SUMMARY
Canonically, GPCR signaling is transient and confined to the plasma membrane (PM). Deviating from this paradigm, the parathyroid hormone receptor (PTHR1) stimulates sustained Gs signaling at endosomes. In addition to Gs, PTHR1 activates Gq signaling; yet, in contrast to the PTHR1–Gs pathway, the spatiotemporal dynamics of the Gq branch of PTHR1 signaling, and its relationship to Gs signaling, remain largely ill-defined. Recognizing that a downstream consequence of Gq signaling is the activation of phospholipase D (PLD) enzymes, we leverage activity-based, bioorthogonal imaging tools for PLD signaling to visualize and quantify the Gq branch of PTHR1 signaling. We establish that PTHR1–Gq signaling is short-lived, exclusively at the PM, and antagonized by PTHR1 endocytosis. Our data support a model wherein Gq and Gs compete for ligand-bound receptors at the PM and more broadly highlight the utility of bioorthogonal tools for imaging PLDs as probes to visualize GPCR–Gq signaling.
eTOC BLURB
The parathyroid hormone receptor (PTHR1) signals via both Gs and Gq, with the former occurring, non-classically, on endosomes. Here, Liang et al. apply a bioorthogonal method for visualizing phospholipase D signaling to reveal that PTHR1–Gq signaling is transient and localized exclusively at the plasma membrane.
Graphical Abstract

INTRODUCTION
The traditional paradigm for GPCR signaling involves transient, agonist-induced activation of heterotrimeric G proteins and second messenger production at the plasma membrane (PM), which terminates upon receptor internalization (Irannejad and Von Zastrow, 2014; L. Mohan et al., 2013; Pavlos and Friedman, 2017). Classically, different second messengers are produced at the PM depending on the Gα subtype that is activated. For example, Gs activates adenylyl cyclase, producing cyclic AMP (cAMP), leading to protein kinase A signaling, whereas Gq/11 stimulates phospholipase Cβ (PLCβ) enzymes, inducing PI(4,5)P2 hydrolysis to produce diacylglycerol (DAG) and inositol trisphosphate (IP3), leading to protein kinase C (PKC) and Ca2+ signaling. Both Gs and Gq are primarily found in the cytosolic leaflet of the PM, though they can also be found at intracellular locations, along with downstream signaling proteins and second messengers (Marrari et al., 2007; Mohammad Nezhady et al., 2020).
Notably, recent work has identified that certain GPCRs, including opioid receptors (Stoeber et al., 2018), the β2-adrenergic receptor (β2AR) (Irannejad et al., 2013), and the parathyroid hormone receptor 1 (PTHR1) (Cheloha et al., 2015; Ferrandon et al., 2009; Sutkeviciute et al., 2019), also signal, via Gs, from intracellular membranes after receptor-mediated endocytosis. In the case of PTHR1, such signaling occurs on endosomes following β-arrestin-induced, clathrin-mediated endocytosis of the ligand-bound GPCR (Luttrell and Lefkowitz, 2002). This non-classical, endosomal signaling persists until V-ATPase-mediated endosomal acidification causes ligand dissociation, and subsequent receptor binding to the retromer complex enables eventual recycling to the PM (Feinstein et al., 2011; Gidon et al., 2014; Vilardaga et al., 2012; Wehbi et al., 2013). Recent efforts to characterize whether GPCRs remain signaling-competent after internalization have used single-domain antibodies that bind to the receptor in its active conformation as biosensors (Cheloha et al., 2020a). Although this approach reveals the subcellular localizations of GPCRs in their active conformations, it does not elucidate whether these GPCRs maintain the ability to signal via different pathways following internalization.
Sustained endosomal Gs signaling by both β2AR and PTHR1 has proven physiologically significant for regulating distinct outcomes compared to when confined at the PM (Tsvetanova and von Zastrow, 2014; White et al., 2019). However, PTHR1, which has numerous functions in physiology of bone, kidney, and other tissues, has a more complex signaling landscape (Sato et al., 2020; Wein and Kronenberg, 2018). In addition to both PM and endosome-localized Gs–cAMP signaling, PTHR1 also activates the Gq–PLC axis, and such signaling is also physiologically relevant (Abou-Samra et al., 1992; Guo et al., 2002). A longstanding question has been the extent of interplay between the Gq and Gs branches of PTHR1 signaling, which is fundamental to the understanding of diverse physiological functions under the control of PTHR1. Some evidence suggests that regulation of PTHR1–Gq signaling occurs within the kidney (Capuano et al., 2007). A recent study revealed that Gq signaling was required for subsequent prolonged, intracellular Gs-signaling, via a mechanism implicating Gβγ intracellular translocation and activation of PI3Kb (White et al., 2020).
Nevertheless, the spatiotemporal dynamics of PTHR1–Gq signaling and its full relationship to the Gs pathway remain incompletely understood. Here, we establish a chemical toolset for visualizing the Gq branch of GPCR signaling and apply it to elucidate the spatiotemporal dynamics of PTHR1–Gq signaling and clarify its relationship to Gs signaling. Our approach capitalizes upon the recognition that Gq/PLC signaling — via production of DAG and IP3 to activate conventional PKCs — leads to stimulation of phospholipase D (PLD) enzymes (Figure 1A). PLDs in turn produce phosphatidic acid (PA), a lipid second messenger that engages several effectors to influence membrane trafficking, cytoskeletal dynamics, and signaling (Nelson and Frohman, 2015; Selvy et al., 2011).
Figure 1. RT-IMPACT is a tool to quantify phospholipase D (PLD) activity elicited by PTHR1–Gq but not PTHR1–Gs signaling.
(A) Schematic of PTHR1 signaling, illustrating how RT-IMPACT could report on PTHR-dependent Gq signaling. (B–C) Schematic of (B) and chemical structures for (C) RT-IMPACT, a tool to visualize endogenous PLD signaling. (D–E) Flow cytometry of HEK 293 cells indicating the RT-IMPACT labeling depends upon PTHR1–Gq signaling. (D) HEK 293 cells stably expressing PTHR1 (hPTHR1–293) were incubated with oxoTCO and the indicated PTHR1 ligand for 5 min prior to a rinse and addition of Tz-BODIPY for 1 min, rinse, and flow cytometry analysis. Shown: mean BODIPY fluorescence (AU, arbitrary units). Where indicated, cells were treated with the Gq inhibitor YM-254890 (YM) or DMSO vehicle for 15 min prior to and during oxoTCO and PTHR1 activation step. PTH: PTH(1–34); Trp (Gs-selective agonist): PTH-Trp. (E) Gs knockout (KO) cells transfected with hPTHR1-HA were stained with AF647-conjugated α-PTHR1 antibody to identify transfected cells. Cells were labeled via RT-IMPACT as in (D). Flow cytometry was performed, gating on PTHR1-positive or -negative cells (left). Shown: mean BODIPY fluorescence. hPTHR1–293 cells (Gs WT) were analyzed concurrently (right). Asterisks above data denote significance compared to PTH without inhibitors. Asterisks above lines denote significance comparing the indicated groups. Error bars represent standard deviation. One-way ANOVA, Games-Howell post-hoc test: *P < 0.05, **P < 0.01 and ***P < 0.001; ns, not significant; n=3 for (D) and (E). See also Figure S1.
