hum Genet
94:91–94. doi:10.1007/BF02272850
Compton LA, Johnson WC Jr (1986) Analysis of protein circular
dichroism spectra for secondary structure using a simple matrix
multiplication. Anal Biochem 155:155–167. doi:10.1016/
0003-2697(86)90241-1
Connelly GP, Bai Y, Jeng MF et al (1993) Isotope effects in peptide
group hydrogen exchange. Proteins 17:87–92. doi:10.1002/
prot.340170111
Del Borgo MP, Hughes RA, Bathgate RA et al (2006) Analogs of
insulin-like peptide 3 (INSL3) B-chain are LGR8 antagonists in
vitro and in vivo. J Biol Chem 281:13068–13074. doi:10.1074/
jbc.M600472200
Feng S, Cortessis VK, Hwang A et al (2004) Mutation analysis of
INSL3 and GREAT/LGR8 genes in familial cryptorchidism.
Urology 64:1032–1036. doi:10.1016/j.urology.2004.06.051
Ferlin A, Pepe A, Gianesello L et al (2008) Mutations in the insulin-
like factor 3 receptor are associated with osteoporosis. J Bone
Miner Res 23:683–693. doi:10.1359/jbmr.080204
Foresta C, Ferlin A (2004) Role of INSL3 and LGR8 in cryptorchi-
dism and testicular functions. Reprod Biomed Online 9:294–298
Hossain MA, Rosengren KJ, Haugaard-Jonsson LM et al (2008) The
A-chain of human relaxin family peptides has distinct roles in
the binding and activation of the different relaxin family peptide
receptors. J Biol Chem 283:17287–17297. doi:10.1074/jbc.
M801911200
Houston ME Jr, Gannon CL, Kay CM et al (1995) Lactam bridge
stabilization of alpha-helical peptides: ring size, orientation and
positional effects. J Pept Sci 1:274–282. doi:10.1002/psc.
310010408
Hsu SY (2003) New insights into the evolution of the relaxin-
LGR signaling system. Trends Endocrinol Metab 14:303–309.
doi:10.1016/S1043-2760(03)00106-1
Ivell R, Einspanier A (2002) Relaxin peptides are new global players.
Trends Endocrinol Metab 13:343–348. doi:10.1016/S1043-
2760(02)00664-1
Kawamura K, Kumagai J, Sudo S et al (2004) Paracrine regulation of
mammalian oocyte maturation and male germ cell survival. Proc
Natl Acad Sci USA 101:7323–7328. doi:10.1073/pnas.
0307061101
Klonisch T, Mustafa T, Bialek J et al (2005) Human medullary
thyroid carcinoma: a source and potential target for relaxin-like
hormones. Ann N Y Acad Sci 1041:449–461. doi:10.1196/
annals.1282.069
Lees JG, Miles AJ, Wien F et al (2006) A reference database for
circular dichroism spectroscopy covering fold and secondary
structure space. Bioinformatics 22:1955–1962. doi:10.1093/
bioinformatics/btl327
Lobley A, Wallace BA (2001) DICHROWEB: a website for the
analysis of protein secondary structure from circular dichroism
spectra. Biophys J 80:373
Lobley A, Whitmore L, Wallace BA (2002) DICHROWEB: an
interactive website for the analysis of protein secondary structure
from circular dichroism spectra. Bioinformatics 18:211–212.
doi:10.1093/bioinformatics/18.1.211
Ma S, Bonaventure P, Ferraro T et al (2007) Relaxin-3 in GABA
projection neurons of nucleus incertus suggests widespread
influence on forebrain circuits via G-protein-coupled receptor-
135 in the rat. Neuroscience 144:165–190. doi:10.1016/j.
neuroscience.2006.08.072
Marshall GR, Hodgkin EE, Langs DA et al (1990) Factors governing
helical preference of peptides containing multiple alpha, alpha-
dialkyl amino acids. Proc Natl Acad Sci USA 87:487–491. doi:
10.1073/pnas.87.1.487
Mimna R, Tuchscherer G, Mutter M (2007) Toward the design of
highly efficient, readily accessible peptide N-caps for the
induction of helical conformations. Int J Pept Res Ther
13:237–244. doi:10.1007/s10989-006-9073-9
Nef S, Parada LF (1999) Cryptorchidism in mice mutant for Insl3.
