1
|
Kume M, Ahmad A, Shiers S, Burton MD, DeFea KA, Vagner J, Dussor G, Boitano S, Price TJ. C781, a β-Arrestin Biased Antagonist at Protease-Activated Receptor-2 (PAR2), Displays in vivo Efficacy Against Protease-Induced Pain in Mice. THE JOURNAL OF PAIN 2023; 24:605-616. [PMID: 36417966 PMCID: PMC10079573 DOI: 10.1016/j.jpain.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/21/2022]
Abstract
Given the limited options and often harmful side effects of current analgesics and the suffering caused by the opioid crisis, new classes of pain therapeutics are needed. Protease-activated receptors (PARs), particularly PAR2, are implicated in a variety of pathologies, including pain. Since the discovery of the role of PAR2 in pain, development of potent and specific antagonists has been slow. In this study, we describe the in vivo characterization of a novel small molecule/peptidomimetic hybrid compound, C781, as a β-arrestin-biased PAR2 antagonist. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. Pharmacokinetic studies were done to assess pharmacokinetic/pharmacodynamic relationship in vivo. We used both prevention and reversal paradigms with protease treatment to determine whether C781 could attenuate protease-evoked pain. C781 effectively prevented and reversed mechanical and spontaneous nociceptive behaviors in response to small molecule PAR2 agonists, mast cell activators, and neutrophil elastase. The ED50 of C781 (intraperitoneal dosing) for inhibition of PAR2 agonist (20.9 ng 2-AT)-evoked nociception was 6.3 mg/kg. C781 was not efficacious in the carrageenan inflammation model. Pharmacokinetic studies indicated limited long-term systemic bioavailability for C781 suggesting that optimizing pharmacokinetic properties could improve in vivo efficacy. Our work demonstrates in vivo efficacy of a biased PAR2 antagonist that selectively inhibits β-arrestin/MAPK signaling downstream of PAR2. Given the importance of this signaling pathway in PAR2-evoked nociception, C781 exemplifies a key pharmacophore for PAR2 that can be optimized for clinical development. PERSPECTIVE: Our work provides evidence that PAR2 antagonists that only block certain aspects of signaling by the receptor can be effective for blocking protease-evoked pain in mice. This is important because it creates a rationale for developing safer PAR2-targeting approaches for pain treatment.
Collapse
Affiliation(s)
- Moeno Kume
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Ayesha Ahmad
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Michael D Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | | | - Josef Vagner
- University of Arizona Bio5 Institute, Tucson, Arizona
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Scott Boitano
- University of Arizona Bio5 Institute, Tucson, Arizona; Asthma and Airway Disease Research Center, University of Arizona Heath Sciences, Tucson, Arizona; Department of Physiology, University of Arizona Heath Sciences, Tucson, Arizona
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
2
|
Schiff HV, Rivas CM, Pederson WP, Sandoval E, Gillman S, Prisco J, Kume M, Dussor G, Vagner J, Ledford JG, Price TJ, DeFea KA, Boitano S. β-Arrestin-biased proteinase-activated receptor-2 antagonist C781 limits allergen-induced airway hyperresponsiveness and inflammation. Br J Pharmacol 2023; 180:667-680. [PMID: 35735078 PMCID: PMC10311467 DOI: 10.1111/bph.15903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/13/2022] [Accepted: 06/18/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Asthma is a heterogenous disease strongly associated with inflammation that has many different causes and triggers. Current asthma treatments target symptoms such as bronchoconstriction and airway inflammation. Despite recent advances in biological therapies, there remains a need for new classes of therapeutic agents with novel, upstream targets. The proteinase-activated receptor-2 (PAR2) has long been implicated in allergic airway inflammation and asthma and it remains an intriguing target for novel therapies. Here, we describe the actions of C781, a newly developed low MW PAR2 biased antagonist, in vitro and in vivo in the context of acute allergen exposure. EXPERIMENTAL APPROACH A human bronchial epithelial cell line expressing PAR2 (16HBE14o- cells) was used to evaluate the modulation in vitro, by C781, of physiological responses to PAR2 activation and downstream β-arrestin/MAPK and Gq/Ca2+ signalling. Acute Alternaria alternata sensitized and challenged mice were used to evaluate C781 as a prophylactically administered modulator of airway hyperresponsiveness, inflammation and mucus overproduction in vivo. KEY RESULTS C781 reduced in vitro physiological signalling in response to ligand and proteinase activation. C781 effectively antagonized β-arrestin/MAPK signalling without significant effect on Gq/Ca2+ signalling in vitro. Given prophylactically, C781 modulated airway hyperresponsiveness, airway inflammation and mucus overproduction of the small airways in an acute allergen-challenged mouse model. CONCLUSION AND IMPLICATIONS Our work demonstrates the first biased PAR2 antagonist for β-arrestin/MAPK signalling. C781 is efficacious as a prophylactic treatment for allergen-induced airway hyperresponsiveness and inflammation in mice. It exemplifies a key pharmacophore for PAR2 that can be optimized for clinical development.
Collapse
Affiliation(s)
- Hillary V. Schiff
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Bio5 Collaborative Research Center, University of Arizona
| | - Candy M. Rivas
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Bio5 Collaborative Research Center, University of Arizona
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona
| | - William P. Pederson
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona
| | - Estevan Sandoval
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Bio5 Collaborative Research Center, University of Arizona
| | - Samuel Gillman
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Bio5 Collaborative Research Center, University of Arizona
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona
| | - Joy Prisco
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
| | - Moeno Kume
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, TX
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, TX
| | - Josef Vagner
- Bio5 Collaborative Research Center, University of Arizona
| | - Julie G. Ledford
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Department of Cellular and Molecular Medicine, University of Arizona
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, TX
| | - Kathryn A. DeFea
- University of California Riverside, Biomedical Sciences and PARMedics, Incorporated
| | - Scott Boitano
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences Center
- Bio5 Collaborative Research Center, University of Arizona
- Department of Physiology, University of Arizona
| |
Collapse
|
3
|
Rivas CM, Schiff H, Moutal A, Khanna R, Kiela PR, Dussor G, Price TJ, Vagner J, DeFea KA, Boitano S. Alternaria alternata-induced airway epithelial signaling and inflammatory responses via protease-activated receptor-2 expression. Biochem Biophys Res Commun 2022; 591:13-19. [PMID: 34990903 PMCID: PMC8792334 DOI: 10.1016/j.bbrc.2021.12.090] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023]
Abstract
Inhalation of the fungus Alternaria alternata is associated with an increased risk of allergic asthma development and exacerbations. Recent work in acute exposure animal models suggests that A. alternata-induced asthma symptoms, which include inflammation, mucus overproduction and airway hyperresponsiveness, are due to A. alternata proteases that act via protease-activated receptor-2 (PAR2). However, because other active components present in A. alternata may be contributing to asthma pathophysiology through alternative signaling, the specific role PAR2 plays in asthma initiation and maintenance remains undefined. Airway epithelial cells provide the first encounter with A. alternata and are thought to play an important role in initiating the physiologic response. To better understand the role for PAR2 airway epithelial signaling we created a PAR2-deficient human bronchial epithelial cell line (16HBEPAR-/-) from a model bronchial parental line (16HBE14o-). Comparison of in vitro physiologic responses in these cell lines demonstrated a complete loss of PAR2 agonist (2at-LIGRL-NH2) response and significantly attenuated protease (trypsin and elastase) and A. alternata responses in the 16HBEPAR-/- line. Apical application of A. alternata to 16HBE14o- and 16HBEPAR2-/- grown at air-liquid interface demonstrated rapid, PAR2-dependent and independent, inflammatory cytokine, chemokine and growth factor basolateral release. In conclusion, the novel human PAR2-deficient cell line allows for direct in vitro examination of the role(s) for PAR2 in allergen challenge with polarized human airway epithelial cells.