We have developed bioorthogonal chemical tools to visualize and quantify PLD activity with organelle-level resolution (Bumpus et al., 2020; Liang et al., 2019). These approaches harness the ability of PLD enzymes, which normally catalyze phosphatidylcholine hydrolysis to produce PA, to also produce phosphatidyl alcohol lipids via transphosphatidylation with exogenous primary alcohols (Brown et al., 2007). Our tools, termed Imaging Phospholipase D Activity with Clickable Alcohols via Transphosphatidylation (IMPACT), use bioorthogonally labeled alcohols as transphosphatidylation substrates, followed by click chemistry tagging of the resultant lipids in live cells with fluorescent groups (Figure 1B) (Liang et al., 2019).
By using an alcohol and click chemistry partner capable of undergoing a rapid and fluorogenic inverse electron-demand Diels-Alder (IEDDA) cycloaddition, such labeling can be performed in real time (RT-IMPACT) (Bumpus et al., 2020; Liang et al., 2019). In RT-IMPACT, the IEDDA-tagged lipids, prior to their trafficking to other organelle membranes, report on the localizations of endogenous PLD activity (Liang et al., 2019). Because of the specific connection between Gq/PLC signaling and PLD activation, we reasoned that RT-IMPACT would enable us to visualize localizations and quantify the extent of PLD activity downstream of PTHR1–Gq signaling, reporting on the spatiotemporal dynamics of this branch of PTHR1 signaling.
Here, we first establish that RT-IMPACT is a selective reporter for PTHR1–Gq signaling, and we show that PTHR1–Gq signaling specifically activates the PLD1 isoform. We then reveal that PLD1 signaling downstream of PTHR1–Gq is transient and occurs exclusively at the PM. The timescale of this signaling matched that of the PM residence of the ligand-bound GPCR, and inhibition of endocytosis prolonged Gq and suppressed Gs signaling. These data suggest that Gq and Gs compete for PTHR1-mediated signaling upon ligand binding at the PM, hence giving rise to maximal levels of cAMP production after the termination of Gq signaling by receptor endocytosis. This work reveals insights into spatiotemporal control of signaling via different Gα subunits from the same GPCR and highlights how bioorthogonal tools to visualize PLD signaling can act as selective reporters of Gq signaling.
RESULTS AND DISCUSSION
Gq, but not Gs, is required for PLD activation downstream of PTHR1
We began by determining the specificity of RT-IMPACT as a reporter for Gq-signaling downstream of PTHR1. During PTHR1 stimulation, we treated cells with a trans-cyclooctene alcohol (oxoTCO, Figure 1C), in the presence or absence of PTHR1 ligands, to generate oxoTCO-containing lipids as transphosphatidylation products. After a rinse and IEDDA tagging with a fluorogenic tetrazine-BODIPY (Tz-BODIPY, Figure 1C), cellular fluorescence was quantified by flow cytometry. To stimulate PTHR1, we first used PTH(1–34), comprising the N-terminal fragment of the parathyroid hormone (PTH), which strongly activates all downstream PTHR1-coupled Gα pathways. We found substantial PTH(1–34)-dependent RT-IMPACT labeling (Figure 1D).
To determine whether RT-IMPACT reports on Gq versus Gs signaling, we treated HEK 293 cells stably expressing PTHR1 with PTH(1–34)-Trp1, a Gs-biased ligand that induces binding of the receptor exclusively to Gs (Gardella and Vilardaga, 2015), and found that this ligand exhibited only a very minimal activation of PLDs (Figure 1D). Alternatively, we applied PTH(1–34) with YM-254890 (Nishimura et al., 2010; Takasaki et al., 2004), a Gq-selective inhibitor, and found no detectable PLD activity (Figure 1D). These results are consistent with our previous work using these probes to distinguish PTHR1-dependent Gq vs. Gs signaling using fluorescence-based Ca2+ measurements as a readout (Roszko et al., 2017; Sato et al., 2020). To selectively activate Gq, we expressed PTHR1 in Gs knockout cells, and upon PTH stimulation, found a comparable level of PLD activity to that from PTH-stimulated, PTHR1-expressing wild-type cells (Figures 1E and S1). These data indicate that Gq, and not Gs, is responsible for the activation of PLDs downstream of PTH–PTHR1 signaling, likely via PLCβ enzymes, which hydrolyze PI(4,5)P2 to generate DAG and IP3 to activate PKCs, which upregulate PLDs.
PTHR1 predominantly activates the PLD1 isoform at the PM
To characterize the nature of the PTHR1–Gq-induced PLD signaling, we used isoform-selective PLD inhibitors to determine which of the two PLD isoforms that generate PA via PC hydrolysis, PLD1 and PLD2, is responsible for PLD activity downstream of PTHR1. Using RT-IMPACT followed by flow cytometry, we found that a PLD1-selective inhibitor (VU0359595, IC50(PLD1) = 3.7 nM; IC50(PLD2) = 6.4 μM) abrogated nearly all of the PTH-induced PLD activity, equivalent to a pan-PLD inhibitor (Figure 2A). By contrast, a PLD2-selective inhibitor (VU0364739, IC50(PLD1) = 1.5 μM; IC50(PLD2) = 20 nM) had only a modest effect (Figure 2A). Thus, PLD1 is responsible for the bulk of the PLD activity downstream of PTHR1, consistent with the established role of PLD1 as inducible by activated PKCs (Brown et al., 2007; Hu and Exton, 2003).
Figure 2. PTHR–Gq-induced PLD signaling is due predominantly to the PLD1 isoform and is localized at the PM.
