scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Structure-Activity Studies Reveal the Molecular Basis for GABAB-Receptor Mediated Inhibition of High Voltage-Activated Calcium Channels by α-Conotoxin Vc1.1.

TL;DR: Findings suggest that selectively targeting GABABR-mediated HVA calcium channel inhibition by α-conotoxins could be effective for the treatment of chronic visceral pain.
Abstract: α-Conotoxins are disulfide-bonded peptides from cone snail venoms and are characterized by their affinity for nicotinic acetylcholine receptors (nAChR). Several α-conotoxins with distinct selectivity for nAChR subtypes have been identified as potent analgesics in animal models of chronic pain. However, a number of α-conotoxins have been shown to inhibit N-type calcium channel currents in rodent dissociated dorsal root ganglion (DRG) neurons via activation of G protein-coupled GABAB receptors (GABABR). Therefore, it is unclear whether activation of GABABR or inhibition of α9α10 nAChRs is the analgesic mechanism. To investigate the mechanisms by which α-conotoxins provide analgesia, we synthesized a suite of Vc1.1 analogues where all residues, except the conserved cysteines, in Vc1.1 were individually replaced by alanine (A), lysine (K), and aspartic acid (D). Our results show that the amino acids in the first loop play an important role in binding of the peptide to the receptor, whereas those in the second...

Summary (1 min read)

Introduction

  • Structure-Activity Studies Reveal the Molecular Basis for GABA(B)-Receptor Mediated Inhibition of High Voltage-Activated Calcium Channels by α-Conotoxin Vc1.1.
  • Structure-activity studies reveal the molecular basis for GABAB-receptor mediated inhibition of high voltage-activated calcium channels by α-conotoxin Vc1.1.

Conus victoriae

  • Testing for antagonist activity against a range of nAChR subtypes revealed that Vc1.1 was a selective inhibitor of the α9α10 nAChR subtype10, 11.
  • Therefore, deliniating the precise molecular mechanisms responsible for their analgesic effects in vivo is important.
  • To develop these conotoxins as potential analgesics it is important to identify the key peptide residues for the interaction with both the α9α10 nAChR and the GABABR.
  • A comprehensive structure-activity study of Vc1.1 at the α9α10 nAChR, where an alanine, lysine and aspartic acid scan was performed, revealed that key regions in Vc1.1 for activity were residues Asp5-Asp7 and Asp11-Ile15.26.
  • To further clarify the roles of the α9α10 nAChR and HVA calcium channel inhibition on the analgesic effects of this sub-family of α-conotoxins, the authors undertook a systematic study of the structure-activity relationships of Vc1.1 inhibition of HVA calcium channel currents.

Results and Discussion

  • Activity of Vc1.1 analogues on HVA calcium channels current in DRG neurons: Representative traces of HVA calcium channel inhibition in mice DRG neurons by Vc1.1 analogues equipotent to Vc1.1 and less effective than Vc1.1 are shown in Figure S2A.
  • Representative traces of IBa inhibition in mice DRG neurons by Vc1.1 analogues equipotent and less potent to Vc1.1 are shown in Figure S2B.
  • Both peptides were characterised by NMR spectroscopy and Hα chemical shift analysis indicated that the structures of Vc1.1[D11A,E14A] and cVc1.1[D11A,E14A] were consistent with the native sequences (Figure S3, Table S1).
  • Therefore, the authors sought to investigate the antinociceptive effects of cVc1.1[D11A,E14A], which is selective only for the GABABR in a mouse model of CVH.

Conclusion

  • A subset of α-conotoxins, including Vc1.1 and RgIA, are thought to produce their pain relieving effects by inhibiting either nicotinic acetylcholine receptors or HVA calcium channels via GABABR activation in mammalian sensory neurons.
  • By demonstrating that an α-conotoxin analogue selective for GABABR mediating HVA calcium channel inhibition is analgesic in a mouse model of chronic visceral pain, this study provides compelling evidence for the role of this analgesic mechanism.
  • This finding opens novel perspectives in the pharmacological intervention of chronic pain management and the potential use of GABABR-targeting α-conotoxins as treatments for chronic pain conditions.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

University of Wollongong
Research Online
::/D/??/3/:A6/<2#3271/:'3@3/?16<@A7ABA3 /1B:AF=4(173<13#32717<3/<23/:A6

