65 results on '"Ragnarsson, Lotten"'
Search Results
52. Novel Superantigens for Immunotherapy of Multiple Myeloma
- Author
-
Ragnarsson, Lotten and Ragnarsson, Lotten
- Published
- 2000
53. Extracellular Surface Residues of the α1B-Adrenoceptor Critical for G Protein–Coupled Receptor Function
- Author
-
Ragnarsson, Lotten, Andersson, Åsa, Thomas, Walter G., and Lewis, Richard J.
- Abstract
Ligand binding and conformational changes that accompany signaling from G protein–coupled receptors (GPCRs) have mostly focused on the role of transmembrane helices and intracellular loop regions. However, recent studies, including several GPCRs cocrystallized with bound ligands, suggest that the extracellular surface (ECS) of GPCRs plays an important role in ligand recognition, selectivity, and binding, as well as potentially contributing to receptor activation and signaling. This study applied alanine-scanning mutagenesis to investigate the role of the complete ECS of the α1B-adrenoreceptor on norepinephrine (NE) potency, affinity, and efficacy. Half (24 of 48) of the ECS mutations significantly decreased NE potency in an inositol 1-phosphate assay. Most mutations reduced NE affinity (17) determined from [3H]prazosin displacement studies, whereas four mutations at the entrance to the NE binding pocket enhanced NE affinity. Removing the influence of NE affinity and receptor expression levels on NE potency gave a measure of NE efficacy, which was significantly decreased for 11 of 48 ECS mutants. These different effects tended to cluster to different regions of the ECS, which is consistent with different regions of the ECS playing discrete functional roles. Exposed ECS residues at the entrance to the NE binding pocket mostly affected NE affinity, whereas buried or structurally significant residues mostly affected NE efficacy. The broad potential for ECS mutations to affect GPCR function has relevance for the increasing number of nonsynonymous single nucleotide polymorphisms now being identified in GPCRs.
- Published
- 2015
- Full Text
- View/download PDF
54. Expression and Pharmacology of Endogenous Cav Channels in SH-SY5Y Human Neuroblastoma Cells.
- Author
-
Sousa, Silmara R., Vetter, Irina, Ragnarsson, Lotten, and Lewis, Richard J.
- Subjects
NEUROBLASTOMA ,GENE expression ,PHARMACOLOGY ,CALCIUM channels ,CANCER cells ,CELL physiology ,MESSENGER RNA ,CELLULAR signal transduction ,THERAPEUTICS - Abstract
SH-SY5Y human neuroblastoma cells provide a useful in vitro model to study the mechanisms underlying neurotransmission and nociception. These cells are derived from human sympathetic neuronal tissue and thus, express a number of the Ca
v channel subtypes essential for regulation of important physiological functions, such as heart contraction and nociception, including the clinically validated pain target Cav 2.2. We have detected mRNA transcripts for a range of endogenous expressed subtypes Cav 1.3, Cav 2.2 (including two Cav 1.3, and three Cav 2.2 splice variant isoforms) and Cav 3.1 in SH-SY5Y cells; as well as Cav auxiliary subunits α2 δ1–3 , β1 , β3 , β4 , γ1 , γ4–5 , and γ7 . Both high- and low-voltage activated Cav channels generated calcium signals in SH-SY5Y cells. Pharmacological characterisation using ω-conotoxins CVID and MVIIA revealed significantly (∼ 10-fold) higher affinity at human versus rat Cav 2.2, while GVIA, which interacts with Cav 2.2 through a distinct pharmacophore had similar affinity for both species. CVID, GVIA and MVIIA affinity was higher for SH-SY5Y membranes vs whole cells in the binding assays and functional assays, suggesting auxiliary subunits expressed endogenously in native systems can strongly influence Cav 2.2 channels pharmacology. These results may have implications for strategies used to identify therapeutic leads at Cav 2.2 channels. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
55. Conopeptide ρ-TIA Defines a New Allosteric Site on the Extracellular Surface of the α1B-Adrenoceptor.
- Author
-
Ragnarsson, Lotten, Ching-I Anderson Wang, Andersson, Åsa, Fajarningsih, Dewi, Monks, Thea, Brust, Andreas, Rosengren, K. Johan, and Lewis, Richard J.