(A). Flow cytometry of PLD activity of hPTHR1–293 cells stimulated by PTH in the presence of pan- or isoform-selective PLD inhibitors. Cells were incubated with the indicated inhibitors (PLD1i, PLD1-selective VU0359595; PLD2i, PLD2-selective VU0364739; PLDi, pan-inhibitor FIPI) or DMSO for 30 min. PTH and oxoTCO were added for 5 min before rinsing, Tz-BODIPY addition, and flow cytometry analysis. Asterisks above lines denote significance comparing the indicated groups. Error bars represent standard deviation. One-way ANOVA, Games-Howell post-hoc test: *P < 0.05, **P < 0.01 and ***P < 0.001; ns, not significant; n=3. (B) Confocal microscopy of hPTHR1–293 cells stimulated with PTH and labeled via RT-IMPACT to reveal localization of PTHR1-derived PLD signaling, showing 9 s post-addition of Tz-BODIPY. Scale bar: 20 μm. See also Figure S2.
We next sought to use RT-IMPACT to reveal the subcellular localization of the PTHR1–Gq-induced PLD signaling. Here, we performed a similar RT-IMPACT labeling with oxoTCO during PTH stimulation, rinsing, and then visualization of the fluorogenic IEDDA tagging reaction in real-time by time-lapse confocal microscopy. Because of the rapid kinetics of this IEDDA reaction, we could detect its fluorescent lipid products within seconds of administering Tz-BODIPY to cells, and such images reveal the localization of PLD-generated lipids prior to their subsequent trafficking. Using this technique, we found that PTHR1-induced PLD activity is exclusively at the PM (Figures 2B and S2).
We have previously shown that platelet-derived growth factor receptor signaling induced PLD activity at intracellular membranes using RT-IMPACT (Liang et al., 2019). Therefore, a lack of similar intracellular fluorescence following PTHR1 activation provides strong evidence of a restricted subcellular localization of PLD1 activation in this context. Though PLD1 localizes to many intracellular sites, it can translocate to the PM upon certain stimuli. In this setting, PTHR1–Gq signaling, which activates PLCβ to generate the PKC agonist diacylglycerol, would be expected to stimulate the activity of PLDs in this membrane.
RT-IMPACT reveals that PTHR1–Gq activation of PLDs is transient
In addition to the spatial aspects of PTHR1–Gq signaling, we also sought to use RT-IMPACT to elucidate the temporal dynamics of this pathway. The temporal resolution of RT-IMPACT is roughly 5 min, encompassing transphosphatidylation, rinsing, and IEDDA reaction. To quantify PTHR1–Gq signaling elicited by PTH agonism over time, we varied the time after PTH addition prior to oxoTCO addition by 5-min intervals. Flow cytometry of cells labeled via RT-IMPACT in this manner revealed that PLD activity downstream of PTHR1–Gq signaling was transient, peaking within the first five minutes and rapidly returned to basal levels within 10 min of exposure to PTH (Figures 3A and S3). This finding aligns with previous work showing that PTHR1–Gq signaling induces a transient release of calcium into the cytoplasm that reverts to basal levels within a few minutes (Cheloha et al., 2020b; Sato et al., 2020).
Figure 3. PLD activation downstream of PTHR1–Gq signaling is transient, in contrast to persistent PTHR1–Gs signaling.
(A) Flow cytometry of hPTHR1–293 cells, showing extent of PLD activity following PTH addition. hPTHR1–293 cells were stimulated with PTH for 0–20 min, followed by oxoTCO addition for 5 min, rinsing, Tz-BODIPY addition, and flow cytometry. Gray: background fluorescence in the absence of PTH. (B) Levels of cAMP from hPTHR1–293 cells stably expressing cAMP GloSensor after addition of PTH (solid line; circles) or control (dashed line; triangles). In (A), asterisks above lines denote significance comparing the indicated groups. In (B), asterisks above groups represent significance comparing the +/− PTH conditions at each timepoint. Error bars represent standard deviation. One-way ANOVA, Games-Howell post-hoc test: *P < 0.05, **P < 0.01 and ***P < 0.001; ns, not significant; n=3 for (A) and n=4 for (B). See also Figure S3.
By contrast, PTHR1–Gs signaling was, as expected, delayed and sustained, peaking at 15 min post-PTH application and persisting for at least 60 min, as measured by a cAMP-based biosensor assay (Figure 3B) (Binkowski et al., 2011). This short-lived Gq activation is in line with the classical model of GPCR signaling, wherein ligand-bound receptors are rapidly phosphorylated and internalized via binding to β-arrestins, which terminates most GPCR signaling processes but prolongs Gs signaling in the case of PTHR1.
Despite such rapid deactivation of PTHR1–Gq signaling within 10 min of PTH stimulation, the observed PLD activity at various timepoints (1, 2, and 5 min) post-addition of either PTH or rhodamine-tagged PTH (PTH-TMR), to visualize PTH–PTHR1 complexes, was predominantly at the PM even before complete desensitization (Figure S2), suggesting that Gq activation of PLD was confined to the PM in this context. Importantly, our findings reinforce and are complementary to a recent report that Gq activation is critical for prolonged endosomal Gs signaling through the recruitment of β-arrestins (White et al., 2020).
Receptor endocytosis antagonizes PTHR1–Gq-elicited PLD activity
The above data reveal a stark contrast between the spatiotemporal dynamics of PTHR1-induced Gq and Gs signaling, with the former transient and at the PM and the latter being sustained and largely from endosomes (Feinstein et al., 2011). Typically, ligand-bound GPCRs are internalized via interaction with β-arrestin and subsequent clathrin-mediated endocytosis (Lefkowitz, 1998; Shukla et al., 2011). We hypothesized that receptor-mediated endocytosis might act as a key control point for the switch from Gq to Gs signaling (Feinstein et al., 2011). To test this idea, we perturbed the endocytosis of PTHR1 and investigated effects on the amplitudes and kinetics of Gq and Gs signaling.
First, we tested the effects of blocking endocytosis by treating cells with Dyngo-4a, an inhibitor of dynamin (Harper et al., 2011; McCluskey et al., 2013; Tsvetanova and von Zastrow, 2014), a GTPase required for clathrin-mediated endocytosis. We established conditions under which Dyngo-4a reduced the endocytosis of PTHR1, by visualizing PTH–PTHR1 complexes by confocal microscopy in cells treated with PTH-TMR. At 5–10 min after PTH-TMR addition, we found that Dyngo-4a led to substantially reduced internalization of PTHR1–PTH-TMR complexes (Figure 4A). We then found that Dyngo-4a treatment led to prolonged Gq activation compared to control, as assessed by flow cytometry of RT-IMPACT-labeled cells (Figure 4B). By contrast, under these same conditions, the magnitude of PTHR1–Gs signaling was significantly reduced (Figure 4C). Moreover, the same inhibitory effect of Dyngo-4a on Gs signaling was confirmed in human osteosarcoma cells, which express physiological levels of PTHR1 (Figure S4A), although we were unable to detect Ca2+ responses by fura-2 or PLD activity by IMPACT, following PTH stimulation in these cells, speaking to a possible combination of factors, including a limited sensitivity of the method, low levels of PTHR1 expression, and a more modest stimulation of Gq, compared to Gs, by PTHR1 (Inoue et al., 2019) (Figure S4B–C).