Structure-Activity Studies Reveal the Molecular
Basis for GABA(B)-Receptor Mediated Inhibition
of High Voltage-Activated Calcium Channels by α-
Conotoxin Vc1.1
Mahsa Sadeghi
University of Wollongong;@/23567B=D32B/B
Bodil B. Carstens
University of Queensland
Brid P. Callaghan
University of Wollongong, University of Queensland
James T. Daniel
University of Queensland
Han Shen Tae
University of Wollongong6@A/3B=D32B/B
See next page for additional authors
'3@3/?16%<:7<37@A63=>3</113@@7<@A7ABA7=</:?3>=@7A=?F4=?A63*<7C3?@7AF=4,=::=<5=<5=?4B?A63?7<4=?;/A7=<1=<A/1AA63*%,"70?/?F
?3@3/?16>B0@B=D32B/B
&B0:71/A7=<3A/7:@
(/23567#/?@A3<@/::/56/<&/<73: ))/3%=<<3::)/@A?= ?73?:3F(#2/;@ ?/79 
:/?9' (A?B1AB?31A7C7AF(AB273@'3C3/:A63#=:31B:/?/@7@4=?'313>A=?#327/A32<6707A7=<=4756
+=:A/531A7C/A32/:17B;6/<<3:@0FH=<=A=E7<+1(63;71/:7=:=5F

Structure-Activity Studies Reveal the Molecular Basis for GABA(B)-
Receptor Mediated Inhibition of High Voltage-Activated Calcium
Channels by α-Conotoxin Vc1.1
Abstract
H=<=A=E7<@/?327@B:L230=<232>3>A723@4?=;1=<3@</7:C3<=;@/<2/?316/?/1A3?7G320FA637?/J<7AF4=?
<71=A7<71/13AF:16=:7<3?313>A=?@<6'(3C3?/:H1=<=A=E7<@D7A627@A7<1A@3:31A7C7AF4=?<6'@B0AF>3@
6/C3033<723<A7L32/@>=A3<A/</:53@71@7</<7;/:;=23:@=416?=<71>/7<=D3C3?/<B;03?=4H
1=<=A=E7<@6/C3033<@6=D<A=7<6707A$AF>31/:17B;16/<<3:1B??3<A@7<?=23<A27@@=17/A322=?@/:?==A
5/<5:7=<'<3B?=<@C7//1A7C/A7=<=4>?=A37<1=B>:32
?313>A=?@
'K3?34=?37A7@
B<1:3/?D63A63?/1A7C/A7=<=4
'=?7<6707A7=<=4HH<6'@7@A63/</:53@71;316/<7@;)=
7<C3@A75/A3A63;316/<7@;@0FD6716H1=<=A=E7<@>?=C723/</:53@7/D3@F<A63@7G32/@B7A3=4+1
/</:=5B3@D63?3/::?3@72B3@3E13>AA631=<@3?C321F@A37<3@7<+1D3?37<27C72B/::F?3>:/1320F/:/<7<3
:F@7<3!/<2/@>/?A71/172%B??3@B:A@@6=DA6/AA63/;7<=/172@7<A63L?@A:==>>:/F/<
7;>=?A/<A?=:37<07<27<5=4A63>3>A723A=A63?313>A=?D63?3/@A6=@37<A63@31=<2:==>>:/F/<7;>=?A/<A
?=:34=?A63@3:31A7C7AF=4A63>3>A7234=?A63
'=C3?HH<6'@,323@75<32/1+1/</:=5B3
A6/A7@4=:2@3:31A7C34=?
';327/A327<6707A7=<=46756C=:A/53/1A7C/A32+1/:17B;
16/<<3:@=C3?HH<6'@/<2@6=DA6/A7A7@/</:53@717</;=B@3;=23:=416?=<71C7@13?/:
6F>3?@3<@7A7C7AF+1+1-.1/B@322=@323>3<23<A7<6707A7=<=41=:=<71<=1713>A=?@D7A6
5?3/A3?3J1/1F7<3EC7C=+1=:=<71<=1713>A=?@?3:/A7C3A=63/:A6F1=:=<71<=1713>A=?@K3@3L<27<5@
@B553@AA6/A@3:31A7C3:FA/?53A7<5
';327/A32+1/:17B;16/<<3:7<6707A7=<0FH1=<=A=E7<@1=B:2
033I31A7C34=?A63A?3/A;3<A=416?=<71C7@13?/:>/7<
Disciplines
#32717<3/<23/:A6(173<13@
Publication Details
(/23567#/?@A3<@/::/56/<&/<73: ))/3%=<<3::)/@A?= ?73?:3F(#
2/;@ ?/79 :/?9' (A?B1AB?31A7C7AF(AB273@'3C3/:A63#=:31B:/?/@7@4=?
'313>A=?#327/A32<6707A7=<=4756+=:A/531A7C/A32/:17B;6/<<3:@0FH=<=A=E7<+1
(63;71/:7=:=5F
Authors
#/6@/(/23567=27:/?@A3<@?72&/::/56/< /;3@)/<73:/<(63<)/3)?/13F%=<<3:: =3:
/@A?=(AB/?A?73?:3F/C72 2/;@/C72 ?/79/<2'716/?2 :/?9
K7@8=B?</:/?A71:37@/C/7:/0:3/A'3@3/?16%<:7<3 6M>@?=B=D32B/B76;?7

