1
|
Patnaik R, Varghese RL, Khan S, Huda B, Bhurka F, Amiri L, Banerjee Y. Targeting PAR-2-driven inflammatory pathways in colorectal cancer: mechanistic insights from atorvastatin and rosuvastatin treatment in cell line models. Transl Cancer Res 2025; 14:1531-1566. [PMID: 40224964 PMCID: PMC11985218 DOI: 10.21037/tcr-24-1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/27/2025] [Indexed: 04/15/2025]
Abstract
Background Colorectal cancer (CRC) is a growing health concern globally and in regions such as the United Arab Emirates, where risk factors like obesity and hyperlipidaemia are prevalent. Chronic inflammation, driven by pathways involving protease-activated receptor 2 (PAR-2), plays a pivotal role in CRC progression, creating a tumour-promoting microenvironment. The overexpression of PAR-2 has been associated with increased tumour aggressiveness and drug resistance. While previous studies have focused on broad inflammatory modulation, this study explores the selective targeting of PAR-2 by atorvastatin (ATV) and rosuvastatin (RSV), highlighting their specificity by assessing minimal impact on PAR-1 expression, which serves as a control. Methods HT-29 and Caco-2 CRC cell lines were employed to investigate the anti-inflammatory effects of ATV and RSV. Inflammation was induced with lipopolysaccharide (LPS), followed by treatment with varying concentrations of ATV and RSV. Western blotting and real-time polymerase chain reaction for quantification (qPCR) were performed to quantify PAR-2 and TNF-α at both the protein and mRNA levels. Enzyme linked immunosorbent assay (ELISA) was used to measure the secretion of TNF-α. Calcium signalling, which plays a crucial role in inflammation, was analysed using Fluo-4 AM dye, with fluorescence imaging capturing the effects of statin treatment on intracellular calcium influx. Results LPS treatment significantly upregulated PAR-2 and TNF-α expression in both cell lines, validating the inflammatory model. Co-treatment with ATV or RSV reduced PAR-2 and TNF-α expression in a dose-dependent manner. The higher concentrations of ATV (50 µg/mL) and RSV (20 µg/mL) produced the most significant reduction in these inflammatory markers at both the protein and mRNA levels. Importantly, the treatment did not substantially alter PAR-1 expression, underlining the specificity of ATV and RSV in modulating PAR-2-mediated pathways. Additionally, statin treatment attenuated LPS-induced calcium influx, with fluorescence intensity decreasing markedly at higher concentrations of both statins. Conclusions This study provides novel insights into the selective targeting of PAR-2 by ATV and RSV, distinguishing their effects from PAR-1. The reduction in PAR-2 expression and TNF-α secretion, along with the suppression of calcium signalling, underscores the potential of these statins as targeted anti-inflammatory agents in CRC. The findings highlight the therapeutic value of ATV and RSV in modulating inflammation through PAR-2-specific pathways, which may contribute to reduced cancer progression. These results pave the way for further preclinical and clinical evaluations to explore statins as adjunctive therapies in the management of CRC.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Riah Lee Varghese
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Sara Khan
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Bintul Huda
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Farida Bhurka
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Layla Amiri
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
- Centre for Medical Education, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| |
Collapse
|
2
|
Oe Y, Tanaka T, Takahashi N. The Many Faces of Protease-Activated Receptor 2 in Kidney Injury. Biomedicines 2025; 13:414. [PMID: 40002827 PMCID: PMC11852827 DOI: 10.3390/biomedicines13020414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Protease-activated receptor 2 (PAR2) is a seven-transmembrane, G-protein-coupled receptor that is activated by coagulation proteases such as factor VIIa and factor Xa and other serine proteases. It is a potential therapeutic target for kidney injury, as it enhances inflammatory and fibrotic responses via the nuclear factor-kappa B and mitogen-activated protein kinase cascades. The body of knowledge regarding the role of PAR2 in kidney disease is currently growing, and its role in various kidney disease models, such as acute kidney injury, renal fibrosis, diabetic kidney disease, aging, and thrombotic microangiopathy, has been reported. Here, we review the literature to better understand the various aspects of PAR2 in kidney disease.