Figure 4. Inhibition of PTHR1 endocytosis prolongs PLD activation through Gq and reduces Gs/cAMP signaling.
(A) Confocal microscopy of hPTHR1–293 cells treated with rhodamine-PTH (PTH-TMR) for 5–10 min. Cells were treated with Dyngo-4a or DMSO vehicle 15 min prior to and during ligand addition. Scale bar: 30 μm. (B) Flow cytometry of hPTHR1–293 cells, showing extent of PLD activity over 20 min after the PTH addition +/− Dyngo addition. hPTHR1–293 cells were incubated with Dyngo or vehicle for 15 min, stimulated with PTH for 0–20 min in the presence of Dyngo or vehicle, followed by addition of oxoTCO (5 min) in the presence of PTH and Dyngo or vehicle, rinsing, addition of Tz-BODIPY, and flow cytometry. Shown: mean BODIPY fluorescence. Gray: background fluorescence in the absence of PTH. (C) Timecourse of cAMP levels in hPTHR1–293 cells stably expressing cAMP GloSensor. Dyngo or vehicle was added 15 min prior to and during the PTH labeling step. (D) Model indicating competition between Gq and Gs signaling downstream of PTHR1. In (B–C), asterisks above data denote significance comparing blue and red data points (+ vs. − Dyngo in the presence of PTH). Error bars represent standard deviation. One-way ANOVA, Games-Howell post-hoc test: *P < 0.05; **P < 0.01; ***P < 0.001; ns, not significant; n=3 for (B) and n=4 for (C). See also Figure S4.
To account for any potential off-target effects of Dyngo-4a (Park et al., 2013; Preta et al., 2015), we corroborated these findings by perturbing endocytosis using two independent methods. First, we treated cells with Bafilomycin A1 (BafA1), an inhibitor of the V-ATPase responsible for endosomal acidification. Prolonged BafA1 treatment, by preventing endosomal maturation to lysosomes, has the ultimate effect of diminishing endocytosis (Gidon et al., 2014; Kozik et al., 2013). Similar to Dyngo-4a, BafA1 potentiated Gq-induced PLD signaling (Figure S4D–E). Alternatively, we perturbed clathrin-mediated endocytosis through acute inhibition of β-arrestins by the pharmacological inhibitor Barbadin, which blocks interactions between β-arrestin and the clathrin adaptor protein AP2 to prevent endocytosis of β2AR (Beautrait et al., 2017). Consistent with the Dyngo-4a and BafA1 experiments, Barbadin treatment prolonged PTHR1-depedent PLD activation (Figure S4F).
Collectively, these results support a model wherein the PTHR1–Gq pathway is restricted to the PM. Under physiological conditions, this branch of PTHR1 signaling is short-lived and is terminated upon endocytosis of PTHR1, within 5–10 min of ligand binding. By contrast, the Gs-cAMP pathway occurs on both membranes. At the PM, a brief burst of Gs signaling, immediately following PTHR1 activation, has been reported, but the bulk of Gs signaling is now appreciated to occur at early endosomes, peaking at ~15 min and lasting for more than 1 hour.
These observations can be explained by a model wherein Gq and Gs compete for binding to PTH-bound PTHR1 receptors at the PM, where both pathways are active upon initial ligand binding (Figure 4D). Phosphorylation and subsequent β-arrestin-mediated endocytosis of PTHR1 terminates PTHR1–Gq signaling, leading ultimately to PTHR1 signaling at endosomes via the Gs-cAMP pathway, such that maximal cAMP levels correlate with the termination of Gq-induced PLD signaling. The pharmacological perturbations of receptor endocytosis, which enhance Gq signaling and attenuate Gs signaling, further supports this model of a competitive relationship between Gq and Gs signaling in this system.
CONCLUSION
We have established that RT-IMPACT, an activity-based, bioorthogonal method for visualizing PLD signaling, can be used to selectively image and quantify GPCR–Gq signaling. We applied RT-IMPACT to elucidate the spatiotemporal dynamics of Gq signaling downstream of PTHR1, an important GPCR in physiology and disease that signals through multiple Gα proteins at diverse subcellular locations. Following PTH binding, PLD activity downstream of PTHR1–Gq signaling occurs transiently and exclusively at the PM. Perturbations of receptor endocytosis prolonged such Gq signaling and reduced Gs signaling, which predominantly occurs on endosomes, supporting a model for competition between Gq and Gs for binding to the activated receptor. These data reinforce that Gq and Gs signaling downstream of PTHR1 activation exhibit vastly different spatiotemporal behavior, and such dynamics may be critical for the distinct physiological outcomes of these signaling pathways. More generally, our study underscores the value of applying bioorthogonal imaging methods for tracking PLD activity to understand the regulation of diverse signaling pathways that intersect with PLD signaling.
SIGNIFICANCE
GPCRs can signal via multiple Gα subtypes, and certain GPCRs, including the parathyroid hormone receptor PTHR1, engage in non-classical Gs signaling from endosomes, following their endocytosis. The localization and duration of signaling via other Gα subtypes such as Gq/11 has remained challenging to determine with traditional approaches. Here, we use bioorthogonal, activity-based tools for visualizing phospholipase D (PLD) signaling as a means to selectively visualize and quantify Gq signaling by PTHR1 and determine that such signaling occurs transiently and exclusively at the plasma membrane, upstream of sustained endosomal Gs signaling. Direct imaging of PLD activation represents a means to reveal the spatiotemporal dynamics of GPCR–Gq signaling.
STAR METHODS
RESOURCE AVAILABILITY
Lead contact.
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Jeremy Baskin (jeremy.baskin@cornell.edu).
Materials availability.
Materials generated in this study are available from the lead contact with a completed Materials Transfer Agreement.
Data and code availability.
The published article includes all datasets generated or analyzed during this study.
