1
|
Quon T, Lin LC, Ganguly A, Hudson BD, Tobin AB, Milligan G. Biased constitutive signaling of the G protein-coupled receptor GPR35 suppresses gut barrier permeability. J Biol Chem 2025; 301:108035. [PMID: 39615676 PMCID: PMC11732441 DOI: 10.1016/j.jbc.2024.108035] [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: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Agonist-independent, or constitutive, activity is an integral feature of G protein-coupled receptors, but its relevance in pathophysiological settings is generally poorly explored. GPR35 is a therapeutic target in inflammatory diseases of the lower gut. In colonic organoids from a human GPR35a-expressing transgenic mouse line, the GPR35 inverse agonist CID-2745687 increased barrier permeability substantially, indicating that constitutive receptor activity contributes to maintaining epithelial barrier integrity. High constitutive activity of GPR35 was also observed in both HT-29 and HEPG2 cells that express GPR35 endogenously. Mechanistic investigations in recombinant in vitro systems revealed that the constitutive activity of GPR35a was biased and not equivalent across signaling pathways. Hence, no constitutive interactions of the receptor with arrestin-adaptor proteins or activation of Gαo-containing G protein heterotrimers were detected while, even at low GPR35a expression levels, substantial constitutive activation of heterotrimers containing either Gα12 or Gα13 was observed. Similar biased constitutive activity was observed for the human GPR35b isoform. The extent of constitutive and agonist-mediated activity was dependent on receptor expression level. At high receptor levels, constitutive activation of Gα12 or Gα13 masked any agonist-induced effects while low expression levels with low constitutive activity allowed measurement of agonist-induced responses. These results highlight roles, selectivity, and the extent of constitutive activity of GPR35 in cells and tissues that express this receptor endogenously and highlight the contribution of its constitutive activity to maintaining the colonic epithelial barrier, potentially limiting the development of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Tezz Quon
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Li-Chiung Lin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Amlan Ganguly
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Brian D Hudson
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom.
| |
Collapse
|
2
|
Inverso D, Tacconi C, Ranucci S, De Giovanni M. The power of many: Multilevel targeting of representative chemokine and metabolite GPCRs in personalized cancer therapy. Eur J Immunol 2024; 54:e2350870. [PMID: 39263783 PMCID: PMC11628915 DOI: 10.1002/eji.202350870] [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: 03/28/2024] [Revised: 08/25/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
G protein-coupled receptors (GPCRs) are vital cell surface receptors that govern a myriad of physiological functions. Despite their crucial role in regulating antitumor immunity and tumorigenesis, therapeutic applications targeting GPCRs in oncology are currently limited. This review offers a focused examination of selected protumorigenic chemokine and metabolite-sensing GPCRs. Specifically, the review highlights five GPCRs able to orchestrate tumor immunobiology at three main levels: tumor immunity, cancer cell expansion, and blood vessel development. The review culminates by illuminating emerging therapies and discussing innovative strategies to harness the full potential of GPCR-targeted treatments, by applying a multireceptor and patient-specific logic.
Collapse
Affiliation(s)
- Donato Inverso
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Carlotta Tacconi
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Serena Ranucci
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Marco De Giovanni
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| |
Collapse
|
3
|
Gupta RA, Higham JP, Pearce A, Urriola-Muñoz P, Barker KH, Paine L, Ghooraroo J, Raine T, Hockley JRF, Rahman T, St John Smith E, Brown AJH, Ladds G, Suzuki R, Bulmer DC. GPR35 agonists inhibit TRPA1-mediated colonic nociception through suppression of substance P release. Pain 2024:00006396-990000000-00727. [PMID: 39382322 DOI: 10.1097/j.pain.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/25/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT The development of nonopioid analgesics for the treatment of abdominal pain is a pressing clinical problem. To address this, we examined the expression of Gi/o-coupled receptors, which typically inhibit nociceptor activation, in colonic sensory neurons. This led to the identification of the orphan receptor GPR35 as a visceral analgesic drug target because of its marked coexpression with transient receptor potential ankyrin 1 (TRPA1), a mediator of noxious mechanotransduction in the bowel. Building on in silico docking simulations, we confirmed that the mast cell stabiliser, cromolyn (CS), and phosphodiesterase inhibitor, zaprinast, are agonists at mouse GPR35, promoting the activation of different Gi/o subunits. Pretreatment with either CS or zaprinast significantly attenuated TRPA1-mediated colonic nociceptor activation and prevented TRPA1-mediated mechanosensitisation. These effects were lost in tissue from GPR35-/- mice and were shown to be mediated by inhibition of TRPA1-evoked substance P (SP) release. This observation highlights the pronociceptive effect of SP and its contribution to TRPA1-mediated colonic nociceptor activation and sensitisation. Consistent with this mechanism of action, we confirmed that TRPA1-mediated colonic contractions evoked by SP release were abolished by CS pretreatment in a GPR35-dependent manner. Our data demonstrate that GPR35 agonists prevent the activation and sensitisation of colonic nociceptors through the inhibition of TRPA1-mediated SP release. These findings highlight the potential of GPR35 agonists to deliver nonopioid analgesia for the treatment of abdominal pain.
Collapse
Affiliation(s)
- Rohit A Gupta
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Abigail Pearce
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Paulina Urriola-Muñoz
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Katie H Barker
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Luke Paine
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Joshua Ghooraroo
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - James R F Hockley
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Alastair J H Brown
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Rie Suzuki
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| |
Collapse
|
4
|
Takkar S, Sharma G, Kaushal JB, Abdullah KM, Batra SK, Siddiqui JA. From orphan to oncogene: The role of GPR35 in cancer and immune modulation. Cytokine Growth Factor Rev 2024; 77:56-66. [PMID: 38514303 DOI: 10.1016/j.cytogfr.2024.03.004] [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: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
G protein-coupled receptors (GPCRs) are well-studied and the most traceable cell surface receptors for drug discovery. One of the intriguing members of this family is G protein-coupled receptors 35 (GPR35), which belongs to the class A rhodopsin-like family of GPCRs identified over two decades ago. GPR35 presents interesting features such as ubiquitous expression and distinct isoforms. Moreover, functional and genome-wide association studies on its widespread expression have linked GPR35 with pathophysiological disease progression. Various pieces of evidence have been accumulated regarding the independent or endogenous ligand-dependent role of GPR35 in cancer progression and metastasis. In the current scenario, the relationship of this versatile receptor and its putative endogenous ligands for the activation of oncogenic signal transduction pathways at the cellular level is an active area of research. These intriguing features offered by GPR35 make it an oncological target, justifying its uniqueness at the physiological and pathophysiological levels concerning other GPCRs. For pharmacologically targeting receptor-induced signaling, few potential competitive antagonists have been discovered that offer high selectivity at a human level. In addition to its fascinating features, targeting GPR35 at rodent and human orthologue levels is distinct, thus contributing to the sub-species selectivity. Strategies to modulate these issues will help us understand and truly target GPR35 at the therapeutic level. In this article, we have provided prospects on each topic mentioned above and suggestions to overcome the challenges. This review discusses the molecular mechanism and signal transduction pathways activated by endogenous ligands or spontaneous auto-activation of GPR35 that contributes towards disease progression. Furthermore, we have highlighted the GPR35 structure, ubiquitous expression, its role in immunomodulation, and at the pathophysiological level, especially in cancer, indicating its status as a versatile receptor. Subsequently, we discussed the various proposed ligands and their mechanism of interaction with GPR35. Additionally, we have summarized the GPR35 antagonist that provides insights into the opportunities for therapeutically targeting this receptor.
Collapse
Affiliation(s)
- Simran Takkar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gunjan Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - K M Abdullah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
5
|
Wu Y, Zhang P, Fan H, Zhang C, Yu P, Liang X, Chen Y. GPR35 acts a dual role and therapeutic target in inflammation. Front Immunol 2023; 14:1254446. [PMID: 38035084 PMCID: PMC10687457 DOI: 10.3389/fimmu.2023.1254446] [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: 07/07/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
GPR35 is a G protein-coupled receptor with notable involvement in modulating inflammatory responses. Although the precise role of GPR35 in inflammation is not yet fully understood, studies have suggested that it may have both pro- and anti-inflammatory effects depending on the specific cellular environment. Some studies have shown that GPR35 activation can stimulate the production of pro-inflammatory cytokines and facilitate the movement of immune cells towards inflammatory tissues or infected areas. Conversely, other investigations have suggested that GPR35 may possess anti-inflammatory properties in the gastrointestinal tract, liver and certain other tissues by curbing the generation of inflammatory mediators and endorsing the differentiation of regulatory T cells. The intricate role of GPR35 in inflammation underscores the requirement for more in-depth research to thoroughly comprehend its functional mechanisms and its potential significance as a therapeutic target for inflammatory diseases. The purpose of this review is to concurrently investigate the pro-inflammatory and anti-inflammatory roles of GPR35, thus illuminating both facets of this complex issue.
