Abstract
Arginine vasopressin (AVP) and oxytocin (OT) are nonapeptides that bind to G-protein coupled receptors and influence social behaviors. Consensus mammalian AVP and OT (Leu8-OT) sequences are highly conserved. In marmosets, an amino acid change in the 8th position of the peptide (Pro8-OT) exhibits unique structural and functional properties. There is ~85% structural homology between the OT receptor (OTR) and vasopressin 1a receptor (V1aR) resulting in significant cross-reactivity between the ligands and receptors. Chinese hamster ovary (CHO) cells expressing marmoset (mV1aR), macaque (qV1aR), or human vasopressin receptor 1a (hV1aR) were used to assess AVP, Leu8-OT and Pro8-OT pharmacological profiles. To assess activation of Gq, functional assays were performed using Fluo-3 to measure ligand-induced Ca2+ mobilization. In all three V1aR-expressing cell lines, AVP was more potent than the OT ligands. To assess ligand-induced hyperpolarization, FLIPR Membrane Potential (FMP) assays were performed. In all three V1aR lines, AVP was more potent than the OT analogs. The distinctive U-shaped concentration-response curve displayed by AVP may reflect enhanced desensitization of the mV1aR and hV1aR, which is not observed with qV1aR. Evaluation of Ca2+-activated potassium (K+) channels using the inhibitors apamin, paxilline, and TRAM-34 demonstrated that both intermediate and large conductance Ca2+-activated K+ channels contributed to membrane hyperpolarization, with different pharmacological profiles identified for distinct ligand-receptor combinations. Taken together, these data suggest differences in ligand-receptor signaling that may underlie differences in social behavior. Integrative studies of behavior, genetics and ligand-receptor interaction will help elucidate the connection between receptor pharmacology and social behaviors.
Keywords: arginine vasopressin, oxytocin, vasopressin receptor 1a, g-protein coupled receptor, calcium-activated potassium channels
Graphical Abstract
1. INTRODUCTION
AVP and OT are peptide ligands synthesized in the hypothalamus and secreted into the circulation affecting a host of peripheral physiological functions, including renal water absorption, vasoconstriction, parturition, and lactation. In the central nervous system, these neuropeptides are expressed in the ‘Social Brain Network’ and contribute to a variety of social behaviors [1-4] with AVP and OT generally exhibiting opposing behavioral roles and physiological functions [5-8]. These central AVP and OT projections are regulated by genetic, epigenetic, physiological, social, and environmental factors that affect mating preferences, parental care, decision making, social recognition, learning, and memory, and perturbations are associated with social behavioral deficits and psychopathologies [5, 9].
The OT-AVP neuropeptide family is highly evolutionarily conserved, with members or homologs identified in invertebrates and vertebrate taxa. AVP is completely conserved in mammals, and differs from the consensus mammalian OT sequence at only amino acids 3 and 8 [10-12]. In New World Monkeys (NWMs), six novel OT-analogs have been identified [11-15]. In mammals, the OT-AVP receptor family is comprised of three distinct vasopressin receptors (V1aR, V1bR, V2R) and one oxytocin receptor (OTR) [16]. In the vertebrate brain, V1aR is the most abundant and widely expressed vasopressin receptor [17] and differences in V1aR receptor genetic variability and expression patterns are associated with various social phenotypes [12, 18-21]. Across the phylogenetic tree, coevolution is observed for OT and AVP ligands and their receptors [11, 12, 22]. With ~85% structural homology between OTR and V1aR, significant cross-reactivity is observed between OT-AVP family ligands and receptors [12, 23-26]. AVP binds with higher affinity than oxytocin analogs at marmoset (mV1aR), macaque (qV1aR) and human (hV1aRs) vasopressin 1a receptors. Interestingly, OT analogs display 10-fold less binding affinity at the mV1aR than hV1aR and qV1aR, which may provide important taxon-specific insights into ligand-receptor signaling [27]. Due to high conservation in both ligands and receptors, the OT-AVP family is an excellent model system for studying the effects of ligand and receptor structure on G-protein coupled receptor (GPCR)-mediated signaling.
GPCRs bind to heterotrimeric G-proteins that are classified into four subfamilies: Gs (stimulation of adenylate cyclase), Gi/o (inhibition of adenylate cyclase and/or opening of potassium currents), Gq (activation of phospholipase C; calcium mobilization) and G12 (activatior of Rho GTPases) [28]. As a GPCR, V1aR couples to G protein(s), activating signaling cascades that can result in Ca2+ mobilization and alteration of voltage and ligand-gated ion channels [6, 29-31]. In rat motorneurons, AVP can affect neuronal excitability by exerting a dual effect that suppresses K+ currents and enhances cation conductance [32]. Ligand binding can have agonistic or antagonistic effects on downstream signaling. At the hV1aR, OT analogs partially antagonize AVP binding, and are partial agonists at inducing Ca2+ influx [27].
In this report, we assess differences in GPCR signaling due to natural genetic variation in V1aRs associated with phenotypic consequences: marmoset (NWM, socially monogamous), macaque (OWM, nonmanogamous), and human (apes, socially monogamous) [11, 12, 21, 27]. We stably transfected CHO cells expressing mV1aR, qV1aR, and hV1aR and characterized AVP- and OT-analog G-protein signaling pathways by monitoring Ca2+ mobilization and membrane hyperpolarization.
2. MATERIALS AND METHODS
2.1. Chinese hamster ovary (CHO) cell cultures.
Transcriptome profiling confirmed that neuronal cell lines ND7/23, F-11 and SH-SY5Y express OT receptor [33]. Likewise, RT-PCR showed neuroblastoma cell lines SK-N-SH, SH-SY5Y, IMR-32 and astrocytoma cell line MOG-G-UVW all express OT receptor [34]. We excluded cell lines that express OT receptors from our studies inasmuch as endogenous expression would contribute to observed ligand-induced signaling. Therefore, we selected CHO cells as a common transfection background since they do not express OT receptors [35]. Wild-type Chinese hamster ovarian-K1 (CHO-K1) cells were purchased from ATCC (CCL-61) and cultured in Ham’s F12 (Hyclone SH30026.01), 10% fetal bovine serum (FBS) (Atlanta Biologicals S11550), 1.5% HEPES 1M Solution (Hyclone SH30231.01), 1% Penicillin-Streptomycin (10,000 U/mL; Life Technologies 15140-163). CHO-K1 cells stably transfected with mV1aR, qV1aR and hV1aR as described [27, 36] and were received from Dr. Myron Toews lab (University of Nebraska Medical Center; UNMC). mV1aR, qV1aR and hV1aR expressing cells were cultured in Ham’s F12 (Hyclone SH30026.01), 10% FBS (Atlanta Biologicals S11550), 1.5% HEPES 1M Solution (Hyclone SH30231.01), 1% Penicillin-Streptomycin (10,000 U/mL; Life Technologies 15140-163) and 400 mg/mL G418 (RPI Corp. G64000-5.0). Cells were cultured at 37°C in 5% CO2 and 90% humidity.