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Cell lines
HEK 293 cell lines (H. sapiens, embryonic kidney epithelial, female) that stably express human PTHR1 and cAMP GloSensor (293-PTHR1 cells) and HEK 293 Gs knockout cells that stably express GloSensor (i.e. Gs KO cells), osteosarcoma cell lines UMR-106 (R. norvegicus, adult, bone epithelial osteosarcoma, sex not available) and SaOS-2 (H. sapiens, 11 year old, bone epithelial osteosarcoma, female), and cell lines derived from these that stably express cAMP GloSensor UGS-56 (from UMR-106) and SGS-72 (from SaOS-2) were grown in DMEM (Dulbecco’s modified Eagle medium) supplemented with 10% FBS and 1% penicillin/streptomycin and were maintained in a 5% CO2, moisture-saturated atmosphere at 37 °C. Cell densities were maintained between 105 and 1.6 × 106 cells/mL, and all cells were used between passages 20–40. For cell labeling experiments, all buffers or media were warmed to 37 °C or room temperature prior to addition to cells unless otherwise noted, and incubations were done at 37 °C unless otherwise specified.
METHOD DETAILS
General materials and methods
See Key Resources Table for sources for reagents, cell lines, etc. All imaging experiments were performed on a Zeiss LSM 800 confocal laser scanning microscope equipped with 40X 1.4 NA Plan Apochromat objectives, 405, 488, 561, and 640 nm solid-state lasers, and two GaAsP PMT detectors, using the Zeiss Zen Blue 2.3 software. All image analysis was performed using FIJI/ImageJ. Flow cytometry experiments, except for Figs. 1E and S1, was performed on a BD Accuri C6 flow cytometer, and analysis was performed using the BD Accuri C6 analysis software. Flow cytometry experiment for Figs. 1E and S1 was performed on a Thermo Fisher Attune NxT Flow Cytometer equipped with 405, 488, 561, 637 nm lasers using 480 and 647 dual lasers. Luminescence measurements were performed using a Tecan Infinite M1000 Microplate Reader (cat # 30034301).
KEY RESOURCES TABLE.
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| Anti-PTHR1-Alexa Fluor 647 nanobody | Laboratory of Ross Cheloha, NIH | VHHPTHR, described in (Cheloha et al., 2020) |
| Chemicals, Peptides, and Recombinant Proteins | ||
| Bafilomycin A1 | Cayman Chemical | Cat# 11038 |
| Barbadin | MedChem Express | Cat# HY-119706 |
| D-luciferin | Gold Bio | Cat# L-123-250 |
| Dyngo-4a | Selleck Chem | Cat# S7163 |
| FIPI | Cayman Chemical | Cat# 13563 |
| PLD1i | Laboratory of Alex Brown, Vanderbilt University | VU0359595 |
| PLD2i | Laboratory of Alex Brown, Vanderbilt University | VU0364739 |
| Phorbol 12-myristate 13-acetate (PMA) | Santa Cruz Biotechnology | Cat# sc-3576 |
| PTH(1-34) (PTH) | GenScript | Cat# RP01001 |
| PTH(1-34) Trp1 (PTH-Trp) | Massachusetts General Hospital Peptide/Protein Core Facility | Sequence: WVSEIQLMHNLGKHLNSMERVEWLRKKLQDVHNF-CO2H |
| PTH-K13-TMR (PTH-TMR) | Massachusetts General Hospital Peptide/Protein Core Facility | Sequence: SVSEIQLMHNLGK(5-carboxytetramethylrhodamine) HLNSMERVEWLRKKLQDVHNF-CO2H |
| YM-254890 | Focus Biomolecules | Cat# 10-1590-0100 |
| Critical Commercial Assays | ||
| GloSensor cAMP responsive luciferase stably expressed in HEK 293 | Promega | pGloSensor™-22F, # E2301, described in (Binkowski et al., 2011) |
| Experimental Models: Cell Lines | ||
| hPTHR1-HEK 293 containing GloSensor (293-PTHR1) | ATCC | CRL-11268 base cell line (HEK 293). Stable hPTHR1-HEK 293 expressing GloSensor plasmid originally described in (Maeda et al., 2013) |
| Gs KO-HEK 293 (Gs knockout achieved using CRISPR-Cas9) | Laboratory of Thomas Gardella, Harvard Medical School | Gs KO-HEK 293 cells, described in (Stallaert et al., 2017) |
| SaOS-2 | ATCC | Cat#HTB-85, human osteosarcoma |
| SGS-72 | ATCC | SaOS-2 cells stably expressing GloSensor (Cheloha et al., 2016) |
| UMR-106 | ATCC | Cat#CRL-1661, Rat osteosarcoma |
| UGS-56 | ATCC | UMR-106 cells stably expressing GloSensor (Daley et al., 2021) |
| Recombinant DNA | ||
| hPTHR1-HA plasmid | Laboratory of Thomas Gardella, Harvard Medical School, custom plasmid (VectorBuilder). HA inserted into exon 2 | Gardella lab plasmid #906 |
| Software and Algorithms | ||
| ImageJ | NIH | https://imagej.nih.gov/ij/ |
RT-IMPACT labeling with oxoTCO for live-cell imaging and flow cytometry analysis
HEK 293-PTHR1 cells or Gs KO cells (150,000 cells) were seeded on 35-mm glass-bottom imaging dishes (MatTek) for 24 h prior to experiments. Rinses after oxoTCO incubation were performed in DMEM supplemented with FBS and P/S. Where indicated, the cells were then transfected with the indicated plasmid using Lipofectamine 2000 as per the manufacturer’s instructions. One day after seeding (or transfection, if any), cells were first treated with the indicated PLD inhibitor (PLDi [FIPI], 750 nM; PLD1i [VU0359595], 250 nM; PLD2i [VU0364739], 350 nM; 30 min), YM-254890 (1 μM, 15 min), Dyngo-4a (30 μM, 15 min, serum-free), or corresponding DMSO vehicle in media at 37 °C. The working concentrations of PTH(1–34) (PTH), PTH-Trp (Trp) and PTH-TMR were 50 nM and that of PMA was 100 nM. The appropriate stimulus was then added to the media for the indicated periods of time (0–20 min). Subsequently, freshly prepared oxoTCO (3 mM) together with the respective stimulus and PLD inhibitor/DMSO in media (100 μL) were carefully added to cover the central glass well.
Cautionary note: oxoTCOs are reported to have limited water stability (Lambert et al., 2017). Therefore, all aqueous oxoTCO solutions (e.g., those in DMEM-containing media) were used within 20 min of their generation. For example, we dissolved oxoTCO in 200 μL of DMEM with PLD inhibitor/DMSO and respective stimulus and used it only for two consecutive replicates rather than make a single stock solution for an entire day of experiments at the beginning of the day.
