Summary

Characterizing Modulators of Protease-Activated Receptors with a Calcium Mobilization Assay Using a Plate Reader

Published: May 24, 2024
doi:

Summary

An improved protocol for a calcium mobilization assay with endothelial cells, used to identify ligands of protease-activated receptors (PARs), has been developed. The new protocol reduces total assay time by 90-120 min and yields reproducible concentration-response curves.

Abstract

Changes in calcium concentration in cells are rapidly monitored in a high-throughput fashion with the use of intracellular, fluorescent, calcium-binding dyes and imaging instruments that can measure fluorescent emissions from up to 1,536 wells simultaneously. However, these instruments are much more expensive and can be challenging to maintain relative to widely available plate readers that scan wells individually. Described here is an optimized plate reader assay for use with an endothelial cell line (EA.hy926) to measure the protease-activated receptor (PAR)-driven activation of Gαq signaling and subsequent calcium mobilization using the calcium-binding dye Fluo-4. This assay has been used to characterize a range of PAR ligands, including the allosteric PAR1-targeting anti-inflammatory “parmodulin” ligands identified in the Dockendorff lab. This protocol obviates the need for an automated liquid handler and permits the medium-throughput screening of PAR ligands in 96-well plates and should be applicable to the study of other receptors that initiate calcium mobilization.

Introduction

Protease-activated receptors (PARs)1,2,3 are a subfamily of class A G protein-coupled receptors (GPCRs) that are expressed in a variety of cell types, including platelets and endothelial cells4,5,6,7. Unlike the majority of GPCRs, PARs have a unique intramolecular mode of activation. Most GPCRs are activated by soluble ligands interacting with a distinct binding pocket, but PARs are activated by the proteolytic cleavage of the N-terminus, which results in a new tethered ligand that can interact with the extracellular loop 2 domain on the surface of a cell6,8,9. This interaction activates the receptor and can initiate several signaling pathways, promoting effects such as inflammation and platelet activation4,10,11,12. Different proteases can activate PARs through cleavage at unique sites on the N-terminus, revealing different tethered ligands (TL) that stabilize receptor conformations, which initiate different signaling pathways9,13,14,15. For example, in the most well-studied member of the subfamily, PAR1, cleavage by thrombin is used to support numerous biological processes, including platelet activation and leukocyte recruitment to the endothelium, but can lead to deleterious effects when the receptor is overexpressed or overactivated4,16,17,18,19,20,21. Conversely, cleavage by activated protein C (aPC) can promote anti-inflammatory effects and maintenance of endothelial barriers15,22,23,24,25,26,27,28,29. PARs can also be activated by peptide analogs of the TLs in an intermolecular fashion13,30,31. These peptides are routinely used to measure PAR inhibition (modulation) in place of PAR-targeting proteases, and they are used in this protocol.

Numerous disorders are associated with pathological PAR1 signaling, including sepsis22,32, cardiovascular disease33,34,35,36,37,38, kidney disease39,40,41,42, sickle cell disease43, fibrosis44, osteoporosis and osteoarthritis45,46, neurodegeneration47,48,49,50,51, and cancer52,53,54,55,56,57,58,59. Antagonists of PAR1 have been studied since the 1990s as antiplatelet agents for cardiovascular disease, and the growing list of diseases associated with the receptor necessitates the identification of novel ligands for use as biological probes (tool compounds) or as potential therapeutics. Currently, there is only one FDA-approved PAR1 antagonist, vorapaxar, which is used to treat coronary artery disease in high-risk patients34,36,37,60. An alternative PAR1 antagonist, the pepducin PZ-128, completed a successful phase II study to prevent thrombosis in cardiac catheterization patients38. The Dockendorff group has focused on the medicinal chemistry and pharmacology of a separate class of small molecules, PAR1 ligands known as parmodulins61,62. Unlike reported PAR1 antagonists such as vorapaxar, parmodulins are allosteric, biased modulators of PAR1 that selectively block the Gαq pathway while promoting cytoprotective effects similar to aPC. Unlike potent orthosteric PAR1 antagonists such as vorapaxar, published parmodulins are also reversible63,64,65.