2.2. Drugs.
AVP, Leu8-OT, and Pro8-OT (Anaspec 58863), 1,2-Bis(2-aminophenoxy)ethane-N,N, N9,N9-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM) (A1076; Sigma-Aldrich), NS-1619 (Sigma-Aldrich N170), Paxilline (Sigma-Aldrich P2928), SKA-31 (Sigma-Aldrich S5573), thapsigargin (Sigma-Aldrich T9033), and TRAM-34 (Sigma-Aldrich T6700) were reconstituted in DMSO. Pertussis toxin (Sigma-Aldrich P7208) was reconstituted in ultrapure water with 5 mg/mL bovine serum albumin (Fisher Scientific BP1600-100). Apamin (Sigma-Aldrich A1289) was reconstituted in 0.05 M acetic acid.
2.3. Ca2+ Mobilization Assay.
The effects of AVP and OT analog addition on Ca2+ mobilization were examined using Fluo3-AM fluorescence (Molecular Probes F1241) monitored with a FLIPR2 plate reader (Molecular Devices, Sunnyvale, CA). FLIPR operates by illuminating the bottom of a 96-well microplate with an air-cooled laser and measuring the fluorescence emissions from cell-permeant dyes in all 96 wells simultaneously using a cooled CCD camera. This instrument is equipped with an automated 96-well pipettor, which can be programmed to deliver precise quantities of solutions simultaneously to all 96 culture wells from two separate 96-well source plates.
Cells were plated at 0.3 million cells/mL in 96-well plates (MidSci P9803) and cultured overnight in culture media at 37°C in 5% CO2 and 95% humidity. On the day of assay, growth medium was aspirated and replaced with 100 μl dye-loading medium per well containing 4 μM Fluo-3 AM and 0.04% pluronic acid (Molecular Probes P3000MP) in Locke's buffer (8.6 mM HEPES, 5.6 mM KCl, 154 mM NaCl, 5.6 mM glucose, 1.0 mM MgCl2, 2.3 mM CaCl2, 0.5 mM probenecid; pH 7.4). Cells were incubated for 1 h at 37°C in 5% CO2 and 95% humidity and then washed four times in 180μl fresh Locke's buffer using an automated microplate washer (Bio-Tek Instruments Inc., VT). Baseline fluorescence was recorded for 60 s, prior to a 20 μl addition of various concentrations of Leu8-OT and Pro8-OT. Cells were excited at 488 nm and Ca2+-bound Fluo-3 emission was recorded at 538 nm at 2 s intervals for an additional 200 s.
To assess the role of [Ca2+]i in OT-mediated mobilization of Ca2+, the sarco/endoplasmic reticulum calcium ATPase (SERCA) inhibitor thapsigargin was used to deplete [Ca2+]i stores. Cells were incubated in 100 μl dye-loading medium per well containing 4μM Fluo-3 AM and 0.04% pluronic acid in Locke's buffer (8.6 mM HEPES, 5.6 mM KCl, 154 mM NaCl, 5.6 mM glucose, 1.0 mM MgCl2, 2.3 mM CaCl2, 0.5 mM probenecid; pH 7.4). Cells were incubated at 37°C in a 5% CO2 and 95% humidity for 60 min prior to washing four times in 180 μl Locke’s buffer and 10 μl addition of thapsigargin (1 μM final concentration) and incubated for an additional five minutes. [Ca2+]i mobilization assays were performed as described above.
2.4. Membrane Potential Assay.
To assess changes in membrane potential the FLIPR Membrane Potential Assay (FMP blue; Molecular Probes F1241) was used. Cells were plated at 0.3 million cells/ml in 96-well plates (MidSci P9803) and cultured overnight in culture media at 37°C in 5% CO2 and 95% humidity. Growth medium was removed and replaced with 190 μl per well of FMP Blue in Locke's buffer (8.6 mM HEPES, 5.6 mM KCl, 154 mM NaCl, 5.6 mM glucose, 1.0 mM MgCl2, 2.3 mM CaCl2 pH 7.4). Cells were incubated at 37°C in 5% CO2 and 95% humidity for 45 min. Baseline fluorescence was recorded for 60 s, prior to a 10 μl addition of log concentrations of AVP, Leu8-OT and Pro8-OT. Cells were excited at 530 nm and emission was recorded at 565 nm at 2 s intervals for an additional 200 s.
To ensure the veracity of comparisons of EC50 and maximum response achievable (EMAX) values of AVP and OT variants, all compounds were evaluated in parallel on the same 96-well plate, with the same split of cells and with identical reagent solutions. This experimental design was used for all AVP and OT peptide comparisons throughout this study, using mV1aR, qV1aR, and hV1aR-expressing cells. Inasmuch as all assays were performed in the same CHO cell line, we can exclude differences in cellular context as a source of observed differences in peptide potency or efficacy.
To assess the role of Gi/o in the OT ligand-induced membrane hyperpolarization, cells were incubated overnight with pertussis toxin (PTX) to inactivate Gi/o [37]. Cells were plated at 125,000 cells/mL in 96-well plates. PTX (150 ng/ml) was added 24 hours after plating and incubated for an additional 24 hours. Membrane potential assay was performed as described above.
BAPTA-AM is an calcium chelator [38]. If changes in calcium are responsible for activation of the Ca2+-activated potassium channels, the response should be BAPTA-AM sensitive. Cells were incubated at 37°C in 5% CO2 and 95% humidity for 35 minutes prior to a 10 ml addition of BAPTA-AM. Cells were incubated for an additional 10 minutes after drug addition.
To assess the role of intracellular Ca2+ in AVP and OT ligand-induced changes in membrane potential, thapsigargin was used. Cells were incubated with FMP in Locke’s buffer at 37°C in a 5% CO2 and 95% humidity for 40 min prior to a 10 μl addition of thapsigargin. Cells were incubated for an additional 5 min after drug addition.
To assess potential OT ligand-induced membrane hyperpolarization through Ca2+-activated K+ channels, we tested three inhibitors targeting Ca2+-activated K+ channel subtypes. Gq-mediated activation of protein kinase-C (PKC) causes an increase in cytosolic Ca2+ [28] with attendant activation of Ca2+ activated K+ channels. Ca2+-activated K+ channels are separated into three subtypes of small (SKCa), intermediate (IKCa), and large conductance (BKCa) channels [39]. Apamin is a selective inhibitor of SKCa channels [40, 41]. TRAM-34 is a selective inhibitor of the IK channel, KCa3.1 [42, 43]. Paxilline is a selective inhibitor of the BKCa channel [44]. Cells were incubated with FMP in Locke’s buffer at 37°C in a 5% CO2 and 95% humidity for 35 min prior to a 10 μl addition of apamin, paxilline, TRAM-34, or paxilline + TRAM-34. Cells were incubated for an additional 10 min after drug addition. Membrane potential assays were performed as described above.
