1
|
Russell IC, Lee D, Wootten D, Sexton PM, Bumbak F. Cryoelectron microscopy as a tool for illuminating activation mechanisms of human class A orphan G protein-coupled receptors. Pharmacol Rev 2025; 77:100056. [PMID: 40286430 DOI: 10.1016/j.pharmr.2025.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are critically important medicinal targets, and the cryogenic electron microscopy (cryo-EM) revolution is providing novel high-resolution GPCR structures at a rapid pace. Orphan G protein-coupled receptors (oGPCRs) are a group of approximately 100 nonolfactory GPCRs for which endogenous ligands are unknown or not validated. The absence of modulating ligands adds difficulties to understanding the physiologic significance of oGPCRs and in the determination of high-resolution structures of isolated receptors that could facilitate drug discovery. Despite the challenges, cryo-EM structures of oGPCR-G protein complexes are emerging. This is being facilitated by numerous developments to stabilize GPCR-G protein complexes such as the use of dominant-negative G proteins, mini-G proteins, complex-stabilizing nanobodies or antibody fragments, and protein tethering methods. Moreover, many oGPCRs are constitutively active, which can facilitate complex formation in the absence of a known activating ligand. Consequently, in addition to providing templates for drug discovery, active oGPCR structures shed light on constitutive GPCR activation mechanisms. These comprise self-activation, whereby mobile extracellular portions of the receptor act as tethered agonists by occupying a canonical orthosteric-binding site in the transmembrane core, constitutive activity due to alterations to conserved molecular switches that stabilize inactive states of GPCRs, as well as receptors activated by cryptic ligands that are copurified with the receptor. Cryo-EM structures of oGPCRs are now being determined at a rapid pace and are expected to be invaluable tools for oGPCR drug discovery. SIGNIFICANCE STATEMENT: Orphan G protein-coupled receptors (GPCRs) provide large untapped potential for development of new medicines. Many of these receptors display constitutive activity, enabling structure determination and insights into observed GPCR constitutive activity including (1) self-activation by mobile receptor extracellular portions that function as tethered agonists, (2) modification of conserved motifs canonically involved in receptor quiescence and/or activation, and (3) activation by cryptic lipid ligands. Collectively, these studies advance fundamental understanding of GPCR function and provide opportunities for novel drug discovery.
Collapse
Affiliation(s)
- Isabella C Russell
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Dongju Lee
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Denise Wootten
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Patrick M Sexton
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Fabian Bumbak
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins and Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
2
|
Chen J, Cai M, Zhan C. Neuronal Regulation of Feeding and Energy Metabolism: A Focus on the Hypothalamus and Brainstem. Neurosci Bull 2025; 41:665-675. [PMID: 39704987 PMCID: PMC11978587 DOI: 10.1007/s12264-024-01335-7] [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: 07/07/2024] [Accepted: 09/19/2024] [Indexed: 12/21/2024] Open
Abstract
In the face of constantly changing environments, the central nervous system (CNS) rapidly and accurately calculates the body's needs, regulates feeding behavior, and maintains energy homeostasis. The arcuate nucleus of the hypothalamus (ARC) plays a key role in this process, serving as a critical brain region for detecting nutrition-related hormones and regulating appetite and energy homeostasis. Agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the ARC are core elements that interact with other brain regions through a complex appetite-regulating network to comprehensively control energy homeostasis. In this review, we explore the discovery and research progress of AgRP neurons in regulating feeding and energy metabolism. In addition, recent advances in terms of feeding behavior and energy homeostasis, along with the redundant neural mechanisms involved in energy metabolism, are discussed. Finally, the challenges and opportunities in the field of neural regulation of feeding and energy metabolism are briefly discussed.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Meiting Cai
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Cheng Zhan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Key Laboratory of Immune Response and Immunotherapy, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
3
|
Raffaele S, Clausen BH, Mannella FC, Wirenfeldt M, Marangon D, Tidgen SB, Corradini S, Madsen K, Lecca D, Abbracchio MP, Lambertsen KL, Fumagalli M. Characterisation of GPR17-expressing oligodendrocyte precursors in human ischaemic lesions and correlation with reactive glial responses. J Pathol 2025; 265:226-243. [PMID: 39703181 PMCID: PMC11717493 DOI: 10.1002/path.6381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/14/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024]
Abstract