- Subjects
- *
G protein coupled receptors , *ALLOSTERIC regulation , *ADRENERGIC receptors , *RADIOLIGAND assay , *HYDROGEN bonding , *MEMBRANE proteins - Abstract
The G protein-coupled receptor (GPCR) superfamily is an important drug target that includes over 1000 membrane receptors that functionally couple extracellular stimuli to intracellular effectors. Despite the potential of extracellular surface (ECS) residues in GPCRs to interact with subtype-specific allosteric modulators, few ECS pharmacophores for class A receptors have been identified. Using the turkey β1-adrenergic receptor crystal structure, we modeled the α1B-adrenoceptor (α1B-AR) to help identify the allosteric site for ρ-conopeptide TIA, an inverse agonist at this receptor. Combining mutational radioligand binding and inositol 1-phosphate signaling studies, together with molecular docking simulations using a refined NMR structure of ρ-TIA, we identified 14 residues on the ECS of the α1B-AR that influenced ρ-TIA binding. Double mutant cycle analysis and docking confirmed that ρ-TIA binding was dominated by a salt bridge and cation-π between Arg-4-ρ-TIA and Asp-327 and Phe-330, respectively, and a T-stacking-π interaction between Trp-3-ρ-TIA and Phe-330. Water-bridging hydrogen bonds between Asn-2-ρ-TIA and Val-197, Trp-3-ρ-TIA and Ser-318, and the positively charged N terminus and Glu-186, were also identified. These interactions reveal that peptide binding to the ECS on transmembrane helix 6 (TMH6) and TMH7 at the base of extracellular loop 3 (ECL3) is sufficient to allosterically inhibit agonist signaling at a GPCR. The ligand-accessible ECS residues identified provide the first view of an allosteric inhibitor pharmacophore for α1-adrenoceptors and mechanistic insight and a new set of structural constraints for the design of allosteric antagonists at related GPCRs. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
56. Glycerotoxin stimulates neurotransmitter release from N-type Ca2+ channel expressing neurons.
- Author
-
Schenning, Mitja, Proctor, Dustin T., Ragnarsson, Lotten, Barbier, Julien, Lavidis, Nickolas A., Molgó, Jordi J., Zamponi, Gerald W., Schiavo, Giampietro, and Meunier, Frederic A.
- Subjects
NEUROTRANSMITTERS ,FROGS as laboratory animals ,SYNAPTOSOMES ,NEURONS ,NERVOUS system - Abstract
Glycerotoxin (GLTx) is capable of stimulating neurotransmitter release at the frog neuromuscular junction by directly interacting with N-type Ca
2+ (Cav 2.2) channels. Here we have utilized GLTx as a tool to investigate the functionality of Cav 2.2 channels in various mammalian neuronal preparations. We first adapted a fluorescent-based high-throughput assay to monitor glutamate release from rat cortical synaptosomes. GLTx potently stimulates glutamate secretion and Ca2+ influx in synaptosomes with an EC50 of 50 pm. Both these effects were prevented using selective Cav 2.2 channel blockers suggesting the functional involvement of Cav 2.2 channels in mediating glutamate release in this system. We further show that both Cav 2.1 (P/Q-type) and Cav 2.2 channels contribute equally to depolarization-induced glutamate release. We then investigated the functionality of Cav 2.2 channels at the neonatal rat neuromuscular junction. GLTx enhances both spontaneous and evoked neurotransmitter release causing a significant increase in the frequency of postsynaptic action potentials. These effects were blocked by specific Cav 2.2 channel blockers demonstrating that either GLTx or its derivatives could be used to selectively enhance the neurotransmitter release from Cav 2.2-expressing mammalian neurons. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
57. Mutations in the NPxxY motif stabilize pharmacologically distinct conformational states of the α1B- and β2-adrenoceptors.
- Author
-
Ragnarsson, Lotten, Andersson, Åsa, Thomas, Walter G., and Lewis, Richard J.