Nat Genet 22:295–299. doi:10.1038/10364
Park JI, Semyonov J, Yi W (2008) Regulation of receptor signaling by
relaxin A chain motifs: derivation of pan-specific and LGR7-
specific human relaxin analogs. J Biol Chem 283(46):32099–
32109
Roche PJ, Butkus A, Wintour EM et al (1996) Structure and
expression of Leydig insulin-like peptide mRNA in the sheep.
Mol Cell Endocrinol 121:171–177. doi:10.1016/0303-7207(96)
03861-0
Rosengren KJ, Zhang S, Lin F et al (2006) Solution structure and
characterization of the LGR8 receptor binding surface of insulin-
like peptide 3. J Biol Chem 281:28287–28295. doi:10.1074/
jbc.M603829200
Scott DJ, Layfield S, Yan Y et al (2006) Characterization of novel
splice variants of LGR7 and LGR8 reveals that receptor
signaling is mediated by their unique low density lipoprotein
class A modules. J Biol Chem 281:34942–34954. doi:10.1074/
jbc.M602728200
Shabanpoor F, Bathgate RA, Hossain MA et al (2007) Design,
synthesis and pharmacological evaluation of cyclic mimetics of
the insulin-like peptide 3 (INSL3) B-chain. J Pept Sci 13:113–
120. doi:10.1002/psc.807
Shabanpoor F, Hughes RA, Bathgate RA et al (2008) Solid-phase
synthesis of europium-labeled human INSL3 as a novel probe for
the study of ligand-receptor interactions. Bioconjug Chem
19:1456–1463. doi:10.1021/bc800127p
Shepherd NE, Hoang HN, Desai VS et al (2006) Modular alpha-
helical mimetics with antiviral activity against respiratory
syncitial virus. J Am Chem Soc 128:13284–13289. doi:10.1021/
ja064058a
Spiess AN, Balvers M, Tena-Sempere M et al (1999) Structure and
expression of the rat relaxin-like factor (RLF) gene. Mol Reprod
Dev 54:319–325. doi:10.1002/(SICI)1098-2795(199912)54:4
\319::AID-MRD1[3.0.CO;2-Z
Tashima LS, Hieber AD, Greenwood FC et al (1995) The human
Leydig insulin-like (hLEY I-L) gene is expressed in the corpus
luteum and trophoblast. J Clin Endocrinol Metab 80:707–710.
doi:10.1210/jc.80.2.707
130 F. Shabanpoor et al.
123
Werle M, Bernkop-Schnu
¨rch A (2006) Strategies to improve plasma
half life time of peptide and protein drugs. Amino Acids 30:351–
367. doi:10.1007/s00726-005-0289-3
Whitmore L, Wallace BA (2004) DICHROWEB, an online server for
protein secondary structure analyses from circular dichroism
spectroscopic data. Nucleic Acids Res 32:W668–W673
Zimmermann S, Steding G, Emmen JM et al (1999) Targeted
disruption of the Insl3 gene causes bilateral cryptorchidism. Mol
Endocrinol 13:681–691. doi:10.1210/me.13.5.681
Effect of helix-promoting strategies on the biological activity 131
123
... Thus, in the first instance, the B chain was constrained by incorporation of disulfide constraint (distanced at position i, i + 4) in the helical region of the B chain as well as between the strand and the helix, as shown by us previously to induce helical structure and increase receptor binding affinity in the B chain of the related peptide INSL3. 31,32 A Glu − −Lys + salt bridge was also placed in the middle of the B chain helix to potentially increase peptide helical stability. A longer constraint was also incorporated along the helix at position i, i + 7 by cross-linking two Cys using 1,6bismaleimidohexane in an attempt to induce α-helical structure, as previously shown to induce α-helicity in amyloid β peptide. ...
