scispace - formally typeset
Open AccessJournal ArticleDOI

Molecular determinants conferring the stoichiometric-dependent activity of α-conotoxins at the human α9α10 nicotinic acetylcholine receptor subtype

TLDR
The molecular determinants of α-conotoxins Vc1.1, RgIA#, and PeIA inhibition at hypothetical stoichiometries of the human α9α10 nAChR are investigated to suggest that only Vc 1.1 exhibits stoichiometric-dependent inhibition at the α9 α10 n AChR.
Abstract
α9α10 nicotinic acetylcholine receptors (nAChRs) putatively exist at different stoichiometries. We systematically investigated the molecular determinants of α-conotoxins Vc1.1, RgIA#, and PeIA inhibition at hypothetical stoichiometries of the human α9α10 nAChR. Our results suggest that only Vc1.1 exhibits stoichiometric-dependent inhibition at the α9α10 nAChR. The hydrogen bond between N154 of α9 and D11 of Vc1.1 at the α9(+)-α9(−) interface is responsible for the stoichiometric-dependent potency of Vc1.1.

read more

Content maybe subject to copyright    Report

University of Wollongong
Research Online
33)=)88)-)3:0)5,-,1+)3"-9-)8+059:1:;:- )+;3:?6.#+1-5+--,1+15-)5,-)3:0

Molecular Determinants Conferring the
Stoichiometric-Dependent Activity of α-
Conotoxins at the Human α9α10 Nicotinic
Acetylcholine Receptor Subtype
Rilei Yu
Qingdao National Laboratory for Marine Science and Technology, Ocean University of China
Han-Shen Tae
University of Wollongong
Nargis Tabassum
Ocean University of China, Qingdao National Laboratory for Marine Science and Technology
Juan Shi
Ocean University of China, Qingdao National Laboratory for Marine Science and Technology
Tao Jiang
Qingdao National Laboratory for Marine Science and Technology, Ocean University of China
See next page for additional authors
"-9-)8+0 5315-19:0-67-5)++-99159:1:;:165)38-7691:68?.68:0-%51<-891:?6.'63365/65/68.;8:0-815.684):165+65:)+::0-% '1*8)8?
8-9-)8+07;*9;6=-,;);
!;*31+):165-:)139
(;"$)-$)*)99;4#011)5/$,)4963-+;3)8-:-8415)5:965.-8815/:0-#:61+0164-:81+
-7-5,-5:+:1<1:?6.@656:6>159)::0-;4)5@@1+6:151++-:?3+06315-"-+-7:68#;*:?7-6;85)36.-,1+15)30-419:8?


Molecular Determinants Conferring the Stoichiometric-Dependent
Activity of α-Conotoxins at the Human α9α10 Nicotinic Acetylcholine
Receptor Subtype
Abstract
@@51+6:151+)+-:?3+06315-8-+-7:68950"97;:):1<-3?->19:):,1B-8-5:9:61+0164-:81-9'-
9?9:-4):1+)33?15<-9:1/):-,:0-463-+;3)8,-:-8415)5:96.@+656:6>159&+"/)5,!-1501*1:165):
0?76:0-:1+)39:61+0164-:81-96.:0-0;4)5@@50" ;88-9;3:99;//-9::0):653?&+->01*1:9
9:61+0164-:81+,-7-5,-5:1501*1:165)::0-@@50"C-0?,86/-5*65,*-:=--56.@)5,
6.&+)::0-@@A15:-8.)+-198-976591*3-.68:0-9:61+0164-:81+,-7-5,-5:76:-5+?6.&+
Disciplines
-,1+15-)5,-)3:0#+1-5+-9
Publication Details
(;"$)-$)*)99;4#011)5/$,)4963-+;3)8-:-8415)5:965.-8815/:0-
#:61+0164-:81+-7-5,-5:+:1<1:?6.@656:6>159)::0-;4)5@@1+6:151++-:?3+06315-"-+-7:68
#;*:?7-6;85)36.-,1+15)30-419:8?
Authors
"13-1(;)5#0-5$)-)8/19$)*)99;4;)5#01$)61)5/)5,)<1,,)49
C1926;85)3)8:1+3-19)<)13)*3-):"-9-)8+0 5315- 0D7986;6=-,;);10481