Collapse
Affiliation(s)
- Yuji Oe
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Tetsuhiro Tanaka
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Sendai 980-0845, Japan
| |
Collapse
|
3
|
Shah H, Fairlie DP, Lim J. Protease-activated receptor 2: A promising therapeutic target for women's cancers. J Pharmacol Exp Ther 2025; 392:100016. [PMID: 39892996 DOI: 10.1124/jpet.124.002176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 01/22/2025] Open
Abstract
Cancers affecting women, such as breast, uterine, ovarian, endometrial, and cervical cancers, have become increasingly prevalent. The growing incidence and death rates associated with these cancers warrant the development of innovative and alternative approaches to current treatments. This article investigates the association of women's cancers with a molecular target known as protease-activated receptor 2 (PAR2), a G protein-coupled receptor that is expressed on the surface of cancer cells. Expression levels of the PAR2 gene were curated from publicly available databases, and PAR2 was found to be significantly overexpressed in tissues from patients with breast, uterine, ovarian, endometrial, or cervical cancer compared with normal tissues. PAR2 overexpression has been previously linked to tumor progression and, in some cases, tumor growth. Activation of PAR2 by either endogenous proteases or synthetic agonists triggers certain downstream intracellular signaling pathways that have been associated with tumor progression, cell migration and invasion, angiogenesis, and apoptosis of cancer cells. Although recent advances have led to identification of several PAR2 antagonists, none has yet been developed for human use. Additionally, PAR2 inhibition has been shown to increase the efficacy of chemotherapeutic drugs, allowing them to be potentially used at less toxic doses in combination therapies for cancer. The present work briefly summarizes the current status of PAR2 as a potential therapeutic target for treating women's cancers. SIGNIFICANCE STATEMENT: This article highlights potential roles for protease-activated receptor 2 (PAR2) in cancers affecting women. Overexpression of the PAR2 gene in women's cancers is associated with various oncogenic processes, such as tumor progression, cell migration, and invasion, ultimately contributing to poorer patient prognoses. Given the increasing incidence of women's cancers, there is an urgent need to develop novel therapeutic drugs, and PAR2 represents a promising target for developing new treatments.
Collapse
Affiliation(s)
- Himani Shah
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| | - Junxian Lim
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
4
|
Barbosa ED, Ma Y, Clift HE, Olson LJ, Zhu L, Liu W. Structural Insights into Dopamine Receptor-Ligand Interactions: From Agonists to Antagonists. ACS Chem Neurosci 2024; 15:4123-4131. [PMID: 39485723 DOI: 10.1021/acschemneuro.4c00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
This study explores the intricacies of dopamine receptor-ligand interactions, focusing on the D1R and D5R subtypes. Using molecular modeling techniques, we investigated the binding of the pan-agonist rotigotine, revealing a universal binding mode at the orthosteric binding pocket. Additionally, we analyze the stability of antagonist-receptor complexes with SKF83566 and SCH23390. By examining the impact of specific mutations on ligand-receptor interactions through computational simulations and thermostability assays, we gain insights into binding stability. Our research also delves into the structural and energetic aspects of antagonist binding to D1R and D5R in their inactive states. These findings enhance our understanding of dopamine receptor pharmacology and hold promise for drug development in central nervous system disorders, opening doors to future research and innovation in this field.
Collapse
MESH Headings
- Dopamine Agonists/pharmacology
- Dopamine Agonists/chemistry
- Humans
- Ligands
- Dopamine Antagonists/pharmacology
- Dopamine Antagonists/chemistry
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D1/agonists
- Tetrahydronaphthalenes/pharmacology
- Tetrahydronaphthalenes/chemistry
- Receptors, Dopamine D5/agonists
- Receptors, Dopamine D5/metabolism
- Thiophenes/pharmacology
- Thiophenes/chemistry
- Protein Binding/physiology
- Binding Sites
- Benzazepines/pharmacology
- Benzazepines/chemistry
- Models, Molecular
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives
Collapse
Affiliation(s)
- Emmanuel D Barbosa
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Yuanyuan Ma
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Heather E Clift
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Linda J Olson
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Lan Zhu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Wei Liu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
5
|
Fan M, Fan X, Lai Y, Chen J, Peng Y, Peng Y, Xiang L, Ma Y. Protease-Activated Receptor 2 in inflammatory skin disease: current evidence and future perspectives. Front Immunol 2024; 15:1448952. [PMID: 39301020 PMCID: PMC11410643 DOI: 10.3389/fimmu.2024.1448952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Protease-activated receptor-2 (PAR2) is a class-A G protein-coupled receptor (GPCR) activated by serine proteases and is expressed by multiple tissues, including the skin. PAR2 is involved in the skin inflammatory response, promoting Th2 inflammation, delaying skin barrier repair, and affecting the differentiation of keratinocytes. It also participates in the transmission of itch and pain sensations in the skin. Increasing evidence indicates that PAR2 plays an important role in the pathogenesis of inflammatory skin diseases such as acne vulgaris, rosacea, psoriasis, and atopic dermatitis. Additional focus will be placed on potential targeted therapies based on PAR2. The Goal of this review is to outline the emerging effects of PAR2 activation in inflammatory skin disease and highlight the promise of PAR2 modulators.