White matter damage and subsequent demyelination significantly contribute to long-term functional impairment after ischaemic stroke. Identifying novel pharmacological targets to restore myelin integrity by promoting the maturation of oligodendrocyte precursor cells (OPCs) into new myelinating oligodendrocytes may open new perspectives for ischaemic stroke treatment. In this respect, previous studies highlighted the role of the G protein-coupled membrane receptor 17 (GPR17) as a key regulator of OPC differentiation in experimental models of brain injury, including ischaemic stroke. To determine the translational value of GPR17 as a possible target in the context of human disease, we exploited immunohistochemistry to characterise the distribution of GPR17-expressing cells in brain tissue samples from ischaemic stroke cases and correlated it with the reactive state of neighbouring glial cells. The results showed that GPR17 specifically decorates a subpopulation of differentiation-committed OPCs, labelled by the peculiar marker breast carcinoma-amplified sequence 1 (BCAS1), that accumulates in the peri-infarct region in the later stages after the ischaemic event. Interestingly, the response of GPR17-expressing cells appears to be paralleled by the switch of reactive microglia/macrophages from a phagocytic to a dystrophic phenotype and by astrocytic scar formation. A negative correlation was found between GPR17-expressing OPCs and reactive microglia/macrophages and astrocytes surrounding chronic ischaemic lesions in female subjects, while the same relationship was less pronounced in males. These results were reinforced by bioinformatic analysis of a publicly available transcriptomic dataset, which implicated a possible role of inflammation and defective neuron-to-OPC communication in remyelination failure after ischaemic damage. Hence, these data strengthen the relevance of GPR17-based remyelinating therapies for the treatment of ischaemic stroke. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
| | - Francesca Carolina Mannella
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Martin Wirenfeldt
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of PathologySouth Denmark University HospitalOdenseDenmark
| | - Davide Marangon
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | - Sarah Boe Tidgen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Silvia Corradini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Kirsten Madsen
- Department of PathologySouth Denmark University HospitalOdenseDenmark
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Davide Lecca
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of NeurologyOdense University HospitalOdenseDenmark
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| |
Collapse
|
4
|
Lee M, Boyce JA, Barrett NA. Cysteinyl Leukotrienes in Allergic Inflammation. ANNUAL REVIEW OF PATHOLOGY 2025; 20:115-141. [PMID: 39374430 PMCID: PMC11759657 DOI: 10.1146/annurev-pathmechdis-111523-023509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
The cysteinyl leukotrienes (CysLTs), LTC4, LTD4, and LTE4, are potent lipid mediators derived from arachidonic acid through the 5-lipoxygenase pathway. These mediators produce both inflammation and bronchoconstriction through three distinct G protein-coupled receptors (GPCRs)-CysLT1, CysLT2, and OXGR1 (also known as CysLT3 or GPR99). While CysLT-mediated functions in the effector phase of allergic inflammation and asthma have been established for some time, recent work has demonstrated novel roles for these mediators and their receptors in the induction and amplification of type 2 inflammation. Additionally, in vitro studies and murine models have uncovered diverse regulatory mechanisms that restrain or amplify CysLT receptor activation and CysLT receptor function. This review provides an overview of CysLT biosynthesis and its regulation, the molecular and functional pharmacology of CysLT receptors, and an overview of the established and emerging roles of CysLTs in asthma, aspirin-exacerbated respiratory disease, and type 2 inflammation.
Collapse
Affiliation(s)
- Minkyu Lee
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Massachusetts, USA; , ,
| | - Joshua A Boyce
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Massachusetts, USA; , ,
| | - Nora A Barrett
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Massachusetts, USA; , ,
| |
Collapse
|
5
|
Conley JM, Jochim A, Evans-Molina C, Watts VJ, Ren H. G Protein-Coupled Receptor 17 Inhibits Glucagon-like Peptide-1 Secretion via a Gi/o-Dependent Mechanism in Enteroendocrine Cells. Biomolecules 2024; 15:9. [PMID: 39858405 PMCID: PMC11762167 DOI: 10.3390/biom15010009] [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: 11/25/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Gut peptides, including glucagon-like peptide-1 (GLP-1), regulate metabolic homeostasis and have emerged as the basis for multiple state-of-the-art diabetes and obesity therapies. We previously showed that G protein-coupled receptor 17 (GPR17) is expressed in intestinal enteroendocrine cells (EECs) and modulates nutrient-induced GLP-1 secretion. However, the GPR17-mediated molecular signaling pathways in EECs have yet to be fully deciphered. Here, we expressed the human GPR17 long isoform (hGPR17L) in GLUTag cells, a murine EEC line, and we used the GPR17 synthetic agonist MDL29,951 together with pharmacological probes and genetic approaches to quantitatively assess the contribution of GPR17 signaling to GLP-1 secretion. Constitutive hGPR17L activity inhibited GLP-1 secretion, and MDL29,951 treatment further inhibited this secretion, which was attenuated by treatment with the GPR17 antagonist HAMI3379. MDL29,951 promoted both Gi/o and Gq protein coupling to mediate cyclic AMP (cAMP) and calcium signaling. hGPR17L regulation of GLP-1 secretion appeared to be Gq-independent and dependent upon Gi/o signaling, but was not correlated with MDL29,951-induced whole-cell cAMP signaling. Our studies revealed key signaling mechanisms underlying the role of GPR17 in regulating GLP-1 secretion and suggest future opportunities for pharmacologically targeting GPR17 with inverse agonists to maximize GLP-1 secretion.
Collapse
Affiliation(s)
- Jason M. Conley
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.M.C.); (A.J.); (C.E.-M.)
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander Jochim
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.M.C.); (A.J.); (C.E.-M.)
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carmella Evans-Molina
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.M.C.); (A.J.); (C.E.-M.)
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Val J. Watts
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA;
| | - Hongxia Ren
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (J.M.C.); (A.J.); (C.E.-M.)