The dish was incubated for 5 min, the treatment media was aspirated, the cells were rinsed with PBS (1 mL) briefly, and the cells were then rinsed in DMEM (500 μL) for 1 min at 37 °C. The media was aspirated and replaced with Tz–BODIPY (0.33 μM) in PBS (100 μL) for 1 min, which was further aspirated and replaced with 100 μL Tyrode’s-HEPES buffer. Cells were imaged immediately afterwards. Multicolor images were obtained in two-channel, line-switching mode. Z stacks were taken with 0.45 μm sectioning. For flow cytometry analysis, cells were instead seeded in 24 well-plates (125,000 cells/well) and labeled as described above. Following the final aspiration of Tz–BODIPY, cells were lifted with trypsin, transferred to 96-well plates, rinse twice with cold PBS + 0.5% FBS by centrifugation at 500 × g, and analyzed by flow cytometry. For the experiment shown in Fig. 1E, an extra step of incubation of anti-PTHR1-Alexa Fluor 647 nanobodies (100 nM) at 4 °C in the dark for 30 min before oxoTCO incubation steps. At least 10,000 live cells were analyzed for each well in all flow cytometry experiments, as determined by forward/side scatter analysis for all experiments and additional gating for the experiment shown in Fig. 1E (see Fig. S1).
For Figure S4B–D, IMPACT labeling for flow cytometry analysis (Bumpus et al., 2020) was performed as described above but substituting azidopropanol (10 mM) and BCN-BODIPY (1 mM) in place of oxoTCO in place of Tz-BODIPY, respectively, and with the BCN-BODIPY incubation being 10 min followed by 10 min rinse-out in full media at 37 °C. Phorbol 12-myristate 13-acetate (PMA, 100 nM), where indicated, was added to the cells together with azidopropanol with no prior incubation as a positive control for PLD activation. Barbadin (100 μM, serum-free), where indicated, was present 30 min before experiment. The incubation of BCN-BODIPY was performed at 37 °C for 10 min followed by rinsing in Tyrode’s-HEPES buffer at 37 °C for 10 min and triple rinses in PBS prior to trypsinization.
Luciferase assays of cAMP production
Procedures were adopted from previously published protocols (Cheloha et al., 2014). Monolayers of HEK 293 cells stably expressing PTHR1 were seeded on a 96-well plate 24 h before experiments. Dyngo-4a (30 μM) or corresponding DMSO vehicle, where indicated, was incubated with the cells in serum-free media for 15 min at 37 °C prior to the experiment. The cells were incubated with D-luciferin (0.5 mM) for 30 min at room temperature until the luminescence achieved a steady baseline. PTH ligands (50 nM) were then added to the wells and luminescence recorded by a Tecan Infinite M1000 Microplate Reader at the indicated time points.
Imaging of PTH-TMR to visualize PTHR1 endocytosis
293-PTHR1 cells (150,000 cells) were seeded on 35-mm glass-bottom imaging dishes (MatTek) for 24 h prior to experiments. Dyngo-4a (30 μM), where indicated, was incubated with the cells in serum-free media for 15 min at 37 °C prior to the experiment. PTH-TMR (50 nM) was added to cells for 20 s, followed by three PBS rinses and imaging, with excitation by the 561 nm laser.
RT-IMPACT imaging of the localization of PLD activity
Real-time IMPACT using oxoTCO ((S)-oxoTCO–C1) and Tz-BODIPY was performed as described previously (Bumpus et al., 2020; Liang et al., 2019). Briefly, freshly prepared oxoTCO (3 mM) together with the PTH(1–34) or PTH-TMR (50 nM) in media (100 μL) were carefully added to cover the central glass well. The dish was incubated for 5 min, the treatment media was aspirated, the cells were rinsed with PBS (1 mL) briefly, and the cells were then rinsed in DMEM (500 μL) for 1 min at 37 °C. The media was replaced with 100 μL of Tyrode’s-HEPES buffer to cover the center of the glass bottom and the dish was mounted on the microscope. The cells to be imaged were quickly located and time-lapse imaging with an interval of 3 s (488 nm, for PTH(1–34) or 6 s (488 nm and 561 nm, for PTH-TMR) and duration of 3 min begun. Tz-BODIPY (100 μL, 1 μM in PBS) was added dropwise but quickly to the center of the dish during acquisition. The image shown in Fig. 2B represents the 9-s timepoint post-addition of Tz-BODIPY. The RT-IMPACT experiment was repeated four times, yielding similar localizations.
QUANTIFICATION AND STATISTICAL ANALYSIS
All imaging experiments show representative images from experiments performed in at least three biological replicates on different days, where each replicate refers to a single dish of cells with approximately 15 cells in the field of view, whose parameters were all determined and averaged for the data point for that dish. Exact numbers of replicate experiments, sample sizes, and p values are provided in each figure legend. For experiments involving quantification of comparisons between more than two independent groups, significance was calculated using one-way ANOVA, followed by Games-Howell post-hoc test (for samples of unequal variance).
Supplementary Material
HIGHLIGHTS.
RT-IMPACT is an activity-based method to image phospholipase D signaling
RT-IMPACT specifically visualizes Gq signaling downstream of PTHR1 activation
PTHR1 signals via Gq transiently and at the plasma membrane, in contrast to Gs
Bioorthogonal tools for imaging PLDs are useful probes to image GPCR–Gq signaling
ACKNOWLEDGMENTS
J.M.B. acknowledges support from NSF CAREER (CHE-1749919), Beckman Young Investigator, and Sloan Research Fellowship awards. T.J.G. acknowledges support from the NIH (P01DK011794). D.L. was supported by a Cornell Fellowship. R.W.C was supported by a CRI Irvington Postdoctoral Fellowship. Schematics were created with BioRender.com. We thank the Baskin lab for helpful discussions, Kane Wu for technical assistance, and the Emr, Fromme, and Yu labs for use of equipment.
Footnotes
DECLARATION OF INTERESTS
The authors declare no competing interests.