Collapse
Affiliation(s)
- Mengjie Fan
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Xiaoyao Fan
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Yangfan Lai
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Jin Chen
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Yifan Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Leihong Xiang
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| | - Ying Ma
- Department of Dermatology, Huashan Hosptial, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Jallouli R, Moreno Salinas AL, Laniel A, Holleran B, Avet C, Jacob J, Hoang T, Lavoie C, Carmon KS, Bouvier M, Leduc R. G protein selectivity profile of GPR56/ADGRG1 and its effect on downstream effectors. RESEARCH SQUARE 2024:rs.3.rs-4869264. [PMID: 39281883 PMCID: PMC11398566 DOI: 10.21203/rs.3.rs-4869264/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
GPR56, an adhesion G-protein coupled receptor (aGPCRs) with constitutive and ligand-promoted activity, is involved in many physiological and pathological processes. Whether the receptor's constitutive or ligand-promoted activation occur through the same molecular mechanism, and whether different activation modes lead to functional selectivity between G proteins is unknown. Here we show that GPR56 constitutively activates both G12 and G13. Unlike constitutive activation and activation with 3-a-acetoxydihydrodeoxygedunin (3αDOG), stimulation with an antibody, 10C7, directed against GPR56's extracellular domain (ECD) led to an activation that favors G13 over G12. An autoproteolytically deficient mutant, GPR56-T383A, was also activated by 10C7 indicating that the tethered agonist (TA) exposed through autocatalytic cleavage, is not required for this activation modality. In contrast, this proteolysis-resistant mutant could not be activated by 3αDOG indicating different modes of activation by the two ligands. We show that an N-terminal truncated GPR56 construct (GPR56-Δ1-385) is devoid of constitutive activity but was activated by 3αDOG. Similarly to 3αDOG, 10C7 promoted the recruitment of b-arrestin-2 but GPR56 internalization was β-arrestin independent. Despite the slow activation mode of 10C7 that favors G13 over G12, it efficiently activated the downstream Rho pathway in BT-20 breast cancer cells. These data show that different GPR56 ligands have different modes of activation yielding differential G protein selectivity but converging on the activation of the Rho pathway both in heterologous expressions system and in cancer cells endogenously expressing the receptor. 10C7 is therefore an interesting tool to study both the processes underlying GPR56 activity and its role in cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Joan Jacob
- The University of Texas MD Anderson Cancer Center
| | - Trang Hoang
- University of Montreal: Universite de Montreal
| | | | | | | | | |
Collapse
|
7
|
Jallouli R, Moreno-Salinas AL, Laniel A, Holleran B, Avet C, Jacob J, Hoang T, Lavoie C, Carmon KS, Bouvier M, Leduc R. G protein selectivity profile of GPR56/ADGRG1 and its effect on downstream effectors. Cell Mol Life Sci 2024; 81:383. [PMID: 39231834 PMCID: PMC11374949 DOI: 10.1007/s00018-024-05416-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/08/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024]
Abstract
GPR56, an adhesion G-protein coupled receptor (aGPCRs) with constitutive and ligand-promoted activity, is involved in many physiological and pathological processes. Whether the receptor's constitutive or ligand-promoted activation occur through the same molecular mechanism, and whether different activation modes lead to functional selectivity between G proteins is unknown. Here we show that GPR56 constitutively activates both G12 and G13. Unlike constitutive activation and activation with 3-α-acetoxydihydrodeoxygedunin (3αDOG), stimulation with an antibody, 10C7, directed against GPR56's extracellular domain (ECD) led to an activation that favors G13 over G12. An autoproteolytically deficient mutant, GPR56-T383A, was also activated by 10C7 indicating that the tethered agonist (TA) exposed through autocatalytic cleavage, is not required for this activation modality. In contrast, this proteolysis-resistant mutant could not be activated by 3αDOG indicating different modes of activation by the two ligands. We show that an N-terminal truncated GPR56 construct (GPR56-Δ1-385) is devoid of constitutive activity but was activated by 3αDOG. Similarly to 3αDOG, 10C7 promoted the recruitment of β-arrestin-2 but GPR56 internalization was β-arrestin independent. Despite the slow activation mode of 10C7 that favors G13 over G12, it efficiently activated the downstream Rho pathway in BT-20 breast cancer cells. These data show that different GPR56 ligands have different modes of activation yielding differential G protein selectivity but converging on the activation of the Rho pathway both in heterologous expressions system and in cancer cells endogenously expressing the receptor. 10C7 is therefore an interesting tool to study both the processes underlying GPR56 activity and its role in cancer cells.
Collapse
Affiliation(s)
- Raida Jallouli
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Ana L Moreno-Salinas
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andréanne Laniel
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Brian Holleran
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Charlotte Avet
- Institute for Research in Immunology and Cancer (IRIC), Department of Pharmacology and Physiology, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada
| | - Joan Jacob
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Trang Hoang
- Institute for Research in Immunology and Cancer (IRIC), Department of Pharmacology and Physiology, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada
| | - Christine Lavoie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kendra S Carmon
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC), Department of Pharmacology and Physiology, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada.
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.
- Institut de Pharmacologie de Sherbrooke, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
8
|
McDuffie EL, Panettieri RA, Scott CP. G 12/13 signaling in asthma. Respir Res 2024; 25:295. [PMID: 39095798 PMCID: PMC11297630 DOI: 10.1186/s12931-024-02920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.