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Boshta NM, Lewash M, Köse M, Namasivayam V, Sarkar S, Voss JH, Liedtke AJ, Junker A, Tian M, Stößel A, Rashed M, Mahal A, Merten N, Pegurier C, Hockemeyer J, Kostenis E, Müller CE. Discovery of Anthranilic Acid Derivatives as Antagonists of the Pro-Inflammatory Orphan G Protein-Coupled Receptor GPR17. J Med Chem 2024; 67:19365-19394. [PMID: 39484825 DOI: 10.1021/acs.jmedchem.4c01755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The G protein-coupled receptor 17 (GPR17) is an orphan receptor involved in inflammatory diseases. GPR17 antagonists have been proposed for the treatment of multiple sclerosis due to their potential to induce remyelination. Potent, selective antagonists are required to enable target validation. In the present study, we describe the discovery of a novel class of GPR17 antagonists based on an anthranilic acid scaffold. The compounds' potencies were evaluated in calcium mobilization and radioligand binding assays, and structure-activity relationships were analyzed. Selected antagonists were additionally studied in cAMP and G protein activation assays. The most potent antagonists were 5-methoxy-2-(5-(3'-methoxy-[1,1'-biphenyl]-2-yl)furan-2-carboxamido)benzoic acid (52, PSB-22269, Ki 8.91 nM) and its 3'-trifluoromethyl analog (54, PSB-24040, Ki 83.2 nM). Receptor-ligand docking studies revealed that the compounds' binding site is characterized by positively charged arginine residues and a lipophilic pocket. These findings yield valuable insights into this poorly characterized receptor providing a basis for future drug development.
Collapse
Affiliation(s)
- Nader M Boshta
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Michael Lewash
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Meryem Köse
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Soumya Sarkar
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Jan H Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Andy J Liedtke
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Anna Junker
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Maoqun Tian
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Anne Stößel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Mahmoud Rashed
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Ahmed Mahal
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Nicole Merten
- Pharmaceutical Biology, University of Bonn, Nußallee 6, Bonn D-53115, Germany
| | | | - Jörg Hockemeyer
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Evi Kostenis
- Pharmaceutical Biology, University of Bonn, Nußallee 6, Bonn D-53115, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| |
Collapse
|
7
|
Conley JM, Jochim A, Evans-Molina C, Watts VJ, Ren H. G protein-coupled receptor 17 inhibits glucagon-like peptide-1 secretion via a Gi/o-dependent mechanism in enteroendocrine cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623413. [PMID: 39605686 PMCID: PMC11601441 DOI: 10.1101/2024.11.13.623413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Gut peptides, including glucagon-like peptide-1 (GLP-1), regulate metabolic homeostasis and have emerged as the basis for multiple state-of-the-art diabetes and obesity therapies. We previously showed that G protein-coupled receptor 17 (GPR17) is expressed in intestinal enteroendocrine cells (EECs) and modulates nutrient-induced GLP-1 secretion. However, the GPR17-mediated molecular signaling pathways in EECs have yet to be fully deciphered. Here, we expressed the human GPR17 long isoform (hGPR17L) in GLUTag cells, a murine EEC line, and we used the GPR17 synthetic agonist MDL29,951 together with pharmacological probes and genetic approaches to quantitatively assess the contribution of GPR17 signaling to GLP-1 secretion. Constitutive hGPR17L activity inhibited GLP-1 secretion, and MDL29,951 treatment further inhibited this secretion, which was attenuated by treatment with the GPR17 antagonist HAMI3379. MDL29,951 promoted both Gi/o and Gq protein coupling to mediate cyclic AMP (cAMP) and calcium signaling. hGPR17L regulation of GLP-1 secretion was Gq-independent and dependent upon Gi/o signaling, but was not correlated with MDL29,951-induced whole-cell cAMP signaling. Our studies revealed key signaling mechanisms underlying the role of GPR17 in regulating GLP-1 secretion and suggest future opportunities for pharmacologically targeting GPR17 with inverse agonists to maximize GLP-1 secretion.
Collapse
Affiliation(s)
- Jason M. Conley
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Alexander Jochim
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Carmella Evans-Molina
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Roudebush VA Medical Center, Indianapolis, IN 46202
| | - Val J. Watts
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907
| | - Hongxia Ren
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
8
|
Nie R, Zhou X, Fu J, Hu S, Zhang Q, Jiang W, Yan Y, Cao X, Yuan D, Long Y, Hong H, Tang S. GPR17 modulates anxiety-like behaviors via basolateral amygdala to ventral hippocampal CA1 glutamatergic projection. Acta Pharm Sin B 2024; 14:4789-4805. [PMID: 39664418 PMCID: PMC11628806 DOI: 10.1016/j.apsb.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/17/2024] [Accepted: 07/26/2024] [Indexed: 12/13/2024] Open
Abstract
Anxiety disorders are one of the most epidemic and chronic psychiatric disorders. An incomplete understanding of anxiety pathophysiology has limited the development of highly effective drugs against these disorders. GPR17 has been shown to be involved in multiple sclerosis and some acute brain injury disorders. However, no study has investigated the role of GPR17 in psychiatric disorders. In a well-established chronic restraint stress (CRS) mouse model, using a combination of pharmacological and molecular biology techniques, viral tracing, in vitro electrophysiology recordings, in vivo fiber photometry, chemogenetic manipulations and behavioral tests, we demonstrated that CRS induced anxiety-like behaviors and increased the expression of GPR17 in basolateral amygdala (BLA) glutamatergic neurons. Inhibition of GPR17 by cangrelor or knockdown of GPR17 by adeno-associated virus in BLA glutamatergic neurons effectively improved anxiety-like behaviors. Overexpression of GPR17 in BLA glutamatergic neurons increased the susceptibility to anxiety-like behaviors. What's more, BLA glutamatergic neuronal activity was required for anxiolytic-like effects of GPR17 antagonist and GPR17 modulated anxiety-like behaviors via BLA to ventral hippocampal CA1 glutamatergic projection. Our study finds for the first and highlights the new role of GPR17 in regulating anxiety-like behaviors and it might be a novel potential target for therapy of anxiety disorders.