Collapse
Affiliation(s)
- Candy M. Rivas
- Department of Physiology, University of Arizona, Tucson, AZ;,Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ;,Bio5 Collaborative Research Institute, University of Arizona, Tucson, AZ
| | - Hillary Schiff
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ;,Bio5 Collaborative Research Institute, University of Arizona, Tucson, AZ;,Department of Biochemistry, University of Arizona, Tucson AZ
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Pawel R. Kiela
- Department of Pediatrics, University of Arizona, Tucson, AZ
| | - Gregory Dussor
- Center for Advanced Pain Studies, University of Texas at Dallas, TX
| | - Theodore J Price
- Center for Advanced Pain Studies, University of Texas at Dallas, TX
| | - Josef Vagner
- Bio5 Collaborative Research Institute, University of Arizona, Tucson, AZ
| | - Kathryn A. DeFea
- University of California Riverside, Biomedical Sciences and PARMedics, Incorporated
| | - Scott Boitano
- Department of Physiology, University of Arizona, Tucson, AZ;,Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ;,Bio5 Collaborative Research Institute, University of Arizona, Tucson, AZ;,Corresponding Author: Scott Boitano, Ph.D., Professor, Physiology, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, Arizona. 85724-5030, , +1 (520) 626-2105
| |
Collapse
|
4
|
Dy ABC, Langlais PR, Barker NK, Addison KJ, Tanyaratsrisakul S, Boitano S, Christenson SA, Kraft M, Meyers D, Bleecker ER, Li X, Ledford JG. Myeloid-associated differentiation marker is a novel SP-A-associated transmembrane protein whose expression on airway epithelial cells correlates with asthma severity. Sci Rep 2021; 11:23392. [PMID: 34862427 PMCID: PMC8642528 DOI: 10.1038/s41598-021-02869-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Surfactant protein A (SP-A) is well-known for its protective role in pulmonary immunity. Previous studies from our group have shown that SP-A mediates eosinophil activities, including degranulation and apoptosis. In order to identify potential binding partners on eosinophils for SP-A, eosinophil lysates were subjected to SP-A pull-down and tandem mass spectrometry (MS/MS) analysis. We identified one membrane-bound protein, myeloid-associated differentiation marker (MYADM), as a candidate SP-A binding partner. Blocking MYADM on mouse and human eosinophils ex vivo prevented SP-A from inducing apoptosis; blocking MYADM in vivo led to increased persistence of eosinophilia and airway hyper-responsiveness in an ovalbumin (OVA) allergy model and increased airways resistance and mucus production in a house dust mite (HDM) asthma model. Examination of a subset of participants in the Severe Asthma Research Program (SARP) cohort revealed a significant association between epithelial expression of MYADM in asthma patients and parameters of airway inflammation, including: peripheral blood eosinophilia, exhaled nitric oxide (FeNO) and the number of exacerbations in the past 12 months. Taken together, our studies provide the first evidence of MYADM as a novel SP-A-associated protein that is necessary for SP-A to induce eosinophil apoptosis and we bring to light the potential importance of this previously unrecognized transmembrane protein in patients with asthma.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Clinical Translational Sciences, University of Arizona Health Sciences, Tucson, AZ, 85721, USA
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Paul R Langlais
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Natalie K Barker
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
| | | | - Scott Boitano
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, 94117, USA
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Deborah Meyers
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Eugene R Bleecker
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Xingnan Li
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
- , 1230 N Cherry Avenue, BSRL Building, Tucson, AZ, 85719, USA.
| |
Collapse
|
5
|
Mwirigi J, Kume M, Hassler SN, Ahmad A, Ray PR, Jiang C, Chamessian A, Mseeh N, Ludwig BP, Rivera BD, Nieman MT, Van de Ven T, Ji RR, Dussor G, Boitano S, Vagner J, Price TJ. A Role for Protease Activated Receptor Type 3 (PAR3) in Nociception Demonstrated Through Development of a Novel Peptide Agonist. THE JOURNAL OF PAIN 2021; 22:692-706. [PMID: 33429107 PMCID: PMC8197731 DOI: 10.1016/j.jpain.2020.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
The protease activated receptor (PAR) family is a group of G-protein coupled receptors (GPCRs) activated by proteolytic cleavage of the extracellular domain. PARs are expressed in a variety of cell types with crucial roles in homeostasis, immune responses, inflammation, and pain. PAR3 is the least researched of the four PARs, with little known about its expression and function. We sought to better understand its potential function in the peripheral sensory nervous system. Mouse single-cell RNA sequencing data demonstrates that PAR3 is widely expressed in dorsal root ganglion (DRG) neurons. Co-expression of PAR3 mRNA with other PARs was identified in various DRG neuron subpopulations, consistent with its proposed role as a coreceptor of other PARs. We developed a lipid tethered PAR3 agonist, C660, that selectively activates PAR3 by eliciting a Ca2+ response in DRG and trigeminal neurons. In vivo, C660 induces mechanical hypersensitivity and facial grimacing in WT but not PAR3-/- mice. We characterized other nociceptive phenotypes in PAR3-/- mice and found a loss of hyperalgesic priming in response to IL-6, carrageenan, and a PAR2 agonist, suggesting that PAR3 contributes to long-lasting nociceptor plasticity in some contexts. To examine the potential role of PAR3 in regulating the activity of other PARs in sensory neurons, we administered PAR1, PAR2, and PAR4 agonists and assessed mechanical and affective pain behaviors in WT and PAR3-/- mice. We observed that the nociceptive effects of PAR1 agonists were potentiated in the absence of PAR3. Our findings suggest a complex role of PAR3 in the physiology and plasticity of nociceptors. PERSPECTIVE: We evaluated the role of PAR3, a G-protein coupled receptor, in nociception by developing a selective peptide agonist. Our findings suggest that PAR3 contributes to nociception in various contexts and plays a role in modulating the activity of other PARs.