Collapse
Affiliation(s)
- Yetian Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Pei Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Hongjie Fan
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Caiying Zhang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Pengfei Yu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yang Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| |
Collapse
|
6
|
Ganguly A, Quon T, Jenkins L, Joseph B, Al-Awar R, Chevigne A, Tobin AB, Uehling DE, Hoffmann C, Drube J, Milligan G. G protein-receptor kinases 5/6 are the key regulators of G protein-coupled receptor 35-arrestin interactions. J Biol Chem 2023; 299:105218. [PMID: 37660910 PMCID: PMC10520886 DOI: 10.1016/j.jbc.2023.105218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Human G protein-coupled receptor 35 is regulated by agonist-mediated phosphorylation of a set of five phospho-acceptor amino acids within its C-terminal tail. Alteration of both Ser300 and Ser303 to alanine in the GPR35a isoform greatly reduces the ability of receptor agonists to promote interactions with arrestin adapter proteins. Here, we have integrated the use of cell lines genome edited to lack expression of combinations of G protein receptor kinases (GRKs), selective small molecule inhibitors of subsets of these kinases, and antisera able to specifically identify either human GPR35a or mouse GPR35 only when Ser300 and Ser303 (orce; the equivalent residues in mouse GPR35) have become phosphorylated to demonstrate that GRK5 and GRK6 cause agonist-dependent phosphorylation of these residues. Extensions of these studies demonstrated the importance of the GRK5/6-mediated phosphorylation of these amino acids for agonist-induced internalization of the receptor. Homology and predictive modeling of the interaction of human GPR35 with GRKs showed that the N terminus of GRK5 is likely to dock in the same methionine pocket on the intracellular face of GPR35 as the C terminus of the α5 helix of Gα13 and, that while this is also the case for GRK6, GRK2 and GRK3 are unable to do so effectively. These studies provide unique and wide-ranging insights into modes of regulation of GPR35, a receptor that is currently attracting considerable interest as a novel therapeutic target in diseases including ulcerative colitis.
Collapse
Affiliation(s)
- Amlan Ganguly
- Centre for Translational Pharmacology, School of Molecular Biosciences, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Tezz Quon
- Centre for Translational Pharmacology, School of Molecular Biosciences, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Laura Jenkins
- Centre for Translational Pharmacology, School of Molecular Biosciences, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Babu Joseph
- Drug Discovery Program, Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario, Canada
| | - Andy Chevigne
- Division of Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - David E Uehling
- Drug Discovery Program, Ontario Institute for Cancer Research, MaRS Centre, Toronto, Ontario, Canada
| | - Carsten Hoffmann
- Institute for Molecular Cell Biology, CMB-Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Julia Drube
- Institute for Molecular Cell Biology, CMB-Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Graeme Milligan
- Centre for Translational Pharmacology, School of Molecular Biosciences, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK.
| |
Collapse
|
7
|
Im DS. Recent advances in GPR35 pharmacology; 5-HIAA serotonin metabolite becomes a ligand. Arch Pharm Res 2023:10.1007/s12272-023-01449-y. [PMID: 37227682 DOI: 10.1007/s12272-023-01449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
GPR35, an orphan receptor, has been waiting for its ligand since its cloning in 1998. Many endogenous and exogenous molecules have been suggested to act as agonists of GPR35 including kynurenic acid, zaprinast, lysophosphatidic acid, and CXCL17. However, complex and controversial responses to ligands among species have become a huge hurdle in the development of therapeutics in addition to the orphan state. Recently, a serotonin metabolite, 5-hydroxyindoleacetic acid (5-HIAA), is reported to be a high potency ligand for GPR35 by investigating the increased expression of GPR35 in neutrophils. In addition, a transgenic knock-in mouse line is developed, in which GPR35 was replaced with a human ortholog, making it possible not only to overcome the different selectivity of agonists among species but also to conduct therapeutic experiments on human GPR35 in mouse models. In the present article, I review the recent advances and prospective therapeutic directions in GPR35 research. Especially, I'd like to draw attention of readers to the finding of 5-HIAA as a ligand of GPR35 and lead to apply the 5-HIAA and human GPR35 knock-in mice to their research fields in a variety of pathophysiological conditions.
Collapse
Affiliation(s)
- Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences and Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02446, Republic of Korea.
- Laboratory of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
8
|
Milligan G. GPR35: from enigma to therapeutic target. Trends Pharmacol Sci 2023; 44:263-273. [PMID: 37002007 DOI: 10.1016/j.tips.2023.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 04/16/2023]
Abstract
The orphan G-protein-coupled receptor 35 (GPR35), although poorly characterised, is attracting considerable interest as a therapeutic target. Marked differences in pharmacology between human and rodent orthologues of the receptor and a dearth of antagonists with affinity for mouse and rat GPR35 have previously restricted the use of preclinical disease models. The development of improved ligands, novel transgenic knock-in mouse lines, and detailed analysis of the disease relevance of single-nucleotide polymorphisms (SNPs) have greatly enhanced understanding of the key roles of GPR35 and have stimulated efforts towards disease-targeted proof-of-concept studies. In this opinion article, new information on the biology of the receptor is considered, whilst insight into how GPR35 is currently being assessed for therapeutic utility - in areas ranging from inflammatory bowel diseases to nonalcoholic steatohepatitis and various cancers - is also provided.
Collapse
Affiliation(s)
- Graeme Milligan
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
9
|
Otkur W, Liu X, Chen H, Li S, Ling T, Lin H, Yang R, Xia T, Qi H, Piao HL. GPR35 antagonist CID-2745687 attenuates anchorage-independent cell growth by inhibiting YAP/TAZ activity in colorectal cancer cells. Front Pharmacol 2023; 14:1126119. [PMID: 37113762 PMCID: PMC10126512 DOI: 10.3389/fphar.2023.1126119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Background and purpose: GPR35, a member of the orphan G-protein-coupled receptor, was recently implicated in colorectal cancer (CRC). However, whether targeting GPR35 by antagonists can inhibit its pro-cancer role has yet to be answered. Experimental approach: We applied antagonist CID-2745687 (CID) in established GPR35 overexpressing and knock-down CRC cell lines to understand its anti-cell proliferation property and the underlying mechanism. Key results: Although GPR35 did not promote cell proliferation in 2D conditions, it promoted anchorage-independent growth in soft-agar, which was reduced by GPR35 knock-down and CID treatment. Furthermore, YAP/TAZ target genes were expressed relatively higher in GPR35 overexpressed cells and lower in GPR35 knock-down cells. YAP/TAZ activity is required for anchorage-independent growth of CRC cells. By detecting YAP/TAZ target genes, performing TEAD4 luciferase reporter assay, and examining YAP phosphorylation and TAZ protein expression level, we found YAP/TAZ activity is positively correlated to GPR35 expression level, which CID disrupted in GPR35 overexpressed cells, but not in GPR35 knock-down cells. Intriguingly, GPR35 agonists did not promote YAP/TAZ activity but ameliorated CID's inhibitory effect; GPR35-promoted YAP/TAZ activity was only partly attenuated by ROCK1/2 inhibitor. Conclusion and implications: GPR35 promoted YAP/TAZ activity partly through Rho-GTPase with its agonist-independent constitutive activity, and CID exhibited its inhibitory effect. GPR35 antagonists are promising anti-cancer agents that target hyperactivation and overexpression of YAP/TAZ in CRC.
Collapse
|
10
|
Schihada H, Klompstra TM, Humphrys LJ, Cervenka I, Dadvar S, Kolb P, Ruas JL, Schulte G. Isoforms of GPR35 have distinct extracellular N-termini that allosterically modify receptor-transducer coupling and mediate intracellular pathway bias. J Biol Chem 2022; 298:102328. [PMID: 35933013 PMCID: PMC9450150 DOI: 10.1016/j.jbc.2022.102328] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Within the intestine, the human G protein–coupled receptor (GPCR) GPR35 is involved in oncogenic signaling, bacterial infections, and inflammatory bowel disease. GPR35 is known to be expressed as two distinct isoforms that differ only in the length of their extracellular N-termini by 31 amino acids, but detailed insights into their functional differences are lacking. Through gene expression analysis in immune and gastrointestinal cells, we show that these isoforms emerge from distinct promoter usage and alternative splicing. Additionally, we employed optical assays in living cells to thoroughly profile both GPR35 isoforms for constitutive and ligand-induced activation and signaling of 10 different heterotrimeric G proteins, ligand-induced arrestin recruitment, and receptor internalization. Our results reveal that the extended N-terminus of the long isoform limits G protein activation yet elevates receptor–β-arrestin interaction. To better understand the structural basis for this bias, we examined structural models of GPR35 and conducted experiments with mutants of both isoforms. We found that a proposed disulfide bridge between the N-terminus and extracellular loop 3, present in both isoforms, is crucial for constitutive G13 activation, while an additional cysteine contributed by the extended N-terminus of the long GPR35 isoform limits the extent of agonist-induced receptor–β-arrestin2 interaction. The pharmacological profiles and mechanistic insights of our study provide clues for the future design of isoform-specific GPR35 ligands that selectively modulate GPR35–transducer interactions and allow for mechanism-based therapies against, for example, inflammatory bowel disease or bacterial infections of the gastrointestinal system.