Collapse
Affiliation(s)
- Ruizhe Nie
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xinting Zhou
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaru Fu
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Shanshan Hu
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qilu Zhang
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Weikai Jiang
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yizi Yan
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xian Cao
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Danhua Yuan
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yan Long
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hao Hong
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Susu Tang
- Department of Pharmacology, College of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
9
|
Di Virgilio F, Vultaggio-Poma V, Tarantini M, Giuliani AL. Overview of the role of purinergic signaling and insights into its role in cancer therapy. Pharmacol Ther 2024; 262:108700. [PMID: 39111410 DOI: 10.1016/j.pharmthera.2024.108700] [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: 01/07/2024] [Revised: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024]
Abstract
Innovation of cancer therapy has received a dramatic acceleration over the last fifteen years thanks to the introduction of the novel immune checkpoint inhibitors (ICI). On the other hand, the conspicuous scientific knowledge accumulated in purinergic signaling since the early seventies is finally being transferred to the clinic. Several Phase I/II clinical trials are currently underway to investigate the effect of drugs interfering with purinergic signaling as stand-alone or combination therapy in cancer. This is supporting the novel concept of "purinergic immune checkpoint" (PIC) in cancer therapy. In the present review we will address a) the basic pharmacology and cell biology of the purinergic system; b) principles of its pathophysiology in human diseases; c) implications for cell death, cell proliferation and cancer; d) novel molecular tools to investigate nucleotide homeostasis in the extracellular environment; e) recent developments in the pharmacology of P1, P2 receptors and related ecto-enzymes; f) P1 and P2 ligands as novel diagnostic tools; g) current issues in PIC-based anti-cancer therapy. This review will provide an appraisal of the current status of purinergic signaling in cancer and will help identify future avenues of development.
Collapse
Affiliation(s)
| | | | - Mario Tarantini
- Department of Medical Sciences, University of Ferrara, Italy
| | | |
Collapse
|
10
|
Pietrantonio F, Serreqi A, Zerbe H, Svenningsson P, Aigner L. The leukotriene receptor antagonist montelukast as a potential therapeutic adjuvant in multiple sclerosis - a review. Front Pharmacol 2024; 15:1450493. [PMID: 39346564 PMCID: PMC11427386 DOI: 10.3389/fphar.2024.1450493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Multiple Sclerosis (MS) is a multifactorial autoimmune disease of the central nervous system (CNS). It is characterized by a heightened activation of the immune system with ensuing inflammation, demyelination and neurodegeneration with consequences such as motor, sensory, cognitive, as well as autonomic dysfunctions. While a range of immune-modulatory drugs have shown certain efficacy in alleviating pathology and symptoms, none of the currently available therapeutics regenerates the damaged CNS to restore function. There is emerging evidence for leukotrienes and leukotriene receptors being involved in the various aspects of the MS pathology including neuroinflammation and de/remyelination. Moreover, leukotriene receptor antagonists such as the asthma drug montelukast diminish inflammation and promote regeneration/remyelination. Indeed, montelukast has successfully been tested in animal models of MS and a recent retrospective case-control study suggests that montelukast treatment reduces relapses in patients with MS. Therefore, we propose montelukast as a therapeutic adjuvant to the standard immune-modulatory drugs with the potential to reduce pathology and promote structural and functional restoration. Here, we review the current knowledge on MS, its pathology, and on the potential of leukotriene receptor antagonists as therapeutics for MS.
Collapse
Affiliation(s)
| | | | | | - Per Svenningsson
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
11
|
Keifi Bajestani A, Alavi MS, Etemad L, Roohbakhsh A. Role of orphan G-protein coupled receptors in tissue ischemia: A comprehensive review. Eur J Pharmacol 2024; 978:176762. [PMID: 38906238 DOI: 10.1016/j.ejphar.2024.176762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/23/2024]
Abstract
Ischemic events lead to many diseases and deaths worldwide. Ischemia/reperfusion (I/R) occurs due to reduced blood circulation in tissues followed by blood reflow. Reoxygenation of ischemic tissues is characterized by oxidative stress, inflammation, energy distress, and endoplasmic reticulum stress. There are still no adequate clinical protocols or pharmacological approaches to address the consequences of I/R damage. G protein-coupled receptors (GPCRs) are important therapeutic targets. They compose a large family of seven transmembrane-spanning proteins that are involved in many biological functions. Orphan GPCRs are a large subgroup of these receptors expressed in different organs. In the present review, we summarized the literature regarding the role of orphan GPCRs in I/R in different organs. We focused on the effect of these receptors on modulating cellular and molecular processes underlying ischemia including apoptosis, inflammation, and autophagy. The study showed that GPR3, GPR4, GPR17, GPR30, GPR31, GPR35, GPR37, GPR39, GPR55, GPR65, GPR68, GPR75, GPR81, and GPR91 are involved in ischemic events, mainly in the brain and heart. These receptors offer new possibilities for treating I/R injuries in the body.