SKA-31 is an activator of IKCa channel KCa3.1 [45, 46]. If changes in [Ca2+]i are responsible for the activation of KCa3.1, the response should be SKA-31 sensitive. Cells were incubated at 37°C in a 5% CO2 and 95% humidity for 35 min prior to a 10 μl addition of TRAM-34. Cells were incubated for an additional 10 min after TRAM-34 addition. Membrane potential assays were performed as described above, with the exception of stimulation with SKA-31 rather than OT analogs.
NS-1619 is a BKCa channel activator [47, 48]. If changes in [Ca2+]i are responsible for activation of the BKCa, the response should be NS-1619 sensitive. Cells were incubated at 37°C in a 5% CO2 and 95% humidity for 35 min prior to a 10 μl addition of paxilline. Cells were incubated for an additional 10 min after paxilline addition. Membrane potential assays were performed as described above, with the exception of stimulation with NS-1619 rather than OT analogs.
2.5. Data Analysis.
All concentration-response data were analyzed and graphs generated using GraphPad Prism (San Diego, CA, U.S.A.) software. EC50 and EMAX values for AVP- and OT peptide-stimulated increases in Fluo-3 fluorescence or IC50 and EMAX values decreases in FMP Blue fluorescence were determined by nonlinear least-squares fitting of a logistic equation to the peptide concentration versus fluorescence area under the curve data. The 95% confidence intervals for all EC50/IC50 and EMAX were used to assess differences in potency/efficacy. R2 was used to assess goodness of fit. A one-way ANOVA was performed with Sidek’s multiple comparisons to determine statistical significance and the adjusted p-values reported.
3. RESULTS
3.1. Comparison of marmoset and macaque V1aR amino acid sequence to human V1aR
In contrast to the natural variation in OT analogs in NWM’s, the AVP amino acid sequence is completely conserved in NWMs, OWMs, and humans. However, variation in V1aRs in NWMs is implicated in social monogamy, and may also contribute to other social phenotypes [11, 12, 21]. Marmoset, macaque and human V1aR amino acid sequences were accessed from the National Center for Biotechnology Information (NCBI). Alignment using the NCBI basic local alignment search tool (BLAST) [49] indicates that human and macaque (Macaca mulatta) V1aR are 97% conserved; and human and marmoset (Callithrix jacchus) are 92% conserved. Physiochemical changes to marmoset and macaque V1aR sequences were classified as radical when the amino acid substitution differed by size, polarity and/or charge, and conservative if the substitution did not differ by these properties (Figure 1; Supplementary Table 1). Radical amino acid substitutions were primarily observed in the mV1aR N-terminus and extracellular regions that may play a role in ligand binding. In intracellular regions and the C-terminus radical substitutions may affect G protein coupling and downstream signaling. Radical changes were more numerous for mV1aR than for qV1aR. Amino acids 3, 245, 264, 319 and 369 in mV1aR and qV1aR both differed from hV1aR. In this manuscript, we assess how this natural variation V1aRs impacts AVP and OT-induced receptor activation and cellular signaling.
Figure 1.
Comparison of marmoset and macaque V1aR amino acid sequence to human V1aR. Identification of amino acid substitutions in marmoset (red), macaque (blue), or both (red and blue striped) V1aRs relative to human V1aR. Numbers represent the location of the amino acid substitution. Radical physiochemical substitutions that differ in size, polarity and/or charge are indicated by diamonds and amino acid substitutions that do not differ by these properties are conservative changes indicated by circles (Supplementary Table 1).
3.2. AVP and OT analogs induce Gq-mediated intracellular Ca2+ Mobilization
Gq activates the phospholipase Cβ (PLC) and inositol 3-phosphate (IP3; PLC-IP3) signaling pathway [50] resulting in calcium mobilization. To compare AVP and OT analog activation of V1aR-coupled Gq, we performed functional assays using the Ca2+ indicator dye Fluo3-AM. At the mV1aR and qV1aR, AVP was significantly more potent, with a subpicomolar EC50, whereas Pro8- and Leu8-OT displayed EC50s in the nanomolar range (Table 1; Figure 2A-B; Supplementary Figure 1A-F) There was no significant difference in efficacy between AVP and OT analogs at these receptors (EMAX; Supplementary Figure 1A-F). At the hV1aR, AVP was 60-175X more potent than Pro8- and Leu8-OT and significantly more efficacious (EMAX) (Table 1; Figure 2C, Supplementary Figure 1G-I). AVP, Leu8-OT, and Pro8-OT did not mobilize Ca2i in non-transfected CHO-K1 cells [35](Pierce et al. 2020). These data demonstrate that the dose-dependent response observed in V1aR-expressing lines is attributable to V1aR expression.
Table 1.
Potency of AVP, Leu8-OT and Pro8-OT at inducing calcium mobilization in mV1aR, qV1aR and hV1aR-expressing CHO cells. Sigmoidal curves (Figure 2). Raw data (Supplementary Figure 1).
Ligand | ||||
---|---|---|---|---|
Line | Parameter | AVP | Leu8-OT | Pro8-OT |
mV1aR | EC50 | 0.77 pM | 7.23 nM | 3.39 nM |
95% CI | 0.25 to 2.25 pM | 2.81 to 18.58 nM | 0.81 to 14.18 nM | |
R2 | 0.81 | 0.88 | 0.78 | |
qV1aR | EC50 | 0.34 pM | 54.98 nM | 19.40 nM |
95% CI | 0.11 to 1.02 pM | 25.77 to 117.30 nM | 10.80 to 34.87 nM | |
R2 | 0.82 | 0.91 | 0.95 | |
hV1aR | EC50 | 1.45 nM | 258.4 nM | 92.78 nM |
95% CI | 0.28 to 7.42 nM | 7.81 to 8556.00 nM | 13.56 to 635.00 nM | |
R2 | 0.68 | 0.55 | 0.62 |
Figure 2.
AVP, Leu8-OT and Pro8-OT induced calcium mobilization in mV1aR, qV1aR and hV1aR expressing CHO cells. AVP, Leu8-OT, and Pro8-OT concentration-response relationships (A) in mV1aR cells. AVP, Leu8-OT, and Pro8-OT concentration-response relationships (B) in qV1aR cells. AVP, Leu8-OT, and Pro8-OT concentration-response relationships (C) in hV1aR cells. N=3 experiments (3-4 replicates per dose per experiment).
Inhibition of the sarco/endoplasmic reticulum calcium ATPase (SERCA) pump results in depletion of intracellular Ca2+ stores, because SERCA is responsible for maintaining the Ca2+ gradient between the cytosol and endoplasmic reticulum [51, 52]. To confirm the role of intracellular Ca2+ stores in AVP- and OT-analog mediated Ca2+ influx, cells were pretreated with thapsigargin, a potent SERCA inhibitor. In control assays AVP and OT analogs produced concentration dependent increases in intracellular Ca2+ in all three V1aR cell lines, however pretreatment with thapsigargin blocked AVP and OT-analog induced calcium mobilization Ca2+ (Figure 3; Supplementary Figure 2), and thus demonstrated AVP and OT-mediated increases in cytosolic Ca2+ levels are dependent on intracellular Ca2+ stores.