Structure-activity studies reveal the molecular basis for GABA
B
-receptor
mediated inhibition of high voltage-activated calcium channels by α
αα
α-conotoxin
Vc1.1.
Mahsa Sadeghi
1
, Bodil B. Carstens
2
, Brid P. Callaghan
3
, James T. Daniel
4
, Han-Shen
Tae
1
, Tracey O’Donnell
5,6
, Joel Castro
5,6
, Stuart M. Brierley
5,6
, David J. Adams
1
,
David J. Craik
2
and Richard J. Clark
4*
.
1
Illawarra Health and Medical Research Institute (IHMRI), University of
Wollongong, Wollongong, NSW 2522, Australia.
2
The University of Queensland, Institute for Molecular Bioscience, Brisbane, Qld
4072, Australia.
3
Department of Anatomy & Neuroscience, University of Melbourne, Parkville,
Victoria 3010, Australia
4
The University of Queensland, School of Biomedical Sciences, Brisbane, Qld 4072,
Australia.
5
Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College
of Medicine and Public Health, Flinders University, Bedford Park, South Australia,
5042, Australia.
6
Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine,
University of Adelaide, South Australian Health and Medical Research Institute
(SAHMRI), North Terrace, Adelaide, Southern Australia 5000, Australia.
Address correspondence to: Dr. Richard J Clark, The University of Queensland,
School of Biomedical Sciences, Brisbane, QLD, 4072, Australia. Phone: +61 7 3365
1527. Fax: +61 7 3365 1766. Email: richard.clark@uq.edu.au
Page 1 of 44
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

Abbreviated title: Molecular basis of calcium channel inhibition by Vc1.1
Page 2 of 44
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

Abstract
α-Conotoxins are disulfide-bonded peptides from cone snail venoms and are
characterized by their affinity for nicotinic acetylcholine receptors (nAChR). Several α-
conotoxins with distinct selectivity for nAChR subtypes have been identified as potent
analgesics in animal models of chronic pain. However, a number of α-conotoxins have
been shown to inhibit N-type
calcium channel currents in rodent dissociated dorsal root
ganglion (DRG) neurons via activation of G protein-coupled GABA
B
receptors
(GABA
B
R). Therefore it is unclear whether activation of GABA
B
R or inhibition of
α9α10 nAChRs is the analgesic mechanism. To investigate the mechanisms by which α-
conotoxins provide analgesia, we synthesised a suite of Vc1.1 analogues where all
residues, except the conserved cysteines, in Vc1.1 were individually replaced by alanine
(A), lysine (K) and aspartic acid (D). Our results show that the amino acids in the first
loop play an important role in binding of the peptide to the receptor whereas those in the
second loop play an important role for the selectivity of the peptide for the GABA
B
R over
α9α10 nAChRs.
We designed a cVc1.1 analogue that is >8000-fold selective for
GABA
B
R-mediated inhibition of high voltage-activated (HVA) calcium channels over
α9α10 nAChRs and show that it is analgesic in a mouse model of chronic visceral
hypersensitivity (CVH). cVc1.1[D11A,E14A] caused dose-dependent inhibition of
colonic nociceptors with greater efficacy in ex vivo CVH colonic nociceptors relative
to healthy colonic nociceptors. These findings suggest that selectively targeting
GABA
B
R-mediated HVA calcium channel inhibition by α-conotoxins could be
effective for the treatment of chronic visceral pain.
Keywords: conotoxin, calcium channels, GPCRs, GABA
B
receptor, chronic pain,
analgesia
Page 3 of 44
ACS Paragon Plus Environment
ACS Chemical Biology
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