1
Molecular determinants conferring the stoichiometric-dependent activity of
α-conotoxins at the human α9α10 nAChR subtype
Rilei Yu
1,3†*
, Han-Shen Tae
2†
, Nargis Tabassum
1,3
, Juan Shi
1,3
, Tao Jiang
1,3
and David J. Adams
2*
1
Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and
Pharmacy, Ocean University of China, Qingdao 266003, China
2
Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong,
New South Wales 2522, Australia
3
Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine
Science and Technology, Qingdao 266003, China
These authors contribute equally to this work.
*
Corresponding authors: djadams@uow.edu.au or ryu@ouc.edu.cn

2
ABSTRACT
α9α10 Nicotinic acetylcholine receptors (nAChRs) putatively exist at different stoichiometries. We
systematically investigated the molecular determinants of α-conotoxins Vc1.1, RgIA#, and PeIA
inhibition at hypothetical stoichiometries of the human α9α10 nAChR. Our results suggest that only
Vc1.1 exhibits strong stoichiometric-dependent inhibition at the α9α10 nAChR. The hydrogen bond
between N154 of α9 and D11 of Vc1.1 at the α9(+)-α9() interface is responsible for the
stoichiometric-dependent potency of Vc1.1.

3
Introduction
Nicotinic acetylcholine receptors (nAChRs) are cation-selective pentameric ligand-gated ion
channels belonging to the family of Cys-loop receptors, which also includes γ-aminobutyric acid
type A (GABA
A
), glycine, and serotonin (5-HT
3
) receptors.
1
The vertebrate nAChRs are
homo/heteromeric assembles of α1-α10, β1-β4, γ, δ or ε subunits. Although the nAChRs are
generally featured in the nervous system, they are also expressed in non-neuronal cells participating
in various physiological events and they are important targets for drug design.
2
The first crystal structure of any heteromeric nAChRs, the human(h) α4β2 nAChR subtype,
reveals the overall architecture consisting of a large extracellular domain (ECD), a transmembrane
domain (TMD) and an intracellular domain (ICD) (Figure 1A).
3
The ligand binding site is located at
the ECD interface of two adjacent subunits, comprising of (+) (principal) and (─) (complementary)
components. For hα4β2 nAChR, the α4 subunit loops A, B and C form the (+) site, and the β2
subunit β-sheet contributes to the (─) site (Figure 1B). Five subunits of the nAChR circle a
conducting pore (Figure 1C) where a gate consisting of hydrophobic residues, such as Leu and Ile,
is located in the middle of the TMD.
Heteromeric α9α10 nAChRs play an important physiological role in mediating olivocochlear
and vestibular neurotransmission.
4,5
Transcripts/protein expression of α9 and/or α10 subunits have
been reported in dorsal root ganglion neurons,
6,7
adrenal medulla,
8
and in other non-neuronal cells,
such as skin keratinocytes, pituitary pars tuberalis, lymphocytes, macrophages and bladder
urothelium.
915
α9-Containing nAChRs are potential targets for the therapy of several disorders or diseases
such as ear disorders, chronic pain and pemphigus vulgaris.
7,9,11,16,17
In addition, α9-containing
nAChRs are responsible for nicotine-induced transformation of normal human breast epithelial cells,
and inducible overexpression of α9-nAChR substantially increased tumor growth.
18
Conotoxins derived from the venom of Conus sea snails are pharmacologically valuable
peptides with selective potency at nAChR subtypes. α-Conotoxin antagonists of α9α10 nAChR such

Citations
More filters
Journal ArticleDOI

Medicinal chemistry, pharmacology, and therapeutic potential of α-conotoxins antagonizing the α9α10 nicotinic acetylcholine receptor.