Collapse
Affiliation(s)
- Elizabeth L McDuffie
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Charles P Scott
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Nag JK, Appasamy P, Malka H, Sedley S, Bar-Shavit R. New Target(s) for RNF43 Regulation: Implications for Therapeutic Strategies. Int J Mol Sci 2024; 25:8083. [PMID: 39125653 PMCID: PMC11311281 DOI: 10.3390/ijms25158083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer cells depend on specific oncogenic pathways or present a genetic alteration that leads to a particular disturbance. Still, personalized and targeted biological therapy remains challenging, with current efforts generally yielding disappointing results. Carefully assessing onco-target molecular pathways can, however, potently assist with such efforts for the selection of patient populations that would best respond to a given drug treatment. RNF43, an E3 ubiquitin ligase that negatively regulates Wnt/frizzled (FZD) receptors by their ubiquitination, internalization, and degradation, controls a key pathway in cancer. Recently, additional target proteins of RNF43 were described, including p85 of the PI3K/AKT/mTOR signaling pathway and protease-activated receptor 2 (PAR2), a G-protein-coupled receptor that potently induces β-catenin stabilization, independent of Wnts. RNF43 mutations with impaired E3 ligase activity were found in several types of cancers (e.g., gastrointestinal system tumors and endometrial and ovarian cancer), pointing to a high dependency on FZD receptors and possibly PAR2 and the PI3K/AKT/mTOR signaling pathway. The development of drugs toward these targets is essential for improved treatment of cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Rachel Bar-Shavit
- Sharett Institute of Oncology, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel; (J.K.N.); (P.A.); (H.M.); (S.S.)
| |
Collapse
|
10
|
McCullock TW, Cardani LP, Kammermeier PJ. Signaling Specificity and Kinetics of the Human Metabotropic Glutamate Receptors. Mol Pharmacol 2024; 105:104-115. [PMID: 38164584 PMCID: PMC10794986 DOI: 10.1124/molpharm.123.000795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 11/08/2023] [Indexed: 01/03/2024] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are obligate dimer G protein coupled receptors that can all function as homodimers. Here, each mGluR homodimer was examined for its G protein coupling profile using a bioluminescence resonance energy transfer-based assay that detects the interaction between a split YFP-tagged Gβ 1γ2 and a Nanoluciferase tagged free Gβγ sensor, MAS-GRK3-ct- nanoluciferase with 14 specific Gα proteins heterologously expressed, representing each family. Canonically, the group II and III mGluRs (2 and 3 and 4, 6, 7, and 8, respectively) are thought to couple to Gi/o exclusively. In addition, the group I mGluRs (1 and 5) are known to couple to the Gq/11 family and generally thought to also couple to the pertussis toxin-sensitive Gi/o family some reports have suggested Gs coupling is possible as cAMP elevations have been noted. In this study, coupling was observed with all eight mGluRs through the Gi/o proteins and only mGluR1 and mGluR5 through Gq/11, and, perhaps surprisingly, not G14 None activated any Gs protein. Interestingly, coupling was seen with the group I and II but not the group III mGluRs to G16 Slow but significant coupling to Gz was also seen with the group II receptors. SIGNIFICANCE STATEMENT: Metabotropic glutamate receptor (mGluR)-G protein coupling has not been thoroughly examined, and some controversy remains about whether some mGluRs can activate Gαs family members. Here we examine the ability of each mGluR to activate representative members of every Gα protein family. While all mGluRs can activate Gαi/o proteins, only the group I mGluRs couple to Gαq/11, and no members of the family can activate Gαs family members, including the group I receptors alone or with positive allosteric modulators.
Collapse
Affiliation(s)
- Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Loren P Cardani
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
11
|
Barbosa E, Clift H, Olson L, Zhu L, Liu W. Structural Insights into Dopamine Receptor-Ligand Interactions: From Agonists to Antagonists. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565579. [PMID: 37961276 PMCID: PMC10635143 DOI: 10.1101/2023.11.03.565579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
This study explores the intricacies of dopamine receptor-ligand interactions, focusing on the D1R and D5R subtypes. Using molecular modeling techniques, we investigate the binding of the pan-agonist rotigotine, revealing a universal binding mode at the orthosteric binding pocket (OBP). Additionally, we analyze the stability of antagonist-receptor complexes with SKF83566 and SCH23390. By examining the impact of specific mutations on ligand-receptor interactions through computational simulations and thermostability assays, we gain insights into binding stability. Our research also delves into the structural and energetic aspects of antagonist binding to D1R and D5R in their inactive states. These findings enhance our understanding of dopamine receptor pharmacology and hold promise for drug development in central nervous system disorders, opening doors to future research and innovation in this field.