... 34,35 We have also shown that the B chain dimer of INSL3 has a receptor binding affinity about 1000-fold greater than the B chain monomer. 31,32 However, the dimerization of H3 relaxin B chain did not improve the level of RXFP3 binding beyond that of the B chain alone. ...
... The CD spectral measurements were carried as previously described. 31 The concentration of peptides used was 0.15 μg/μL, except for H3 relaxin, which was at 0.10 μg/μL. ...
Article
Relaxin-3 is a neuropeptide that is implicated in the regulation of stress responses and memory. The elucidation of its precise physiological role(s) has, however, been hampered by cross-activation of the relaxin-2 receptor, RXFP1, in the brain. The current study undertook to develop analogues of human relaxin-3 (H3 relaxin) that can selectively bind and activate its receptor, RXFP3. We developed a high-affinity selective agonist (analogue 2) by removal of the intra-A chain disulfide bond and deletion of 10 residues from the N terminus of the A chain. Further truncation of this analogue from the C terminus of the B chain to Cys(B22) and addition of an Arg(B23) led to a high-affinity, RXFP3-selective, competitive antagonist (analogue 3). Central administration of analogue 2 in rats increased food intake, which was blocked by prior coadministration of analogue 3. These novel RXFP3-selective peptides represent valuable pharmacological tools to study the physiological roles of H3 relaxin/RXFP3 systems in the brain and important leads for the development of novel compounds for the treatment of affective and cognitive disorders.
... The constraints were introduced to mimic one turn of an α-helix (i, i+4). Similar stapling motif has been used for helix induction of a single-chain relaxin-3 agonist using lactam and hydrocarbon staples (23) and also in the related peptide INSL3 (35). Although the affinity and activity of single-chain relaxin-3 agonists were successfully improved with helical stapling compared to linear B-chain analogs, no improvement was seen for the relaxin-3 antagonist. ...
Article
Full-text available
The neuropeptide relaxin-3 and its receptor relaxin family peptide receptor-3 (RXFP3) play key roles in modulating behavior such as memory and learning, food intake and reward-seeking. A linear relaxin-3 antagonist (R3 B1-22R) based on a modified and truncated relaxin-3 B-chain was recently developed. R3 B1-22R is unstructured in solution, thus the binding conformation and determinants of receptor binding are unclear. Here we have designed, chemically synthesized and pharmacologically characterized more than 60 analogues of R3 B1-22R to develop an extensive understanding of its structure-activity relationships. We show that the key driver for affinity is the non-native C-terminal Arg23. Additional contributors to binding include amino acid residues that are important also for relaxin-3 binding, including Arg12, Ile15 and Ile19. Intriguingly, amino acid residues that are not exposed in native relaxin-3, including Phe14 and Ala17, also interact with RXFP3. We show that R3 B1-22R has a propensity to form a helical structure and modifications that support a helical conformation are functionally well tolerated while helix breakers such as proline residues disrupt binding. These data suggest the peptide adopts a helical conformation, like relaxin-3, upon binding to RXFP3 but that its smaller size allows it to penetrate deeper into the orthosteric binding site creating more extensive contacts with the receptor.
... The INSL3 B-chain alone is a RXFP2 antagonist although it only has low affinity for the receptor (Del Borgo et al. 2006;Shabanpoor et al. 2007). Modifications of the single chain structure result in modest gains in affinity, but larger gains are obtained by linking two B-chains using the native cysteine residues ( Shabanpoor et al. 2010) to produce antagonists with affinities only slightly lower than INSL3 ( Shabanpoor et al. 2011). ...
... The secondary structural changes of the peptides was measured by recording their CD spectra on JASCO model J815 spectropolarimeter as previously described [25]. The CD spectra were recorded in phosphate buffer saline (10 mM) with peptide concentrations made up to 0.2 mg/ml. ...