Collapse
Affiliation(s)
- Hannes Schihada
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany.
| | - Thomas M Klompstra
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Laura J Humphrys
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Igor Cervenka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Shamim Dadvar
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
11
|
Agonist-induced phosphorylation of orthologues of the orphan receptor GPR35 functions as an activation sensor. J Biol Chem 2022; 298:101655. [PMID: 35101446 PMCID: PMC8892012 DOI: 10.1016/j.jbc.2022.101655] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/29/2022] Open
Abstract
G protein-coupled receptor 35 (GPR35) is poorly characterized but nevertheless has been revealed to have diverse roles in areas including lower gut inflammation and pain. The development of novel reagents and tools will greatly enhance analysis of GPR35 functions in health and disease. Here, we used mass spectrometry, mutagenesis, and [32P] orthophosphate labeling to identify that all five hydroxy-amino acids in the C-terminal tail of human GPR35a became phosphorylated in response to agonist occupancy of the receptor and that, apart from Ser294, each of these contributed to interactions with arretin-3, which inhibits further G protein-coupled receptor signaling. We found that Ser303 was key to such interactions; the serine corresponding to human GPR35a residue 303 also played a dominant role in arrestin-3 interactions for both mouse and rat GPR35. We also demonstrated that fully phospho-site–deficient mutants of human GPR35a and mouse GPR35 failed to interact effectively with arrestin-3, and the human phospho-deficient variant was not internalized from the surface of cells in response to agonist treatment. Even in cells stably expressing species orthologues of GPR35, a substantial proportion of the expressed protein(s) was determined to be immature. Finally, phospho-site–specific antisera targeting the region encompassing Ser303 in human (Ser301 in mouse) GPR35a identified only the mature forms of GPR35 and provided effective sensors of the activation status of the receptors both in immunoblotting and immunocytochemical studies. Such antisera may be useful tools to evaluate target engagement in drug discovery and target validation programs.
Collapse
|
12
|
Lin LC, Quon T, Engberg S, Mackenzie AE, Tobin AB, Milligan G. G Protein-Coupled Receptor GPR35 Suppresses Lipid Accumulation in Hepatocytes. ACS Pharmacol Transl Sci 2021; 4:1835-1848. [PMID: 34927014 DOI: 10.1021/acsptsci.1c00224] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Indexed: 02/07/2023]
Abstract
Although prevalent, nonalcoholic fatty liver disease is not currently treated effectively with medicines. Initially, using wild-type and genome-edited clones of the human hepatocyte cell line HepG2, we show that activation of the orphan G protein-coupled receptor GPR35 is both able and sufficient to block liver X-receptor-mediated lipid accumulation. Studies on hepatocytes isolated from both wild-type and GPR35 knock-out mice were consistent with a similar effect of GPR35 agonists in these cells, but because of marked differences in the pharmacology of GPR35 agonists and antagonists at the mouse and human orthologues, as well as elevated basal lipid levels in hepatocytes from the GPR35 knock-out mice, no definitive conclusion could be reached. To overcome this, we generated and characterized a transgenic knock-in mouse line in which the corresponding human GPR35 splice variant replaced the mouse orthologue. In hepatocytes from these humanized GPR35 mice, activation of this receptor was shown conclusively to prevent, and also reverse, lipid accumulation induced by liver X-receptor stimulation. These studies highlight the potential to target GPR35 in the context of fatty liver diseases.
Collapse
Affiliation(s)
- Li-Chiung Lin
- The Centre for Translational Pharmacology, Institute of Molecular, Cellular and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Tezz Quon
- The Centre for Translational Pharmacology, Institute of Molecular, Cellular and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Susanna Engberg
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Amanda E Mackenzie
- The Centre for Translational Pharmacology, Institute of Molecular, Cellular and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Andrew B Tobin
- The Centre for Translational Pharmacology, Institute of Molecular, Cellular and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Graeme Milligan
- The Centre for Translational Pharmacology, Institute of Molecular, Cellular and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
13
|
Kaya B, Melhem H, Niess JH. GPR35 in Intestinal Diseases: From Risk Gene to Function. Front Immunol 2021; 12:717392. [PMID: 34790192 PMCID: PMC8591220 DOI: 10.3389/fimmu.2021.717392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Diet and gut microbial metabolites mediate host immune responses and are central to the maintenance of intestinal health. The metabolite-sensing G-protein coupled receptors (GPCRs) bind metabolites and trigger signals that are important for the host cell function, survival, proliferation and expansion. On the contrary, inadequate signaling of these metabolite-sensing GPCRs most likely participate to the development of diseases including inflammatory bowel diseases (IBD). In the intestine, metabolite-sensing GPCRs are highly expressed by epithelial cells and by specific subsets of immune cells. Such receptors provide an important link between immune system, gut microbiota and metabolic system. Member of these receptors, GPR35, a class A rhodopsin-like GPCR, has been shown to be activated by the metabolites tryptophan-derived kynurenic acid (KYNA), the chemokine CXCL17 and phospholipid derivate lysophosphatidic acid (LPA) species. There have been studies on GPR35 in the context of intestinal diseases since its identification as a risk gene for IBD. In this review, we discuss the pharmacology of GPR35 including its proposed endogenous and synthetic ligands as well as its antagonists. We elaborate on the risk variants of GPR35 implicated in gut-related diseases and the mechanisms by which GPR35 contribute to intestinal homeostasis.
Collapse
Affiliation(s)
- Berna Kaya
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Hassan Melhem
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Gastroenterology/Hepatology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| |
Collapse
|
14
|
Boleij A, Fathi P, Dalton W, Park B, Wu X, Huso D, Allen J, Besharati S, Anders RA, Housseau F, Mackenzie AE, Jenkins L, Milligan G, Wu S, Sears CL. G-protein coupled receptor 35 (GPR35) regulates the colonic epithelial cell response to enterotoxigenic Bacteroides fragilis. Commun Biol 2021; 4:585. [PMID: 33990686 PMCID: PMC8121840 DOI: 10.1038/s42003-021-02014-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/18/2021] [Indexed: 02/03/2023] Open
Abstract
G protein-coupled receptor (GPR)35 is highly expressed in the gastro-intestinal tract, predominantly in colon epithelial cells (CEC), and has been associated with inflammatory bowel diseases (IBD), suggesting a role in gastrointestinal inflammation. The enterotoxigenic Bacteroides fragilis (ETBF) toxin (BFT) is an important virulence factor causing gut inflammation in humans and animal models. We identified that BFT signals through GPR35. Blocking GPR35 function in CECs using the GPR35 antagonist ML145, in conjunction with shRNA knock-down and CRISPRcas-mediated knock-out, resulted in reduced CEC-response to BFT as measured by E-cadherin cleavage, beta-arrestin recruitment and IL-8 secretion. Importantly, GPR35 is required for the rapid onset of ETBF-induced colitis in mouse models. GPR35-deficient mice showed reduced death and disease severity compared to wild-type C57Bl6 mice. Our data support a role for GPR35 in the CEC and mucosal response to BFT and underscore the importance of this molecule for sensing ETBF in the colon.
Collapse
Affiliation(s)
- Annemarie Boleij
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA.