Citations
More filters
Journal ArticleDOI
TL;DR: This review highlights the diversity of new conotoxins uncovered since 2014, their three-dimensional structures and folds, novel chemical approaches to their syntheses, and their value as pharmacological tools to unravel complex biology.
Abstract: The venom of the marine predatory cone snails (genus Conus) has evolved for prey capture and defense, providing the basis for survival and rapid diversification of the now estimated 750+ species. A typical Conus venom contains hundreds to thousands of bioactive peptides known as conotoxins. These mostly disulfide-rich and well-structured peptides act on a wide range of targets such as ion channels, G protein-coupled receptors, transporters, and enzymes. Conotoxins are of interest to neuroscientists as well as drug developers due to their exquisite potency and selectivity, not just against prey but also mammalian targets, thereby providing a rich source of molecular probes and therapeutic leads. The rise of integrated venomics has accelerated conotoxin discovery with now well over 10,000 conotoxin sequences published. However, their structural and pharmacological characterization lags considerably behind. In this review, we highlight the diversity of new conotoxins uncovered since 2014, their three-dimensional structures and folds, novel chemical approaches to their syntheses, and their value as pharmacological tools to unravel complex biology. Additionally, we discuss challenges and future directions for the field.

148 citations

Journal ArticleDOI
TL;DR: The case for toxins as 'friends' is set out that are providing the basis of novel medicines, insecticides, and even nucleic acid sequencing technologies and emerging toxins are discussed that are becoming increasingly prevalent in a range of contexts through climate change and the globalisation of food supply chains.

41 citations

Journal ArticleDOI
TL;DR: A series of recent cryo-EM studies revealed critical details of the activation mechanism of the G protein-coupled receptors (GABAB) and provided comprehensive insights into the activation of the GABAB receptor which not only broaden our understanding of its structure, pharmacology, and physiological effects but also will ultimately facilitate the discovery of new therapeutic drugs and neuromodulators as mentioned in this paper.
Abstract: Metabotropic γ-aminobutyric acid G protein-coupled receptors (GABAB) represent one of the two main types of inhibitory neurotransmitter receptors in the brain. These receptors act both pre- and postsynaptically by modulating the transmission of neuronal signals and are involved in a range of neurological diseases, from alcohol addiction to epilepsy. A series of recent cryo-EM studies revealed critical details of the activation mechanism of GABAB Structures are now available for the receptor bound to ligands with different modes of action, including antagonists, agonists, and positive allosteric modulators, and captured in different conformational states from the inactive apo to the fully active state bound to a G protein. These discoveries provide comprehensive insights into the activation of the GABAB receptor, which not only broaden our understanding of its structure, pharmacology, and physiological effects but also will ultimately facilitate the discovery of new therapeutic drugs and neuromodulators.

34 citations

Journal ArticleDOI
TL;DR: This review describes the successes and limitations of seven cysteine‐rich scaffolds, their bioactive epitopes, and the resulting grafted peptides.
Abstract: Cysteine-rich peptides (CRPs) are small proteins of less than 100 amino acids in length characterized by the presence of disulfide bridges and common end-to-end macrocyclization. These properties confer hyperstability against high temperatures, salt concentration, serum presence, and protease degradation to CRPs. Moreover, their intercysteine domains (loops) are susceptible to residue hypervariability. CRPs have been successfully applied as stable scaffolds for molecular grafting, a protein engineering process in which cysteine-rich structures provide higher thermodynamic and metabolic stability to an epitope and acquire new biological function(s). This review describes the successes and limitations of seven cysteine-rich scaffolds, their bioactive epitopes, and the resulting grafted peptides.