Figure 3.
Effects of pretreatment with thapsigargin (Tg) on AVP, Leu8-OT and Pro8-OT induced changes in intracellular Ca2+ mobilization in mV1aR-, qV1aR-, and hV1aR-expressing CHO cells. In mV1aR-expressing cells, control and Tg-pretreated concentration response relationships with AVP (A), Leu8-OT (B), and Pro8-OT (C). In qV1aR-expressing cells, control and Tg-pretreated concentration response relationships with AVP (D), Leu8-OT (E), and Pro8-OT (F). In hV1aR-expressing cells, control and Tg-pretreated concentration response relationships with AVP (G), Leu8-OT (H), and Pro8-OT (I). N=3 experiments (five replicates per dose per experiment).
3.3. AVP and OT Analog-Induced Changes in Membrane Potential are Dependent on Gq Mediated Ca2+ Moblization
Functional assays using FMP Blue, a membrane potential-sensitive dye, were used to assess AVP and OT-induced membrane hyperpolarization [53, 54]. In all three V1aR expressing cell lines, AVP and both OT-analogs produced concentration-dependent decreases in fluorescence consistent with a hyperpolarizing response (Table 2; Figure 4; Supplementary Figure 3). In mV1aR and hV1aR expressing cells, AVP-induced membrane hyperpolarization displayed a distinctive U-shaped concentration response curve, which was not observed with qV1aR (Figure 4) or with OT-analogs at mV1aR-, qV1aR- and hV1aR-expressing cells. In mV1aR, qV1aR and hV1aR-expressing cell lines, there was no difference in potency between Leu8- and Pro8-OT-induced hyperpolarizing responses (Table 2; Figure 4; Supplementary Table 3). At the hV1aR, no significant difference in efficacy is observed between the three ligands (Supplementary Figure 3), suggesting the OT analogs function as full agonists in this signaling pathway. The absence of AVP or OT-analog induced hyperpolarizing response in untransfected CHO-K1 cells demonstrated the requirement for V1aR transfection in the observed hyperpolarizing responses [35](Pierce et al. 2020).
Table 2.
Potency of AVP, Leu8-OT and Pro8-OT at inducing membrane hyperpolarization in mV1aR, qV1aR and hV1aR CHO cells. Sigmoidal curves (Figure 4). Raw data (Supplementary Figure 4).
Ligand | ||||
---|---|---|---|---|
Line | Parameter | AVP | Leu8-OT | Pro8-OT |
mV1aR | IC50_1 | 274.17 pM | 61.88 pM | 68.26 pM |
95% CI | 10.01 to 382.20 pM | 14.62 to 318.7 pM | ||
IC50_2 | 2.51 nM | |||
R2 | 0.64 | 0.65 | 0.73 | |
qV1aR | IC50 | 0.56 pM | 125.00 pM | 203.00 pM |
95% CI | 0.17 to 1.86 pM | 50.30 to 310.60 pM | 62.56 to 658.70 pM | |
R2 | 0.62 | 0.84 | 0.78 | |
hV1aR | IC50_1 | 0.70 nM | 22.70 nM | 12.26 nM |
95% CI | 1.98 to 260.70 nM | 1.57 to 95.89 nM | ||
IC50_2 | 1.70 nM | |||
R2 | 0.57 | 0.55 | 0.62 |
Figure 4.
AVP, Leu8-OT and Pro8-OT induced membrane hyperpolarization in mV1aR, qV1aR and hV1aR expressing CHO cells. AVP, Leu8-OT, and Pro8-OT concentration-response relationships in mV1aR cells(A), in qV1aR cells (B), and in hV1aR cells (C). N=3 experiments (3-4 replicates per dose per experiment).
To assess if Gi/o contributes to the AVP and OT-analog induced hyperpolarizing response, cells were pretreated with PTX, which catalyzes the ADP-ribosylation of the Gαi/o subunit, preventing this subunit from interacting with a GPCR [55]. Pretreatment with PTX did not inhibit AVP or OT-analog induced hyperpolarizing responses in any of the V1aR-expressing cell lines (Figure 5; Supplementary Figure 4; Supplementary Table 2), suggesting Gi/o does not contribute to the hyperpolarizing response. As a positive control, we used a human kappa-opioid-receptor expressing CHO cell line, which couples to Gi, and demonstrated that its agonist DynorphinA 1-13-NH2 produced a robust hyperpolarizing response that was abolished in cells pretreated with PTX (data not shown) [35].
Figure 5.
Lack of effect for pretreatment with PTX on AVP, Leu8-OT and Pro8-OT induced changes in membrane hyperpolarization in mV1aR-, qV1aR-, and hV1aR-expressing CHO cells. In mV1aR-expressing cells, control and PTX-pretreated concentration response relationships with AVP (A), Leu8-OT (B), and Pro8-OT (C). In qV1aR-expressing cells, control and PTX-pretreated concentration response relationships with AVP (D), Leu8-OT, (E), and Pro8-OT (F). In hV1aR-expressing cells, control and PTX-pretreated concentration response relationships with AVP (G), Leu8-OT (H), and Pro8-OT (I). N=3 experiments (five replicates per dose per experiment).
To directly assess the role of calcium in AVP and OT-mediated membrane hyperpolarization, cells were pretreated with the calcium chelator BAPTA-AM. In all three V1aR-expressing cell lines, BAPTA-AM exposure blocked AVP and OT-analog induced hyperpolarization. Interestingly, in BAPTA-AM treated V1aR-expressing cells, an AVP and OT-analog induced depolarization was observed (Supplementary Figure 5), indicating a possible dual modulation of ion channels [32]. To confirm the role of intracellular calcium stores in OT-mediated changes in membrane potential, we pretreated cells with the SERCA inhibitor thapsigargin. As expected, pretreatment with thapsigargin eliminated hyperpolarization produced by AVP and OT-analogs in V1aR-expressing cells. Notably, in thapsigargin pretreated V1aR-expressing cells, AVP and OT-analogs again produced a depolarization response (Supplementary Figure 6).