- Radboud University Medical Center (Radboudumc), Department of Pathology, Radboud Institute for Molecular Life sciences (RIMLS), Nijmegen, The Netherlands.
| | - Payam Fathi
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - William Dalton
- Johns Hopkins University, Department of Oncology Center-Hematologic Malignancies, Baltimore, MD, USA
| | - Ben Park
- Johns Hopkins University, Department of Oncology Center-Hematologic Malignancies, Baltimore, MD, USA
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology and Oncology, Nashville, Tenessee, USA
| | - Xinqun Wu
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - David Huso
- Johns Hopkins University, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Jawara Allen
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - Sepideh Besharati
- Johns Hopkins University, Department of Pathobiology, Baltimore, MD, USA
| | - Robert A Anders
- Johns Hopkins University, Department of Pathobiology, Baltimore, MD, USA
| | - Franck Housseau
- Johns Hopkins University, Department of Oncology Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Amanda E Mackenzie
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Laura Jenkins
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Shaoguang Wu
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - Cynthia L Sears
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| |
Collapse
|
15
|
Quon T, Lin LC, Ganguly A, Tobin AB, Milligan G. Therapeutic Opportunities and Challenges in Targeting the Orphan G Protein-Coupled Receptor GPR35. ACS Pharmacol Transl Sci 2020; 3:801-812. [PMID: 33073184 PMCID: PMC7551713 DOI: 10.1021/acsptsci.0c00079] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Indexed: 02/07/2023]
Abstract
GPR35 is a class A, rhodopsin-like G protein-coupled receptor (GPCR) first identified more than 20 years ago. In the intervening period, identification of strong expression in the lower intestine and colon, in a variety of immune cells including monocytes and a variety of dendritic cells, and in dorsal root ganglia has suggested potential therapeutic opportunities in targeting this receptor in a range of conditions. GPR35 is, however, unusual in a variety of ways that challenge routes to translation. These include the following: (i) Although a substantial range and diversity of endogenous ligands have been suggested as agonist partners for this receptor, it officially remains defined as an "orphan" GPCR. (ii) Humans express two distinct protein isoform sequences, while rodents express only a single form. (iii) The pharmacologies of the human and rodent orthologues of GPR35 are very distinct, with variation between rat and mouse GPR35 being as marked as that between either of these species and the human forms. Herein we provide perspectives on each of the topics above as well as suggesting ways to overcome the challenges currently hindering potential translation. These include a better understanding of the extent and molecular basis for species selective GPR35 pharmacology and the production of novel mouse models in which both "on-target" and "off-target" effects of presumptive GPR35 ligands can be better defined, as well as a clear understanding of the human isoform expression profile and its significance at both tissue and individual cell levels.
Collapse
Affiliation(s)
- Tezz Quon
- Centre for Translational Pharmacology,
Institute of Molecular Cell and Systems Biology, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Glasgow G12 8QQ, United Kingdom of Great
Britain and Northern Ireland
| | - Li-Chiung Lin
- Centre for Translational Pharmacology,
Institute of Molecular Cell and Systems Biology, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Glasgow G12 8QQ, United Kingdom of Great
Britain and Northern Ireland
| | - Amlan Ganguly
- Centre for Translational Pharmacology,
Institute of Molecular Cell and Systems Biology, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Glasgow G12 8QQ, United Kingdom of Great
Britain and Northern Ireland
| | - Andrew B. Tobin
- Centre for Translational Pharmacology,
Institute of Molecular Cell and Systems Biology, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Glasgow G12 8QQ, United Kingdom of Great
Britain and Northern Ireland
| | - Graeme Milligan
- Centre for Translational Pharmacology,
Institute of Molecular Cell and Systems Biology, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Glasgow G12 8QQ, United Kingdom of Great
Britain and Northern Ireland
| |
Collapse
|
16
|
Yokoda RT, Rodriguez EA. Review: Pathogenesis of cholestatic liver diseases. World J Hepatol 2020; 12:423-435. [PMID: 32952871 PMCID: PMC7475774 DOI: 10.4254/wjh.v12.i8.423] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/07/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cholestatic liver diseases (CLD) begin to develop after an impairment of bile flow start to affect the biliary tree. Cholangiocytes actively participate in the liver response to injury and repair and the intensity of this reaction is a determinant factor for the development of CLD. Progressive cholangiopathies may ultimately lead to end-stage liver disease requiring at the end orthotopic liver transplantation. This narrative review will discuss cholangiocyte biology and pathogenesis mechanisms involved in four intrahepatic CLD: Primary biliary cholangitis, primary sclerosing cholangitis, cystic fibrosis involving the liver, and polycystic liver disease.
Collapse
Affiliation(s)
- Raquel T Yokoda
- Department of Anatomic and Clinical Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, United States
| | - Eduardo A Rodriguez
- Department of Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, UT 84132, United States
| |
Collapse
|
17
|
Kim MJ, Park SJ, Nam SY, Im DS. Lodoxamide Attenuates Hepatic Fibrosis in Mice: Involvement of GPR35. Biomol Ther (Seoul) 2019; 28:92-97. [PMID: 31189299 PMCID: PMC6939691 DOI: 10.4062/biomolther.2018.227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/07/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
A previous pharmacogenomic analysis identified cromolyn, an anti-allergic drug, as an effective anti-fibrotic agent that acts on hepatocytes and stellate cells. Furthermore, cromolyn was shown to be a G protein-coupled receptor 35 (GPR35) agonist. However, it has not been studied whether anti-fibrotic effects are mediated by GPR35. Therefore, in this study, the role of GPR35 in hepatic fibrosis was investigated through the use of lodoxamide, another anti-allergic drug and a potent GPR35 agonist. Long-term treatment with carbon tetrachloride induced hepatic fibrosis, which was inhibited by treatment with lodoxamide. Furthermore, CID2745687, a specific GPR35 antagonist, reversed lodoxamide-mediated anti-fibrotic effects. In addition, lodoxamide treatment showed significant effects on the mRNA expression of collagen Iα1, collagen Iα2, and TGF-β1 in the extracellular matrix. However, a transforming growth factor α (TGF-α) shedding assay revealed lodoxamide not to be a potent agonist of mouse GPR35 in vitro. Therefore, these results showed anti-fibrotic effects of lodoxamide in mice and raise concerns how lodoxamide protects against liver fibrosis in vivo and whether GPR35 is involved in the action.
Collapse
Affiliation(s)
- Mi-Jeong Kim
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Soo-Jin Park
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - So-Yeon Nam
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Dong-Soon Im
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
18
|
Mancini SJ, Mahmud ZA, Jenkins L, Bolognini D, Newman R, Barnes M, Edye ME, McMahon SB, Tobin AB, Milligan G. On-target and off-target effects of novel orthosteric and allosteric activators of GPR84. Sci Rep 2019; 9:1861. [PMID: 30755705 PMCID: PMC6372602 DOI: 10.1038/s41598-019-38539-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/20/2018] [Indexed: 12/03/2022] Open
Abstract
Many members of the G protein-coupled receptor family, including examples with clear therapeutic potential, remain poorly characterised. This often reflects limited availability of suitable tool ligands with which to interrogate receptor function. In the case of GPR84, currently a target for the treatment of idiopathic pulmonary fibrosis, recent times have seen the description of novel orthosteric and allosteric agonists. Using 2-(hexylthiol)pyrimidine-4,6 diol (2-HTP) and di(5,7-difluoro-1H-indole-3-yl)methane (PSB-16671) as exemplars of each class, in cell lines transfected to express either human or mouse GPR84, both ligands acted as effective on-target activators and with high co-operativity in their interactions. This was also the case in lipopolysaccharide-activated model human and mouse immune cell lines. However in mouse bone-marrow-derived neutrophils, where expression of GPR84 is particularly high, the capacity of PSB-16671 but not of 2-HTP to promote G protein activation was predominantly off-target because it was not blocked by an antagonist of GPR84 and was preserved in neutrophils isolated from GPR84 deficient mice. These results illustrate the challenges of attempting to study and define functions of poorly characterised receptors using ligands that have been developed via medicinal chemistry programmes, but where assessed activity has been limited largely to the initially identified target.
Collapse
Affiliation(s)
- Sarah J Mancini
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Zobaer Al Mahmud
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Laura Jenkins
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Daniele Bolognini
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Robert Newman
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, United Kingdom
| | - Matt Barnes
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, United Kingdom
| | - Michelle E Edye
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, United Kingdom
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
19
|
Mackenzie AE, Quon T, Lin LC, Hauser AS, Jenkins L, Inoue A, Tobin AB, Gloriam DE, Hudson BD, Milligan G. Receptor selectivity between the G proteins Gα 12 and Gα 13 is defined by a single leucine-to-isoleucine variation. FASEB J 2019; 33:5005-5017. [PMID: 30601679 PMCID: PMC6436656 DOI: 10.1096/fj.201801956r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite recent advances in structural definition of GPCR-G protein complexes, the basis of receptor selectivity between G proteins remains unclear. The Gα12 and Gα13 subtypes together form the least studied group of heterotrimeric G proteins. G protein-coupled receptor 35 (GPR35) has been suggested to couple efficiently to Gα13 but weakly to Gα12. Using combinations of cells genome-edited to not express G proteins and bioluminescence resonance energy transfer-based sensors, we confirmed marked selectivity of GPR35 for Gα13. Incorporating Gα12/Gα13 chimeras and individual residue swap mutations into these sensors defined that selectivity between Gα13 and Gα12 was imbued largely by a single leucine-to-isoleucine variation at position G.H5.23. Indeed, leucine could not be substituted by other amino acids in Gα13 without almost complete loss of GPR35 coupling. The critical importance of leucine at G.H5.23 for GPR35-G protein interaction was further demonstrated by introduction of this leucine into Gαq, resulting in the gain of coupling to GPR35. These studies demonstrate that Gα13 is markedly the most effective G protein for interaction with GPR35 and that selection between Gα13 and Gα12 is dictated largely by a single conservative amino acid variation.-Mackenzie, A. E., Quon, T., Lin, L.-C., Hauser, A. S., Jenkins, L., Inoue, A., Tobin, A. B., Gloriam, D. E., Hudson, B. D., Milligan, G. Receptor selectivity between the G proteins Gα12 and Gα13 is defined by a single leucine-to-isoleucine variation.