Collapse
Affiliation(s)
- Alireza Keifi Bajestani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Sood R, Anoopkumar-Dukie S, Rudrawar S, Hall S. Neuromodulatory effects of leukotriene receptor antagonists: A comprehensive review. Eur J Pharmacol 2024; 978:176755. [PMID: 38909933 DOI: 10.1016/j.ejphar.2024.176755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Cysteinyl leukotrienes (CysLTs) are central to the pathophysiology of asthma and various inflammatory disorders. Leukotriene receptor antagonists (LTRAs) effectively treat respiratory conditions by targeting cysteinyl leukotriene receptors, CysLT1 and CysLT2 subtypes. This review explores the multifaceted effects of LTs, extending beyond bronchoconstriction. CysLT receptors are not only present in the respiratory system but are also crucial in neuronal signaling pathways. LTRAs modulate these receptors, influencing downstream signaling, calcium levels, inflammation, and oxidative stress (OS) within neurons hinting at broader implications. Recent studies identify novel molecular targets, sparking interest in repurposing LTRAs for therapeutic use. Clinical trials are investigating their potential in neuroinflammation control, particularly in Alzheimer's disease (AD) and Parkinson's diseases (PD). However, montelukast, a long-standing LTRA since 1998, raises concerns due to neuropsychiatric adverse drug reactions (ADRs). Despite widespread use, understanding montelukast's metabolism and underlying ADR mechanisms remains limited. This review comprehensively examines LTRAs' diverse biological effects, emphasizing non-bronchoconstrictive activities. It also analyses plausible mechanisms behind LTRAs' neuronal effects, offering insights into their potential as neurodegenerative disease modulators. The aim is to inform clinicians, researchers, and pharmaceutical developers about LTRAs' expanding roles, particularly in neuroinflammation control and their promising repurposing for neurodegenerative disease management.
Collapse
Affiliation(s)
- Radhika Sood
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia
| | | | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia; Institute for Glycomics, Griffith University, Queensland, 4222, Australia
| | - Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia.
| |
Collapse
|
13
|
Bergstrom JJD, Fu MM. Dysregulation of myelination-related genes in schizophrenia. J Neurochem 2024; 168:2227-2242. [PMID: 39086020 PMCID: PMC11449665 DOI: 10.1111/jnc.16152] [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: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024]
Abstract
Schizophrenic individuals display disrupted myelination patterns, altered oligodendrocyte distribution, and abnormal oligodendrocyte morphology. Schizophrenia is linked with dysregulation of a variety of genes involved in oligodendrocyte function and myelin production. Single-nucleotide polymorphisms (SNPs) and rare mutations in myelination-related genes are observed in certain schizophrenic populations, representing potential genetic risk factors. Downregulation of myelination-related RNAs and proteins, particularly in frontal and limbic regions, is consistently associated with the disorder across multiple studies. These findings support the notion that disruptions in myelination may contribute to the cognitive and behavioral impairments experienced in schizophrenia, although further evidence of causation is needed.
Collapse
Affiliation(s)
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
14
|
Wang Y, Yu Z, Cheng M, Hu E, Yan Q, Zheng F, Guo X, Zhang W, Li H, Li Z, Zhu W, Wu Y, Tang T, Li T. Buyang huanwu decoction promotes remyelination via miR-760-3p/GPR17 axis after intracerebral hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118126. [PMID: 38556140 DOI: 10.1016/j.jep.2024.118126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/02/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The repairment of myelin sheaths is crucial for mitigating neurological impairments of intracerebral hemorrhage (ICH). However, the current research on remyelination processes in ICH remains limited. A representative traditional Chinese medicine, Buyang Huanwu decoction (BYHWD), shows a promising therapeutic strategy for ICH treatment. AIM OF THE STUDY To investigate the pro-remyelination effects of BYHWD on ICH and explore the underlying mechanisms. MATERIALS AND METHODS The collagenase-induced mice ICH model was created for investigation. BYHWD's protective effects were assessed by behavioral tests and histological staining. Transmission electron microscopy was used for displaying the structure of myelin sheaths. The remyelination and oligodendrocyte differentiation were evaluated by the expressions of myelin proteolipid protein (PLP), myelin basic protein (MBP), MBP/TAU, Olig2/CC1, and PDGFRα/proliferating cell nuclear antigen (PCNA) through RT-qPCR and immunofluorescence. Transcriptomics integrated with disease database analysis and experiments in vivo and in vitro revealed the microRNA-related underlying mechanisms. RESULTS Here, we reported that BYHWD promoted the neurological function of ICH mice and improved remyelination by increasing PLP, MBP, and TAU, as well as restoring myelin structure. Besides, we showed that BYHWD promoted remyelination by boosting the differentiation of PDGFRα+ oligodendrocyte precursor cells into olig2+/CC1+ oligodendrocytes. Additionally, we demonstrated that the remyelination effects of BYHWD worked by inhibiting G protein-coupled receptor 17 (GPR17). miRNA sequencing integrated with miRNA database prediction screened potential miRNAs targeting GPR17. By applying immunofluorescence, RNA in situ hybridization and dual luciferase reporter gene assay, we confirmed that BYHWD suppressed GPR17 and improved remyelination by increasing miR-760-3p. CONCLUSIONS BYHWD improves remyelination and neurological function in ICH mice by targeting miR-760-3p to inhibit GPR17. This study may shed light on the orchestration of remyelination mechanisms after ICH, thus providing novel insights for developing innovative prescriptions with brain-protective properties.