TL;DR: The structure and function of the α9α10 nAChR are highlighted and studies of α-conotoxins targeting it are reviewed, including their three-dimensional structures, structure optimization strategies, and binding modes at the α 9α10nA chR, as well as their therapeutic potential.
Journal ArticleDOI

Mutagenesis of α-Conotoxins for Enhancing Activity and Selectivity for Nicotinic Acetylcholine Receptors

TL;DR: This review will focus on structure-activity relationship studies involving native α-CTxs that have been rationally mutated and molecular interactions that underlie binding between ligand and nAChR isoform.
Journal ArticleDOI

d-Amino Acid Substitution of α-Conotoxin RgIA Identifies its Critical Residues and Improves the Enzymatic Stability

TL;DR: The findings suggest that a d-amino acid scan is a useful strategy for investigating how the side-chain chirality of amino acids affects the structure and function of peptides and may facilitate the development of more stable analogues to increase therapeutic potential.
Journal ArticleDOI

Dimerization of α-Conotoxins as a Strategy to Enhance the Inhibition of the Human α7 and α9α10 Nicotinic Acetylcholine Receptors

TL;DR: It is proposed that the dimerization of α-conotoxins is a simpler and efficient alternative strategy to dendrimers for enhancing the activity ofα-conooxins.
References
More filters
Journal ArticleDOI

Particle mesh Ewald: An N⋅log(N) method for Ewald sums in large systems

TL;DR: An N⋅log(N) method for evaluating electrostatic energies and forces of large periodic systems is presented based on interpolation of the reciprocal space Ewald sums and evaluation of the resulting convolutions using fast Fourier transforms.
Journal ArticleDOI

ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB

TL;DR: Together, these backbone and side chain modifications (hereafter called ff14SB) not only better reproduced their benchmarks, but also improved secondary structure content in small peptides and reproduction of NMR χ1 scalar coupling measurements for proteins in solution.
Journal ArticleDOI

SETTLE: an analytical version of the SHAKE and RATTLE algorithm for rigid water models

TL;DR: In this article, an analytical algorithm called SETTLE for resetting the positions and velocities to satisfy the holonomic constraints on the rigid water model is presented, which is based on the Cartesian coordinate system and can be used in place of SHAKE and RATTLE.
Journal ArticleDOI

PROPKA3: Consistent Treatment of Internal and Surface Residues in Empirical pKa Predictions

TL;DR: The rules and parameters for one of the most commonly used empirical pKa predictors, PROPKA, are revised based on better physical description of the desolvation and dielectric response for the protein, and a new and consistent approach to interpolate the description between the previously distinct classifications into internal and surface residues is introduced.
Journal ArticleDOI

MMPBSA.py: An Efficient Program for End-State Free Energy Calculations.

TL;DR: MMPBSA.py is a program written in Python for streamlining end-state free energy calculations using ensembles derived from molecular dynamics or Monte Carlo simulations, including the Poisson-Boltzmann Model and several implicit solvation models.
Related Papers (5)
Frequently Asked Questions (18)
Q1. What are the contributions mentioned in the paper "Molecular determinants conferring the stoichiometric-dependent activity of î±-conotoxins at the human î±9î±10 nicotinic acetylcholine receptor subtype" ?

The authors systematically investigated the molecular determinants of α-conotoxins Vc1. 1, RgIA #, and PeIA inhibition at hypothetical stoichiometries of the human α9α10 nAChR. This journal article is available at Research Online: https: //ro. uow. edu. au/ihmri/1269 Their results suggest that only Vc1. 1 exhibits stoichiometric-dependent inhibition at the α9α10 nAChR. 