23 citations

Journal ArticleDOI
06 Aug 2020-Toxins
TL;DR: This review scrutinises the N-terminal domain of the α-conotoxin family of peptides, a region defined by an invariant disulfide bridge, a turn-inducing proline residue and multiple polar sidechain residues, and focusses on structural features that provide analgesia through inhibition of high-voltage-activated Ca2+ channels.
Abstract: Several analgesic α-conotoxins have been isolated from marine cone snails. Structural modification of native peptides has provided potent and selective analogues for two of its known biological targets-nicotinic acetylcholine and γ-aminobutyric acid (GABA) G protein-coupled (GABAB) receptors. Both of these molecular targets are implicated in pain pathways. Despite their small size, an incomplete understanding of the structure-activity relationship of α-conotoxins at each of these targets has hampered the development of therapeutic leads. This review scrutinises the N-terminal domain of the α-conotoxin family of peptides, a region defined by an invariant disulfide bridge, a turn-inducing proline residue and multiple polar sidechain residues, and focusses on structural features that provide analgesia through inhibition of high-voltage-activated Ca2+ channels. Elucidating the bioactive conformation of this region of these peptides may hold the key to discovering potent drugs for the unmet management of debilitating chronic pain associated with a wide range of medical conditions.

20 citations

References
More filters
PatentDOI
04 May 1995-Science
TL;DR: The technique of native chemical ligation is employable for chemically synthesizing full length proteins as discussed by the authors, which are chemically identical to proteins produced by cell free synthesis, and can be refolded and/or oxidized to form native disulfide-containing protein molecules.
Abstract: Proteins of moderate size having native peptide backbones are produced by a method of native chemical ligation. Native chemical ligation employs a chemoselective reaction of two unprotected peptide segments to produce a transient thioester-linked intermediate. The transient thioester-linked intermediate then spontaneously undergoes a rearrangement to provide the full length ligation product having a native peptide bond at the ligation site. Full length ligation products are chemically identical to proteins produced by cell free synthesis. Full length ligation products may be refolded and/or oxidized, as allowed, to form native disulfide-containing protein molecules. The technique of native chemical ligation is employable for chemically synthesizing full length proteins.

3,347 citations

Journal ArticleDOI
TL;DR: Test calculations starting from conformers with random torsion angle values showed that DYANA is capable of efficient calculation of high-quality protein structures with up to 400 amino acid residues, and of nucleic acid structures.

2,768 citations

Journal ArticleDOI
TL;DR: Extension of the original 20-protein database to 200 proteins increased the fraction of residues for which backbone angles could be predicted from 65 to 74%, while reducing the error rate from 3 to 2.5%, and addition of a two-layer neural network filter to the database fragment selection process forms the basis for a new program, TALOS+, which further enhances the prediction rate to 88.5%.
Abstract: NMR chemical shifts in proteins depend strongly on local structure. The program TALOS establishes an empirical relation between 13C, 15N and 1H chemical shifts and backbone torsion angles ϕ and ψ (Cornilescu et al. J Biomol NMR 13 289–302, 1999). Extension of the original 20-protein database to 200 proteins increased the fraction of residues for which backbone angles could be predicted from 65 to 74%, while reducing the error rate from 3 to 2.5%. Addition of a two-layer neural network filter to the database fragment selection process forms the basis for a new program, TALOS+, which further enhances the prediction rate to 88.5%, without increasing the error rate. Excluding the 2.5% of residues for which TALOS+ makes predictions that strongly differ from those observed in the crystalline state, the accuracy of predicted ϕ and ψ angles, equals ±13°. Large discrepancies between predictions and crystal structures are primarily limited to loop regions, and for the few cases where multiple X-ray structures are available such residues are often found in different states in the different structures. The TALOS+ output includes predictions for individual residues with missing chemical shifts, and the neural network component of the program also predicts secondary structure with good accuracy.