Collapse
Affiliation(s)
- Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Zhe Yu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Menghan Cheng
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - En Hu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Qiuju Yan
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Fei Zheng
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Xiaohang Guo
- School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Haigang Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, PR China
| | - Zhilin Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Wenxin Zhu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yao Wu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Tao Tang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Teng Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China.
| |
Collapse
|
15
|
Öz-Arslan D, Yavuz M, Kan B. Exploring orphan GPCRs in neurodegenerative diseases. Front Pharmacol 2024; 15:1394516. [PMID: 38895631 PMCID: PMC11183337 DOI: 10.3389/fphar.2024.1394516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Neurodegenerative disorders represent a significant and growing health burden worldwide. Unfortunately, limited therapeutic options are currently available despite ongoing efforts. Over the past decades, research efforts have increasingly focused on understanding the molecular mechanisms underlying these devastating conditions. Orphan receptors, a class of receptors with no known endogenous ligands, emerge as promising druggable targets for diverse diseases. This review aims to direct attention to a subgroup of orphan GPCRs, in particular class A orphans that have roles in neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Multiple sclerosis. We highlight the diverse roles orphan receptors play in regulating critical cellular processes such as synaptic transmission, neuronal survival and neuro-inflammation. Moreover, we discuss the therapeutic potential of targeting orphan receptors for the treatment of neurodegenerative disorders, emphasizing recent advances in drug discovery and preclinical studies. Finally, we outline future directions and challenges in orphan receptor research.
Collapse
Affiliation(s)
- Devrim Öz-Arslan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| | - Melis Yavuz
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
- Department of Pharmacology, Acibadem MAA University, School of Pharmacy, Istanbul, Türkiye
| | - Beki Kan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| |
Collapse
|
16
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
17
|
Birgül Iyison N, Abboud C, Abboud D, Abdulrahman AO, Bondar AN, Dam J, Georgoussi Z, Giraldo J, Horvat A, Karoussiotis C, Paz-Castro A, Scarpa M, Schihada H, Scholz N, Güvenc Tuna B, Vardjan N. ERNEST COST action overview on the (patho)physiology of GPCRs and orphan GPCRs in the nervous system. Br J Pharmacol 2024. [PMID: 38825750 DOI: 10.1111/bph.16389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 06/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that play a critical role in nervous system function by transmitting signals between cells and their environment. They are involved in many, if not all, nervous system processes, and their dysfunction has been linked to various neurological disorders representing important drug targets. This overview emphasises the GPCRs of the nervous system, which are the research focus of the members of ERNEST COST action (CA18133) working group 'Biological roles of signal transduction'. First, the (patho)physiological role of the nervous system GPCRs in the modulation of synapse function is discussed. We then debate the (patho)physiology and pharmacology of opioid, acetylcholine, chemokine, melatonin and adhesion GPCRs in the nervous system. Finally, we address the orphan GPCRs, their implication in the nervous system function and disease, and the challenges that need to be addressed to deorphanize them.