Collapse
Affiliation(s)
- Juliet Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Moeno Kume
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Shayne N Hassler
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Ayesha Ahmad
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Changyu Jiang
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Alexander Chamessian
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Nakleh Mseeh
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Breya P Ludwig
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Benjamin D Rivera
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio
| | - Thomas Van de Ven
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Ru-Rong Ji
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Scott Boitano
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Josef Vagner
- University of Arizona, Bio5 Research Institute, Tucson, Arizona
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas.
| |
Collapse
|
6
|
Majewski MW, Gandhi DM, Holyst T, Wang Z, Hernandez I, Rosas R, Zhu J, Weiler H, Dockendorff C. Synthesis and initial pharmacology of dual-targeting ligands for putative complexes of integrin αVβ3 and PAR2. RSC Med Chem 2020; 11:940-949. [PMID: 33479689 PMCID: PMC7496306 DOI: 10.1039/d0md00098a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/24/2020] [Indexed: 11/21/2022] Open
Abstract
Unpublished data from our labs led us to hypothesize that activated protein C (aPC) may initiate an anti-inflammatory signal in endothelial cells by modulating both the integrin αVβ3 and protease-activated receptor 2 (PAR2), which may exist in close proximity on the cellular surface. To test this hypothesis and to probe the possible inflammation-related pathway, we designed and synthesized dual-targeting ligands composed of modified versions of two αVβ3 ligands and two agonists of PAR2. These novel ligands were connected via copper-catalyzed alkyne-azide cycloadditions with polyethylene glycol (PEG) spacers of variable length. Initial in vitro pharmacology with EA.hy926 and HUVEC endothelial cells indicated that these ligands are effective binders of αVβ3 and potent agonists of PAR2. These were also used in preliminary studies investigating their effects on PAR2 signaling in the presence of inflammatory agents, and represent the first examples of ligands targeting both PARs and integrins, though concurrent binding to αVβ3 and PAR2 has not yet been demonstrated.
Collapse
Affiliation(s)
- Mark W Majewski
- Department of Chemistry , Marquette University , P.O. Box 1881 , Milwaukee , WI 53201-1881 , USA . ; Tel: +1 414 288 1617
| | - Disha M Gandhi
- Department of Chemistry , Marquette University , P.O. Box 1881 , Milwaukee , WI 53201-1881 , USA . ; Tel: +1 414 288 1617
| | - Trudy Holyst
- Blood Research Institute , Versiti , Milwaukee , WI 53226 , USA
| | - Zhengli Wang
- Blood Research Institute , Versiti , Milwaukee , WI 53226 , USA
| | - Irene Hernandez
- Blood Research Institute , Versiti , Milwaukee , WI 53226 , USA
| | - Ricardo Rosas
- Department of Chemistry , Marquette University , P.O. Box 1881 , Milwaukee , WI 53201-1881 , USA . ; Tel: +1 414 288 1617
| | - Jieqing Zhu
- Blood Research Institute , Versiti , Milwaukee , WI 53226 , USA
- Department of Biochemistry , Medical College of Wisconsin , Milwaukee , WI 53226 , USA
| | - Hartmut Weiler
- Blood Research Institute , Versiti , Milwaukee , WI 53226 , USA
- Department of Physiology , Medical College of Wisconsin , Milwaukee , WI 53226 , USA
| | - Chris Dockendorff
- Department of Chemistry , Marquette University , P.O. Box 1881 , Milwaukee , WI 53201-1881 , USA . ; Tel: +1 414 288 1617
| |
Collapse
|
7
|
Dy ABC, Arif MZ, Addison KJ, Que LG, Boitano S, Kraft M, Ledford JG. Genetic Variation in Surfactant Protein-A2 Delays Resolution of Eosinophilia in Asthma. THE JOURNAL OF IMMUNOLOGY 2019; 203:1122-1130. [PMID: 31350355 DOI: 10.4049/jimmunol.1900546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/01/2019] [Indexed: 01/21/2023]
Abstract
Surfactant protein-A (SP-A) is an important mediator of pulmonary immunity. A specific genetic variation in SP-A2, corresponding to a glutamine (Q) to lysine (K) amino acid substitution at position 223 of the lectin domain, was shown to alter the ability of SP-A to inhibit eosinophil degranulation. Because a large subgroup of asthmatics have associated eosinophilia, often accompanied by inflammation associated with delayed clearance, our goal was to define how SP-A mediates eosinophil resolution in allergic airways and whether genetic variation affects this activity. Wild-type, SP-A knockout (SP-A KO) and humanized (SP-A2 223Q/Q, SP-A2 223K/K) C57BL/6 mice were challenged in an allergic OVA model, and parameters of inflammation were examined. Peripheral blood eosinophils were isolated to assess the effect of SP-A genetic variation on apoptosis and chemotaxis. Five days postchallenge, SP-A KO and humanized SP-A2 223K/K mice had persistent eosinophilia in bronchoalveolar lavage fluid compared with wild-type and SP-A2 223Q/Q mice, suggesting an impairment in eosinophil resolution. In vitro, human SP-A containing either the 223Q or the 223K allele was chemoattractant for eosinophils whereas only 223Q resulted in decreased eosinophil viability. Our results suggest that SP-A aids in the resolution of allergic airway inflammation by promoting eosinophil clearance from lung tissue through chemotaxis, independent of SP-A2 Q223K, and by inducing apoptosis of eosinophils, which is altered by the polymorphism.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Clinical Translational Sciences, University of Arizona Health Sciences, Tucson, AZ 85721.,Asthma and Airway Disease Research Center, Tucson, AZ 85724
| | - Muhammad Z Arif
- Department of Medicine, University of Arizona, Tucson, AZ 85724
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, Tucson, AZ 85724.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Loretta G Que
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710; and
| | - Scott Boitano
- Asthma and Airway Disease Research Center, Tucson, AZ 85724.,Department of Physiology, University of Arizona, Tucson, AZ 85724
| | - Monica Kraft
- Asthma and Airway Disease Research Center, Tucson, AZ 85724.,Department of Medicine, University of Arizona, Tucson, AZ 85724
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, Tucson, AZ 85724; .,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
8
|
Stolwijk JA, Skiba M, Kade C, Bernhardt G, Buschauer A, Hübner H, Gmeiner P, Wegener J. Increasing the throughput of label-free cell assays to study the activation of G-protein-coupled receptors by using a serial agonist exposure protocol. Integr Biol (Camb) 2019; 11:99-108. [PMID: 31083709 DOI: 10.1093/intbio/zyz010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/12/2019] [Accepted: 05/03/2019] [Indexed: 12/25/2022]
Abstract
Label-free, holistic assays, monitoring, for example, the impedance of cells on electrodes, are gaining increasing popularity in the evaluation of G-protein-coupled receptor (GPCR) ligands. It is the strength of these approaches to provide the integrated cellular response non-invasively, highly automated and with a device-dependent time resolution down to several milliseconds. With an increasing number of samples to be studied in parallel, the available time resolution is, however, reduced and the cost for the disposable sensor arrays may become limiting. Inspired by protocols from organ pharmacology, we investigated a simple serial agonist addition assay that circumvents these limitations in impedance-based cellular assays. Using a serial addition of increasing concentrations of a GPCR agonist while continuously monitoring the sample's impedance, we were able to establish a full concentration-response curve for the endogenous agonist histamine on a single layer of U-373 MG cells endogenously expressing the histamine 1 receptor (H1R). This approach is validated with respect to conventional, parallel agonist addition protocols and studies using H1R antagonists such as mepyramine. Applicability of the serial agonist addition assay was shown for other GPCRs known for their signaling via one of the canonical G-protein pathways, Gq, Gi/0 or Gs as well. The serial agonist addition protocol has the potential to further strengthen the output of label-free analysis of GPCR activation.