To explore the role of Ca2+-activated K+ channels in AVP and OT-analog induced membrane hyperpolarization, which involves activating the Gq/phosphoinositide-phospholipase C pathway, we used a pharmacological approach with inhibitors that discriminate between subtypes of Ca2+-activated K+ channels; small conductance (SKCa), intermediate conductance (IKCa), and large conductance (BKCa) channels [39]. Cells were pretreated with the SKCa-selective blocker apamin [41] to assess the role of SKCa channels in AVP and OT-analog induced membrane hyperpolarization. In mV1aR and qV1aR-expressing cells, apamin did not substantially affect (≤10%) AVP or OT-analog induced membrane hyperpolarization (Figure 6A-F; Supplementary Table 3; Supplementary Figures 7A-C and 8A-C). In hV1aR-expressing cells, apamin did not markedly affect AVP or Pro8-OT-induced hyperpolarizing response, however, did significantly reduce Leu8-OT-induced membrane hyperpolarization by ~50% (Figure 6G-I, Supplementary Table 3; Supplementary Figure 9A-C). V1aR cells were treated with the IKCa channel blocker TRAM-34, which specifically blocks KCa3.1 [42]. In all three V1aR-expressing cell lines, TRAM-34 produced the most effective inhibition of AVP and OT-analog induced membrane hyperpolarization (Figure 6; Supplementary Table 3; Supplementary Figures 7G-I, 8G-I, 9G-I). To confirm the contribution of KCa3.1 in AVP and OT-analog induced membrane hyperpolarization, we used KCa3.1 activator SKA-31. In all three V1aR-expressing cell lines, TRAM-34 inhibited SKA-31-induced membrane hyperpolarization in a dose-dependent manner (Supplementary Figure 10; Supplementary Table 4), supporting a role for KCa3.1 in the AVP and OT-induced membrane hyperpolarization in V1aR-expressing cells. V1aR-expressing cells were pretreated with the BKCa-blocker paxilline to assess the role of BKCa channels in AVP and OT-induced hyperpolarizing responses. In mV1aR and hV1aR-expressing cells, pretreatment with paxilline had little to no effect on AVP or OT-analog induced hyperpolarizing responses (Figure 6A-C, G-I; Supplementary Table 3; Supplementary Figures 7D-F, 9D-F), whereas in qV1aR-expressing cells, paxilline modestly inhibited AVP and OT-analog induced hyperpolarizing responses by ~15-30% (Figure 8D-F, Supplementary Table 3, Supplementary Figure 6D-F). To confirm the contribution of BKCa channels in AVP and OT-analog induced hyperpolarizing response, we used the BKCa activator NS-1619. In all three V1aR-expressing cell lines, paxilline inhibited NS-1619-induced membrane hyperpolarization in a dose-dependent manner (Supplementary Figure 11; Supplementary Table 4), supporting a role for the regulation of membrane potential by BKCa channels in qV1aR-expressing cells. To determine the combined contribution of BKCa and KCa3.1 channels in AVP and OT-induced changes in membrane potential, cells were pretreated with both paxilline and TRAM-34. In mV1aR and hV1aR expressing cells, the combined pretreatment of paxilline and TRAM-34 robustly inhibited AVP and OT-analog induced hyperpolarizing responses, which was primarily attributable to TRAM-34. However, in qV1aR-expressing cells, the combined pretreatment of paxilline and TRAM-34 demonstrated an additive effect of these inhibitors (Figure 6; Supplementary Table 3; Supplementary Figures 7J-L, 8J-L, 9J-L), suggesting that BKCa and KCa3.1 both contribute to the AVP and OT-analog hyperpolarizing response in qV1aR-expressing cells.
Figure 6.
Effects of pretreatment with Ca2+-activated K+ channel inhibitors on AVP and/or Leu8-OT or Pro8-OT induced changes in membrane potential in mV1aR-, qV1aR-, and hV1aR-expressing CHO cells. Apamin (asparagus), paxilline (light blue), TRAM-34 (maroon), and paxilline + TRAM-34 (lavender) inhibitor fluorescence was normalized to AVP (green; A, D, G), Leu8-OT (blue; B, E, H), or Pro8-OT-induced (red; C, F, I) membrane hyperpolarization in mV1aR- (A-C), qV1aR- (D-F), and hV1aR expressing cells. Area under the curve (negative peaks only) were assessed and a one-way Anova was performed with Sidek’s multiple comparisons to determine statistical significance. N=3 experiments for each inhibitor (10 replicates per dose per experiment). Adjusted P-values (Supplementary Table 2).
4. DISCUSSION
In this study, we compared the pharmacological profiles of AVP, Leu8-OT, and Pro8-OT at the marmoset, macaque and human V1aRs. Previous studies have demonstrated modest differences in primate OT ligand and receptor binding and signaling profiles [35, 36], as well as differences in AVP and OT binding properties and Ca2+ mobilization at these V1aRs [27]. The natural variation in V1aR across NWM, OWM, and humans provides a unique opportunity to assess signaling cascades in relation to genetic variation in receptors. Comparison of mV1aR, qV1aR and hV1aR amino acid sequences demonstrated that variation was most commonly observed in the N-terminus, which may affect ligand binding, and the C-terminus, which has implications in G-protein coupling. By comparing these receptors expressed in a common cell line (CHO cells), we exclude differences in cellular context for variations in cellular signaling. Effects on intracellular signaling cascades can result in changes in cellular function, as well as physiological and behavioral effects at the organismal level [56, 57].
Our results confirm that all three ligands activated Gq signaling in concentration-dependent increases in intracellular Ca2+ through all three V1aRs. Consistent with binding data, AVP was much more potent at mobilizing intracellular Ca2+ than either Leu8-OT and Pro8-OT at all three V1aRs. Notably, in hV1aR expressing cells, AVP was also significantly more efficacious than either OT analog [27], suggesting that receptor variation in hV1aR may contribute to partial agonism by OT ligands. In hV1aR, amino acids at positions 3, 245, 264, 319 and 369 differ from both mV1aR and qV1aR, any or all of which may contribute to this effect. Notably, differences in Leu8-OT and Pro8-OT ligand structure did not show appreciable differences in signaling profiles at each primate V1aR.
In all three V1aR expressing cell lines, AVP, Leu8-OT, and Pro8-OT induced a dose-dependent hyperpolarizing response. AVP was much more potent at inducing a hyperpolarizing response in all three cell lines than either Leu8-OT and Pro8-OT. Although efficacy of both OT analogs trended lower at the human V1aR, these differences did not reach statistical significance as previously observed with the Fluo3-AM calcium mobilization assays. Notably, AVP and OT-analog hyperpolarizing response in qV1aR cells did not return to baseline as quickly as mV1aR and hV1aR. Additionally, AVP-induced membrane hyperpolarization at mV1aR and hV1aR demonstrated a u-shaped dose response curve that wasn’t observed in qV1aR-expressing cells, which may be reflective of enhanced desensitization with higher doses at mV1aR and hV1aR. AVP and OT-analog induced hyperpolarizing responses were insensitive to pretreatment with the Gi/o inhibitor PTX in all three V1aR expressing cell lines, suggesting Gi/o coupling does not contribute to these responses. Pretreatment with BAPTA-AM blocked AVP and OT-analog induced hyperpolarizing responses, demonstrating the effect is dependent on Ca2+ mobilization. Moreover, pretreatment with thapsigargin blocked AVP and OT-analog induced hyperpolarizing responses in all three V1aR expressing cell lines, establishing that the response is dependent on intracellular Ca2+ mobilization.