- Subjects
MOLECULAR structure of G protein coupled receptors ,BETA adrenoceptors ,ALPHA adrenoceptors ,GENETIC mutation ,MUTAGENESIS - Abstract
Inactive conformations of α
1B -AR and β2 -AR are structurally similar but exhibit distinct pharmacological properties. Pharmacological heterogeneity in GPCRs: G protein–coupled receptors (GPCRs) share a common overall structure that switches between inactive and active conformations, with the latter being stabilized by ligand binding. Ragnarsson et al. investigated the role of a switch formed by hydrogen bonds between the NPxxY motif in transmembrane helix 7 (TMH7) and a tyrosine residue in TMH5 in activation of the α1B - and β2 -adrenoceptors (ARs). Mutating the switch stabilized the inactive receptor conformations, resulting in reduced signaling. Although the mutations reduced the agonist affinity of the β2 -AR without affecting signaling efficacy, they enhanced the agonist affinity and reduced the signaling efficacy of the α1B -AR. These findings show that inactive conformations of individual GPCRs have different pharmacological properties, which may help with designing new drugs or improving the efficacy and reducing the side effects of existing drugs. G protein–coupled receptors (GPCRs) convert extracellular stimuli to intracellular responses that regulate numerous physiological processes. Crystallographic and biophysical advances in GPCR structural analysis have aided investigations of structure-function relationships that clarify our understanding of these dynamic receptors, but the molecular mechanisms associated with activation and signaling for individual GPCRs may be more complex than was previously appreciated. Here, we investigated the proposed water-mediated, hydrogen-bonded activation switch between the conserved NPxxY motif on transmembrane helix 7 (TMH7) and a conserved tyrosine in TMH5, which contributes to α1B -adrenoceptor (α1B -AR) and β2 -AR activation. Disrupting this bond by mutagenesis stabilized the α1B -AR and the β2 -AR in inactive-state conformations, which displayed decreased agonist potency for stimulating downstream IP1 and cAMP signaling, respectively. Compared to that for wild-type receptors, agonist-mediated β-arrestin recruitment was substantially reduced or abolished for all α1B -AR and β2 -AR inactive-state mutants. However, the inactive-state β2 -ARs exhibited decreased agonist affinity, whereas the inactive-state α1B -ARs had enhanced agonist affinity. Conversely, antagonist affinity was unchanged for inactive-state conformations of both α1B -AR and β2 -AR. Removing the influence of agonist affinity on agonist potency gave a measure of signaling efficacy, which was markedly decreased for the α1B -AR mutants but little altered for the β2 -AR mutants. These findings highlight the pharmacological heterogeneity of inactive-state GPCR conformations, which may facilitate the rational design of drugs that target distinct conformational states of GPCRs. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
58. Mutations in the NPxxY motif stabilize pharmacologically distinct conformational states of the α1B- and β2-adrenoceptors
- Author
-
Ragnarsson, Lotten, Andersson, Åsa, Thomas, Walter G., and Lewis, Richard J.
- Abstract
Inactive conformations of α1B-AR and β2-AR are structurally similar but exhibit distinct pharmacological properties.
- Published
- 2019
- Full Text
- View/download PDF
59. The structural conformation of the tachykinin domain drives the anti-tumoural activity of an octopus peptide in melanoma BRAFV600E.
- Author
-
Moral‐Sanz, Javier, Fernandez‐Rojo, Manuel A., Colmenarejo, Gonzalo, Kurdyukov, Sergey, Brust, Andreas, Ragnarsson, Lotten, Andersson, Åsa, Vila, Sabela F., Cabezas‐Sainz, Pablo, Wilhelm, Patrick, Vela‐Sebastián, Ana, Fernández‐Carrasco, Isabel, Chin, Yanni K. Y., López‐Mancheño, Yaiza, Smallwood, Taylor B., Clark, Richard J., Fry, Bryan G., King, Glenn F., Ramm, Grant A., and Alewood, Paul F.