Collapse
|
12
|
McCullock TW, Cardani LP, Kammermeier PJ. Signaling specificity and kinetics of the human metabotropic glutamate receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550373. [PMID: 37546908 PMCID: PMC10402105 DOI: 10.1101/2023.07.24.550373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are obligate dimer G protein coupled receptors that can all function as homodimers. Here, each mGluR homodimer was examined for its G protein coupling profile using a BRET based assay that detects the interaction between a split YFP-tagged Gβ1γ2 and a Nanoluc tagged free Gβγ sensor, MAS-GRK3-ct-NLuc with 14 specific Ga proteins heterologously expressed, representing each family. Canonically, the group II and III mGluRs (2&3, and 4, 6, 7&8, respectively) are thought to couple to Gi/o exclusively. In addition, the group I mGluRs (1&5) are known to couple to the Gq/11 family, and generally thought to also couple to the PTX-sensitive Gi/o family; some reports have suggested Gs coupling is possible as cAMP elevations have been noted. In this study, coupling was observed with all 8 mGluRs through the Gi/o proteins, and only mGluR1&5 through Gq/11, and perhaps surprisingly, not G14. None activated any Gs protein. Interestingly, coupling was seen with the group I and II, but not the group III mGluRs to G16. Slow but significant coupling to Gz was also seen with the group II receptors.
Collapse
Affiliation(s)
- Tyler W. McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Loren P. Cardani
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Paul J. Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
13
|
Abdel-Bakky MS, Aldakhili ASA, Ali HM, Babiker AY, Alhowail AH, Mohammed SAA. Evaluation of Cisplatin-Induced Acute Renal Failure Amelioration Using Fondaparinux and Alteplase. Pharmaceuticals (Basel) 2023; 16:910. [PMID: 37513824 PMCID: PMC10383028 DOI: 10.3390/ph16070910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023] Open
Abstract
Acute renal failure (ARF) is a deleterious condition with increased mortality or healthcare costs or dialysis-dependent end-stage renal disease. The study aims to compare prophylaxis with fondaparinux (Fund) vs. treatment with alteplase (Alt) in ameliorating cisplatin (Cis)-induced ARF. Sixty male mice were equally divided randomly into six groups of control, Cis, Alt, and Cis + Alt groups receiving normal saline for 10 days. All four groups except for the control received Cis (30 mg/kg, i.p.) on day 7, and 6 h later, both the Alt groups received Alt (0.9 mg/kg, i.v.). The animal groups Fund and Fund + Cis received Fund (5 mg/kg, i.p.) for 10 days, and the Fund + Cis group on day 7 received Cis. All the animal groups were euthanized 72 h after the Cis dose. The Fund + Cis group showed significantly increased expression levels of platelet count, retinoid X receptor alpha (RXR-α) and phosphorylated Akt (p-Akt) in addition to decreased levels of urea, blood urea nitrogen (BUN), uric acid, white blood cells (WBCs), red blood cells (RBCs), relative kidney body weight, kidney injury score, glucose, prothrombin (PT), A Disintegrin And Metalloproteinases-10 (ADAM10), extracellular matrix deposition, protease-activated receptor 2 (PAR-2), and fibrinogen expression when compared to the Cis-only group. Meanwhile, the Cis + Alt group showed increased caspase-3 expression in addition to decreased levels of urea, BUN, uric acid, WBCs, RBCs, glucose, platelet count and PT expression with a marked decrease in PAR-2 protein expression compared to the Cis group. The creatinine levels for both the Fund + Cis and Cis + Alt groups were found to be comparable to those of the Cis-only group. The results demonstrate that the coagulation system's activation through the stimulation of PAR-2 and fibrinogen due to Cis-induced ADAM10 protein expression mediated the apoptotic pathway, as indicated by caspase-3 expression through the p-Akt pathway. This is normally accompanied by the loss of RXR-α distal and proximal tubules as lipid droplets. When the animals were pre-treated with the anticoagulant, Fund, the previous deleterious effect was halted while the fibrinolytic agent, Alt, most of the time failed to treat Cis-induced toxicity.
Collapse
Affiliation(s)
- Mohamed S Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Anas S A Aldakhili
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Hussein M Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
- Department of Biochemistry, Faculty of Medicine, Al-Azhar University, Assiut 71524, Egypt
| | - Ali Y Babiker
- Department of Medical Laboratories, College of Applied Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Ahmad H Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Salman A A Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
14
|
Gaitonde SA, Bouvier M. Enhanced Bystander BRET (ebBRET) Biosensors as Biophysical Tools to Map the Signaling Profile of Neuropsychiatric Drugs Targeting GPCRs. Methods Mol Biol 2023; 2687:15-30. [PMID: 37464159 DOI: 10.1007/978-1-0716-3307-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Bioluminescence resonance energy transfer (BRET) is a non-radiative energy transfer between a bioluminescent donor and a fluorescent acceptor with far-reaching applications in detecting physiologically relevant protein-protein interactions. The recently developed enhanced bystander BRET (ebBRET) biosensors have made it possible to rapidly determine the signaling profile of a series of ligands across a large number of GPCRs and their signaling repertoires, which has tremendous implications in the drug discovery process. Here we describe BRET and the ebBRET biosensors as investigational tools in establishing functional selectivity downstream of GPCRs.