Collapse
Affiliation(s)
- Necla Birgül Iyison
- Department of Molecular Biology and Genetics, University of Bogazici, Istanbul, Turkey
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | | | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Magurele, Romania
- Forschungszentrum Jülich, Institute for Computational Biomedicine (IAS-5/INM-9), Jülich, Germany
| | - Julie Dam
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Christos Karoussiotis
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Alba Paz-Castro
- Molecular Pharmacology of GPCRs research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago, Spain
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilge Güvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
18
|
Golia MT, Frigerio R, Pucci S, Sironi F, Margotta C, Pasetto L, Testori C, Berrone E, Ingravalle F, Chiari M, Gori A, Duchi R, Perota A, Bergamaschi L, D'Angelo A, Cagnotti G, Galli C, Corona C, Bonetto V, Bendotti C, Cretich M, Colombo SF, Verderio C. Changes in glial cell activation and extracellular vesicles production precede the onset of disease symptoms in transgenic hSOD1 G93A pigs. Exp Neurol 2024; 374:114716. [PMID: 38331161 DOI: 10.1016/j.expneurol.2024.114716] [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: 10/13/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
SOD1 gene is associated with progressive motor neuron degeneration in the familiar forms of amyotrophic lateral sclerosis. Although studies on mutant human SOD1 transgenic rodent models have provided important insights into disease pathogenesis, they have not led to the discovery of early biomarkers or effective therapies in human disease. The recent generation of a transgenic swine model expressing the human pathological hSOD1G93A gene, which recapitulates the course of human disease, represents an interesting tool for the identification of early disease mechanisms and diagnostic biomarkers. Here, we analyze the activation state of CNS cells in transgenic pigs during the disease course and investigate whether changes in neuronal and glial cell activation state can be reflected by the amount of extracellular vesicles they release in biological fluids. To assess the activation state of neural cells, we performed a biochemical characterization of neurons and glial cells in the spinal cords of hSOD1G93A pigs during the disease course. Quantification of EVs of CNS cell origin was performed in cerebrospinal fluid and plasma of transgenic pigs at different disease stages by Western blot and peptide microarray analyses. We report an early activation of oligodendrocytes in hSOD1G93A transgenic tissue followed by astrocyte and microglia activation, especially in animals with motor symptoms. At late asymptomatic stage, EV production from astrocytes and microglia is increased in the cerebrospinal fluid, but not in the plasma, of transgenic pigs reflecting donor cell activation in the spinal cord. Estimation of EV production by biochemical analyses is corroborated by direct quantification of neuron- and microglia-derived EVs in the cerebrospinal fluid by a Membrane Sensing Peptide enabled on-chip analysis that provides fast results and low sample consumption. Collectively, our data indicate that alteration in astrocytic EV production precedes the onset of disease symptoms in the hSODG93A swine model, mirroring donor cell activation in the spinal cord, and suggest that EV measurements from the cells first activated in the ALS pig model, i.e. OPCs, may further improve early disease detection.
Collapse
Affiliation(s)
- Maria Teresa Golia
- National Research Council of Italy, Institute of Neuroscience (IN-CNR), Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| | - Roberto Frigerio
- National Research Council of Italy, Institute of Chemical Science and Technologies (SCITEC-CNR), Via Mario Bianco 9, 20131 Milan, Italy
| | - Susanna Pucci
- National Research Council of Italy, Institute of Neuroscience (IN-CNR), Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| | - Francesca Sironi
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156 Milano, Italy
| | - Cassandra Margotta
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156 Milano, Italy
| | - Laura Pasetto
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156 Milano, Italy
| | - Camilla Testori
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta (IZSPLV), Via Bologna 148, 10154 Torino, Italy
| | - Elena Berrone
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta (IZSPLV), Via Bologna 148, 10154 Torino, Italy
| | - Francesco Ingravalle
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta (IZSPLV), Via Bologna 148, 10154 Torino, Italy
| | - Marcella Chiari
- National Research Council of Italy, Institute of Chemical Science and Technologies (SCITEC-CNR), Via Mario Bianco 9, 20131 Milan, Italy
| | - Alessandro Gori
- National Research Council of Italy, Institute of Chemical Science and Technologies (SCITEC-CNR), Via Mario Bianco 9, 20131 Milan, Italy
| | - Roberto Duchi
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Andrea Perota
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Luca Bergamaschi
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Antonio D'Angelo
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy
| | - Giulia Cagnotti
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy
| | - Cesare Galli
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta (IZSPLV), Via Bologna 148, 10154 Torino, Italy
| | - Valentina Bonetto
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156 Milano, Italy
| | - Caterina Bendotti
- Research Center for ALS, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri, 2, 20156 Milano, Italy
| | - Marina Cretich
- National Research Council of Italy, Institute of Chemical Science and Technologies (SCITEC-CNR), Via Mario Bianco 9, 20131 Milan, Italy
| | - Sara Francesca Colombo
- National Research Council of Italy, Institute of Neuroscience (IN-CNR), Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| | - Claudia Verderio
- National Research Council of Italy, Institute of Neuroscience (IN-CNR), Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy.
| |
Collapse
|
19
|
Muluhie M, Castiglioni L, Rzemieniec J, Mercuriali B, Gelosa P, Sironi L. Montelukast, an available and safe anti-asthmatic drug, prevents maladaptive remodelling and maintains cardiac functionality following myocardial infarction. Sci Rep 2024; 14:3371. [PMID: 38337010 PMCID: PMC10858037 DOI: 10.1038/s41598-024-53936-x] [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: 01/02/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Preclinical and clinical data indicate that the 5-lipoxygenase pathway becomes activated in cardiovascular diseases suggesting an important role of CysLTs in atherosclerosis and in its ischemic complications. This study aims to investigate the effects of montelukast, a CysLTR-1 antagonist, in a mouse model of myocardial infarction (MI). C57BL/6N female mice were subjected to coronary artery ligation and received montelukast (10 mg/kg/day, intraperitoneal) or vehicle. Montelukast exerted beneficial effects in the infarcted area, decreasing mRNA expression of inflammatory genes, such Il1β and Ccl2 (p < 0.05), at 48 h after MI, and reducing infarct size and preventing ischemic wall thinning (p < 0.05) at 4 weeks. Furthermore, montelukast counteracted maladaptive remodelling of whole heart. Indeed, montelukast reduced LV mass (p < 0.05) and remote wall thickening (p < 0.05), and improved cardiac pumping function, as evidenced by increased global ejection fraction (p < 0.01), and regional contractility in infarcted (p < 0.05) and in remote non-infarcted (p < 0.05) myocardium. Finally, montelukast prevented cardiomyocytes hypertrophy (p < 0.05) in remote myocardium, reducing the phosphorylation of GSK3β, a regulator of hypertrophic pathway (p < 0.05). Our data strongly demonstrate the ability of montelukast to contrast the MI-induced maladaptive conditions, thus sustaining cardiac contractility. The results provide evidences for montelukast "repurposing" in cardiovascular diseases and in particular in myocardial infarction.