- Subjects
- *
ADENOSINE triphosphate , *GENETIC mutation , *MELANOMA , *MOLECULAR models , *RNA , *FISHES , *TRANSFERASES , *RESEARCH funding , *MOLLUSKS , *CELL lines , *CALCIUM , *REACTIVE oxygen species , *ANIMALS , *MICE - Abstract
Background and Purpose: Over past decades, targeted therapies and immunotherapy have improved survival and reduced the morbidity of patients with BRAF-mutated melanoma. However, drug resistance and relapse hinder overall success. Therefore, there is an urgent need for novel compounds with therapeutic efficacy against BRAF-melanoma. This prompted us to investigate the antiproliferative profile of a tachykinin-peptide from the Octopus kaurna, Octpep-1 in melanoma.Experimental Approach: We evaluated the cytotoxicity of Octpep-1 by MTT assay. Mechanistic insights on viability and cellular damage caused by Octpep-1 were gained via flow cytometry and bioenergetics. Structural and pharmacological characterization was conducted by molecular modelling, molecular biology, CRISPR/Cas9 technology, high-throughput mRNA and calcium flux analysis. In vivo efficacy was validated in two independent xerograph animal models (mice and zebrafish).Key Results: Octpep-1 selectively reduced the proliferative capacity of human melanoma BRAFV600E -mutated cells with minimal effects on fibroblasts. In melanoma-treated cells, Octpep-1 increased ROS with unaltered mitochondrial membrane potential and promoted non-mitochondrial and mitochondrial respiration with inefficient ATP coupling. Molecular modelling revealed that the cytotoxicity of Octpep-1 depends upon the α-helix and polyproline conformation in the C-terminal region of the peptide. A truncated form of the C-terminal end of Octpep-1 displayed enhanced potency and efficacy against melanoma. Octpep-1 reduced the progression of tumours in xenograft melanoma mice and zebrafish.Conclusion and Implications: We unravel the intrinsic anti-tumoural properties of a tachykinin peptide. This peptide mediates the selective cytotoxicity in BRAF-mutated melanoma in vitro and prevents tumour progression in vivo, providing a foundation for a therapy against melanoma. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
60. Neurotoxic and cytotoxic peptides underlie the painful stings of the tree nettle Urtica ferox.
- Author
-
Jing Xie, Robinson, Samuel D., Gilding, Edward K., Jami, Sina, Deuis, Jennifer R., Rehm, Fabian B. H., Yap, Kuok, Ragnarsson, Lotten, Lai Yue Chan, Hamilton, Brett R., Harvey, Peta J., Craik, David J., Vetter, Irina, and Durek, Thomas
- Subjects
- *
STINGING nettle , *PEPTIDES , *SODIUM channels , *ACTIVATION energy , *CELL membranes , *TOXINS - Abstract
The stinging hairs of plants from the family Urticaceae inject compounds that inflict pain to deter herbivores. The sting of the New Zealand tree nettle (Urtica ferox) is among the most painful of these and can cause systemic symptoms that can even be life-threatening; however, the molecular species effecting this response have not been elucidated. Here we reveal that two classes of peptide toxin are responsible for the symptoms of U. ferox stings: Δ-Uf1a is a cytotoxic thionin that causes pain via disruption of cell membranes, while β/δ-Uf2a defines a new class of neurotoxin that causes pain and systemic symptoms via modulation of voltage-gated sodium (NaV) channels. We demonstrate using whole-cell patch-clamp electrophysiology experiments that β/δ-Uf2a is a potent modulator of human NaV1.5 (EC50: 55 nM), NaV1.6 (EC50: 0.86 nM), and NaV1.7 (EC50: 208 nM), where it shifts the activation threshold to more negative potentials and slows fast inactivation. We further found that both toxin classes are widespread among members of the Urticeae tribe within Urticaceae, suggesting that they are likely to be pain-causing agents underlying the stings of other Urtica species. Comparative analysis of nettles of Urtica, and the recently described pain-causing peptides from nettles of another genus, Dendrocnide, indicates that members of tribe Urticeae have developed a diverse arsenal of pain-causing peptides. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