Collapse
Affiliation(s)
- Amanda E Mackenzie
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tezz Quon
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Li-Chiung Lin
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; and
| | - Laura Jenkins
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; and
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
20
|
Protective effect of lodoxamide on hepatic steatosis through GPR35. Cell Signal 2019; 53:190-200. [DOI: 10.1016/j.cellsig.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022]
|
21
|
Matysik-Woźniak A, Wnorowski A, Turski WA, Jóźwiak K, Jünemann A, Rejdak R. The presence and distribution of G protein-coupled receptor 35 (GPR35) in the human cornea - Evidences from in silico gene expression analysis and immunodetection. Exp Eye Res 2018; 179:188-192. [PMID: 30445046 DOI: 10.1016/j.exer.2018.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
We provide the evidence for G protein-coupled receptor 35 (GPR35) presence and distribution in the human cornea. The initial data on GPR35 gene expression were retrieved from microarray repositories and were further confirmed by western blotting and immunohistochemical analysis. Immunoblotting suggested that GPR35 exists predominantly as a dimer in corneal tissue. Moreover, corneal tissues were significantly richer in GPR35 compared to the adjacent sclera. Immunoreactivity for GPR35 was detected in normal corneas, keratoconus and Fuchs' dystrophy, mainly in the corneal epithelium and endothelium. In corneas with Fuchs' dystrophy, less intensive immunoreactivity for GPR35 in endothelium was revealed. The physiological relevance of this phenomenon requires further investigation.
Collapse
Affiliation(s)
- Anna Matysik-Woźniak
- Department of General Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079, Lublin, Poland.
| | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, Chodźki 4A, 20-093, Lublin, Poland.
| | - Waldemar A Turski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, K. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, Chodźki 4A, 20-093, Lublin, Poland.
| | - Anselm Jünemann
- Department of General Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079, Lublin, Poland; Department of Ophthalmology, University of Rostock, Doberaner Strasse, 140, 18057, Rostock, Germany.
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079, Lublin, Poland; Mossakowski Medical Research Centre Polish Academy of Sciences, Pawinskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
22
|
Divorty N, Milligan G, Graham D, Nicklin SA. The Orphan Receptor GPR35 Contributes to Angiotensin II-Induced Hypertension and Cardiac Dysfunction in Mice. Am J Hypertens 2018; 31:1049-1058. [PMID: 29860395 PMCID: PMC6077831 DOI: 10.1093/ajh/hpy073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/23/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The orphan receptor G protein–coupled receptor 35 (GPR35) has been associated with a range of diseases, including cancer, inflammatory bowel disease, diabetes, hypertension, and heart failure. To assess the potential for GPR35 as a therapeutic target in cardiovascular disease, this study investigated the cardiovascular phenotype of a GPR35 knockout mouse under both basal conditions and following pathophysiological stimulation. METHODS Blood pressure was monitored in male wild-type and GPR35 knockout mice over 7–14 days using implantable telemetry. Cardiac function and dimensions were assessed using echocardiography, and cardiomyocyte morphology evaluated histologically. Two weeks of angiotensin II (Ang II) infusion was used to investigate the effects of GPR35 deficiency under pathophysiological conditions. Gpr35 messenger RNA expression in cardiovascular tissues was assessed using quantitative polymerase chain reaction. RESULTS There were no significant differences in blood pressure, cardiac function, or cardiomyocyte morphology in GPR35 knockout mice compared with wild-type mice. Following Ang II infusion, GPR35 knockout mice were protected from significant increases in systolic, diastolic, and mean arterial blood pressure or impaired left ventricular systolic function, in contrast to wild-type mice. There were no significant differences in Gpr35 messenger RNA expression in heart, kidney, and aorta following Ang II infusion in wild-type mice. CONCLUSIONS Although GPR35 does not appear to influence basal cardiovascular regulation, these findings demonstrate that it plays an important pathological role in the development of Ang II–induced hypertension and impaired cardiac function. This suggests that GPR35 is a potential novel drug target for therapeutic intervention in hypertension.
Collapse
Affiliation(s)
- Nina Divorty
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
23
|
Binti Mohd Amir NAS, Mackenzie AE, Jenkins L, Boustani K, Hillier MC, Tsuchiya T, Milligan G, Pease JE. Evidence for the Existence of a CXCL17 Receptor Distinct from GPR35. THE JOURNAL OF IMMUNOLOGY 2018; 201:714-724. [PMID: 29875152 PMCID: PMC6036231 DOI: 10.4049/jimmunol.1700884] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 04/29/2018] [Indexed: 11/19/2022]
Abstract
The chemokine CXCL17 is associated with the innate response in mucosal tissues but is poorly characterized. Similarly, the G protein–coupled receptor GPR35, expressed by monocytes and mast cells, has been implicated in the immune response, although its precise role is ill-defined. A recent manuscript reported that GPR35 was able to signal in response to CXCL17, which we set out to confirm in this study. GPR35 was readily expressed using transfection systems but failed to signal in response to CXCL17 in assays of β-arrestin recruitment, inositol phosphate production, calcium flux, and receptor endocytosis. Similarly, in chemotaxis assays, GPR35 did not confirm sensitivity to a range of CXCL17 concentrations above that observed in the parental cell line. We subsequently employed a real time chemotaxis assay (TAXIScan) to investigate the migratory responses of human monocytes and the monocytic cell line THP-1 to a gradient of CXCL17. Freshly isolated human monocytes displayed no obvious migration to CXCL17. Resting THP-1 cells showed a trend toward directional migration along a CXCL17 gradient, which was significantly enhanced by overnight incubation with PGE2. However, pretreatment of PGE2-treated THP-1 cells with the well-characterized GPR35 antagonist ML145 did not significantly impair their migratory responses to CXCL17 gradient. CXCL17 was susceptible to cleavage with chymase, although this had little effect its ability to recruit THP-1 cells. We therefore conclude that GPR35 is unlikely to be a bona fide receptor for CXCL17 and that THP-1 cells express an as yet unidentified receptor for CXCL17.
Collapse
Affiliation(s)
- Nurul A S Binti Mohd Amir
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom.,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Amanda E Mackenzie
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom; and
| | - Laura Jenkins
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom; and
| | - Karim Boustani
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Marston C Hillier
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Tomoko Tsuchiya
- Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom; and
| | - James E Pease
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom; .,Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| |
Collapse
|
24
|
Masjosthusmann S, Becker D, Petzuch B, Klose J, Siebert C, Deenen R, Barenys M, Baumann J, Dach K, Tigges J, Hübenthal U, Köhrer K, Fritsche E. A transcriptome comparison of time-matched developing human, mouse and rat neural progenitor cells reveals human uniqueness. Toxicol Appl Pharmacol 2018; 354:40-55. [PMID: 29753005 DOI: 10.1016/j.taap.2018.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
It is widely accepted that human brain development has unique features that cannot be represented by rodents. Obvious reasons are the evolutionary distance and divergent physiology. This might lead to false predictions when rodents are used for safety or pharmacological efficacy studies. For a better translation of animal-based research to the human situation, human in vitro systems might be useful. In this study, we characterize developing neural progenitor cells from prenatal human and time-matched rat and mouse brains by analyzing the changes in their transcriptome profile during neural differentiation. Moreover, we identify hub molecules that regulate neurodevelopmental processes like migration and differentiation. Consequences of modulation of three of those hubs on these processes were studied in a species-specific context. We found that although the gene expression profiles of the three species largely differ qualitatively and quantitatively, they cluster in similar GO terms like cell migration, gliogenesis, neurogenesis or development of multicellular organism. Pharmacological modulation of the identified hub molecules triggered species-specific cellular responses. This study underlines the importance of understanding species differences on the molecular level and advocates the use of human based in vitro models for pharmacological and toxicological research.