Collapse
Affiliation(s)
- J A Stolwijk
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - M Skiba
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - C Kade
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - G Bernhardt
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - A Buschauer
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - H Hübner
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg FAU, Erlangen, Germany
| | - P Gmeiner
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg FAU, Erlangen, Germany
| | - J Wegener
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
- Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Munich, Germany
| |
Collapse
|
9
|
Stolwijk JA, Wegener J. Impedance-Based Assays Along the Life Span of Adherent Mammalian Cells In Vitro: From Initial Adhesion to Cell Death. BIOANALYTICAL REVIEWS 2019. [DOI: 10.1007/11663_2019_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Cotter ML, Boitano S, Vagner J, Burt JM. Lipidated connexin mimetic peptides potently inhibit gap junction-mediated Ca 2+-wave propagation. Am J Physiol Cell Physiol 2018; 315:C141-C154. [PMID: 29631365 DOI: 10.1152/ajpcell.00156.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Connexin (Cx) mimetic peptides (e.g., Gap27: SRPTEKTIFII; Peptide5: VDCFLSRPTEKT) reversibly inhibit hemichannel (HCh) and gap junction channel (GJCh) function in a concentration- and time-dependent manner (HCh: ~5 µM, <1 h; GJCh: ~100 µM, > 1 h). We hypothesized that addition of a hexadecyl tail to SRPTEKT (SRPTEKT- Hdc) would improve its ability to concentrate in the plasma membrane and consequently increase its inhibitory efficacy. We show that SRPTEKT- Hdc inhibited intercellular Ca2+-wave propagation in Cx43-expressing MDCK and rabbit tracheal epithelial cells in a time (61-75 min)- and concentration (IC50: 66 pM)-dependent manner, a concentration efficacy five orders of magnitude lower than observed for the nonlipidated Gap27. HCh-mediated dye uptake was inhibited by SRPTEKT- Hdc with similar efficacy. Following peptide washout, HCh-mediated dye uptake was restored to control levels, whereas Ca2+-wave propagation was only partially restored. Scrambled and reverse sequence lipidated peptides had no detectable inhibitory effect on Ca2+-wave propagation or dye uptake. Cx43 expression was unchanged by SRPTEKT- Hdc incubation; however, Triton-insoluble Cx43 was reduced by SRPTEKT- Hdc exposure and reversed following washout. In summary, our results show that SRPTEKT- Hdc blocked HCh function and intercellular Ca2+ signaling at concentrations that minimally affected dye coupling. Selective inhibition of intercellular Ca2+ signaling, likely indicative of channel conformation-specific SRPTEKT- Hdc binding, could contribute significantly to the protective effects of these mimetic peptides in settings of injury. Our data also demonstrate that lipidation represents a paradigm for development of highly potent, efficacious, and selective mimetic peptide inhibitors of hemichannel and gap junction channel-mediated signaling.
Collapse
Affiliation(s)
- Maura L Cotter
- Department of Physiology, University of Arizona , Tucson, Arizona
| | - Scott Boitano
- Department of Physiology, University of Arizona , Tucson, Arizona.,Asthma and Airway Disease Research Center, University of Arizona , Tucson, Arizona.,Bio5 Collaborative Research Institute, University of Arizona , Tucson, Arizona
| | - Josef Vagner
- Bio5 Collaborative Research Institute, University of Arizona , Tucson, Arizona.,Department of Pharmacology, University of Arizona , Tucson, Arizona
| | - Janis M Burt
- Department of Physiology, University of Arizona , Tucson, Arizona
| |
Collapse
|
11
|
Suen J, Adams M, Lim J, Madala P, Xu W, Cotterell A, He Y, Yau M, Hooper J, Fairlie D. Mapping transmembrane residues of proteinase activated receptor 2 (PAR 2 ) that influence ligand-modulated calcium signaling. Pharmacol Res 2017; 117:328-342. [DOI: 10.1016/j.phrs.2016.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022]
|
12
|
Palygin O, Ilatovskaya DV, Staruschenko A. Protease-activated receptors in kidney disease progression. Am J Physiol Renal Physiol 2016; 311:F1140-F1144. [PMID: 27733370 DOI: 10.1152/ajprenal.00460.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/07/2016] [Indexed: 01/05/2023] Open
Abstract
Protease-activated receptors (PARs) are members of a well-known family of transmembrane G protein-coupled receptors (GPCRs). Four PARs have been identified to date, of which PAR1 and PAR2 are the most abundant receptors, and have been shown to be expressed in the kidney vascular and tubular cells. PAR signaling is mediated by an N-terminus tethered ligand that can be unmasked by serine protease cleavage. The receptors are activated by endogenous serine proteases, such as thrombin (acts on PARs 1, 3, and 4) and trypsin (PAR2). PARs can be involved in glomerular, microvascular, and inflammatory regulation of renal function in both normal and pathological conditions. As an example, it was shown that human glomerular epithelial and mesangial cells express PARs, and these receptors are involved in the pathogenesis of crescentic glomerulonephritis, glomerular fibrin deposition, and macrophage infiltration. Activation of these receptors in the kidney also modulates renal hemodynamics and glomerular filtration rate. Clinical studies further demonstrated that the concentration of urinary thrombin is associated with glomerulonephritis and type 2 diabetic nephropathy; thus, molecular and functional mechanisms of PARs activation can be directly involved in renal disease progression. We briefly discuss here the recent literature related to activation of PAR signaling in glomeruli and the kidney in general and provide some examples of PAR1 signaling in glomeruli podocytes.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
13
|
Pera T, Penn RB. Bronchoprotection and bronchorelaxation in asthma: New targets, and new ways to target the old ones. Pharmacol Ther 2016; 164:82-96. [PMID: 27113408 PMCID: PMC4942340 DOI: 10.1016/j.pharmthera.2016.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023]
Abstract
Despite over 50years of inhaled beta-agonists and corticosteroids as the default management or rescue drugs for asthma, recent research suggests that new therapeutic options are likely to emerge. This belief stems from both an improved understanding of what causes and regulates airway smooth muscle (ASM) contraction, and the identification of new targets whose inhibition or activation can relax ASM. In this review we discuss the recent findings that provide new insight into ASM contractile regulation, a revolution in pharmacology that identifies new ways to "tune" G protein-coupled receptors to improve therapeutic efficacy, and the discovery of several novel targets/approaches capable of effecting bronchoprotection or bronchodilation.