61. Erythromelalgia caused by the missense mutation p.Arg220Pro in an alternatively spliced exon of SCN9A (NaV1.7).
- Author
-
Deuis JR, Kumble S, Keramidas A, Ragnarsson L, Simons C, Pais L, White SM, and Vetter I
- Subjects
- Humans, Mutation, Missense genetics, NAV1.7 Voltage-Gated Sodium Channel genetics, Pain genetics, Mutation, Exons genetics, Erythromelalgia genetics
- Abstract
Erythromelalgia (EM), is a familial pain syndrome characterized by episodic 'burning' pain, warmth, and erythema. EM is caused by monoallelic variants in SCN9A, which encodes the voltage-gated sodium channel (NaV) NaV1.7. Over 25 different SCN9A mutations attributed to EM have been described to date, all identified in the SCN9A transcript utilizing exon 6N. Here we report a novel SCN9A missense variant identified in seven related individuals with stereotypic episodes of bilateral lower limb pain presenting in childhood. The variant, XM_011511617.3:c.659G>C;p.(Arg220Pro), resides in the exon 6A of SCN9A, an exon previously shown to be selectively incorporated by developmentally regulated alternative splicing. The mutation is located in the voltage-sensing S4 segment of domain I, which is important for regulating channel activation. Functional analysis showed the p.Arg220Pro mutation altered voltage-dependent activation and delayed channel inactivation, consistent with a NaV1.7 gain-of-function molecular phenotype. These results demonstrate that alternatively spliced isoforms of SCN9A should be included in all genomic testing of EM., (© The Author(s) 2023. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
62. Changes in Potency and Subtype Selectivity of Bivalent Na V Toxins are Knot-Specific.
- Author
-
Tran P, Tran HNT, McMahon KL, Deuis JR, Ragnarsson L, Norman A, Sharpe SJ, Payne RJ, Vetter I, and Schroeder CI
- Subjects
- Humans, Peptides pharmacology
- Abstract
Disulfide-rich peptide toxins have long been studied for their ability to inhibit voltage-gated sodium channel subtype Na
V 1.7, a validated target for the treatment of pain. In this study, we sought to combine the pore blocking activity of conotoxins with the gating modifier activity of spider toxins to design new bivalent inhibitors of NaV 1.7 with improved potency and selectivity. To do this, we created an array of heterodimeric toxins designed to target human NaV 1.7 by ligating a conotoxin to a spider toxin and assessed the potency and selectivity of the resulting bivalent toxins. A series of spider-derived gating modifier toxins (GpTx-1, ProTx-II, gHwTx-IV, JzTx-V, CcoTx-1, and Pn3a) and two pore-blocker μ-conotoxins, SxIIIC and KIIIA, were used for this study. We employed either enzymatic ligation with sortase A for C- to N-terminal ligation or click chemistry for N- to N-terminal ligation. The bivalent peptide resulting from ligation of ProTx-II and SxIIIC (Pro[LPATG6 ]Sx) was shown to be the best combination as native ProTx-II potency at hNaV 1.7 was conserved following ligation. At hNaV 1.4, a synergistic effect between the pore blocker and gating modifier toxin moieties was observed, resulting in altered sodium channel subtype selectivity compared to the parent peptides. Further studies including mutant bivalent peptides and mutant hNaV 1.7 channels suggested that gating modifier toxins have a greater contribution to the potency of the bivalent peptides than pore blockers. This study delineated potential benefits and drawbacks of designing pharmacological hybrid peptides targeting hNaV 1.7.- Published
- 2023
- Full Text
- View/download PDF
63. Low potency inhibition of Na V 1.7 by externally applied QX-314 via a depolarizing shift in the voltage-dependence of activation.
- Author
-
Klasfauseweh T, Israel MR, Ragnarsson L, Cox JJ, Durek T, Carter DA, Leffler A, Vetter I, and Deuis JR
- Subjects
- HEK293 Cells, Humans, Sodium metabolism, Sodium Channel Blockers pharmacology, Anesthetics, Local pharmacology, Lidocaine analogs & derivatives, Lidocaine pharmacology
- Abstract
QX-314 is a quaternary permanently charged lidocaine derivative that inhibits voltage-gated sodium channels (Na
V ). As it is membrane impermeable, it is generally considered that QX-314 applied externally is inactive, unless it can gain access to the internal local anesthetic binding site via another entry pathway. Here, we characterized the electrophysiological effects of QX-314 on NaV 1.7 heterologously expressed in HEK293 cells, and found that at high concentrations, external QX-314 inhibited NaV 1.7 current (IC50 2.0 ± 0.3 mM) and shifted the voltage-dependence to more depolarized potentials (ΔV50 +10.6 mV). Unlike lidocaine, the activity of external QX-314 was not state- or use-dependent. The effect of externally applied QX-314 on NaV 1.7 channel biophysics differed to that of internally applied QX-314, suggesting QX-314 has an additional externally accessible site of action. In line with this hypothesis, disruption of the local anesthetic binding site in a [F1748A]NaV 1.7 mutant reduced the potency of lidocaine by 40-fold, but had no effect on the potency or activity of externally applied QX-314. Therefore, we conclude, using an expression system where QX-314 was unable to cross the membrane, that externally applied QX-314 is able to inhibit NaV 1.7 peak current at low millimolar concentrations., (Copyright © 2022 Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
64. The Tarantula Venom Peptide Eo1a Binds to the Domain II S3-S4 Extracellular Loop of Voltage-Gated Sodium Channel Na V 1.8 to Enhance Activation.