Collapse
Affiliation(s)
- Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Daniel Becker
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany
| | - Barbara Petzuch
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Clara Siebert
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Rene Deenen
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| | - Marta Barenys
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Jenny Baumann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany
| | - Katharina Dach
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany; Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Ulrike Hübenthal
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Karl Köhrer
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany; Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| |
Collapse
|
25
|
Peng J, Zhao Y, Hong Y, Burkhalter RS, Hogue CL, Tran E, Wei L, Romeo L, Dolley-Sonneville P, Melkoumian Z, Liang X, Fang Y. Chemical Identity and Mechanism of Action and Formation of a Cell Growth Inhibitory Compound from Polycarbonate Flasks. Anal Chem 2018. [DOI: 10.1021/acs.analchem.7b05102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
| | - Yaopeng Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China
| | | | | | | | | | - Lai Wei
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China
| | | | | | | | - Xinmiao Liang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China
| | | |
Collapse
|
26
|
Park S, Lee S, Nam S, Im D. GPR35 mediates lodoxamide-induced migration inhibitory response but not CXCL17-induced migration stimulatory response in THP-1 cells; is GPR35 a receptor for CXCL17? Br J Pharmacol 2018; 175:154-161. [PMID: 29068046 PMCID: PMC5740256 DOI: 10.1111/bph.14082] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/29/2017] [Accepted: 10/07/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE GPR35 has long been considered an orphan GPCR, because no endogenous ligand of GPR35 has been discovered. CXCL17 (a chemokine) has been reported to be an endogenous ligand of GPR35, and it has even been suggested that it be called CXCR8. However, at present there is no supporting evidence that CXCL17 does interact with GPR35. EXPERIMENTAL APPROACH We applied two assay systems to explore the relationship between CXCL17 and GPR35. An AP-TGF-α shedding assay in GPR35 over-expressing HEK293 cells was used as a gain-of-function assay. GPR35 knock-down by siRNA transfection was performed in endogenously GPR35-expressing THP-1 cells. KEY RESULTS In the AP-TGF-α shedding assay, lodoxamide, a well-known synthetic GPR35 agonist, was confirmed to be the most potent agonist among other reported agonists. However, neither human nor mouse CXCL17 had an effect on GPR35. Consistent with previous findings, G proteins Gαi/o and Gα12/13 were found to couple with GPR35. Furthermore, lodoxamide-induced activation of GPR35 was concentration-dependently inhibited by CID2745687 (a selective GPR35 antagonist). In endogenously GPR35-expressing THP-1 cells, lodoxamide concentration-dependently inhibited migration and this inhibitory effect was blocked by CID2745687 treatment or GPR35 siRNA transfection. However, even though CXCL17 stimulated the migration of THP-1 cells, which is consistent with a previous report, this stimulatory effect of CXCL17 was not blocked by CID2745687 or GPR35 siRNA. CONCLUSIONS AND IMPLICATIONS The present findings suggest that GPR35 functions as a migration inhibitory receptor, but CXCL17-stimulated migration of THP-1 cells is not dependent on GPR35.
Collapse
Affiliation(s)
- Soo‐Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - Seung‐Jin Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - So‐Yeon Nam
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| | - Dong‐Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of PharmacyPusan National UniversityBusanKorea
| |
Collapse
|
27
|
Milligan G. G protein-coupled receptors not currently in the spotlight: free fatty acid receptor 2 and GPR35. Br J Pharmacol 2017; 175:2543-2553. [PMID: 28940377 PMCID: PMC6003633 DOI: 10.1111/bph.14042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/17/2017] [Accepted: 08/30/2017] [Indexed: 01/05/2023] Open
Abstract
It is widely appreciated that G protein‐coupled receptors have been the most successfully exploited class of targets for the development of small molecule medicines. Despite this, to date, less than 15% of the non‐olfactory G protein‐coupled receptors in the human genome are the targets of a clinically used medicine. In many cases, this is likely to reflect a lack of understanding of the basic underpinning biology of many G protein‐coupled receptors that are not currently in the spotlight, as well as a paucity of pharmacological tool compounds and appropriate animal models to test in vivo function of such G protein‐coupled receptors in both normal physiology and in the context of disease. ‘Open Innovation’ arrangements, in which pharmaceutical companies and public–private partnerships provide wider access to tool compounds identified from ligand screening programmes, alongside enhanced medicinal chemistry support to convert such screening ‘hits’ into useful ‘tool’ compounds will provide important routes to improved understanding. However, in parallel, novel approaches to define and fully appreciate the selectivity and mode of action of such tool compounds, as well as better understanding of potential species orthologue variability in the pharmacology and/or signalling profile of a wide range of currently poorly understood and understudied G protein‐coupled receptors, will be vital to fully exploit the therapeutic potential of this large target class. I consider these themes using as exemplars two G protein‐coupled receptors, free fatty acid receptor 2 and GPR35.
Collapse
Affiliation(s)
- Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
28
|
Sturchler E, Li X, de Lourdes Ladino M, Kaczanowska K, Cameron M, Griffin PR, Finn MG, Markou A, McDonald P. GABA B receptor allosteric modulators exhibit pathway-dependent and species-selective activity. Pharmacol Res Perspect 2017; 5:e00288. [PMID: 28357120 PMCID: PMC5368958 DOI: 10.1002/prp2.288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/02/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Positive modulation of the GABAB receptor (GABABR) represents a potentially useful therapeutic approach for the treatment of nicotine addiction. The positive allosteric modulators (PAMs) of GABABR GS39783 and BHF177 enhance GABA‐stimulated [35S]GTPγS‐binding, and have shown efficacy in a rodent nicotine self‐administration procedure reflecting aspects of nicotine dependence. Interestingly, the structural related analog, NVP998, had no effect on nicotine self‐administration in rats despite demonstrating similar pharmacokinetic properties. Extensive in vitro characterization of GS39783, BHF177, and NVP998 activity on GABABR‐regulated signaling events, including modulation of cAMP, intracellular calcium levels, and ERK activation, revealed that these structurally related molecules display distinct pathway‐specific signaling activities that correlate with the dissimilarities observed in rodent models and may be predictive of in vivo efficacy. Furthermore, these GABABR allosteric modulators exhibit species‐dependent activity. Collectively, these data will be useful in guiding the development of GABABR allosteric modulators that display optimal in vivo efficacy in a preclinical model of nicotine dependence, and will identify those that have the potential to lead to novel antismoking therapies.
Collapse
Affiliation(s)
- Emmanuel Sturchler
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Xia Li
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Maria de Lourdes Ladino
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458; Present address: School of Medicine Vanderbilt University 2215 Garland Ave Nashville Tennessee 37232
| | - Kasia Kaczanowska
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Department of Chemistry University of California San Diego, 9500 Gilman Drive La Jolla California 92093
| | - Michael Cameron
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Patrick R Griffin
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - M G Finn
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Georgia Institute of Technology School of Chemistry and Biochemistry 901 Atlantic Drive Atlanta Georgia 30332
| | - Athina Markou
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Patricia McDonald
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| |
Collapse
|
29
|
Mackenzie AE, Milligan G. The emerging pharmacology and function of GPR35 in the nervous system. Neuropharmacology 2017; 113:661-671. [DOI: 10.1016/j.neuropharm.2015.07.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/20/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
|
30
|
Resta F, Masi A, Sili M, Laurino A, Moroni F, Mannaioni G. Kynurenic acid and zaprinast induce analgesia by modulating HCN channels through GPR35 activation. Neuropharmacology 2016; 108:136-43. [PMID: 27131920 DOI: 10.1016/j.neuropharm.2016.04.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/11/2016] [Accepted: 04/26/2016] [Indexed: 12/30/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have a key role in the control of cellular excitability. HCN2, a subgroup of the HCN family channels, are heavily expressed in small dorsal root ganglia (DRG) neurons and their activation seems to be important in the determination of pain intensity. Intracellular elevation of cAMP levels activates HCN-mediated current (Ih) and small DRG neurons excitability. GPR35, a Gi/o coupled receptor, is highly expressed in small DRG neurons, and we hypothesized that its activation, mediated by endogenous or exogenous ligands, could lead to pain control trough a reduction of Ih current. Patch clamp recordings were carried out in primary cultures of rat DRG neurons and the effects of GPR35 activation on Ih current and neuronal excitability were studied in control conditions and after adenylate cyclase activation with either forskolin or prostaglandin E2 (PGE2). We found that both kynurenic acid (KYNA) and zaprinast, the endogenous and synthetic GPR35 agonist respectively, were able to antagonize the forskolin-induced depolarization of resting membrane potential by reducing Ih-mediated depolarization. Similar results were obtained when PGE2 was used to activate adenylate cyclase and to increase Ih current and the overall neuronal excitability. Finally, we tested the analgesic effect of both GPR35 agonists in an in vivo model of PGE2-induced thermal hyperalgesia. In accord with the hypothesis, both KYNA and zaprinast showed a dose dependent analgesic effect. In conclusion, GPR35 activation leads to a reduced excitability of small DRG neurons in vitro and causes a dose-dependent analgesia in vivo. GPR35 agonists, by reducing adenylate cyclase activity and inhibiting Ih in DRG neurons may represent a promising new group of analgesic drugs.