Initially, parmodulins were identified by Flaumenhaft and coworkers for their ability to inhibit P-selectin expression or granule secretion in platelets61,66. However, an alternative method was required to study the effects of parmodulins on endothelial cells. One common method to monitor GPCR-related signaling is to measure intracellular Ca2+ mobilization, an important secondary messenger that can be measured using a suitable intracellular calcium-binding dye67,68. Substantial evidence has been provided showing that calcium mobilization induced by PAR1 is through the activation of Gαq69,70. Once activated by its tethered ligand (or a suitable exogenous ligand), PAR1 undergoes a conformational change which causes guanosine diphosphate (GDP) bound to the Gαq subunit to be replaced by guanosine triphosphate (GTP)68. The Gαq subunit then activates phospholipase Cβ (PLC-β), which catalyzes the hydrolysis of phosphatidylinositol 4,5 bisphosphate (PIP2), forming 1,4,5-inositol triphosphate (IP3) and diacylglycerol (DAG). Finally, IP3 binds to IP3-sensitive Ca2+ channels in the membrane of the endoplasmic reticulum, allowing Ca2+ to be released into the cytoplasm, where it can bind to Ca2+-dependent fluorescent dyes, such as Fluo-4, that are added to the cells71. This process occurs within seconds and can increase the concentration of Ca2+ 100-fold, leading to a drastic change in the amount of calcium-bound dye and a robust fluorescence signal.

In 2018, the Dockendorff group disclosed a medium-throughput Ca2+ mobilization assay that could be used to identify antagonists of the Gαq pathway of PAR172. The assay used EA.hy92673, a hybrid human endothelial cell line, which can be used for multiple passages without a noticeable change in PAR1 expression, and is established for in vitro measurements of cytoprotective effects. 

The original protocol used EA.hy926 cells in 96-well plates and loaded with Fluo-4/AM dye, which was chosen due to its intense fluorescence at 488 nm and high cell permeability. Once the dye was loaded into the cells, lengthy washing steps were performed with an automated 8-channel liquid handler (faster methods of liquid handling, such as a 96-channel washer, were inaccessible). The reproducibility of this assay was superior to that without the careful, automated robotic media changes. Antagonists were then incubated with the cells, PAR1 was activated through the sequential addition of a selective agonist (16 wells at a time), and changes in fluorescence resulting from calcium mobilization and dye binding were measured to determine activity.

While this protocol allows for the measurement of PAR1-mediated calcium mobilization, it is limited by the time required to assay each 96-well plate. Long experiment times are problematic not only because the number of compounds that can be screened each day is limited, but also because dye efflux occurs over time, narrowing the assay window by increasing the basal fluorescence. One contributor to the long experiment time is the use of an 8-channel liquid handler for plate washing, which adds over 30 min to each experiment. The required tips also became difficult to obtain due to supply chain problems. Here an updated protocol for the PAR-mediated calcium mobilization assay that does not require a liquid handler, and therefore can be run in higher throughput, is reported. This protocol should also be suitable for measuring signaling with other GPCRs that lead to intracellular calcium mobilization. This updated plate reader protocol is ideal for academic and small industrial labs that do not have the resources for expensive cell imaging instruments but have a need to rapidly screen numerous compounds. For an example of a calcium mobilization assay using a plate imager, see Caers et al.74.