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
- Abou-Samra AB, Juppner H, Force T, Freeman MW, Kong XF, Schipani E, Urena P, Richards J, Bonventre JV, Potts JT, et al. (1992). Expression cloning of a common receptor for parathyroid hormone and parathyroid hormone-related peptide from rat osteoblast-like cells: A single receptor stimulates intracellular accumulation of both cAMP and inositol trisphosphates and increases intracel. Proc. Natl. Acad. Sci. U. S. A 89, 2732–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beautrait A, Paradis JS, Zimmerman B, Giubilaro J, Nikolajev L, Armando S, Kobayashi H, Yamani L, Namkung Y, Heydenreich FM, et al. (2017). A new inhibitor of the β-arrestin/AP2 endocytic complex reveals interplay between GPCR internalization and signalling. Nat. Commun 8, 15054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binkowski BF, Butler BL, Stecha PF, Eggers CT, Otto P, Zimmerman K, Vidugiris G, Wood MG, Encell LP, Fan F, et al. (2011). A luminescent biosensor with increased dynamic range for intracellular cAMP. ACS Chem. Biol 6, 1193–1197. [DOI] [PubMed] [Google Scholar]
- Brown HA, Henage LG, Preininger AM, Xiang Y, and Exton JH (2007). Biochemical Analysis of Phospholipase D. Methods Enzymol. 434, 49–87. [DOI] [PubMed] [Google Scholar]
- Bumpus TW, Liang D, and Baskin JM (2020). IMPACT: Imaging phospholipase D activity with clickable alcohols via transphosphatidylation. Methods Enzymol. 641, 75–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capuano P, Bacic D, Roos M, Gisler SM, Stange G, Biber J, Kaissling B, Weinman EJ, Shenolikar S, Wagner CA, et al. (2007). Defective coupling of apical PTH receptors to phospholipase C prevents internalization of the Na+-phosphate cotransporter NaP i-IIa in Nherf1-deficient mice. Am. J. Physiol. - Cell Physiol 292. [DOI] [PubMed] [Google Scholar]
- Cheloha RW, Maeda A, Dean T, Gardella TJ, and Gellman SH (2014). Backbone modification of a polypeptide drug alters duration of action in vivo. Nat. Biotechnol 32, 653–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheloha RW, Gellman SH, Vilardaga JP, and Gardella TJ (2015). PTH receptor-1 signalling - Mechanistic insights and therapeutic prospects. Nat. Rev. Endocrinol 11, 712–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheloha RW, Watanabe T, Dean T, Gellman SH, and Gardella TJ (2016). Backbone Modification of a Parathyroid Hormone Receptor-1 Antagonist/Inverse Agonist. ACS Chem. Biol 11, 2752–2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheloha RW, Harmand TJ, Wijne C, Schwartz TU, and Ploegh HL (2020a). Exploring cellular biochemistry with nanobodies. J. Biol. Chem 295, 15307–15327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheloha RW, Fischer FA, Woodham AW, Daley E, Suminski N, Gardella TJ, and Ploegh HL (2020b). Improved GPCR ligands from nanobody tethering. Nat. Commun 11, 2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daley EJ, Khatri A, Dean T, Vilardaga JP, Zaidi SA, Katritch V, and Gardella TJ (2021). Ligand-dependent effects of methionine-8 oxidation in parathyroid hormone peptide analogues. Endocrinol. (United States) 162, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feinstein TN, Wehbi VL, Ardura JA, Wheeler DS, Ferrandon S, Gardella TJ, and Vilardaga JP (2011). Retromer terminates the generation of cAMP by internalized PTH receptors. Nat. Chem. Biol 7, 278–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrandon S, Feinstein TN, Castro M, Wang B, Bouley R, Potts JT, Gardella TJ, and Vilardaga JP (2009). Sustained cyclic AMP production by parathyroid hormone receptor endocytosis. Nat. Chem. Biol 5, 734–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardella TJ, and Vilardaga JP (2015). International union of basic and clinical pharmacology. XCIII. The parathyroid hormone receptors—family B G protein–coupled receptors. Pharmacol. Rev 67, 310–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gidon A, Al-Bataineh MM, Jean-Alphonse FG, Stevenson HP, Watanabe T, Louet C, Khatri A, Calero G, Pastor-Soler NM, Gardella TJ, et al. (2014). Endosomal GPCR signaling turned off by negative feedback actions of PKA and v-ATPase. Nat. Chem. Biol 10, 707–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo J, Chung U Il, Kondo H, Bringhurst FR, and Kronenberg HM (2002). The PTH/PTHrP receptor can delay chondrocyte hypertrophy in vivo without activating phospholipase C. Dev. Cell 3, 183–194. [DOI] [PubMed] [Google Scholar]
- Harper CB, Martin S, Nguyen TH, Daniels SJ, Lavidis NA, Popoff MR, Hadzic G, Mariana A, Chau N, McCluskey A, et al. (2011). Dynamin inhibition blocks botulinum neurotoxin type A endocytosis in neurons and delays botulism. J. Biol. Chem 286, 35966–35976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu T, and Exton JH (2003). Mechanisms of regulation of phospholipase D1 by protein kinase Ca. J. Biol. Chem 278, 2348–2355. [DOI] [PubMed] [Google Scholar]
- Inoue A, Raimondi F, Kadji FMN, Singh G, Kishi T, Uwamizu A, Ono Y, Shinjo Y, Ishida S, Arang N, et al. (2019). Illuminating G-Protein-Coupling Selectivity of GPCRs. Cell 177, 1933–1947.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irannejad R, and Von Zastrow M (2014). GPCR signaling along the endocytic pathway. Curr. Opin. Cell Biol 27, 109–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irannejad R, Tomshine JC, Tomshine JR, Chevalier M, Mahoney JP, Steyaert J, Rasmussen SGF, Sunahara RK, El-Samad H, Huang B, et al. (2013). Conformational biosensors reveal GPCR signalling from endosomes. Nature 495, 534–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kozik P, Hodson NA, Sahlender DA, Simecek N, Soromani C, Wu J, Collinson LM, and Robinson MS (2013). A human genome-wide screen for regulators of clathrin-coated vesicle formation reveals an unexpected role for the V-ATPase. Nat. Cell Biol 15, 50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohan L, M., Vasudevan T, N., Gupta K, M., Martelli E, E., and Naga Prasad V, S. (2013). G-Protein Coupled Receptor Resensitization - Appreciating the Balancing Act of Receptor Function. Curr. Mol. Pharmacol 5, 350–361. [PMC free article] [PubMed] [Google Scholar]