Collapse
Affiliation(s)
- Tonio Pera
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Raymond B Penn
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
14
|
Harpel K, Baker RD, Amirsolaimani B, Mehravar S, Vagner J, Matsunaga TO, Banerjee B, Kieu K. Imaging of targeted lipid microbubbles to detect cancer cells using third harmonic generation microscopy. BIOMEDICAL OPTICS EXPRESS 2016; 7:2849-60. [PMID: 27446711 PMCID: PMC4948635 DOI: 10.1364/boe.7.002849] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 05/19/2023]
Abstract
The use of receptor-targeted lipid microbubbles imaged by ultrasound is an innovative method of detecting and localizing disease. However, since ultrasound requires a medium between the transducer and the object being imaged, it is impractical to apply to an exposed surface in a surgical setting where sterile fields need be maintained and ultrasound gel may cause the bubbles to collapse. Multiphoton microscopy (MPM) is an emerging tool for accurate, label-free imaging of tissues and cells with high resolution and contrast. We have recently determined a novel application of MPM to be used for detecting targeted microbubble adherence to the upregulated plectin-receptor on pancreatic tumor cells. Specifically, the third-harmonic generation response can be used to detect bound microbubbles to various cell types presenting MPM as an alternative and useful imaging method. This is an interesting technique that can potentially be translated as a diagnostic tool for the early detection of cancer and inflammatory disorders.
Collapse
Affiliation(s)
- Kaitlin Harpel
- Department of Biomedical Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, Arizona, 85721, USA
- Department of Medical Imaging, College of Medicine, University of Arizona, 1609 N. Warren Ave., Tucson, Arizona, 85719, USA
| | - Robert Dawson Baker
- College of Optical Sciences, University of Arizona, 1603 E. University Blvd., Tucson, AZ, 85721, USA
| | - Babak Amirsolaimani
- College of Optical Sciences, University of Arizona, 1603 E. University Blvd., Tucson, AZ, 85721, USA
| | - Soroush Mehravar
- College of Optical Sciences, University of Arizona, 1603 E. University Blvd., Tucson, AZ, 85721, USA
| | - Josef Vagner
- Ligand Discovery Laboratory, BIO5 Institute, University of Arizona, 1657 E. Helen Street, Tucson, AZ, 85721, USA
| | - Terry O. Matsunaga
- Department of Biomedical Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, Arizona, 85721, USA
- Department of Medical Imaging, College of Medicine, University of Arizona, 1609 N. Warren Ave., Tucson, Arizona, 85719, USA
| | - Bhaskar Banerjee
- Department of Biomedical Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, Arizona, 85721, USA
- College of Optical Sciences, University of Arizona, 1603 E. University Blvd., Tucson, AZ, 85721, USA
- Department of Medicine, College of Medicine, University of Arizona, 1501 N. Campbell, Tucson, Arizona, 85724, USA
| | - Khanh Kieu
- College of Optical Sciences, University of Arizona, 1603 E. University Blvd., Tucson, AZ, 85721, USA
| |
Collapse
|
15
|
Sherwood CL, Boitano S. Airway epithelial cell exposure to distinct e-cigarette liquid flavorings reveals toxicity thresholds and activation of CFTR by the chocolate flavoring 2,5-dimethypyrazine. Respir Res 2016. [PMID: 27184162 DOI: 10.1186/s12931‐016‐0369‐9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The potential for adverse respiratory effects following exposure to electronic (e-) cigarette liquid (e-liquid) flavorings remains largely unexplored. Given the multitude of flavor permutations on the market, identification of those flavor constituents that negatively impact the respiratory tract is a daunting task. In this study we examined the impact of common e-liquid flavoring chemicals on the airway epithelium, the cellular monolayer that provides the first line of defense against inhaled particulates, pathogens, and toxicants. METHODS We used the xCELLigence real-time cell analyzer (RTCA) as a primary high-capacity screening tool to assess cytotoxicity thresholds and physiological effects of common e-liquid flavoring chemicals on immortalized human bronchial epithelial cells (16HBE14o-). The RTCA was used secondarily to assess the capability of 16HBE14o- cells to respond to cellular signaling agonists following a 24 h exposure to select flavoring chemicals. Finally, we conducted biophysical measurements of well-differentiated primary mouse tracheal epithelial (MTE) cells with an Ussing chamber to measure the effects of e-cigarette flavoring constituents on barrier function and ion conductance. RESULTS In our high-capacity screens five of the seven flavoring chemicals displayed changes in cellular impedance consistent with cell death at concentrations found in e-liquid. Vanillin and the chocolate flavoring 2,5-dimethylpyrazine caused alterations in cellular physiology indicative of a cellular signaling event. At subcytotoxic levels, 24 h exposure to 2,5-dimethylpyrazine compromised the ability of airway epithelial cells to respond to signaling agonists important in salt and water balance at the airway surface. Biophysical measurements of 2,5-dimethylpyrazine on primary MTE cells revealed alterations in ion conductance consistent with an efflux at the apical airway surface that was accompanied by a transient loss in transepithelial resistance. Mechanistic studies confirmed that the increases in ion conductance evoked by 2,5-dimethylpyrazine were largely attributed to a protein kinase A-dependent (PKA) activation of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel. CONCLUSIONS Data from our high-capacity screening assays demonstrates that individual e-cigarette liquid flavoring chemicals vary in their cytotoxicity profiles and that some constituents evoke a cellular physiological response on their own independent of cell death. The activation of CFTR by 2,5-dimethylpyrazine may have detrimental consequences for airway surface liquid homeostasis in individuals that use e-cigarettes habitually.