Article
Full-text available
Human relaxin-3 is a neuropeptide that is structurally similar to human insulin with two chains (A and B) connected by three disulfide bonds. It is expressed primarily in the brain and has modulatory roles in stress and anxiety, feeding and metabolism, and arousal and behavioural activation. Structure-activity relationship studies have shown that relaxin-3 interacts with its cognate receptor RXFP3 primarily through its B-chain and that its A-chain does not have any functional role. In this study, we have investigated the effect of modification of the B-chain C-terminus on the binding and activity of the peptide. We have chemically synthesised and characterized H3 relaxin as C-termini acid (both A and B chains having free C-termini; native form) and amide forms (both chains' C-termini were amidated). We have confirmed that the acid form of the peptide is more potent than its amide form at both RXFP3 and RXFP4 receptors. We further investigated the effects of amidation at the C-terminus of individual chains. We report here for the first time that amidation at the C-terminus of the B-chain of H3 relaxin leads to significant drop in the binding and activity of the peptide at RXFP3/RXFP4 receptors. However, modification of the A-chain C-terminus does not have any effect on the activity. We have confirmed using circular dichroism spectroscopy that there is no secondary structural change between the acid and amide form of the peptide, and it is likely that it is the local C-terminal carboxyl group orientation that is crucial for interacting with the receptors.
Article
Insulin-like 3 peptide (INSL3) is a member of the insulin-like peptide superfamily and is the only known physiological ligand of relaxin family peptide receptor 2 (RXFP2), a G protein-coupled receptor (GPCR). In mammals INSL3 is primarily produced both in testicular Leydig cells and in ovarian theca cells, but circulating levels of the hormone are much higher in males than in females. The INSL3/RXFP2 system has an essential role in the development of the gubernaculum for the initial transabdominal descent of the testis and in maintaining proper reproductive health in men. Although its function in female physiology has been less well-characterized, it was reported that INSL3 deletion affects antral follicle development during the follicular phase of the menstrual cycle and uterus function. Since the discovery of its role in the reproductive system, the study of INSL3/RXFP2 has expanded to others organs such as skeletal muscle, bone, kidney, thyroid, brain, and eye. This review aims to summarize the various advances in understanding the physiological function of this ligand-receptor pair since its first discovery and elucidate its future therapeutic potential in the management of various diseases.
Chapter
Insulin-like peptide 3 (INSL3) belongs to the insulin/relaxin family of structurally related peptides, and mediates its effects via the G protein-coupled receptor, RXFP2 (relaxin family peptide receptor 2). Extensive mutagenesis studies have revealed the molecular determinants of INSL3/RXFP2 binding, and developed several INSL3-based antagonists. The major physiological effects of INSL3 are observed within the reproductive system, where the peptide/receptor pair is essential for testes descent during development. Additional endocrine roles for INSL3 are emerging in bone metabolism and cancer metastasis.
Article
Relaxin is essential for trophoblast development during pregnancy. Evidence shows that relaxin increases trophoblast cell migration capacity. Here, we show the effect of relaxin on protein kinase B (AKT) activation and glycogen synthase kinase 3-beta (GSK3β) inactivation as well as on the proliferation of HTR-8/SVneo cells, a model of human extravillous trophoblast (EVT). HTR-8/SVneo cells were treated with different doses of human recombinant (rH2) relaxin in serum-deprived conditions and treated for increasing time with 1 ng/mL of rH2 relaxin. Western blot analysis was performed to detect pAKT, AKT, pGSK3β, GSK3β, and actin expression. Proliferation of HTR-8/SVneo cells was analyzed by MTS assay. rH2 relaxin treatment increased the ratio of pAKT/AKT, pGSK3β/GSK3β, and proliferation in HTR-8/SVneo cells. Furthermore, AKT and GSK3β activation by rH2 relaxin was inhibited by a phosphoinositide 3-kinase (PI3K) inhibitor. This study suggests that rH2 relaxin induces AKT and GSK3β phosphorylation as well as proliferation in HTR-8/SVneo cells.