However, the authors can not exclude the possibility of the α9 ( + ) -α9 ( ─ ) as an energetically favourable binding site of RgIA # at ( α9 ) 3 ( α10 ) 2 nAChR. Instead, their study suggests that the sensitivity of both α10 ( + ) -α9 ( ─ ) and α9 ( + ) -α9 ( ─ ) interfaces to RgIA # might be comparable. Regardless, stoichiometric-selective inhibitors of α9α10 nAChR would be useful neurochemical tools for further elucidating the functional differences between the stoichiometries in native cells. Conversely, α9α10 nAChR antagonists that do not discriminate between the stoichiometries, may be suitable candidates to use under conditions where different stoichiometries exist. 

In addition, α9-containing nAChRs are responsible for nicotine-induced transformation of normal human breast epithelial cells, and inducible overexpression of α9-nAChR substantially increased tumor growth. 

The hydrogen bondbetween N154 of α9 and D11 of Vc1.1 at the α9(+)-α9() interface is responsible for thestoichiometric-dependent potency of Vc1.1. 

The DPR (Asp-Pro-Arg) motif is conserved in α-conotoxins ImI, RgIA# (or RgIA) and Vc1.1,and essential to their binding affinities. 

at α9:α10 mRNA ratio of 1:3, presumably forming the (α9)2(α10)3 nAChR stoichiometry, 1 μM Vc1.1 inhibited ACh-evoked currents by 25.5 ± 1.8% (n = 15). 

Using thermodynamic mutant cycles, 32 the coupling coefficient (Ω) gave a reciprocal of 28.57 suggesting a relatively strong coupling between Vc1.1 D11 and α9 N154 . 

Injections of higher α9 to α10 mRNA ratios resulted in greater Vc1.1 inhibition at the expressed hα9α10 nAChRs compared to injections with excess α10 mRNA, alluding to the presence of Vc1.1-highsensitivity α9(+)-α9() site/s in the (α9)3(α10)2 or (α9)4(α10)1 configurations. 

a high-sensitivity Vc1.1 binding site was proposed at the α9(+)-α9() interfacepresent only in the (α9)3(α10)2 stoichiometry, and a low–sensitivity Vc1.1 binding site contributed by the α10(+)-α9() interface that exists in both stoichiometries. 

in their model the aromatic residues of the binding site form compact van der Waals contacts with the α-helix of the α-conotoxins, and the C192-C193 disulfide bond stacks with the C2-C8 disulfide bond of the α-conotoxins. 

Thehydrogen bond formed between the side chains of α9 N154 and Vc1.1 D11 confers the specificity of Vc1.1 at α9(+)-α9(─) interface, and is responsible for the higher sensitivity of the α9α10 nAChR expressed with higher abundance of the α9 mRNA to Vc1.1. 

Although the nAChRs are generally featured in the nervous system, they are also expressed in non-neuronal cells participating in various physiological events and they are important targets for drug design. 

Subsequent experiments were carried out using 1 μM Vc1.1, 300 nM RgIA# and 30 nM PeIA, close to the IC50 values of the α-conotoxins at the hα9α10 nAChR. 

To validate their prediction that the hydrogen bond interaction between Vc1.1 D11-α9 and N154is the determinant of Vc1.1 binding preference at the hα9(+)-α9() interface, residue N154 of thehα9 subunit was mutated to the corresponding hα10 subunit G154. 

The stoichiometry of rat(r) α9α10 nAChR heterologously expressed in Xenopus laevis oocytes was initially suggested to consist of two α9 and three α10 subunits ((α9)2(α10)3). 

In this study, the authors used the R13-deleted analogue of RgIA (RgIA#) to probe its sensitivity todifferent stoichiometries of α9α10 nAChRs. 

24,25 Both Vc1.1 binding sites share a common α9 subunit (─) component, therefore, the determinants of Vc1.1 binding should be contributed by non-conserved residues of the α9 and α10 subunit (+) components. 

As shown in PeIA-α9(+)α9(─) model (Figure 3F), residues from the second loop of PeIA form few contacts with the side chain of α9 N154.