2,407 citations

Journal ArticleDOI
03 Jan 1980-Nature
TL;DR: GABA clearly decreased the evoked release of accumulated 3H-noradrenaline from rat atria in vitro and3H-acetylcholine from preganglionic terminals in the rat superior cervical ganglion in vitro without affecting the basal release of tritium.
Abstract: The existence of a receptor for γ-aminobutyric acid (GABA) on neurones of the mammalian central nervous system (CNS) is now firmly established1–3. It is generally accepted that bicuculline (and its methohalide salts) is an antagonist of the actions of GABA4,5, although resistance to bicuculline has been described6,7. The view that bicuculline prevents GABA from interacting with a membrane recognition site is supported by results obtained in radiolabelled ligand binding studies8,9. Bicuculline-sensitive GABA receptors are not confined to neurones in the CNS but are also present on neurones and axons of the peripheral nervous system10,11. Their existence on neurones in sympathetic ganglia made us consider the possibility that they are also present on the terminals of such neurones. This has recently been tested12,13 by assuming that if GABA depolarises the terminals in a manner similar to the cell bodies, evoked transmitter output might be decreased (see ref. 14). GABA (ED50, 4µM) clearly decreased the evoked release of accumulated 3H-noradrenaline from rat atria in vitro and 3H-acetylcholine from preganglionic terminals in the rat superior cervical ganglion in vitro without affecting the basal release of tritium. In neither system was the effect of GABA antagonised by bicuculline methobromide, even though the ganglion terminal depolarisation was13. This suggested that the two phenomena, depolarisation and inhibition of transmitter release, were separate. The decrease in transmitter release not surprisingly leads to a decrease in the postsynaptic response15. Again, the decrease in response was not prevented by bicuculline or other GABA antagonists. We believe that these results indicate the presence of a novel GABA receptor on nerve terminals, a theory supported by results obtained with a variety of GABA analogues. For example, 3-aminopropane sulphonic acid (3-APS) which is at least as active as GABA at bicuculline sensitive sites10,16,17 is inactive at the terminal receptor. By contrast, the analogue baclofen (β-chlorophenyl GABA) is inactive at bicuculline-sensitive sites18,19 but is as active as GABA in reducing evoked transmitter output15. This effect of baclofen is stereospecific, the (−) isomer being > 100-fold more active than the (+) isomer15. We now report the presence of the novel GABA receptor within the mammalian CNS.

1,063 citations

Journal ArticleDOI
12 Mar 1981-Nature
TL;DR: It is reported that high-affinity saturable binding of 3H-baclof en and3H-G AB A to the GABAB site can be detected in fragments of crude synaptic membranes prepared from rat brain and that GABA and baclofen can compete for the same recognition site.
Abstract: The presence of a novel receptor for the neurotransmitter gamma-aminobutyric acid (GABA) on peripheral autonomic nerve terminals and in mammalian brain slices has been described recently. This receptor differs from the classical GABA site as it is unaffected by recognized GABA antagonists such as bicuculline and is not sensitive to the majority of accepted GABA-mimetics such as 3-aminopropanesulphonic acid (3-APS) or isoguvacine. We propose to designate the classical site as the GABA A and the novel site as the GABA B receptor. The beta-p-chlorophenyl derivative of GABA, baclofen, is stereospecifically active at the GABA B site whereas it is devoid of activity at the classical GABA A3 site. We now report that high-affinity saturable binding of 3H-baclofen and 3H-GABA to the GABA B site can be detected in fragments of crude synaptic membranes prepared from rat brain. The results support the concept of a novel GABA receptor within the mammalian brain and show that GABA and baclofen can compete for the same recognition site.

1,007 citations

Related Papers (5)
Frequently Asked Questions (18)
Q1. What contributions have the authors mentioned in the paper "Structure-activity studies reveal the molecular basis for gaba(b)-receptor mediated inhibition of high voltage-activated calcium channels by î±-conotoxin vc1.1" ?

To investigate the mechanisms by which α-conotoxins provide analgesia, the authors synthesized a suite of Vc1. The authors designed a cVc1. 1 analogue that is > 8000-fold selective for GABABR-mediated inhibition of high voltage-activated ( HVA ) calcium channels over α9α10 nAChRs and show that it is analgesic in a mouse model of chronic visceral hypersensitivity ( CVH ). This journal article is available at Research Online: https: //ro. uow. edu. au/ihmri/1263 These findings suggest that selectively targeting GABABR-mediated HVA calcium channel inhibition by α-conotoxins could be effective for the treatment of chronic visceral pain. 