The Gq/PI-PLC pathway is PTX insensitive and can contribute to hyperpolarizing responses through Ca2+ activated K+ channels, so we assessed this pathway by using blockers specific for SKCa, BKCa(KCa1.1), and IKCa (KCa3.1) channels. Apamin selectively blocks SKCa channels, and pretreatment with this inhibitor resulted in minimal inhibition (≤ 10%) of AVP and OT analogs in all three lines with the exception of Leu8-OT in hV1aR-expressing cells. These data demonstrate that SKCa channels provide minimal contribution to AVP and OT-analog induced membrane hyperpolarization. Paxilline selectively blocks BKCa channels, and pretreatment with this inhibitor demonstrated a modest reduction in q-V1aR-expressing cells with AVP and OT-analog induced membrane hyperpolarization, suggesting that BKCa channels contribute to this response. Paxilline also inhibited the hyperpolarizing response to BKCa channel opener NS-1619, further supporting a role for BKCa channels in the hyperpolarizing response. TRAM-34 selectively blocks the IKCa channel KCa3.1, and pretreatment with TRAM-34 demonstrated a modest inhibition of AVP and OT-analog induced hyperpolarizing responses in qV1aR-expressing cells, whereas it robustly inhibited these responses in mV1aR and hV1aR-expressing cells. TRAM-34 also inhibited the hyperpolarizing response to KCa3.1 channel activator SKA-31, supporting a role for KCa3.1 channel contribution to the hyperpolarizing response in all three V1aR expressing cell lines. In qV1aR-expressing cells pretreatment with paxilline + TRAM-34 demonstrated an additive effect, suggesting that BKCa and KCa3.1 channels contribute to the hyperpolarizing response. Notably, the qV1aR sequence is more similar to the hV1aR than the mV1aR, so a study to assess qV1aR-specific variation at amino acid P17L in the N-terminus or I414L in the C-terminus affected AVP and OT-analog induced signaling responses would be interesting to parse out whether one or both amino acid substitution contributes to these signaling differences.
We recognize the importance of cellular context in GPCR signaling and understand that signaling profiles in the heterologous expression system (CHO cells) that we selected likely differs from those of intact neurons. However, primary neuronal cultures expressing the V1a receptors from the species we are evaluating are not currently available. Established human neuroblastoma and glioma cell lines reportedly express the endogenous OT receptor [34], which would cross react with AVP and OT ligands. Thus, we believe that these initial in vitro experiments to define the signaling profiles of AVP and OT variants are a fundamental first step to understanding ligand-receptor function.
Identification of the effects of natural variation on V1aR-mediated cellular signaling is crucial to translating pharmacological profiles at the cellular level to sociobehavioral processes at the organismal level. Species-specific differences in selectivity and affinity, as well as in vitro and in vivo differences complicate drug development [26]. The findings OT analogs have partial antagonist binding properties and partial agonist signaling properties at the hV1aR [27] has important implications for analyzing behavioral outcomes and for therapeutic development. Characterization of additional AVP and OT agonists may provide insight into functionally selective elements of the V1aR and how that may contribute to species-specific responses in AVP and OT-mediated cellular signaling. Together, these factors likely contribute to a disparate outcomes between animal studies and human clinical trials. Although there have been substantial increases in peptide and non-peptide agonists and antagonists for OT-AVP family as research tools [8, 26, 58-62], therapeutic development has been slow [26].
The OT-AVP neuropeptide family mediates social behavior and physiological processes, with perturbations associated with social behavioral deficits [5]. One major challenge is connecting ligand-receptor activation and cellular signaling to physiological and behavioral changes at the organismal level [57]. The present results show that AVP displays functionally distinct responses from OT analogs at the primate V1aRs examined. Together, these data provide insights into species-specific differences in V1aR sequence, resulting in differences in AVP, Leu8-OT and Pro8-OT pharmacological profiles, which likely contribute to differences in social behavior. Integration these pharmacological profiles, as well as genetic, physiological, and behavioral data is essential for advancing AVP and OT mediated therapeutic development.
Supplementary Material
AKNOWLEDGMENTS
This work was supported by the National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development [Grant R01HD089147]. We thank Dr. Myron Toews and Nancy Schulte for providing the stably transfected mV1aR, qV1aR, and hV1aR CHO cell lines. We thank Dr. Jeff French for providing the AVP and OT peptides. We thank Ms. Bridget Sefranek for her critical reading of this work. We thank Dr. Suneet Mehrotra for his thoughtful discussions.
ABBREVIATIONS
- AVP
arginine vasopressin
- BAPTA-AM
1,2-Bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis (acetoxymethyl ester)
- BSA
bovine serum albumin
- Ca2+
calcium
- CHO
Chinese hamster ovary
- CI
confidence interval
- CNS
central nervous system
- EC50
the half-maximal response
- EMAX
is the maximum response achievable
- FMP
Flipr membrane potential
- GPCR
G protein-coupled receptor
- hV1aR
human vasopressin receptor 1a
- IC50
is the half-inhibitory response
- K+
potassium
- Leu8-OT
consensus mammalian oxytocin sequence
- mV1aR
marmoset vasopressin receptor 1a
- NWM
New World Monkeys
- OT
oxytocin
- OTR
oxytocin receptor
- OWM
Old World Monkeys
- Pro8-OT
oxytocin sequence with proline in 8th position
- PKC
protein kinase C
- PTX
pertussis toxin
- qV1aR
macaque vasopressin receptor 1a
- V1aR
vasopressin receptor 1a
Footnotes
CONFLICT OF INTEREST STATEMENT
The authors declare that there are no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- [1].Huber D, Veinante P, Stoop R, Vasopressin and oxytocin excite distinct neuronal populations in the central amygdala, Science 308(5719) (2005) 245–8. [DOI] [PubMed] [Google Scholar]
- [2].Gainer H, Cell-type specific expression of oxytocin and vasopressin genes: an experimental odyssey, J Neuroendocrinol 24(4) (2012) 528–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Heinrichs M, Domes G, Neuropeptides and social behaviour: effects of oxytocin and vasopressin in humans, Prog Brain Res 170 (2008) 337–50. [DOI] [PubMed] [Google Scholar]
- [4].Heinrichs M, von Dawans B, Domes G, Oxytocin, vasopressin, and human social behavior, Front Neuroendocrinol 30(4) (2009) 548–57. [DOI] [PubMed] [Google Scholar]