Article
Relaxin, insulin-like peptide 3 (INSL3), relaxin-3, and INSL5 are the cognate ligands for the relaxin family peptide (RXFP) receptors 1-4, respectively. RXFP1 activates pleiotropic signaling pathways including the signalosome protein complex that facilitates high-sensitivity signaling; coupling to Gαs, Gαi, and Gαo proteins; interaction with glucocorticoid receptors; and the formation of hetero-oligomers with distinctive pharmacological properties. In addition to relaxin-related ligands, RXFP1 is activated by Clq-tumor necrosis factor-related protein 8 and by small-molecular-weight agonists, such as ML290 [2-isopropoxy-N-(2-(3-(trifluoromethylsulfonyl)phenylcarbamoyl)phenyl)benzamide], that act allosterically. RXFP2 activates only the Gαs- and Gαo-coupled pathways. Relaxin-3 is primarily a neuropeptide, and its cognate receptor RXFP3 is a target for the treatment of depression, anxiety, and autism. A variety of peptide agonists, antagonists, biased agonists, and an allosteric modulator target RXFP3. Both RXFP3 and the related RXFP4 couple to Gαi/Gαo proteins. INSL5 has the properties of an incretin; it is secreted from the gut and is orexigenic. The expression of RXFP4 in gut, adipose tissue, and β-islets together with compromised glucose tolerance in INSL5 or RXFP4 knockout mice suggests a metabolic role. This review focuses on the many advances in our understanding of RXFP receptors in the last 5 years, their signal transduction mechanisms, the development of novel compounds that target RXFP1-4, the challenges facing the field, and current prospects for new therapeutics. Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.
Article
The peptide hormone INSL3 and its receptor RXFP2 have co-evolved alongside relaxin and its receptor RXFP1. Both RXFP1 and RXFP2 are G protein-coupled receptors (GPCRs) containing the hallmark seven transmembrane (TM) helices in addition to a distinct ecto-domain of Leucine-rich repeats (LRRs) and a single Low Density Lipoprotein Class-A (LDLa) module at the N-terminus. RXFP1 and RXFP2 are the only mammalian GPCRs described as containing an LDLa, and its removal does not perturb primary ligand binding to the LRRs, however signaling is abolished. This presents a general mechanism whereby ligand binding induces a conformational change of the receptor to position the LDLa to elicit a signal response. Although the LDLa interaction site has not been identified the residues important to the action have been mapped within RXFP1 LDLa. In this study we comprehensively study the RXFP2 LDLa module. We solve its structure using Nuclear Magnetic Resonance and concurrently investigate the signaling of an RXFP2 with the LDLa removed (RXFP2-short), confirming that the LDLa is essential to signaling. We then replaced the LDLa with the second ligand-binding module from the LDL receptor, LB2, creating RXFP2-LB2. Unlike the equivalent RXFP1-LB2 chimera, signaling is rescued albeit modestly. Guided by the NMR structure we dissected regions of the RXFP2 LDLa to identify specific residues that are important to signal activation. We determine that although the module is important to the activation of RXFP2, unlike the RXFP1 receptor, specific residues in the N-terminus of the domain are not involved in signal activation.
Article
Full-text available
One of the most recognised and studied family of peptide hormones is the insulin superfamily. Within this family is the relaxin subfamily which comprises seven members: relaxin-1, -2 and -3 and insulin-like peptides 3, 4, 5 and 6. Besides exhibiting sequence similarities, each member exists as an active A-B heterodimer linked by three disulfide bonds. This mini-review is divided into three broad themes: an overview of all insulin superfamily members (including structural similarities); roles of each superfamily member and finally, a focus on the pleiotropic peptide hormone, human relaxin-2. In addition to promoting vasodilatory effects leading to evaluation in Phase III clinical trials for the treatment of acute heart failure, relaxin has recently been shown to be highly expressed by cancer cells, aiding in their proliferation, invasiveness and metastasis. These contrary effects of relaxin are discussed together with current efforts in the development of relaxin antagonists that may possess future therapeutic potential for the treatment of certain cancers.