Protocol

All media exchanges/additions made in steps 1 and 2 of the following protocol are performed in a sterile hood. Unless otherwise noted, all plasticware used in the sterile hood must be purchased sterilized and sealed or sterilized appropriately via autoclave. 1. Initiation of EA.hy926 cell line Acquire EA.hy926 cells. Store vial(s) of cells in the vapor phase of a liquid nitrogen tank. Prepare DMEM complete medium by first warming DMEM, FBS, and …

Representative Results

The purpose of this assay is generally to produce concentration-response curves (CRCs) for three to four new parmodulins. On each assay plate, an additional CRC for a known compound, such as NRD-21, is often generated that acts as a quality check for the experiment due to its known IC50. To generate CRCs, a plate map such as the one depicted in Table 1 should be planned. If single-point concentration-responses are desired instead, compounds at 10 µM final concentrations (or other preferre…

Discussion

While the previously reported protocol72 was generally reliable and allowed us to identify a new lead parmodulin, NRD-21,62 a more efficient protocol was desired. The assay was further compromised during the supply shortage caused by the COVID-19 pandemic. Acquiring tips for the automated liquid handler became difficult, and attempting to wash, sterilize, and reuse the tips produced CRCs with significant variance. This facilitated an urgent series of experiments designed to…

Divulgations

The authors have nothing to disclose.

Acknowledgements

We thank Irene Hernandez, Trudy Holyst, Dr. Hartmut Weiler (Versiti Blood Research Institute), and Dr. Leggy Arnold (University of Wisconsin-Milwaukee) for providing space and indirect support of this project, and Dr. John McCorvy (Medical College of Wisconsin) for pertinent advice. We thank the National Heart, Lung, and Blood Institute (R15HL127636), the U.S. Dept. of Defense (W81XWH22101), and the National Science Foundation (2223225) for grant support.

Materials

Cell Culture Reagents
Adherent EA.hy926 cells ATCC CRL-2922
CellStripper cell dissociation reagent Corning 25-056-CI Trypsin can optionally be used, but should definitely be avoided with PAR2 assays.
Dulbecco's Modified Eagle Medium (DMEM) w/phenol red Corning 10-013-CV
Fetal Bovine Serum (FBS) Avantor 97068-091
Gelatin from porcine skin MilliporeSigma G2500 Use to make an aqueous 0.4% (w/v) solution with deionized water. Autoclave before use to sterilize.
Pen/Strep (100X) Corning 30-002-CI
Phosphate-buffered saline (PBS) Corning 21-040-CV
Trypan Blue (0.4% w/v) Corning 25-900-CI
Calcium Mobilization Reagents
4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) Thermo 172571000
Bovine serum albumin (BSA) Avantor 97061-420
Calcium chloride dihydrate Thermo 42352-0250
Dimethyl sulfoxide Thermo J66650-AD
Fluo-4/AM Invitrogen F14201
Hank's balanced salt solution (Ca/Mg/phenol-red free) Corning 21-022-CV
Magnesium chloride hexahydrate MilliporeSigma M2393
Pluronic F-127 (Poloxamer 407) Spectrum Chemical P1166
Probenecid TCI America P1975
Sodium hydroxide VWR International BDH9292
TFLLRN-NH2 (TFA salt) Prepared by Trudy Holyst at the Versiti Blood Research Institute
Materials
96-well culture-treated, black-walled, clear bottom assay plate Corning 3603 with transparent lids
Centrifuge tube, 15 mL Avantor 89039-664
Centrifuge tube, 50 mL Avantor 89039-656
Culture flask, T-75 Corning 353136 tissue culture treated
Disposable reagent reservoir, 50 mL Corning RES-V-50-S
Enspire plate reader Perkin Elmer Discontinued
Microcentrifuge tube, 1.5 mL Avantor 20170-038
Pasteur pipette, 9" Fisher 13-678-6B must be sterilized
PCR tube strip with separate flat cap strips Avantor 76318-802
Pipette tips, 20 µL Biotix 63300042 sterile, filtered tips
Pipette tips, 200 µL Biotix 63300044 sterile, filtered tips
Pipette tips, 1250 µL Biotix 63300047 sterile, filtered tips
Prism GraphPad volume 6 used
Serological pipette, 5 mL Tradewinds Direct  07-5005
Serological pipette, 10 mL Tradewinds Direct  07-5010
Serological pipette, 25 mL Tradewinds Direct  07-5025