Collapse
Affiliation(s)
- Supriya A Gaitonde
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada.
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
15
|
Hussain Z, Park H. Inflammation and Impaired Gut Physiology in Post-operative Ileus: Mechanisms and the Treatment Options. J Neurogastroenterol Motil 2022; 28:517-530. [PMID: 36250359 PMCID: PMC9577567 DOI: 10.5056/jnm22100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/08/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022] Open
Abstract
Post-operative ileus (POI) is the transient cessation of coordinated gastrointestinal motility after abdominal surgical intervention. It decreases quality of life, prolongs length of hospital stay, and increases socioeconomic costs. The mechanism of POI is complex and multifactorial, and has been broadly categorized into neurogenic and inflammatory phase. Neurogenic phase mediated release of corticotropin-releasing factor (CRF) plays a central role in neuroinflammation, and affects both central autonomic response as well hypothalamic-pituitary-adrenal (HPA) axis. HPA-stress axis associated cortisol release adversely affects gut microbiota and permeability. Peripheral CRF (pCRF) is a key player in stress induced gastric emptying and colonic transit. It functions as a local effector and interacts with the CRF receptors on the mast cell to release chemical mediators of inflammation. Mast cells proteases disrupt epithelial barrier via protease activated receptor-2 (PAR-2). PAR-2 facilitates cytoskeleton contraction to reorient tight junction proteins such as occludin, claudins, junctional adhesion molecule, and zonula occludens-1 to open epithelial barrier junctions. Barrier opening affects the selectivity, and hence permeation of luminal antigens and solutes in the gastrointestinal tract. Translocation of luminal antigens perturbs mucosal immune system to further exacerbate inflammation. Stress induced dysbiosis and decrease in production of short chain fatty acids add to the inflammatory response and barrier disintegration. This review discusses potential mechanisms and factors involved in the pathophysiology of POI with special reference to inflammation and interlinked events such as epithelial barrier dysfunction and dysbiosis. Based on this review, we recommend CRF, mast cells, macrophages, and microbiota could be targeted concurrently for efficient POI management.
Collapse
Affiliation(s)
- Zahid Hussain
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Kurose H. [Biased Signaling through G Protein-coupled Receptors]. YAKUGAKU ZASSHI 2022; 142:1091-1101. [PMID: 36184444 DOI: 10.1248/yakushi.22-00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is well-established that G protein-coupled receptors (GPCRs) transduce signals into cells using G proteins as intermediary molecules. β-Arrestins are molecules involved in regulating GPCRs; however, it has recently been reported that β-arrestins can also mediate signaling through GPCRs. Signaling through G proteins or β-arrestins can be activated selectively using specific agonists; of the latter, those that can selectively activate either G proteins or β-arrestins are called biased agonists. The clinical use of biased agonists could potentially induce fewer side effects. However, partial agonists can also explain the mechanism of G protein-biased agonists; thus, appropriate assay systems must be considered. Endogenous agonists are known to bind to orthosteric and allosteric sites in the agonist binding site, and the allosteric site is associated with the activity of biased agonists. This current review presents a detailed discussion of biased agonists.
Collapse
Affiliation(s)
- Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University
| |
Collapse
|
17
|
Abstract
It has been 30 years since the first member of the protease-activated receptor (PAR) family was discovered. This was followed by the discovery of three other receptors, including PAR2. PAR2 is a G protein-coupled receptor activated by trypsin site-specific proteolysis. The process starts with serine proteases acting between arginine and serine, creating an N-terminus that functions as a tethered ligand that binds, after a conformational change, to the second extracellular loop of the receptor, leading to activation of G-proteins. The physiological and pathological functions of this ubiquitous receptor are still elusive. This review focuses on PAR2 activation and its distribution under physiological and pathological conditions, with a particular focus on the pancreas, a significant producer of trypsin, which is the prototype activator of the receptor. The role in acute or chronic pancreatitis, pancreatic cancer, and diabetes mellitus will be highlighted.