Article
Full-text available
Relaxin peptides are important hormones for the regulation of reproductive tissue remodeling and the renal cardiovascular system during pregnancy. Recent studies demonstrated that two of the seven human relaxin family peptides, relaxin H2 (RLN2) and INSL3, signal exclusively through leucine-rich repeat-containing G protein-coupled receptors, LGR7 and LGR8. Although it was well characterized that an RXXXRXXI motif at the RLN2 B chain confers receptor activation activity, it is not clear what roles RLN2 A chain plays in receptor interaction. Analyses of relaxin family genes on syntenic regions of model tetrapods showed that the A chain of RLN2 orthologs exhibited a greater sequence divergence as compared with the receptor-binding domain-containing B chain, foreshadowing a potential role in receptor interactions; hence, defining receptor selectivity in this fast evolving peptide hormone. To test our hypothesis that select residues in the human RLN2 A chain play key roles in receptor interaction, we studied mutant peptides with residue substitution(s) in the A chain. Here, we showed that alanine substitution at the A16 and A17 positions enhances LGR8-activation activity of RLN2, whereas mutation at the A22-23 region (RLN2A22-23) ablates LGR8, but not LGR7, activation activity. In addition, we demonstrated that the functional characteristics of the RLN2A22-23 mutant are mainly attributed to modifications at the PheA23 position. Taken together, our studies indicated that ThrA16, LysA17, and PheA23 constitute part of the receptor-binding interface of human RLN2, and that modification of these residues has led to the generation of novel human RLN2 analogs that would allow selective activation of human LGR7, but not LGR8, in vivo.
Article
Full-text available
The presence of multiple alpha,alpha-dialkyl amino acids such as alpha-methylalanine (alpha-aminoisobutyric acid, Aib) leads to predominantly helical structures, either with alpha-helical or 3(10)-helical hydrogen bonding patterns. The crystal structure of emerimicin-(1-9) benzyl ester (Ac-Phe-Aib-Aib-Aib-Val-Gly-Leu-Aib-Aib-OBzl) reported here shows essentially pure alpha-helical character, whereas other similar compounds show predominantly 3(10)-helical structures. The factors that govern helical preference include the inherent relative stability of the alpha-helix compared with the 3(10)-helix, the extra hydrogen bond seen with 3(10)-helices, and the enhanced electrostatic dipolar interaction of the 3(10)-helix when packed in a crystalline lattice. The balance of these forces, when combined with the steric requirements of the amino acid side chains, determines the relative stability of the two helical conformations under a given set of experimental conditions.
Article
Full-text available
According to Burkhardt et al. (Burkhardt, E., Adham, I. M., Brosig, B., Gastmann, A., Mattei, M. G., and Engel, W. (1994) Genomics 20, 13-19) Leydig cells contain the message for a protein of the insulin/relaxin family which was named Leydig cell insulin-like protein (LEY I-L). We have synthesized the human LEY I-L according to the amino acid sequence deduced from the published cDNA structure and obtained preliminary results concerning its potential target organs and its biological activity. Leydig cell insulin-like protein binds specifically to crude membrane preparations of mouse uterus and brain and shows cross-reactivity with the relaxin receptor, but not the insulin receptor. On the basis of these observations, together with the results of earlier structure-function considerations, we suggest that the new protein is a relaxin-like factor. By itself the new factor shows no obvious effect, but when given together with relaxin it significantly enhances relaxin-mediated widening of the mouse symphysis pubis.