References

  1. Rasmussen, U. B., et al. cDNA cloning and expression of a hamster alpha-thrombin receptor coupled to Ca2+ mobilization. FEBS Lett. 288 (1-2), 123-128 (1991).
  2. Vu, T. K., Hung, D. T., Wheaton, V. I., Coughlin, S. R. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell. 64 (6), 1057-1068 (1991).
  3. Vu, T. K., Wheaton, V. I., Hung, D. T., Charo, I., Coughlin, S. R. Domains specifying thrombin-receptor interaction. Nature. 353 (6345), 674-677 (1991).
  4. Coughlin, S. R. Thrombin signalling and protease-activated receptors. Nature. 407 (6801), 258-264 (2000).
  5. Adams, M. N., et al. Structure, function and pathophysiology of protease activated receptors. Pharmacol. Ther. 130 (3), 248-282 (2011).
  6. Heuberger, D. M., Schuepbach, R. A. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb. J. 17, 4 (2019).
  7. Han, X., Nieman, M. T., Kerlin, B. A. Protease-activated receptors: An illustrated review. Res. Pract. Thromb. Haemost. 5 (1), 17-26 (2021).
  8. Gerszten, R. E., et al. Specificity of the thrombin receptor for agonist peptide is defined by its extracellular surface. Nature. 368 (6472), 648-651 (1994).
  9. Ludeman, M. J., Kataoka, H., Srinivasan, Y., Esmon, N. L., Esmon, C. T., Coughlin, S. R. PAR1 cleavage and signaling in response to activated protein C and thrombin. J. Biol. Chem. 280 (13), 13122-13128 (2005).
  10. Rezaie, A. R. Protease-activated receptor signalling by coagulation proteases in endothelial cells. Thromb. Haemost. 112 (5), 876-882 (2014).
  11. Déry, O., Corvera, C. U., Steinhoff, M., Bunnett, N. W. Proteinase-activated receptors: novel mechanisms of signaling by serine proteases. Am. J. Physiol. 274 (6), (1998).
  12. Peach, C. J., Edgington-Mitchell, L. E., Bunnett, N. W., Schmidt, B. L. Protease-activated receptors in health and disease. Physiol. Rev. 103 (1), 717-785 (2023).
  13. Chen, J., Ishii, M., Wang, L., Ishii, K., Coughlin, S. R. Thrombin receptor activation. Confirmation of the intramolecular tethered liganding hypothesis and discovery of an alternative intermolecular liganding mode. J. Biol. Chem. 269 (23), 16041-16045 (1994).
  14. Scarborough, R. M., et al. Tethered ligand agonist peptides. Structural requirements for thrombin receptor activation reveal mechanism of proteolytic unmasking of agonist function. J. Biol. Chem. 267 (19), 13146-13149 (1992).
  15. Schuepbach, R. A., Madon, J., Ender, M., Galli, P., Riewald, M. Protease-activated receptor-1 cleaved at R46 mediates cytoprotective effects. J. Thromb. Haemost. 10 (8), 1675-1684 (2012).
  16. Davey, M. G., Lüscher, E. F. Actions of thrombin and other coagulant and proteolytic enzymes on blood platelets. Nature. 216 (5118), 857-858 (1967).
  17. Sambrano, G. R., Weiss, E. J., Zheng, Y. W., Huang, W., Coughlin, S. R. Role of thrombin signalling in platelets in haemostasis and thrombosis. Nature. 413 (6851), 74-78 (2001).
  18. Nieman, M. T., Schmaier, A. H. Interaction of thrombin with PAR1 and PAR4 at the thrombin cleavage site. Biochimie. 46 (29), 8603-8610 (2007).
  19. Andersen, H., Greenberg, D. L., Fujikawa, K., Xu, W., Chung, D. W., Davie, E. W. Protease-activated receptor 1 is the primary mediator of thrombin-stimulated platelet procoagulant activity. Proc. Natl. Acad. Sci. U.S.A. 96 (20), 11189-11193 (1999).