Collapse
Affiliation(s)
- Francesco Resta
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; Azienda Ospedaliero-Universitaria Careggi, Toxicology Unit, Largo Brambilla 1, 50139, Florence, Italy.
| | - Maria Sili
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | - Annunziatina Laurino
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | - Flavio Moroni
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; Azienda Ospedaliero-Universitaria Careggi, Toxicology Unit, Largo Brambilla 1, 50139, Florence, Italy.
| |
Collapse
|
31
|
McCallum JE, Mackenzie AE, Divorty N, Clarke C, Delles C, Milligan G, Nicklin SA. G-Protein-Coupled Receptor 35 Mediates Human Saphenous Vein Vascular Smooth Muscle Cell Migration and Endothelial Cell Proliferation. J Vasc Res 2016; 52:383-95. [PMID: 27064272 PMCID: PMC4959467 DOI: 10.1159/000444754] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/14/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) migration and proliferation is central to neointima formation in vein graft failure following coronary artery bypass. However, there are currently no pharmacological interventions that prevent vein graft failure through intimal occlusion. It is hence a therapeutic target. Here, we investigated the contribution of GPR35 to human VSMC and endothelial cell (EC) migration, using a scratch-wound assay, and also the contribution to proliferation, using MTS and BrdU assays, in in vitro models using recently characterized human GPR35 ortholog-selective small-molecule agonists and antagonists. Real-time PCR studies showed GPR35 to be robustly expressed in human VSMCs and ECs. Stimulation of GPR35, with either the human-selective agonist pamoic acid or the reference agonist zaprinast, promoted VSMC migration in the scratch-wound assay. These effects were blocked by coincubation with either of the human GPR35-specific antagonists, CID-2745687 or ML-145. These GPR35-mediated effects were produced by inducing alterations in the actin cytoskeleton via the Rho A/Rho kinase signaling axis. Additionally, the agonist ligands stimulated a proliferative response in ECs. These studies highlight the potential that small molecules that stimulate or block GPR35 activity can modulate vascular proliferation and migration. These data propose GPR35 as a translational therapeutic target in vascular remodeling.
Collapse
Affiliation(s)
- Jennifer E McCallum
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The immune-mediated hepatobiliary diseases, primary biliary cirrhosis and primary sclerosing cholangitis are relatively rare, albeit and account for a significant amount of liver transplant activity and liver-related mortality globally. Precise disease mechanisms are yet to be described although a contributory role of genetic predisposition is firmly established. In addition to links with the major histocompatibility complex, a number of associations outside this region harbor additional loci which underscore the fundamental role of breaks in immune tolerance and mucosal immunogenicity in the pathogenesis of autoimmune biliary disease. We provide an overview of these key discoveries before discussing putative avenues of therapeutic exploitation based on existing findings.
Collapse
|
33
|
Fritsche E, Alm H, Baumann J, Geerts L, Håkansson H, Masjosthusmann S, Witters H. Literature review on in vitro and alternative Developmental Neurotoxicity (DNT) testing methods. ACTA ACUST UNITED AC 2015. [DOI: 10.2903/sp.efsa.2015.en-778] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine (IUF), Group of Sphere Models and Risk Assessment, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Henrik Alm
- Leibniz Research Institute for Environmental Medicine (IUF), Group of Sphere Models and Risk Assessment, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Jenny Baumann
- Leibniz Research Institute for Environmental Medicine (IUF), Group of Sphere Models and Risk Assessment, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Lieve Geerts
- Flemish Institute for Technological Research (VITO), Environmental Risk & Health, Boeretang 200, B‐2400 Mol, Belgium
| | - Helen Håkansson
- Karolinska Institute (KI), Institute of Environmental Medicine (IMM), Unit of Environmental Health Risk Assessment, SE‐171 77 Stockholm, Sweden
| | - Stefan Masjosthusmann
- Leibniz Research Institute for Environmental Medicine (IUF), Group of Sphere Models and Risk Assessment, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Hilda Witters
- Flemish Institute for Technological Research (VITO), Environmental Risk & Health, Boeretang 200, B‐2400 Mol, Belgium
| |
Collapse
|
34
|
Divorty N, Mackenzie AE, Nicklin SA, Milligan G. G protein-coupled receptor 35: an emerging target in inflammatory and cardiovascular disease. Front Pharmacol 2015; 6:41. [PMID: 25805994 PMCID: PMC4354270 DOI: 10.3389/fphar.2015.00041] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/13/2015] [Indexed: 01/13/2023] Open
Abstract
G protein-coupled receptor 35 (GPR35) is an orphan receptor, discovered in 1998, that has garnered interest as a potential therapeutic target through its association with a range of diseases. However, a lack of pharmacological tools and the absence of convincingly defined endogenous ligands have hampered the understanding of function necessary to exploit it therapeutically. Although several endogenous molecules can activate GPR35 none has yet been confirmed as the key endogenous ligand due to reasons that include lack of biological specificity, non-physiologically relevant potency and species ortholog selectivity. Recent advances have identified several highly potent synthetic agonists and antagonists, as well as agonists with equivalent potency at rodent and human orthologs, which will be useful as tool compounds. Homology modeling and mutagenesis studies have provided insight into the mode of ligand binding and possible reasons for the species selectivity of some ligands. Advances have also been made in determining the role of the receptor in disease. In the past, genome-wide association studies have associated GPR35 with diseases such as inflammatory bowel disease, type 2 diabetes, and coronary artery disease. More recent functional studies have implicated it in processes as diverse as heart failure and hypoxia, inflammation, pain transduction and synaptic transmission. In this review, we summarize the progress made in understanding the molecular pharmacology, downstream signaling and physiological function of GPR35, and discuss its emerging potential applications as a therapeutic target.
Collapse
Affiliation(s)
- Nina Divorty
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow UK ; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow UK
| | - Amanda E Mackenzie
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow UK
| | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow UK
| | - Graeme Milligan
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow UK
| |
Collapse
|
35
|
Foster SR, Roura E, Molenaar P, Thomas WG. G protein-coupled receptors in cardiac biology: old and new receptors. Biophys Rev 2015; 7:77-89. [PMID: 28509979 DOI: 10.1007/s12551-014-0154-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/25/2014] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are seven-transmembrane-spanning proteins that mediate cellular and physiological responses. They are critical for cardiovascular function and are targeted for the treatment of hypertension and heart failure. Nevertheless, current therapies only target a small fraction of the cardiac GPCR repertoire, indicating that there are many opportunities to investigate unappreciated aspects of heart biology. Here, we offer an update on the contemporary view of GPCRs and the complexities of their signalling, and review the roles of the 'classical' GPCRs in cardiovascular physiology and disease. We then provide insights into other GPCRs that have been less extensively studied in the heart, including orphan, odorant and taste receptors. We contend that these novel cardiac GPCRs contribute to heart function in health and disease and thereby offer exciting opportunities to therapeutically modulate heart function.
Collapse
Affiliation(s)
- Simon R Foster
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia
| | - Eugeni Roura
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia.,Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia Campus, Brisbane, Australia
| | - Peter Molenaar
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, St Lucia Campus, Brisbane, Australia.,School of Medicine, University of Queensland, St Lucia Campus, Brisbane, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia.
| |
Collapse
|
36
|
Alkondon M, Pereira EFR, Todd SW, Randall WR, Lane MV, Albuquerque EX. Functional G-protein-coupled receptor 35 is expressed by neurons in the CA1 field of the hippocampus. Biochem Pharmacol 2014; 93:506-18. [PMID: 25542997 DOI: 10.1016/j.bcp.2014.12.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 12/17/2022]
Abstract
The G-protein-coupled receptor 35 (GPR35) was de-orphanized after the discovery that kynurenic acid (KYNA), an endogenous tryptophan metabolite, acts as an agonist of this receptor. Abundant evidence supports that GPR35 exists primarily in peripheral tissues. Here, we tested the hypothesis that GPR35 exists in the hippocampus and influences the neuronal activity. Fluorescence immunohistochemical staining using an antibody anti-NeuN (a neuronal marker), an antibody anti-GFAP (a glial marker), and an antibody anti-GPR35 revealed that neurons in the stratum oriens, stratum pyramidale, and stratum radiatum of the CA1 field of the hippocampus express GPR35. To determine the presence of functional GPR35 in the neurocircuitry, we tested the effects of various GPR35 agonists on the frequency of spontaneous action potentials recorded as fast current transients (CTs) from stratum radiatum interneurons (SRIs) under cell-attached configuration in rat hippocampal slices. Bath application of the GPR35 agonists zaprinast (1-10 μM), dicumarol (50-100 μM), pamoic acid (500-1000 μM), and amlexanox (3 μM) produced a concentration- and time-dependent reduction in the frequency of CTs. Superfusion of the hippocampal slices with the GPR35 antagonist ML145 (1 μM) increased the frequency of CTs and reduced the inhibitory effect of zaprinast. Bath application of phosphodiesterase 5 inhibitor sildenafil (1 or 5 μM) was ineffective, whereas a subsequent application of zaprinast was effective in reducing the CT frequency. The present results demonstrate for the first time that functional GPR35s are expressed by CA1 neurons and suggest that these receptors can be molecular targets for controlling neuronal activity in the hippocampus.