- Author
-
Deuis JR, Ragnarsson L, Robinson SD, Dekan Z, Chan L, Jin AH, Tran P, McMahon KL, Li S, Wood JN, Cox JJ, King GF, Herzig V, and Vetter I
- Abstract
Venoms from cone snails and arachnids are a rich source of peptide modulators of voltage-gated sodium (Na
V ) channels, however relatively few venom-derived peptides with activity at the mammalian NaV 1.8 subtype have been isolated. Here, we describe the discovery and functional characterisation of β-theraphotoxin-Eo1a, a peptide from the venom of the Tanzanian black and olive baboon tarantula Encyocratella olivacea that modulates NaV 1.8. Eo1a is a 37-residue peptide that increases NaV 1.8 peak current (EC50 894 ± 146 nM) and causes a large hyperpolarising shift in both the voltage-dependence of activation (ΔV50 -20.5 ± 1.2 mV) and steady-state fast inactivation (ΔV50 -15.5 ± 1.8 mV). At a concentration of 10 μM, Eo1a has varying effects on the peak current and channel gating of NaV 1.1-NaV 1.7, although its activity is most pronounced at NaV 1.8. Investigations into the binding site of Eo1a using NaV 1.7/NaV 1.8 chimeras revealed a critical contribution of the DII S3-S4 extracellular loop of NaV 1.8 to toxin activity. Results from this work may form the basis for future studies that lead to the rational design of spider venom-derived peptides with improved potency and selectivity at NaV 1.8., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Deuis, Ragnarsson, Robinson, Dekan, Chan, Jin, Tran, McMahon, Li, Wood, Cox, King, Herzig and Vetter.)- Published
- 2022
- Full Text
- View/download PDF
65. Characterisation of d-Conotoxin TxVIA as a Mammalian T-Type Calcium Channel Modulator.
- Author
-
Wang D, Himaya SWA, Giacomotto J, Hasan MM, Cardoso FC, Ragnarsson L, and Lewis RJ
- Subjects
- Animals, Calcium Channels, T-Type, Cell Line, Humans, Models, Molecular, Protein Binding, Protein Conformation, Rats, Zebrafish, Conotoxins pharmacology
- Abstract
The 27-amino acid (aa)-long d-conotoxin TxVIA, originally isolated from the mollusc-hunting cone snail Conus textile , slows voltage-gated sodium (Na
V ) channel inactivation in molluscan neurons, but its mammalian ion channel targets remain undetermined. In this study, we confirmed that TxVIA was inactive on mammalian NaV 1.2 and NaV 1.7 even at high concentrations (10 µM). Given the fact that invertebrate NaV channel and T-type calcium channels (CaV 3.x) are evolutionarily related, we examined the possibility that TxVIA may act on CaV 3.x. Electrophysiological characterisation of the native TxVIA on CaV 3.1, 3.2 and 3.3 revealed that TxVIA preferentially inhibits CaV 3.2 current (IC50 = 0.24 mM) and enhances CaV 3.1 current at higher concentrations. In fish bioassays TxVIA showed little effect on zebrafish behaviours when injected intramuscular at 250 ng/100 mg fish. The binding sites for TxVIA at NaV 1.7 and CaV 3.1 revealed that their channel binding sites contained a common epitope.- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.