  20. Negrier, C., Shima, M., Hoffman, M. The central role of thrombin in bleeding disorders. Blood Rev. 38, 100582 (2019).
  21. Larsen, J. B., Hvas, A. -. M. Thrombin: A Pivotal Player in Hemostasis and. Semin. Thromb. and Hemost. 47 (7), 759-774 (2021).
  22. Riewald, M., Petrovan, R. J., Donner, A., Mueller, B. M., Ruf, W. Activation of endothelial cell protease activated receptor 1 by the protein C pathway. Science. 296 (5574), 1880-1882 (2002).
  23. Riewald, M., Petrovan, R. J., Donner, A., Ruf, W. Activated protein C signals through the thrombin receptor PAR1 in endothelial cells. J. Endotoxin Res. 9 (5), 317-321 (2003).
  24. Mosnier, L. O., Griffin, J. H. Inhibition of staurosporine-induced apoptosis of endothelial cells by activated protein C requires protease-activated receptor-1 and endothelial cell protein C receptor. Biochem. J. 373 (Pt 1), 65-70 (2003).
  25. Mosnier, L. O., Zlokovic, B. V., Griffin, J. H. The cytoprotective protein C pathway. Blood. 109 (8), 3161-3172 (2007).
  26. Mosnier, L. O., Sinha, R. K., Burnier, L., Bouwens, E. A., Griffin, J. H. Biased agonism of protease-activated receptor 1 by activated protein C caused by noncanonical cleavage at Arg46. Blood. 120 (26), 5237-5246 (2012).
  27. Soh, U. J. K., Trejo, J. Activated protein C promotes protease-activated receptor-1 cytoprotective signaling through β-arrestin and dishevelled-2 scaffolds. Proc. Natl. Acad. Sci. U.S.A. 108 (50), E1372-E1380 (2011).
  28. Birch, C. A., Wedegaertner, H., Orduña-Castillo, L. B., Gonzalez Ramirez, M. L., Qin, H., Trejo, J. Endothelial APC/PAR1 distinctly regulates cytokine-induced pro-inflammatory VCAM-1 expression. Front. Mol. Biosci. 10, 1211597 (2023).
  29. Shahzad, K., Kohli, S., Al-Dabet, M. M., Isermann, B. Cell biology of activated protein. C. Curr. Opin. Hematol. 26 (1), 41-50 (2019).
  30. Hollenberg, M. D., Saifeddine, M., al-Ani, B., Kawabata, A. Proteinase-activated receptors: structural requirements for activity, receptor cross-reactivity, and receptor selectivity of receptor-activating peptides. Can. J. Physiol. Pharmacol. 75 (7), 832-841 (1997).
  31. Kawabata, A., Saifeddine, M., Al-Ani, B., Leblond, L., Hollenberg, M. D. Evaluation of proteinase-activated receptor-1 (PAR1) agonists and antagonists using a cultured cell receptor desensitization assay: activation of PAR2 by PAR1-targeted ligands. J. Pharmacol. Exp. Ther. 288 (1), 358-370 (1999).
  32. Kerschen, E. J., et al. Endotoxemia and sepsis mortality reduction by non-anticoagulant activated protein. C. J. Exp. Med. 204 (10), 2439-2448 (2007).
  33. Andrade-Gordon, P., et al. Design, synthesis, and biological characterization of a peptide-mimetic antagonist for a tethered-ligand receptor. Proc. Natl. Acad. Sci. U.S.A. 96 (22), 12257-12262 (1999).
  34. Chackalamannil, S., et al. Discovery of a novel, orally active himbacine-based thrombin receptor antagonist (SCH 530348) with potent antiplatelet activity. J. Med. Chem. 51 (11), 3061-3064 (2008).