Article
The relaxin-like factor (RLF) is a novel member of the insulin-IGF-relaxin family of growth factors and hormones, and its mRNA is expressed very specifically in the Leydig cells of the testis and in the theca and luteal cells of the ovary. Here we report the cloning of the RLF gene and cDNA from the rat. The 0.8kb mRNA is produced from a small gene comprising two exons situated less than 1 kb downstream of the gene for the signalling factor JAK3. Northern hybridization confirms high RLF mRNA expression in the adult rat testis, and low expression in the ovary, but in no other tissues examined. Northern analysis of fetal and neonatal gonadal tissues showed that RLF mRNA is highly upregulated in the testes of day 19 embryos, but not in later neonatal stages, nor in any ovarian tissue from this period. This would indicate that RLF is a marker for the mature fetal as well as the adult-type Leydig cell, but is not expressed in premature, precursor, or dedifferentiated Leydig cells of either cell type. Finally, RNA was analysed from the testes of rats which had been treated with ethylene dimethane sulfonate (EDS), an alkylating agent that specifically destroys rat Leydig cells. RLF mRNA was absent from the acutely treated testes, but became detectable between 15 and 20 days post-treatment, concomitant with the repopulation of the testes by new Leydig cells. Continuous testosterone substitution of EDS-treated rats suppressed the production of gonadotropins, and LH-dependent Leydig cell differentiation, with the result that RLF mRNA remained undetectable throughout the study period. In conclusion, RLF is a very specific marker for the mature Leydig cell phenotype in both the adult-type and fetal Leydig cell populations of the rat testis. Mol. Reprod. Dev. 54:319–325, 1999. © 1999 Wiley-Liss, Inc.
Article
A series of novel peptide N-caps was designed with an emphasis on ease of synthesis and an abundance of hydrogen bond acceptors. Different scaffolds based on sugars, cyclic hydrocarbons, and amino acids are developed with a variety of hydrogen bond acceptors including esters, carboxyls, amides and a sulfonic acid. The efficient use in solid-phase peptide synthesis was demonstrated by incorporating the N-caps to a resin-bound model peptide. Their differential helix nucleating power in aq. buffer was detd. by CD studies. Increases in peptide helicity to a significant extent are obsd., leading to a discussion of N-capping efficiency vs. ease of synthesis. The potential of the elaborated N-caps for the reversal of beta-sheet to alpha-helix conformations in the context of fibrillogenesis is discussed.
Article
In this report we describe a general survey of all helices found in 57 of the known protein crystal structures, together with a detailed analysis of 48 alpha-helices found in 16 of the structures that are determined to high resolution. The survey of all helices reveals a total of 291 alpha-helices, 71 3(10)-helices and no examples of pi-helices. The conformations of the observed helices are significantly different from the "ideal" linear structures. The mean phi, psi angles for the alpha- and 3(10)-helices found in proteins are, respectively, (-62 degrees, -41 degrees) and (-71 degrees, -18 degrees). A computer program, HBEND, is used to characterize and to quantify the different types of helix distortion. alpha-Helices are classified as regular or irregular, linear, curved or kinked. Of the 48 alpha-helices analysed, only 15% are considered to be linear; 17% are kinked, and 58% are curved. The curvature of helices is caused by differences in the peptide hydrogen bonding on opposite faces of the helix, reflecting carbonyl-solvent/side-chain interactions for the exposed residues, and packing constraints for residues involved in the hydrophobic core. Kinked helices arise either as a result of included proline residues, or because of conflicting requirements for the optimal packing of the helix side-chains. In alpha-helices where there are kinks caused by proline residues, we show that the angle of kink is relatively constant (approximately 26 degrees), and that there is minimal disruption of the helix hydrogen bonding. The proline residues responsible for the kinks are highly conserved, suggesting that these distortions may be structurally/functionally important.
Article
Inverse circular dichroism (CD) spectra are presented for each of the five major secondary structures of proteins: alpha-helix, antiparallel and parallel beta-sheet, beta-turn, and other (random) structures. The fraction of the each secondary structure in a protein is predicted by forming the dot product of the corresponding inverse CD spectrum, expressed as a vector, with the CD spectrum of the protein digitized in the same way. We show how this method is based on the construction of the generalized inverse from the singular value decomposition of a set of CD spectra corresponding to proteins whose secondary structures are known from X-ray crystallography. These inverse spectra compute secondary structure directly from protein CD spectra without resorting to least-squares fitting and standard matrix inversion techniques. In addition, spectra corresponding to the individual secondary structures, analogous to the CD spectra of synthetic polypeptides, are generated from the five most significant CD eigenvectors.