- Lambert WD, Scinto SL, Dmitrenko O, Boyd SJ, Magboo R, Mehl RA, Chin JW, Fox JM, and Wallace S (2017). Computationally guided discovery of a reactive, hydrophilic: Trans −5-oxocene dienophile for bioorthogonal labeling. Org. Biomol. Chem 15, 6640–6644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lefkowitz RJ (1998). G protein-coupled receptors: III. New roles for receptor kinases and β- arrestins in receptor signaling and desensitization. J. Biol. Chem 273, 18677–18680. [DOI] [PubMed] [Google Scholar]
- Liang D, Wu K, Tei R, Bumpus TW, Ye J, and Baskin JM (2019). A real-time click chemistry imaging approach reveals stimulus-specific subcellular locations of phospholipase D activity. Proc. Natl. Acad. Sci. U. S. A 116, 15453–15462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luttrell LM, and Lefkowitz RJ (2002). The role of β-arrestins in the termination and transduction of G-protein-coupled receptor signals. J. Cell Sci 115, 455–465. [DOI] [PubMed] [Google Scholar]
- Maeda A, Okazaki M, Baron DM, Dean T, Khatri A, Mahon M, Segawa H, Abou-Samra AB, Jüppner H, Bloch KD, et al. (2013). Critical role of parathyroid hormone (PTH) receptor-1 phosphorylation in regulating acute responses to PTH. Proc. Natl. Acad. Sci. U. S. A 110, 5864–5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marrari Y, Crouthamel M, Irannejad R, and Wedegaertner PB (2007). Assembly and trafficking of heterotrimeric G proteins. Biochemistry 46, 7665–7677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCluskey A, Daniel JA, Hadzic G, Chau N, Clayton EL, Mariana A, Whiting A, Gorgani NN, Lloyd J, Quan A, et al. (2013). Building a better dynasore: The dyngo compounds potently inhibit dynamin and endocytosis. Traffic 14, 1272–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohammad Nezhady MA, Rivera JC, and Chemtob S (2020). Location Bias as Emerging Paradigm in GPCR Biology and Drug Discovery. IScience 23, 101643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson RK, and Frohman MA (2015). Physiological and pathophysiological roles for phospholipase D. J. Lipid Res 56, 2229–2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nishimura A, Kitano K, Takasaki J, Taniguchi M, Mizuno N, Tago K, Hakoshima T, and Itoh H (2010). Structural basis for the specific inhibition of heterotrimeric Gq protein by a small molecule. Proc. Natl. Acad. Sci. U. S. A 107, 13666–13671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park RJ, Shen H, Liu L, Liu X, Ferguson SM, and De Camilli P (2013). Dynamin triple knockout cells reveal off target effects of commonly used dynamin inhibitors. J. Cell Sci 126, 5305–5312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavlos NJ, and Friedman PA (2017). GPCR Signaling and Trafficking: The Long and Short of It. Trends Endocrinol. Metab 28, 213–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preta G, Cronin JG, and Sheldon IM (2015). Dynasore - Not just a dynamin inhibitor. Cell Commun. Signal 13, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roszko KL, Bi R, Gorvin CM, Bräuner-Osborne H, Xiong XF, Inoue A, Thakker RV, Strømgaard K, Gardella T, and Mannstadt M (2017). Knockin mouse with mutant Gα11 mimics human inherited hypocalcemia and is rescued by pharmacologic inhibitors. JCI Insight 2, 3, e91079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato T, Verma S, Khatri A, Dean T, Goransson O, Gardella TJ, and Wein MN (2020). Comparable initial engagement of intracellular signaling pathways by parathyroid hormone receptor ligands teriparatide, abaloparatide, and long- acting PTH. JBMR Plus, 5, e10441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selvy PE, Lavieri RR, Lindsley CW, and Brown HA (2011). Phospholipase D: enzymology, functionality, and chemical modulation. Chem. Rev 111, 6064–6119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shukla AK, Xiao K, and Lefkowitz RJ (2011). Emerging paradigms of β-arrestin-dependent seven transmembrane receptor signaling. Trends Biochem. Sci 36, 457–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stallaert W, Van Der Westhuizen ET, Schönegge AM, Plouffe B, Hogue M, Lukashova V, Inoue A, Ishida S, Aoki J, Le Gouill C, et al. (2017). Purinergic receptor transactivation by the β2-adrenergic receptor increases intracellular Ca2+ in nonexcitable cells. Mol. Pharmacol 91, 533–544. [DOI] [PubMed] [Google Scholar]
- Stoeber M, Jullié D, Lobingier BT, Laeremans T, Steyaert J, Schiller PW, Manglik A, and von Zastrow M (2018). A Genetically Encoded Biosensor Reveals Location Bias of Opioid Drug Action. Neuron 98, 963–976.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sutkeviciute I, Clark LJ, White AD, Gardella TJ, and Vilardaga JP (2019). PTH/PTHrP Receptor Signaling, Allostery, and Structures. Trends Endocrinol. Metab. 30, 860–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takasaki J, Saito T, Taniguchi M, Kawasaki T, Moritani Y, Hayashi K, and Kobori M (2004). A novel Gαq/11-selective inhibitor. J. Biol. Chem 279, 47438–47445. [DOI] [PubMed] [Google Scholar]
- Tsvetanova NG, and von Zastrow M (2014). Spatial encoding of cyclic AMP signaling specificity by GPCR endocytosis. Nat. Chem. Biol 10, 1061–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vilardaga JP, Gardella TJ, Wehbi VL, and Feinstein TN (2012). Noncanonical signaling of the PTH receptor. Trends Pharmacol. Sci 33, 423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wehbi VL, Stevenson HP, Feinstein TN, Calero G, Romero G, and Vilardaga JP (2013). Noncanonical GPCR signaling arising from a PTH receptor-arrestin-Gβγ complex. Proc. Natl. Acad. Sci. U. S. A 110, 1530–1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wein MN, and Kronenberg HM (2018). Regulation of bone remodeling by parathyroid hormone. Cold Spring Harb. Perspect. Med 8, a031237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White AD, Fang F, Jean-Alphonse FG, Clark LJ, An HJ, Liu H, Zhao Y, Reynolds SL, Lee S, Xiao K, et al. (2019). Ca2+ allostery in PTH-receptor signaling. Proc. Natl. Acad. Sci. U. S. A 116, 3294–3299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White AD, Jean-Alphonse FG, Fang F, Peña KA, Liu S, König GM, Inoue A, Aslanoglou D, Gellman SH, Kostenis E, et al. (2020). Gq/11-dependent regulation of endosomal cAMP generation by parathyroid hormone class B GPCR. Proc. Natl. Acad. Sci. U. S. A 117, 7455–7460. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The published article includes all datasets generated or analyzed during this study.