  35. Wiviott, S. D., et al. Randomized trial of atopaxar in the treatment of patients with coronary artery disease: the lessons from antagonizing the cellular effect of Thrombin-Coronary Artery Disease Trial. Circulation. 123 (17), 1854-1863 (2011).
  36. Tricoci, P., et al. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. N. Engl. J. Med. 366 (1), 20-33 (2012).
  37. Morrow, D. A., et al. Vorapaxar in the secondary prevention of atherothrombotic events. N. Engl. J. Med. 366 (15), 1404-1413 (2012).
  38. Kuliopulos, A., et al. PAR1 (Protease-Activated Receptor 1) Pepducin Therapy Targeting Myocardial Necrosis in Coronary Artery Disease and Acute Coronary Syndrome Patients Undergoing Cardiac Catheterization: A Randomized, Placebo-Controlled, Phase 2 study. Arterioscler. Thromb. Vasc. Biol. 40 (12), 2990-3003 (2020).
  39. Gupta, A., Williams, M. D., Macias, W. L., Molitoris, B. A., Grinnell, B. W. Activated protein C and acute kidney injury: Selective targeting of PAR-1. Curr. Drug Targets. 10 (12), 1212-1226 (2009).
  40. Dong, W., et al. Activated Protein C Ameliorates Renal Ischemia-Reperfusion Injury by Restricting Y-Box Binding Protein-1 Ubiquitination. J. Am. Soc. Nephrol. 26 (11), 2789-2799 (2015).
  41. Al-Dabet, M. M., et al. Reversal of the renal hyperglycemic memory in diabetic kidney disease by targeting sustained tubular p21 expression. Nat. Comm. 13 (1), 5062 (2022).
  42. El Eter, E. A., Aldrees, A. Inhibition of proinflammatory cytokines by SCH79797, a selective protease-activated receptor 1 antagonist, protects rat kidney against ischemia-reperfusion injury. Shock. 37 (6), 639-644 (2012).
  43. Sparkenbaugh, E. M., et al. Thrombin activation of PAR-1 contributes to microvascular stasis in mouse models of sickle cell disease. Blood. 135 (20), 1783-1787 (2020).
  44. Lin, C., et al. High endogenous activated protein C levels attenuates bleomycin-induced pulmonary fibrosis. J. of Cell. Mol. Med. 20 (11), 2029-2035 (2016).
  45. Zhang, Y., Wang, H., Zhu, G., Qian, A., Chen, W. F2r negatively regulates osteoclastogenesis through inhibiting the Akt and NFκB signaling pathways. Int. J. Biol. Sci. 16 (9), 1629-1639 (2020).
  46. Chandrabalan, A., Firth, A., Litchfield, R. B., Appleton, C. T., Getgood, A., Ramachandran, R. Human osteoarthritis knee joint synovial fluids cleave and activate Proteinase-Activated Receptor (PAR) mediated signaling. Sci. Rep. 13 (1), 1124 (2023).
  47. Festoff, B. W., et al. Neuroprotective effects of recombinant thrombomodulin in controlled contusion spinal cord injury implicates thrombin signaling. J. Neurotrauma. 21 (7), 907-922 (2004).
  48. Zhong, Z., et al. Activated protein C therapy slows ALS-like disease in mice by transcriptionally inhibiting SOD1 in motor neurons and microglia cells. J. Clin. Investig. 119 (11), 3437-3449 (2009).
  49. Griffin, J. H., Mosnier, L. O., Fernández, J. A., Zlokovic, B. V. 2016 Scientific Sessions Sol Sherry Distinguished Lecturer in Thrombosis: Thrombotic Stroke: Neuroprotective Therapy by Recombinant-Activated Protein C. Arterioscler. Thromb. Vasc. Biol. 36 (11), 2143-2151 (2016).
  50. Yoon, H., et al. Blocking the Thrombin Receptor Promotes Repair of Demyelinated Lesions in the Adult Brain. J.Neurosci. 40 (7), 1483-1500 (2020).