Collapse
Affiliation(s)
- Cara L Sherwood
- Asthma and Airway Disease Research Center, Arizona Health Sciences Center, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5030, USA. .,Bio5 Collaborative Research Institute, Arizona Health Sciences Center, Tucson, AZ, USA.
| | - Scott Boitano
- Asthma and Airway Disease Research Center, Arizona Health Sciences Center, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5030, USA.,Bio5 Collaborative Research Institute, Arizona Health Sciences Center, Tucson, AZ, USA.,Department of Physiology, Arizona Health Sciences Center, Tucson, AZ, USA
| |
Collapse
|
16
|
Sherwood CL, Boitano S. Airway epithelial cell exposure to distinct e-cigarette liquid flavorings reveals toxicity thresholds and activation of CFTR by the chocolate flavoring 2,5-dimethypyrazine. Respir Res 2016; 17:57. [PMID: 27184162 PMCID: PMC4869201 DOI: 10.1186/s12931-016-0369-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/13/2015] [Indexed: 01/20/2023] Open
Abstract
Background The potential for adverse respiratory effects following exposure to electronic (e-) cigarette liquid (e-liquid) flavorings remains largely unexplored. Given the multitude of flavor permutations on the market, identification of those flavor constituents that negatively impact the respiratory tract is a daunting task. In this study we examined the impact of common e-liquid flavoring chemicals on the airway epithelium, the cellular monolayer that provides the first line of defense against inhaled particulates, pathogens, and toxicants. Methods We used the xCELLigence real-time cell analyzer (RTCA) as a primary high-capacity screening tool to assess cytotoxicity thresholds and physiological effects of common e-liquid flavoring chemicals on immortalized human bronchial epithelial cells (16HBE14o-). The RTCA was used secondarily to assess the capability of 16HBE14o- cells to respond to cellular signaling agonists following a 24 h exposure to select flavoring chemicals. Finally, we conducted biophysical measurements of well-differentiated primary mouse tracheal epithelial (MTE) cells with an Ussing chamber to measure the effects of e-cigarette flavoring constituents on barrier function and ion conductance. Results In our high-capacity screens five of the seven flavoring chemicals displayed changes in cellular impedance consistent with cell death at concentrations found in e-liquid. Vanillin and the chocolate flavoring 2,5-dimethylpyrazine caused alterations in cellular physiology indicative of a cellular signaling event. At subcytotoxic levels, 24 h exposure to 2,5-dimethylpyrazine compromised the ability of airway epithelial cells to respond to signaling agonists important in salt and water balance at the airway surface. Biophysical measurements of 2,5-dimethylpyrazine on primary MTE cells revealed alterations in ion conductance consistent with an efflux at the apical airway surface that was accompanied by a transient loss in transepithelial resistance. Mechanistic studies confirmed that the increases in ion conductance evoked by 2,5-dimethylpyrazine were largely attributed to a protein kinase A-dependent (PKA) activation of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel. Conclusions Data from our high-capacity screening assays demonstrates that individual e-cigarette liquid flavoring chemicals vary in their cytotoxicity profiles and that some constituents evoke a cellular physiological response on their own independent of cell death. The activation of CFTR by 2,5-dimethylpyrazine may have detrimental consequences for airway surface liquid homeostasis in individuals that use e-cigarettes habitually.
Collapse
Affiliation(s)
- Cara L Sherwood
- Asthma and Airway Disease Research Center, Arizona Health Sciences Center, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5030, USA. .,Bio5 Collaborative Research Institute, Arizona Health Sciences Center, Tucson, AZ, USA.
| | - Scott Boitano
- Asthma and Airway Disease Research Center, Arizona Health Sciences Center, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5030, USA.,Bio5 Collaborative Research Institute, Arizona Health Sciences Center, Tucson, AZ, USA.,Department of Physiology, Arizona Health Sciences Center, Tucson, AZ, USA
| |
Collapse
|
17
|
Yau MK, Lim J, Liu L, Fairlie DP. Protease activated receptor 2 (PAR2) modulators: a patent review (2010-2015). Expert Opin Ther Pat 2016; 26:471-83. [PMID: 26936077 DOI: 10.1517/13543776.2016.1154540] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Protease activated receptor 2 (PAR2) is a self-activated G protein-coupled receptor that has been implicated in several diseases, including inflammatory, gastrointestinal, respiratory, metabolic diseases, cancers and others, making it an important prospective drug target. No known endogenous ligands are available for PAR2, so having potent exogenous agonists and antagonists can be helpful for studying physiological functions of PAR2. AREAS COVERED This review covers agonist-, antagonist-, antibody- and pepducin-based modulators of PAR2 reported in patent applications between 2010-2015, along with their available structure-activity relationships, biological activities and potential uses for studying PAR2. EXPERT OPINION In the last six years, substantial efforts were made towards developing PAR2 modulators, but most lack potency or selectivity or have poor pharmacokinetic profiles. Many PAR2 modulators were assessed by measuring Gαq protein-mediated calcium release in cells. This may be insufficient to fully characterize ligand function, since different ligands signal through PAR2 via multiple signaling pathways. It may be feasible to develop biased ligands as drugs that can selectively modulate one or more specific signaling pathways linking PAR2 to a specific diseased state. Accordingly, potent, orally bioavailable, pathway- and receptor-selective PAR2 modulators may be an achievable goal to realizing effective drugs that can treat PAR2-mediated diseases.
Collapse
Affiliation(s)
- Mei-Kwan Yau
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - Junxian Lim
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - Ligong Liu
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - David P Fairlie
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| |
Collapse
|
18
|
Characterization and Functions of Protease-Activated Receptor 2 in Obesity, Diabetes, and Metabolic Syndrome: A Systematic Review. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3130496. [PMID: 27006943 PMCID: PMC4781943 DOI: 10.1155/2016/3130496] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
Proteinase-activated receptor 2 (PAR2) is a cell surface receptor activated by serine proteinases or specific synthetic compounds. Interest in PAR2 as a pharmaceutical target for various diseases is increasing. Here we asked two questions relevant to endothelial dysfunction and diabetes: How is PAR2 function affected in blood vessels? What role does PAR2 have in promoting obesity, diabetes, and/or metabolic syndrome, specifically via the endothelium and adipose tissues? We conducted a systematic review of the published literature in PubMed and Scopus (July 2015; search terms: par2, par-2, f2lr1, adipose, obesity, diabetes, and metabolic syndrome). Seven studies focused on PAR2 and vascular function. The obesity, diabetes, or metabolic syndrome animal models differed amongst studies, but each reported that PAR2-mediated vasodilator actions were preserved in the face of endothelial dysfunction. The remaining studies focused on nonvascular functions and provided evidence supporting the concept that PAR2 activation promoted obesity. Key studies showed that PAR2 activation regulated cellular metabolism, and PAR2 antagonists inhibited adipose gain and metabolic dysfunction in rats. We conclude that PAR2 antagonists for treatment of obesity indeed show early promise as a therapeutic strategy; however, endothelial-specific PAR2 functions, which may offset mechanisms that produce vascular dysfunction in diabetes, warrant additional study.