Collapse
Affiliation(s)
- Petr SUHAJ
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, Prague, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas OLEJAR
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, Prague, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radoslav MATEJ
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, Prague, Czech Republic,Department of Pathology, University Hospital Kralovske Vinohrady, Prague, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
18
|
Sedki D, Cho A, Cao Y, Nikolajev L, Atmuri NDP, Lubell WD, Laporte SA. Prostaglandin F2α and angiotensin II type 1 receptors exhibit differential cognate G protein coupling regulation. J Biol Chem 2022; 298:102294. [PMID: 35872018 PMCID: PMC9418914 DOI: 10.1016/j.jbc.2022.102294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Promiscuous G protein–coupled receptors (GPCRs) engage multiple Gα subtypes with different efficacies to propagate signals in cells. A mechanistic understanding of Gα selectivity by GPCRs is critical for therapeutic design, since signaling can be restrained by ligand–receptor complexes to preferentially engage specific G proteins. However, details of GPCR selectivity are unresolved. Here, we investigated cognate G protein selectivity using the prototypical promiscuous Gαq/11 and Gα12/13 coupling receptors, angiotensin II type I receptor (AT1R) and prostaglandin F2α receptor (FP), bioluminescence resonance energy transfer–based G protein and pathway-selective sensors, and G protein knockout cells. We determined that competition between G proteins for receptor binding occurred in a receptor- and G protein–specific manner for AT1R and FP but not for other receptors tested. In addition, we show that while Gα12/13 competes with Gαq/11 for AT1R coupling, the opposite occurs for FP, and Gαq-mediated signaling regulated G protein coupling only at AT1R. In cells, the functional modulation of biased ligands at FP and AT1R was contingent upon cognate Gα availability. The efficacy of AT1R-biased ligands, which poorly signal through Gαq/11, increased in the absence of Gα12/13. Finally, we show that a positive allosteric modulator of Gαq/11 signaling that also allosterically decreases FP–Gα12/13 coupling, lost its negative modulation in the absence of Gαq/11 coupling to FP. Together, our findings suggest that despite preferential binding of similar subsets of G proteins, GPCRs follow distinct selectivity rules, which may contribute to the regulation of ligand-mediated G protein bias of AT1R and FP.
Collapse
Affiliation(s)
- Dana Sedki
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Aaron Cho
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Yubo Cao
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ljiljana Nikolajev
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - N D Prasad Atmuri
- Department of Chemistry, University of Montréal, Montréal, Québec H3A 0B8, Canada
| | - William D Lubell
- Department of Chemistry, University of Montréal, Montréal, Québec H3A 0B8, Canada
| | - Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
19
|
Abstract
Agonists are defined as the ligands that activate intracellular signaling and evoke cellular responses. Synthetic and endogenous agonists should bind specific amino acids to activate G protein-coupled receptor (GPCR). Agonists that induce maximal responses are full agonists. Partial agonists cannot induce full responses unlike full agonists. In definition, antagonists inhibit agonist-stimulated responses by binding to orthosteric or allosteric sites. Antagonists modulate agonist-induced responses and are often related with inverse agonist activity. However, the relationship between antagonists and partial agonists is complex. An antagonist behaves as a partial agonist when the constitutive activity of the GPCR is high. In contrast, a partial agonist with very weak intrinsic activity may be classified as an antagonist. Thus, antagonisms of the compounds are influenced by constitutive activity of GPCRs, intrinsic activity and differences in the binding sites of GPCRs. Since "antagonism" has been revealed to have multiple aspects and more complex than previously thought, it may be difficult to classify each compound as simply "agonist" or "antagonist" as before. In this review, we discuss the recent findings and perspectives on the pharmacology of GPCR-binding antagonists, inverse agonists, and signaling.
Collapse
Affiliation(s)
- Hitoshi Kurose
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Sang Geon Kim
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul
| |
Collapse
|
20
|
Rivas CM, Yee MC, Addison KJ, Lovett M, Pal K, Ledford JG, Dussor G, Price TJ, Vagner J, DeFea KA, Boitano S. Proteinase-activated receptor-2 antagonist C391 inhibits Alternaria-induced airway epithelial signalling and asthma indicators in acute exposure mouse models. Br J Pharmacol 2022; 179:2208-2222. [PMID: 34841515 DOI: 10.1111/bph.15745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/19/2021] [Accepted: 11/04/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Despite the availability of a variety of treatment options, many asthma patients have poorly controlled disease with frequent exacerbations. Proteinase-activated receptor-2 (PAR2) has been identified in preclinical animal models as important to asthma initiation and progression following allergen exposure. Proteinase activation of PAR2 raises intracellular Ca2+ , inducing MAPK and β-arrestin signalling in the airway, leading to inflammatory and protective effects. We have developed C391, a potent PAR2 antagonist effective in blocking peptidomimetic- and trypsin-induced PAR2 signalling in vitro as well as reducing inflammatory PAR2-associated pain in vivo. We hypothesized that PAR2 antagonism by C391 would attenuate allergen-induced acutely expressed asthma indicators in murine models. EXPERIMENTAL APPROACH We evaluated the ability of C391 to alter Alternaria alternata-induced PAR2 signalling pathways in vitro using a human airway epithelial cell line that naturally expresses PAR2 (16HBE14o-) and a transfected embryonic cell line (HEK 293). We next evaluated the ability for C391 to reduce A. alternata-induced acutely expressed asthma indicators in vivo in two murine strains. KEY RESULTS C391 blocked A. alternata-induced, PAR2-dependent Ca2+ and MAPK signalling in 16HBE14o- cells, as well as β-arrestin recruitment in HEK 293 cells. C391 effectively attenuated A. alternata-induced inflammation, mucus production, mucus cell hyperplasia and airway hyperresponsiveness in acute allergen-challenged murine models. CONCLUSIONS AND IMPLICATIONS To our best knowledge, this is the first demonstration of pharmacological intervention of PAR2 to reduce allergen-induced asthma indicators in vivo. These data support further development of PAR2 antagonists as potential first-in-class allergic asthma drugs.