  51. Kanki, H., et al. β-arrestin-2 in PAR-1-biased signaling has a crucial role in endothelial function via PDGF-β in stroke. Cell Death Dis. 10 (2), 100 (2019).
  52. Even-Ram, S., et al. Thrombin receptor overexpression in malignant and physiological invasion processes. Nat. Med. 4 (8), 909-914 (1998).
  53. Kamath, L., Meydani, A., Foss, F., Kuliopulos, A. Signaling from protease-activated receptor-1 inhibits migration and invasion of breast cancer cells. Cancer Res. 61 (15), 5933-5940 (2001).
  54. Shi, X., Gangadharan, B., Brass, L. F., Ruf, W., Mueller, B. M. Protease-activated receptors (PAR1 and PAR2) contribute to tumor cell motility and metastasis. Mol. Cancer Res. 2 (7), 395-402 (2004).
  55. Yang, E., et al. Blockade of PAR1 signaling with cell-penetrating pepducins inhibits Akt survival pathways in breast cancer cells and suppresses tumor survival and metastasis. Cancer Res. 69 (15), 6223-6231 (2009).
  56. McEachron, T. A., Pawlinski, R., Richards, K. L., Church, F. C., Mackman, N. Protease-activated receptors mediate crosstalk between coagulation and fibrinolysis. Blood. 116 (23), 5037-5044 (2010).
  57. Adams, G. N., et al. Protease-activated receptor-1 impedes prostate and intestinal tumor progression in mice. J. Thromb. Haemost. 16 (11), 2258-2269 (2018).
  58. Arakaki, A. K. S., Pan, W. -. A., Lin, H., Trejo, J. The α-arrestin ARRDC3 suppresses breast carcinoma invasion by regulating G protein-coupled receptor lysosomal sorting and signaling. J. Biol. Chem. 293 (9), 3350-3362 (2018).
  59. Schweickert, P. G., et al. Thrombin-PAR1 signaling in pancreatic cancer promotes an immunosuppressive microenvironment. J. Thromb. Haemost. 19 (1), 161-172 (2021).
  60. Chackalamannil, S., et al. Discovery of potent orally active thrombin receptor (protease activated receptor 1) antagonists as novel antithrombotic agents. J. Med. Chem. 48 (19), 5884-5887 (2005).
  61. Dockendorff, C., et al. Discovery of 1,3-Diaminobenzenes as Selective Inhibitors of Platelet Activation at the PAR1 Receptor. ACS Med. Chem. Lett. 3 (3), 232-237 (2012).
  62. Gandhi, D. M., et al. The parmodulin NRD-21 is an allosteric inhibitor of PAR1 Gq signaling with improved anti-inflammatory activity and stability. Bioorg. Med. Chem. 27 (17), 3788-3796 (2019).
  63. Aisiku, O., et al. Parmodulins inhibit thrombus formation without inducing endothelial injury caused by vorapaxar. Blood. 125 (12), 1976-1985 (2015).
  64. De Ceunynck, K., et al. PAR1 agonists stimulate APC-like endothelial cytoprotection and confer resistance to thromboinflammatory injury. Proc. Natl. Acad. Sci. U.S.A. 115 (5), E982-E991 (2018).
  65. Aisiku, O., Peters, C. G., Gunnink, S., Dilks, J. R., Dockendorff, C., Flaumenhaft, R. Effects of Biased PAR1 Ligands On Platelets and Endothelial Cells. Blood. 122 (21), 23 (2013).
  66. Dowal, L., et al. Identification of an antithrombotic allosteric modulator that acts through helix 8 of PAR1. Proc. Natl. Acad. Sci. U.S.A. 108 (7), 2951-2956 (2011).
  67. Tsien, R. Y. A non-disruptive technique for loading calcium buffers and indicators into cells. Nature. 290 (5806), 527-528 (1981).
  68. Ma, Q., Ye, L., Liu, H., Shi, Y., Zhou, N. An overview of Ca2+ mobilization assays in GPCR drug discovery. Expert Opin. Drug Discov. 12 (5), 511-523 (2017).