- [5].Neumann ID, Landgraf R, Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors, Trends Neurosci 35(11) (2012) 649–59. [DOI] [PubMed] [Google Scholar]
- [6].Stoop R, Neuromodulation by oxytocin and vasopressin, Neuron 76(1) (2012) 142–59. [DOI] [PubMed] [Google Scholar]
- [7].Stoop R, Neuromodulation by oxytocin and vasopressin in the central nervous system as a basis for their rapid behavioral effects, Curr Opin Neurobiol 29 (2014) 187–93. [DOI] [PubMed] [Google Scholar]
- [8].Chini B, Manning M, Agonist selectivity in the oxytocin/vasopressin receptor family: new insights and challenges, Biochem Soc Trans 35(Pt 4) (2007) 737–41. [DOI] [PubMed] [Google Scholar]
- [9].Koshimizu TA, Nakamura K, Egashira N, Hiroyama M, Nonoguchi H, Tanoue A, Vasopressin V1a and V1b receptors: from molecules to physiological systems, Physiol Rev 92(4) (2012) 1813–64. [DOI] [PubMed] [Google Scholar]
- [10].Wallis M, Molecular evolution of the neurohypophysial hormone precursors in mammals: Comparative genomics reveals novel mammalian oxytocin and vasopressin analogues, Gen Comp Endocrinol 179(2) (2012) 313–8. [DOI] [PubMed] [Google Scholar]
- [11].Ren D, Lu G, Moriyama H, Mustoe AC, Harrison EB, French JA, Genetic diversity in oxytocin ligands and receptors in New World monkeys, PLoS One 10(5) (2015) e0125775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].French JA, Taylor JH, Mustoe AC, Cavanaugh J, Neuropeptide diversity and the regulation of social behavior in New World primates, Front Neuroendocrinol 42 (2016) 18–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Lee AG, Cool DR, Grunwald WC Jr., Neal DE, Buckmaster CL, Cheng MY, Hyde SA, Lyons DM, Parker KJ, A novel form of oxytocin in New World monkeys, Biol Lett 7(4) (2011) 584–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Vargas-Pinilla P, Paixao-Cortes VR, Pare P, Tovo-Rodrigues L, Vieira CM, Xavier A, Comas D, Pissinatti A, Sinigaglia M, Rigo MM, Vieira GF, Lucion AB, Salzano FM, Bortolini MC, Evolutionary pattern in the OXT-OXTR system in primates: coevolution and positive selection footprints, Proc Natl Acad Sci U S A 112(1) (2015) 88–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Mustoe A, Taylor JH, French JA, Oxytocin structure and function in New World monkeys: from pharmacology to behavior, Integr Zool 13(6) (2018) 634–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Zingg HH, Laporte SA, The oxytocin receptor, Trends Endocrinol Metab 14(5) (2003) 222–7. [DOI] [PubMed] [Google Scholar]
- [17].Albers HE, Species, sex and individual differences in the vasotocin/vasopressin system: relationship to neurochemical signaling in the social behavior neural network, Front Neuroendocrinol 36 (2015) 49–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Phelps SM, Okhovat M, Berrio A, Individual Differences in Social Behavior and Cortical Vasopressin Receptor: Genetics, Epigenetics, and Evolution, Front Neurosci 11 (2017) 537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Lim MM, Wang Z, Olazabal DE, Ren X, Terwilliger EF, Young LJ, Enhanced partner preference in a promiscuous species by manipulating the expression of a single gene, Nature 429(6993) (2004) 754–7. [DOI] [PubMed] [Google Scholar]
- [20].Fink S, Excoffier L, Heckel G, High variability and non-neutral evolution of the mammalian avpr1a gene, BMC Evol Biol 7 (2007) 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Ren D, Chin KR, French JA, Molecular variation in AVP and AVPR1a in New World monkeys (Primates, Platyrrhini): evolution and implications for social monogamy, PLoS One 9(10) (2014) e111638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Koehbach J, Gruber CW, From ethnopharmacology to drug design, Commun Integr Biol 6(6) (2013) e27583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Jurek B, Neumann ID, The Oxytocin Receptor: From Intracellular Signaling to Behavior, Physiol Rev 98(3) (2018) 1805–1908. [DOI] [PubMed] [Google Scholar]
- [24].Song Z, Albers HE, Cross-talk among oxytocin and arginine-vasopressin receptors: Relevance for basic and clinical studies of the brain and periphery, Front Neuroendocrinol (2017). [DOI] [PMC free article] [PubMed]
- [25].Gimpl G, Fahrenholz F, The oxytocin receptor system: structure, function, and regulation, Physiol Rev 81(2) (2001) 629–83. [DOI] [PubMed] [Google Scholar]
- [26].Manning M, Misicka A, Olma A, Bankowski K, Stoev S, Chini B, Durroux T, Mouillac B, Corbani M, Guillon G, Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics, J Neuroendocrinol 24(4) (2012) 609–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Mustoe A, Schulte NA, Taylor JH, French JA, Toews ML, Leu(8) and Pro(8) oxytocin agonism differs across human, macaque, and marmoset vasopressin 1a receptors, Sci Rep 9(1) (2019) 15480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Ritter SL, Hall RA, Fine-tuning of GPCR activity by receptor-interacting proteins, Nat Rev Mol Cell Biol 10(12) (2009) 819–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Zhao L, Brinton RD, Vasopressin-induced cytoplasmic and nuclear calcium signaling in embryonic cortical astrocytes: dynamics of calcium and calcium-dependent kinase translocation, J Neurosci 23(10) (2003) 4228–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Bone EA, Fretten P, Palmer S, Kirk CJ, Michell RH, Rapid accumulation of inositol phosphates in isolated rat superior cervical sympathetic ganglia exposed to V1-vasopressin and muscarinic cholinergic stimuli, Biochem J 221(3) (1984) 803–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Easaw JC, Jassar BS, Jhamandas JH, Vasopressin receptor subtypes differentially modulate calcium-activated potassium currents in the horizontal limb of the diagonal band of Broca, Neuroscience 81(1) (1997) 57–67. [DOI] [PubMed] [Google Scholar]
- [32].Ogier R, Tribollet E, Suarez P, Raggenbass M, Identified motoneurons involved in sexual and eliminative functions in the rat are powerfully excited by vasopressin and tachykinins, J Neurosci 26(42) (2006) 10717–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Yin K, Baillie GJ, Vetter I, Neuronal cell lines as model dorsal root ganglion neurons: A transcriptomic comparison, Mol Pain 12 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Cassoni P, Sapino A, Stella A, Fortunati N, Bussolati G, Presence and significance of oxytocin receptors in human neuroblastomas and glial tumors, Int J Cancer 77(5) (1998) 695–700. [DOI] [PubMed] [Google Scholar]