1. 18 The importance of both loops for α9α10 nAChR inhibition by α-conotoxins is further supported by the structure-activity studies on RgIA, which have shown that mutations in both loops can have significant impact on potency. 27, 30 Therefore, the difference in the importance of the loop sequences on biological activity suggests that introducing a combination of mutations into loop 2 of Vc1. 21 These results suggest that GABABR-dependant HVA calcium channel inhibition underlies the analgesic effects of Vc1. Therefore, as predicted from the mutational Page 13 of 44 ACS Paragon Plus Environment ACS Chemical Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 data, introduction of the two alanine mutations caused a significant reduction in potency, which was further reduced by introduction of the cyclic backbone. 

alanine substitution at other residues resulted in loss of inhibitory activity on HVA calcium channels in mice DRG neurons. 

RgIA analogues selective for only α9α10 nAChR inhibition have been shown to be effective in animal models of chemotherapy-induced neuropathic pain. 

In cells showing HVA calcium channel current (IBa) inhibition in response to α-conotoxin exposure, subsequent application of baclofen (50 µM) suppressed IBa further depending on the tested analogues. 

Whilst both cVc1.1 and cVc1.1[D11A,E14A] cause greater inhibition of colonic nociceptors in CVH states compared with healthy states, they evoke equivalent inhibition in CVH states, again implicating GABABR activation in this anti-nociceptive action. 

Although theα9α10 nAChR subtype was originally proposed to be the primary analgesic target ofVc1.1 and RgIA, several observations have cast doubt on its importance for α-conotoxin analgesia instead supporting a more prominent role of HVA calcium channel inhibition via GABABR activation. 

inhibiting HVA calcium channel currents either directly with selective antagonists or indirectly via GPCR modulation causes analgesia in animals and humans8. 

Vc1.1[D11A,E14A] and cVc1.1[D11A, E14A] were 3,700-fold and 8,800-fold more selective for HVA calcium channel inhibition over inhibition of α9α10 nAChRs, respectively. 

Given that Vc1.1 and RgIA have been shown to inhibit both α9α10 nAChRs and GABABR-mediated HVA calcium currents, the challenge has been to determine the contribution of these two mechanisms to the analgesic effects of these peptides. 

The authors have previously shown an increased expression of CaV2.2 within colon-innervating DRG neurons from CVH mice,21 which is likely to explain the enhanced anti-nociceptive actions of both cVc1.1 and cVc1.1[D11A,E14A] following GABABR activation. 

these individual Vc1.1 mutations substantially reduced inhibition of α9α10 nAChRs26 and modelling of the interaction between Vc1.1 and α9α10 predicted that Asp11 and Glu14 form key salt bridge interactions with arginine residues on the α9α10 nAChR.27 Furthermore, during their previous development of an orally active, backbone cyclised analogue of Vc1.1, the authors observed a five-fold decrease and 12-fold increase in potency for α9α10 nAChR and HVA calcium channel inhibition, respectively, when Vc1.1 was cyclised.31 

it was subsequently shown that Vc1.1 and the α9α10 nAChR-selective αconotoxin RgIA12 were both able to also potently inhibit HVA calcium channel currents in rat dorsal root ganglion (DRG) neurons and that this inhibition was dependent on GABABR activation 13-15. 

cVc1.1[D11A,E14A] caused dose-dependent inhibition of ex vivo colonic nociceptors from healthy (Figure 5A) and CVH (Figure 5B) mice. 

This substantial selectivity for HVA calcium channel inhibition suggests that cVc1.1[D11A, E14A] is an excellent molecule for investigating the role of this pathway in the analgesic action of Vc1.1. 

a subset of α-conotoxins have been shown to inhibit high voltage-activated(HVA) calcium channel currents via activation of the GABAB receptor (GABABR) (Figure 1B) . 

Representative traces of HVA calcium channel inhibition in mice DRG neurons by Vc1.1 analogues equipotent to Vc1.1 and less effective than Vc1.1 are shown in Figure S2A. 

Although the estimated IC50 for these analogues (13.1, 6.8, 23.1 and 170.7 nM, respectively) are similar to that of Vc1.1 (2.5 nM), the maximum inhibition of IBa at 100 nM concentration of these peptides is greater than those with Vc1.1, highlighting that the efficacy of these peptides is greater on inhibiting IBa in DRG neurons.