  69. Baffy, G., Yang, L., Raj, S., Manning, D. R., Williamson, J. R. G protein coupling to the thrombin receptor in Chinese hamster lung fibroblasts. J. Biol. Chem. 269 (11), 8483-8487 (1994).
  70. McLaughlin, J. N., Shen, L., Holinstat, M., Brooks, J. D., Dibenedetto, E., Hamm, H. E. Functional selectivity of G protein signaling by agonist peptides and thrombin for the protease-activated receptor-1. J. Biol. Chem. 280 (26), 25048-25059 (2005).
  71. Berridge, M. J., Lipp, P., Bootman, M. D. The versatility and universality of calcium signalling. Nat. Rev. Mol. Cell Biol. 1 (1), 11-21 (2000).
  72. Gandhi, D. M., et al. Characterization of Protease-Activated Receptor (PAR) ligands: Parmodulins are reversible allosteric inhibitors of PAR1-driven calcium mobilization in endothelial cells. Bioorg. Med. Chem. 26 (9), 2514-2529 (2018).
  73. Ahn, K., Pan, S., Beningo, K., Hupe, D. A permanent human cell line (EA.hy926) preserves the characteristics of endothelin converting enzyme from primary human umbilical vein endothelial cells. Life Sci. 56 (26), 2331-2341 (1995).
  74. Caers, J., et al. Characterization of G protein-coupled receptors by a fluorescence-based calcium mobilization assay. J. Vis. Exp. (89), e51516 (2014).
  75. Robbins, N., Koch, S. E., Tranter, M., Rubinstein, J. The history and future of probenecid. Cardiovasc. Toxicol. 12 (1), 1-9 (2012).
  76. McKinney, S. E., Peck, H. M., Bochey, J. M., Byham, B. B., Schuchardt, G. S., Beyer, K. H. Benemid, p-(Di-n-propylsulfamyl)-benzoic acid; toxicologic properties. J. Pharmacol. Exp. Ther. 102 (3), 208-214 (1951).
  77. Di Virgilio, F., Steinberg, T. H., Silverstein, S. C. Inhibition of Fura-2 sequestration and secretion with organic anion transport blockers. Cell Calcium. 11 (2-3), 57-62 (1990).
  78. Liu, K., et al. A multiplex calcium assay for identification of GPCR agonists and antagonists. Assay Drug Dev. Technol. 8 (3), 367-379 (2010).
  79. Klein, A. K., et al. Investigation of the Structure-Activity Relationships of Psilocybin Analogues. ACS Pharmacol. Transl. Sci. 4 (2), 533-542 (2021).
  80. Hawes, B. E., et al. In vitro pharmacological characterization of vorapaxar, a novel platelet thrombin receptor antagonist. Eur. J. Pharmacol. 762, 221-228 (2015).
  81. Bokoch, M. P., et al. Entry from the Lipid Bilayer: A Possible Pathway for Inhibition of a Peptide G Protein-Coupled Receptor by a Lipophilic Small Molecule. Biochimie. 57 (39), 5748-5758 (2018).
  82. Zhang, J. H., Chung, T. D., Oldenburg, K. R. A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays. J. Biomol. Screen. 4 (2), 67-73 (1999).
  83. Majewski, M. W., Gandhi, D. M., Rosas, R., Kodali, R., Arnold, L. A., Dockendorff, C. Design and Evaluation of Heterobivalent PAR1-PAR2 Ligands as Antagonists of Calcium Mobilization. ACS Med. Chem. Lett. 10 (1), 121-126 (2019).
This article has been published
Video Coming Soon
Keep me updated:

.

Citer Cet Article
DeRousse, J. T., Dockendorff, C. Characterizing Modulators of Protease-Activated Receptors with a Calcium Mobilization Assay Using a Plate Reader. J. Vis. Exp. (207), e66507, doi:10.3791/66507 (2024).

View Video