- [35].Pierce ML, Mehrotra S, Mustoe AC, French JA, Murray TF, A Comparison of the Ability of Leu(8)- and Pro(8)-Oxytocin to Regulate Intracellular Ca(2+) and Ca(2+)-Activated K(+) Channels at Human and Marmoset Oxytocin Receptors, Mol Pharmacol 95(4) (2019) 376–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Taylor JH, Schulte NA, French JA, Toews ML, Binding Characteristics of Two Oxytocin Variants and Vasopressin at Oxytocin Receptors from Four Primate Species with Different Social Behavior Patterns, J Pharmacol Exp Ther (2018). [DOI] [PMC free article] [PubMed]
- [37].Zhou XB, Lutz S, Steffens F, Korth M, Wieland T, Oxytocin receptors differentially signal via Gq and Gi proteins in pregnant and nonpregnant rat uterine myocytes: implications for myometrial contractility, Mol Endocrinol 21(3) (2007) 740–52. [DOI] [PubMed] [Google Scholar]
- [38].Strayer DS, Hoek JB, Thomas AP, White MK, Cellular activation by Ca2+ release from stores in the endoplasmic reticulum but not by increased free Ca2+ in the cytosol, Biochem J 344 Pt 1 (1999) 39–46. [PMC free article] [PubMed] [Google Scholar]
- [39].Vergara C, Latorre R, Marrion NV, Adelman JP, Calcium-activated potassium channels, Curr Opin Neurobiol 8(3) (1998) 321–9. [DOI] [PubMed] [Google Scholar]
- [40].Blatz AL, Magleby KL, Single apamin-blocked Ca-activated K+ channels of small conductance in cultured rat skeletal muscle, Nature 323(6090) (1986) 718–20. [DOI] [PubMed] [Google Scholar]
- [41].Lamy C, Goodchild SJ, Weatherall KL, Jane DE, Liegeois JF, Seutin V, Marrion NV, Allosteric block of KCa2 channels by apamin, J Biol Chem 285(35) (2010) 27067–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Nguyen HM, Singh V, Pressly B, Jenkins DP, Wulff H, Yarov-Yarovoy V, Structural Insights into the Atomistic Mechanisms of Action of Small Molecule Inhibitors Targeting the KCa3.1 Channel Pore, Mol Pharmacol 91(4) (2017) 392–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Staal RG, Khayrullina T, Zhang H, Davis S, Fallon SM, Cajina M, Nattini ME, Hu A, Zhou H, Poda SB, Zorn S, Chandrasena G, Dale E, Cambpell B, Biilmann Ronn LC, Munro G, Mller T, Inhibition of the potassium channel KCa3.1 by senicapoc reverses tactile allodynia in rats with peripheral nerve injury, Eur J Pharmacol 795 (2017) 1–7. [DOI] [PubMed] [Google Scholar]
- [44].Sanchez M, McManus OB, Paxilline inhibition of the alpha-subunit of the high-conductance calcium-activated potassium channel, Neuropharmacology 35(7) (1996) 963–8. [DOI] [PubMed] [Google Scholar]
- [45].Sankaranarayanan A, Raman G, Busch C, Schultz T, Zimin PI, Hoyer J, Kohler R, Wulff H, Naphtho[1,2-d]thiazol-2-ylamine (SKA-31), a new activator of KCa2 and KCa3.1 potassium channels, potentiates the endothelium-derived hyperpolarizing factor response and lowers blood pressure, Mol Pharmacol 75(2) (2009) 281–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Christophersen P, Wulff H, Pharmacological gating modulation of small- and intermediate-conductance Ca(2+)-activated K(+) channels (KCa2.x and KCa3.1), Channels (Austin) 9(6) (2015) 336–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Lee K, Rowe IC, Ashford ML, NS 1619 activates BKCa channel activity in rat cortical neurones, Eur J Pharmacol 280(2) (1995) 215–9. [DOI] [PubMed] [Google Scholar]
- [48].Edwards G, Niederste-Hollenberg A, Schneider J, Noack T, Weston AH, Ion channel modulation by NS 1619, the putative BKCa channel opener, in vascular smooth muscle, Br J Pharmacol 113(4) (1994) 1538–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Boratyn GM, Schaffer AA, Agarwala R, Altschul SF, Lipman DJ, Madden TL, Domain enhanced lookup time accelerated BLAST, Biol Direct 7 (2012) 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Reversi A, Cassoni P, Chini B, Oxytocin receptor signaling in myoepithelial and cancer cells, J Mammary Gland Biol Neoplasia 10(3) (2005) 221–9. [DOI] [PubMed] [Google Scholar]
- [51].Dravid SM, Murray TF, Spontaneous synchronized calcium oscillations in neocortical neurons in the presence of physiological [Mg(2+)]: involvement of AMPA/kainate and metabotropic glutamate receptors, Brain Res 1006(1) (2004) 8–17. [DOI] [PubMed] [Google Scholar]
- [52].Quynh Doan NT, Christensen SB, Thapsigargin, Origin, Chemistry, Structure-Activity Relationships and Prodrug Development, Curr Pharm Des 21(38) (2015) 5501–17. [DOI] [PubMed] [Google Scholar]
- [53].Baxter DF, Kirk M, Garcia AF, Raimondi A, Holmqvist MH, Flint KK, Bojanic D, Distefano PS, Curtis R, Xie Y, A novel membrane potential-sensitive fluorescent dye improves cell-based assays for ion channels, J Biomol Screen 7(1) (2002) 79–85. [DOI] [PubMed] [Google Scholar]
- [54].Whiteaker KL, Gopalakrishnan SM, Groebe D, Shieh CC, Warrior U, Burns DJ, Coghlan MJ, Scott VE, Gopalakrishnan M, Validation of FLIPR membrane potential dye for high throughput screening of potassium channel modulators, J Biomol Screen 6(5) (2001) 305–12. [DOI] [PubMed] [Google Scholar]
- [55].Murray TF, Siebenaller JF, Differential Susceptibility of Guanine Nucleotide-binding Proteins to Pertussis Toxin-catalyzed ADP-ribosylation in Brain Membranes of Two Congeneric Marine Fishes, Biol Bull 185(3) (1993) 346–354. [DOI] [PubMed] [Google Scholar]
- [56].Luttrell LM, Maudsley S, Bohn LM, Fulfilling the Promise of "Biased" G Protein-Coupled Receptor Agonism, Mol Pharmacol 88(3) (2015) 579–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Luttrell LM, Maudsley S, Gesty-Palmer D, Translating in vitro ligand bias into in vivo efficacy, Cell Signal 41 (2018) 46–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Adachi Y, Sakimura K, Shimizu Y, Nakayama M, Terao Y, Yano T, Asami T, Potent and selective oxytocin receptor agonists without disulfide bridges, Bioorganic & Medicinal Chemistry Letters. [DOI] [PubMed]
- [59].Beard R, Stucki A, Schmitt M, Py G, Grundschober C, Gee AD, Tate EW, Building bridges for highly selective, potent and stable oxytocin and vasopressin analogs, Bioorg Med Chem 26(11) (2018) 3039–3045. [DOI] [PubMed] [Google Scholar]
- [60].Busnelli M, Kleinau G, Muttenthaler M, Stoev S, Manning M, Bibic L, Howell LA, McCormick PJ, Di Lascio S, Braida D, Sala M, Rovati GE, Bellini T, Chini B, Design and Characterization of Superpotent Bivalent Ligands Targeting Oxytocin Receptor Dimers via a Channel-Like Structure, J Med Chem 59(15) (2016) 7152–66. [DOI] [PubMed] [Google Scholar]
- [61].Busnelli M, Sauliere A, Manning M, Bouvier M, Gales C, Chini B, Functional selective oxytocin-derived agonists discriminate between individual G protein family subtypes, J Biol Chem 287(6) (2012) 3617–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Manning M, Stoev S, Chini B, Durroux T, Mouillac B, Guillon G, Peptide and non-peptide agonists and antagonists for the vasopressin and oxytocin V1a, V1b, V2 and OT receptors: research tools and potential therapeutic agents, Prog Brain Res 170 (2008) 473–512. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.