Collapse
Affiliation(s)
- Manickavasagom Alkondon
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine Street, Suite 900, Baltimore, MD 21201, USA
| | - Edna F R Pereira
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine Street, Suite 900, Baltimore, MD 21201, USA
| | - Spencer W Todd
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine Street, Suite 900, Baltimore, MD 21201, USA
| | - William R Randall
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Malcolm V Lane
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine Street, Suite 900, Baltimore, MD 21201, USA
| | - Edson X Albuquerque
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine Street, Suite 900, Baltimore, MD 21201, USA.
| |
Collapse
|
37
|
MacKenzie AE, Caltabiano G, Kent TC, Jenkins L, McCallum JE, Hudson BD, Nicklin SA, Fawcett L, Markwick R, Charlton SJ, Milligan G. The antiallergic mast cell stabilizers lodoxamide and bufrolin as the first high and equipotent agonists of human and rat GPR35. Mol Pharmacol 2013; 85:91-104. [PMID: 24113750 DOI: 10.1124/mol.113.089482] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lack of high potency agonists has restricted analysis of the G protein-coupled receptor GPR35. Moreover, marked variation in potency and/or affinity of current ligands between human and rodent orthologs of GPR35 has limited their productive use in rodent models of physiology. Based on the reported modest potency of the antiasthma and antiallergic ligands cromolyn disodium and nedocromil sodium, we identified the related compounds lodoxamide and bufrolin as high potency agonists of human GPR35. Unlike previously identified high potency agonists that are highly selective for human GPR35, both lodoxamide and bufrolin displayed equivalent potency at rat GPR35. Further synthetic antiallergic ligands, either sharing features of the standard surrogate agonist zaprinast, or with lodoxamide and bufrolin, were also shown to display agonism at either human or rat GPR35. Because both lodoxamide and bufrolin are symmetric di-acids, their potential mode of binding was explored via mutagenesis based on swapping between the rat and human ortholog nonconserved arginine residues within proximity of a key conserved arginine at position 3.36. Computational modeling and ligand docking predicted the contributions of different arginine residues, other than at 3.36, in human GPR35 for these two ligands and were consistent with selective loss of potency of either bufrolin or lodoxamide at distinct arginine mutants. The computational models also suggested that bufrolin and lodoxamide would display reduced potency at a low-frequency human GPR35 single nucleotide polymorphism. This prediction was confirmed experimentally.
Collapse
Affiliation(s)
- Amanda E MacKenzie
- Molecular Pharmacology Group, Institute of Molecular, Cell, and Systems Biology (A.E.M., G.C., L.J., J.E.M., B.D.H., G.M.) and Institute of Cardiovascular and Medical Sciences, (J.E.M., S.A.N.), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom; Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain (G.C.); and Novartis Institutes for Biomedical Research, Horsham, United Kingdom (T.C.K., L.F., R.M., S.J.C.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Thimm D, Funke M, Meyer A, Müller CE. 6-Bromo-8-(4-[3H]methoxybenzamido)-4-oxo-4H-chromene-2-carboxylic Acid: A Powerful Tool for Studying Orphan G Protein-Coupled Receptor GPR35. J Med Chem 2013; 56:7084-99. [DOI: 10.1021/jm4009373] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dominik Thimm
- PharmaCenter Bonn, Pharmaceutical
Institute, Pharmaceutical
Chemistry I, University of Bonn, An der
Immenburg 4, D-53121 Bonn, Germany
| | - Mario Funke
- PharmaCenter Bonn, Pharmaceutical
Institute, Pharmaceutical
Chemistry I, University of Bonn, An der
Immenburg 4, D-53121 Bonn, Germany
| | - Anne Meyer
- PharmaCenter Bonn, Pharmaceutical
Institute, Pharmaceutical
Chemistry I, University of Bonn, An der
Immenburg 4, D-53121 Bonn, Germany
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical
Institute, Pharmaceutical
Chemistry I, University of Bonn, An der
Immenburg 4, D-53121 Bonn, Germany
| |
Collapse
|
39
|
Funke M, Thimm D, Schiedel AC, Müller CE. 8-Benzamidochromen-4-one-2-carboxylic acids: potent and selective agonists for the orphan G protein-coupled receptor GPR35. J Med Chem 2013; 56:5182-97. [PMID: 23713606 DOI: 10.1021/jm400587g] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
8-Amido-chromen-4-one-2-carboxylic acid derivatives were identified as novel agonists at the G protein-coupled orphan receptor GPR35. They were characterized by a β-arrestin recruitment assay and optimized to obtain agonists with nanomolar potency for the human GPR35. The compounds were found to exhibit high selectivity versus the related GPR55. The most potent agonists were 6-bromo-8-(4-methoxybenzamido)-4-oxo-4H-chromene-2-carboxylic acid (85, EC50 12.1 nM) and 6-bromo-8-(2-chloro-4-methoxybenzamido)-4-oxo-4H-chromene-2-carboxylic acid (90, EC50 11.1 nM), both of which were >1700-fold selective versus GPR55. Most compounds were considerably less potent at rat and mouse than at human GPR35. 6-Bromo-8-(2-methoxybenzamido)-4-oxo-4H-chromene-2-carboxylic acid (87) was the only derivative that activated GPR35 of all three species at similar, low micromolar concentration. Compounds 85 and 90 are the most potent agonists at the human GPR35 known to date and might thus serve as powerful pharmacological tools to further elucidate the receptor's (patho)physiological role and its potential as a future drug target.
Collapse
Affiliation(s)
- Mario Funke
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | | | | | | |
Collapse
|
40
|
Deng H, Fang Y. The Three Catecholics Benserazide, Catechol and Pyrogallol are GPR35 Agonists. Pharmaceuticals (Basel) 2013; 6:500-9. [PMID: 24276120 PMCID: PMC3816702 DOI: 10.3390/ph6040500] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 03/20/2013] [Accepted: 04/01/2013] [Indexed: 11/16/2022] Open
Abstract
Nearly 1% of all clinically used drugs are catecholics, a family of catechol-containing compounds. Using label-free dynamic mass redistribution and Tango β-arrestin translocation assays, we show that several catecholics, including benserazide, catechol, 3-methoxycatechol, pyrogallol, (+)-taxifolin and fenoldopam, display agonistic activity against GPR35.
Collapse
Affiliation(s)
- Huayun Deng
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, NY 14831, USA.
| | | |
Collapse
|
41
|
Neetoo-Isseljee Z, MacKenzie AE, Southern C, Jerman J, McIver EG, Harries N, Taylor DL, Milligan G. High-throughput identification and characterization of novel, species-selective GPR35 agonists. J Pharmacol Exp Ther 2012; 344:568-78. [PMID: 23262279 DOI: 10.1124/jpet.112.201798] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Drugs targeting the orphan receptor GPR35 have potential therapeutic application in a number of disease areas, including inflammation, metabolic disorders, nociception, and cardiovascular disease. Currently available surrogate GPR35 agonists identified from pharmacologically relevant compound libraries have limited utility due to the likelihood of off-target effects in vitro and in vivo and the variable potency that such ligands exhibit across species. We sought to identify and characterize novel GPR35 agonists to facilitate studies aimed at defining the physiologic role of GPR35. PathHunter β-arrestin recruitment technology was validated as a human GPR35 screening assay, and a high-throughput screen of 100,000 diverse low molecular weight compounds was conducted. Confirmed GPR35 agonists from five distinct chemotypes were selected for detailed characterization using both β-arrestin recruitment and G protein-dependent assays and each of the human, mouse, and rat GPR35 orthologs. These studies identified 4-{(Z)-[(2Z)-2-(2-fluorobenzylidene)-4-oxo-1,3-thiazolidin-5-ylidene]methyl}benzoic acid (compound 1) as the highest potency full agonist of human GPR35 yet described. As with certain other GPR35 agonists, compound 1 was markedly selective for human GPR35, but displayed elements of signal bias between β-arrestin-2 and G protein-dependent assays. Compound 1 also displayed competitive behavior when assessed against the human GPR35 antagonist, ML-145 (2-hydroxy-4-[4-(5Z)-5-[(E)-2-methyl-3-phenylprop-2-enylidene]-4-oxo-2-sulfanylidene-1,3-thiazolidin-3-yl]butanoylamino]benzoic acid). Of the other chemotypes studied, compounds 2 and 3 were selective for the human receptor, but compounds 4 and 5 demonstrated similar activity at human, rat, and mouse GPR35 orthologs. Further characterization of these compounds and related analogs is likely to facilitate a better understanding of GPR35 in health and disease.
Collapse
Affiliation(s)
- Zaynab Neetoo-Isseljee
- Medical Research Council Technology Centre for Therapeutics Discovery, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|