Collapse
|
19
|
Protease-activated receptor 2 activation is sufficient to induce the transition to a chronic pain state. Pain 2016; 156:859-867. [PMID: 25734998 DOI: 10.1097/j.pain.0000000000000125] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protease-activated receptor type 2 (PAR2) is known to play an important role in inflammatory, visceral, and cancer-evoked pain based on studies using PAR2 knockout (PAR2(-/-)) mice. We have tested the hypothesis that specific activation of PAR2 is sufficient to induce a chronic pain state through extracellular signal-regulated kinase (ERK) signaling to protein synthesis machinery. We have further tested whether the maintenance of this chronic pain state involves a brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (trkB)/atypical protein kinase C (aPKC) signaling axis. We observed that intraplantar injection of the novel highly specific PAR2 agonist, 2-aminothiazol-4-yl-LIGRL-NH2 (2-at), evokes a long-lasting acute mechanical hypersensitivity (median effective dose ∼12 pmoles), facial grimacing, and causes robust hyperalgesic priming as revealed by a subsequent mechanical hypersensitivity and facial grimacing to prostaglandin E2 (PGE2) injection. The promechanical hypersensitivity effect of 2-at is completely absent in PAR2(-/-) mice as is hyperalgesic priming. Intraplantar injection of the upstream ERK inhibitor, U0126, and the eukaryotic initiation factor (eIF) 4F complex inhibitor, 4EGI-1, prevented the development of acute mechanical hypersensitivity and hyperalgesic priming after 2-at injection. Systemic injection of the trkB antagonist ANA-12 similarly inhibited PAR2-mediated mechanical hypersensitivity, grimacing, and hyperalgesic priming. Inhibition of aPKC (intrathecal delivery of ZIP) or trkB (systemic administration of ANA-12) after the resolution of 2-at-induced mechanical hypersensitivity reversed the maintenance of hyperalgesic priming. Hence, PAR2 activation is sufficient to induce neuronal plasticity leading to a chronic pain state, the maintenance of which is dependent on a BDNF/trkB/aPKC signaling axis.
Collapse
|
20
|
Yau MK, Suen JY, Xu W, Lim J, Liu L, Adams MN, He Y, Hooper JD, Reid RC, Fairlie DP. Potent Small Agonists of Protease Activated Receptor 2. ACS Med Chem Lett 2016; 7:105-10. [PMID: 26819675 DOI: 10.1021/acsmedchemlett.5b00429] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022] Open
Abstract
Many proteases cut the PAR2 N-terminus resulting in conformational changes that activate cells. Synthetic peptides corresponding to newly exposed N-terminal sequences of PAR2 also activate the receptor at micromolar concentrations. PAR2-selective small molecules reported here induce PAR2-mediated intracellular calcium signaling at nanomolar concentrations (EC50 = 15-100 nM, iCa(2+), CHO-hPAR2 cells). These are the most potent and efficient small molecule ligands to activate PAR2-mediated calcium release and chemotaxis, including for human breast and prostate cancer cells.
Collapse
Affiliation(s)
- Mei-Kwan Yau
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Jacky Y. Suen
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Weijun Xu
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Junxian Lim
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Ligong Liu
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Mark N. Adams
- Translational
Research Institute, Mater Research Institute, The University of Queensland, Woolloongabba, Qld 4102, Australia
| | - Yaowu He
- Translational
Research Institute, Mater Research Institute, The University of Queensland, Woolloongabba, Qld 4102, Australia
| | - John D. Hooper
- Translational
Research Institute, Mater Research Institute, The University of Queensland, Woolloongabba, Qld 4102, Australia
| | - Robert C. Reid
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - David P. Fairlie
- Institute
for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| |
Collapse
|
21
|
Boitano S, Hoffman J, Flynn AN, Asiedu MN, Tillu DV, Zhang Z, Sherwood CL, Rivas CM, DeFea KA, Vagner J, Price TJ. The novel PAR2 ligand C391 blocks multiple PAR2 signalling pathways in vitro and in vivo. Br J Pharmacol 2015; 172:4535-4545. [PMID: 26140338 DOI: 10.1111/bph.13238] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 06/18/2015] [Accepted: 06/28/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Proteinase-activated receptor-2 (PAR2) is a GPCR linked to diverse pathologies, including acute and chronic pain. PAR2 is one of the four PARs that are activated by proteolytic cleavage of the extracellular amino terminus, resulting in an exposed, tethered peptide agonist. Several peptide and peptidomimetic agonists, with high potency and efficacy, have been developed to probe the functions of PAR2, in vitro and in vivo. However, few similarly potent and effective antagonists have been described. EXPERIMENTAL APPROACH We modified the peptidomimetic PAR2 agonist, 2-furoyl-LIGRLO-NH2 , to create a novel PAR2 peptidomimetic ligand, C391. C391 was evaluated for PAR2 agonist/antagonist activity to PAR2 across Gq signalling pathways using the naturally expressing PAR2 cell line 16HBE14o-. For antagonist studies, a highly potent and specific peptidomimetic agonist (2-aminothiazo-4-yl-LIGRL-NH2 ) and proteinase agonist (trypsin) were used to activate PAR2. C391 was also evaluated in vivo for reduction of thermal hyperalgesia, mediated by mast cell degranulation, in mice. KEY RESULTS C391 is a potent and specific peptidomimetic antagonist, blocking multiple signalling pathways (Gq -dependent Ca2+ , MAPK) induced following peptidomimetic or proteinase activation of human PAR2. In a PAR2-dependent behavioural assay in mice, C391 dose-dependently (75 μg maximum effect) blocked the thermal hyperalgesia, mediated by mast cell degranulation. CONCLUSIONS AND IMPLICATIONS C391 is the first low MW antagonist to block both PAR2 Ca2+ and MAPK signalling pathways activated by peptidomimetics and/or proteinase activation. C391 represents a new molecular structure for PAR2 antagonism and can serve as a basis for further development for this important therapeutic target.
Collapse
Affiliation(s)
- Scott Boitano
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Justin Hoffman
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Andrea N Flynn
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Marina N Asiedu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Dipti V Tillu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Zhenyu Zhang
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Cara L Sherwood
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Candy M Rivas
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, USA
| | - Kathryn A DeFea
- Biomedical Sciences Division, University of California, Riverside, CA, USA
| | - Josef Vagner
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Theodore J Price
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology, University of Arizona, Tucson, AZ, USA.,School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| |
Collapse
|
22
|
Sherwood CL, Daines MO, Price TJ, Vagner J, Boitano S. A highly potent agonist to protease-activated receptor-2 reveals apical activation of the airway epithelium resulting in Ca2+-regulated ion conductance. Am J Physiol Cell Physiol 2014; 307:C718-26. [PMID: 25143347 DOI: 10.1152/ajpcell.00257.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The airway epithelium provides a barrier that separates inhaled air and its various particulates from the underlying tissues. It provides key physiological functions in both sensing the environment and initiating appropriate innate immune defenses to protect the lung. Protease-activated receptor-2 (PAR2) is expressed both apically and basolaterally throughout the airway epithelium. One consequence of basolateral PAR2 activation is the rapid, Ca(2+)-dependent ion flux that favors secretion in the normally absorptive airway epithelium. However, roles for apically expressed PAR2 activation have not been demonstrated, in part due to the lack of specific, high-potency PAR2 ligands. In the present study, we used the newly developed PAR2 ligand 2at-LIGRLO(PEG3-Pam)-NH2 in combination with well-differentiated, primary cultured airway epithelial cells from wild-type and PAR2 (-/-) mice to examine the physiological role of PAR2 in the conducting airway after apical activation. Using digital imaging microscopy of intracellular Ca(2+) concentration changes, we verified ligand potency on PAR2 in primary cultured airway cells. Examination of airway epithelial tissue in an Ussing chamber showed that apical activation of PAR2 by 2at-LIGRLO(PEG3-Pam)-NH2 resulted in a transient decrease in transepithelial resistance that was due to increased apical ion efflux. We determined pharmacologically that this increase in ion conductance was through Ca(2+)-activated Cl(-) and large-conductance K(+) channels that were blocked with a Ca(2+)-activated Cl(-) channel inhibitor and clotrimazole, respectively. Stimulation of Cl(-) efflux via PAR2 activation at the airway epithelial surface can increase airway surface liquid that would aid in clearing the airway of noxious inhaled agents.