Collapse
Affiliation(s)
- Candy M Rivas
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA.,Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Michael C Yee
- Biomedical Sciences, University of California Riverside, Riverside, California, USA
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Marissa Lovett
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
| | - Kasturi Pal
- Biomedical Sciences, University of California Riverside, Riverside, California, USA
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| | - Josef Vagner
- Bio5 Collaborative Research Institute, University of Arizona, Tucson, Arizona, USA
| | - Kathryn A DeFea
- Biomedical Sciences, University of California Riverside, Riverside, California, USA.,Corporate Headquarters, PARMedics, Inc., Temecula, California, USA
| | - Scott Boitano
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA.,Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA.,Bio5 Collaborative Research Institute, University of Arizona, Tucson, Arizona, USA.,Department of Physiology, University of Arizona Health Sciences, Tucson, Arizona, USA
| |
Collapse
|
21
|
Avet C, Mancini A, Breton B, Le Gouill C, Hauser AS, Normand C, Kobayashi H, Gross F, Hogue M, Lukasheva V, St-Onge S, Carrier M, Héroux M, Morissette S, Fauman EB, Fortin JP, Schann S, Leroy X, Gloriam DE, Bouvier M. Effector membrane translocation biosensors reveal G protein and βarrestin coupling profiles of 100 therapeutically relevant GPCRs. eLife 2022; 11:74101. [PMID: 35302493 PMCID: PMC9005190 DOI: 10.7554/elife.74101] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/17/2022] [Indexed: 11/25/2022] Open
Abstract
The recognition that individual GPCRs can activate multiple signaling pathways has raised the possibility of developing drugs selectively targeting therapeutically relevant ones. This requires tools to determine which G proteins and βarrestins are activated by a given receptor. Here, we present a set of BRET sensors monitoring the activation of the 12 G protein subtypes based on the translocation of their effectors to the plasma membrane (EMTA). Unlike most of the existing detection systems, EMTA does not require modification of receptors or G proteins (except for Gs). EMTA was found to be suitable for the detection of constitutive activity, inverse agonism, biased signaling and polypharmacology. Profiling of 100 therapeutically relevant human GPCRs resulted in 1500 pathway-specific concentration-response curves and revealed a great diversity of coupling profiles ranging from exquisite selectivity to broad promiscuity. Overall, this work describes unique resources for studying the complexities underlying GPCR signaling and pharmacology.
Collapse
Affiliation(s)
- Charlotte Avet
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | | | - Billy Breton
- Domain Therapeutics North America, Montréal, Canada
| | - Christian Le Gouill
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | | | | | - Hiroyuki Kobayashi
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | | | - Mireille Hogue
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | - Viktoriya Lukasheva
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | - Stéphane St-Onge
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | - Marilyn Carrier
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | - Madeleine Héroux
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| | | | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Cambridge, United States
| | | | | | | | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montréal, Canada
| |
Collapse
|
22
|
Guo P, Tai Y, Wang M, Sun H, Zhang L, Wei W, Xiang YK, Wang Q. Gα 12 and Gα 13: Versatility in Physiology and Pathology. Front Cell Dev Biol 2022; 10:809425. [PMID: 35237598 PMCID: PMC8883321 DOI: 10.3389/fcell.2022.809425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/17/2022] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs), as the largest family of receptors in the human body, are involved in the pathological mechanisms of many diseases. Heterotrimeric G proteins represent the main molecular switch and receive cell surface signals from activated GPCRs. Growing evidence suggests that Gα12 subfamily (Gα12/13)-mediated signaling plays a crucial role in cellular function and various pathological processes. The current research on the physiological and pathological function of Gα12/13 is constantly expanding, Changes in the expression levels of Gα12/13 have been found in a wide range of human diseases. However, the mechanistic research on Gα12/13 is scattered. This review briefly describes the structural sequences of the Gα12/13 isoforms and introduces the coupling of GPCRs and non-GPCRs to Gα12/13. The effects of Gα12/13 on RhoA and other signaling pathways and their roles in cell proliferation, migration, and immune cell function, are discussed. Finally, we focus on the pathological impacts of Gα12/13 in cancer, inflammation, metabolic diseases, fibrotic diseases, and circulatory disorders are brought to focus.
Collapse
Affiliation(s)
- Paipai Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Manman Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Hanfei Sun
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA, United States.,VA Northern California Health Care System, Mather, CA, United States
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
23
|
Pera T, Loblundo C, Penn RB. Pharmacological Management of Asthma and COPD. COMPREHENSIVE PHARMACOLOGY 2022:762-802. [DOI: 10.1016/b978-0-12-820472-6.00095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|