Collapse
Affiliation(s)
- Cara L Sherwood
- Arizona Respiratory Center, University of Arizona, Tucson, Arizona; The BIO5 Collaborative Research Institute, University of Arizona, Tucson, Arizona
| | - Michael O Daines
- Arizona Respiratory Center, University of Arizona, Tucson, Arizona; Department of Pediatrics, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona; The BIO5 Collaborative Research Institute, University of Arizona, Tucson, Arizona
| | - Theodore J Price
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona; and The BIO5 Collaborative Research Institute, University of Arizona, Tucson, Arizona
| | - Josef Vagner
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, Arizona
| | - Scott Boitano
- Arizona Respiratory Center, University of Arizona, Tucson, Arizona; Department of Physiology, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona; The BIO5 Collaborative Research Institute, University of Arizona, Tucson, Arizona
| |
Collapse
|
23
|
Development and evaluation of small peptidomimetic ligands to protease-activated receptor-2 (PAR2) through the use of lipid tethering. PLoS One 2014; 9:e99140. [PMID: 24927179 PMCID: PMC4057235 DOI: 10.1371/journal.pone.0099140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/09/2014] [Indexed: 02/05/2023] Open
Abstract
Protease-activated receptor-2 (PAR2) is a G-Protein Coupled Receptor (GPCR) activated by proteolytic cleavage to expose an attached, tethered ligand (SLIGRL). We evaluated the ability for lipid-tethered-peptidomimetics to activate PAR2 with in vitro physiological and Ca2+ signaling assays to determine minimal components necessary for potent, specific and full PAR2 activation. A known PAR2 activating compound containing a hexadecyl (Hdc) lipid via three polyethylene glycol (PEG) linkers (2at-LIGRL-PEG3-Hdc) provided a potent agonist starting point (physiological EC50 = 1.4 nM; 95% CI: 1.2-2.3 nM). In a set of truncated analogs, 2at-LIGR-PEG3-Hdc retained potency (EC50 = 2.1 nM; 1.3-3.4 nM) with improved selectivity for PAR2 over Mas1 related G-protein coupled receptor type C11, a GPCR that can be activated by the PAR2 peptide agonist, SLIGRL-NH2. 2at-LIG-PEG3-Hdc was the smallest full PAR2 agonist, albeit with a reduced EC50 (46 nM; 20-100 nM). 2at-LI-PEG3-Hdc retained specific activity for PAR2 with reduced EC50 (310 nM; 260-360 nM) but displayed partial PAR2 activation in both physiological and Ca2+ signaling assays. Further truncation (2at-L-PEG3-Hdc and 2at-PEG3-Hdc) eliminated in vitro activity. When used in vivo, full and partial PAR2 in vitro agonists evoked mechanical hypersensitivity at a 15 pmole dose while 2at-L-PEG3-Hdc lacked efficacy. Minimum peptidomimetic PAR2 agonists were developed with known heterocycle substitutes for Ser1 (isoxazole or aminothiazoyl) and cyclohexylalanine (Cha) as a substitute for Leu2. Both heterocycle-tetrapeptide and heterocycle-dipeptides displayed PAR2 specificity, however, only the heterocycle-tetrapeptides displayed full PAR2 agonism. Using the lipid-tethered-peptidomimetic approach we have developed novel structure activity relationships for PAR2 that allows for selective probing of PAR2 function across a broad range of physiological systems.
Collapse
|
24
|
Walker JKL, DeFea KA. Role for β-arrestin in mediating paradoxical β2AR and PAR2 signaling in asthma. Curr Opin Pharmacol 2014; 16:142-7. [PMID: 24907413 DOI: 10.1016/j.coph.2014.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) utilize (at least) two signal transduction pathways to elicit cellular responses including the classic G protein-dependent, and the more recently discovered β-arrestin-dependent, signaling pathways. In human and murine models of asthma, agonist-activation of β2-adrenergic receptor (β2AR) or Protease-activated-receptor-2 (PAR2) results in relief from bronchospasm via airway smooth muscle relaxation. However, chronic activation of these receptors, leads to pro-inflammatory responses. One plausible explanation underlying the paradoxical effects of β2AR and PAR2 agonism in asthma is that the beneficial and harmful effects are associated with distinct signaling pathways. Specifically, G protein-dependent signaling mediates relaxation of airway smooth muscle, whereas β-arrestin-dependent signaling promotes inflammation. This review explores the evidence supporting the hypothesis that β-arrestin-dependent signaling downstream of β2AR and PAR2 is detrimental in asthma and examines the therapeutic opportunities for selectively targeting this pathway.
Collapse
Affiliation(s)
- Julia K L Walker
- Duke University School of Nursing, Duke University Medical Center, Durham, NC 27710, USA.
| | - Katherine A DeFea
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| |
Collapse
|
25
|
Yau MK, Liu L, Fairlie DP. Toward drugs for protease-activated receptor 2 (PAR2). J Med Chem 2013; 56:7477-97. [PMID: 23895492 DOI: 10.1021/jm400638v] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PAR2 has a distinctive functional phenotype among an unusual group of GPCRs called protease activated receptors, which self-activate after cleavage of their N-termini by mainly serine proteases. PAR2 is the most highly expressed PAR on certain immune cells, and it is activated by multiple proteases (but not thrombin) in inflammation. PAR2 is expressed on many types of primary human cells and cancer cells. PAR2 knockout mice and PAR2 agonists and antagonists have implicated PAR2 as a promising target in inflammatory conditions; respiratory, gastrointestinal, metabolic, cardiovascular, and neurological dysfunction; and cancers. This article summarizes salient features of PAR2 structure, activation, and function; opportunities for disease intervention via PAR2; pharmacological properties of published or patented PAR2 modulators (small molecule agonists and antagonists, pepducins, antibodies); and some personal perspectives on limitations of assessing their properties and on promising new directions for PAR2 modulation.
Collapse
Affiliation(s)
- Mei-Kwan Yau
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland , Brisbane, Queensland 4072, Australia
| | | | | |
Collapse
|