Collapse
Affiliation(s)
- Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Benedetta Mercuriali
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
20
|
Fang M, Chen L, Tang T, Qiu M, Xu X. The committed oligodendrocyte precursor cell, a newly-defined intermediate progenitor cell type in oligodendroglial lineage. Glia 2023; 71:2499-2510. [PMID: 37278537 DOI: 10.1002/glia.24426] [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: 03/16/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/07/2023]
Abstract
In the central nervous system, oligodendrocytes (OLs) produce myelin sheaths that provide trophic support to neuronal axons and increase the propagation speed of action potential. OLs are constantly generated from OL precursor cells (OPCs) throughout life span. The production of myelinating OLs consists of three canonical stages: OPCs, newly-formed OLs (NFOs), and mature myelinating OLs. Recently, single-cell RNA transcriptomic analyses identified a new population of oligodendroglial cells, namely differentiation committed OPCs (COPs). COPs represent a critical intermediate population between OPCs and NFOs, as revealed by specific expression of G-protein coupled receptor 17 (GPR17). The dysregulation of COPs leads to the remyelination failure in demyelinating diseases and impairs the replacement of lost myelin sheaths due to aging. Hence, understanding the development of COPs and their underlying regulatory network will be helpful in establishing new strategies for promoting myelin repair in demyelinating diseases. This review summarizes the current knowledge on the development and functions of COPs under both physiological and pathological conditions. Overall, COPs function as "checkpoints" to prevent inappropriate precocious OL differentiation and myelination through expressing distinct regulatory factors. Deepening our understanding of COPs may not only advance our knowledge of how OL lineage progresses during development, but also open the door to new treatments for demyelinating diseases.
Collapse
Affiliation(s)
- Minxi Fang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Lixia Chen
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Tao Tang
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
- School of Basic Medical Science, Hangzhou Normal University, Hangzhou, China
| | - Xiaofeng Xu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
21
|
Boccazzi M, Macchiarulo G, Lebon S, Janowska J, Le Charpentier T, Faivre V, Hua J, Marangon D, Lecca D, Fumagalli M, Mani S, Abbracchio MP, Gressens P, Schang AL, Van Steenwinckel J. G protein-coupled receptor 17 is regulated by WNT pathway during oligodendrocyte precursor cell differentiation. Neurobiol Dis 2023; 187:106315. [PMID: 37783234 DOI: 10.1016/j.nbd.2023.106315] [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: 07/21/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
G protein-coupled receptor 17 (GPR17) and the WNT pathway are critical players of oligodendrocyte (OL) differentiation acting as essential timers in developing brain to achieve fully-myelinating cells. However, whether and how these two systems are related to each other is still unknown. Of interest, both factors are dysregulated in developing and adult brain diseases, including white matter injury and cancer, making the understanding of their reciprocal interactions of potential importance for identifying new targets and strategies for myelin repair. Here, by a combined pharmacological and biotechnological approach, we examined regulatory mechanisms linking WNT signaling to GPR17 expression in OLs. We first analyzed the relative expression of mRNAs encoding for GPR17 and the T cell factor/Lymphoid enhancer-binding factor-1 (TCF/LEF) transcription factors of the canonical WNT/β-CATENIN pathway, in PDGFRα+ and O4+ OLs during mouse post-natal development. In O4+ cells, Gpr17 mRNA level peaked at post-natal day 14 and then decreased concomitantly to the physiological uprise of WNT tone, as shown by increased Lef1 mRNA level. The link between WNT signaling and GPR17 expression was further reinforced in vitro in primary PDGFRα+ cells and in Oli-neu cells. High WNT tone impaired OL differentiation and drastically reduced GPR17 mRNA and protein levels. In Oli-neu cells, WNT/β-CATENIN activation repressed Gpr17 promoter activity through both putative WNT response elements (WRE) and upregulation of the inhibitor of DNA-binding protein 2 (Id2). We conclude that the WNT pathway influences OL maturation by repressing GPR17, which could have implications in pathologies characterized by dysregulations of the OL lineage including multiple sclerosis and oligodendroglioma.
Collapse
Affiliation(s)
- Marta Boccazzi
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | | | - Sophie Lebon
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Justyna Janowska
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Jennifer Hua
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, 20133 Milan, Italy
| | - Shyamala Mani
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Anne-Laure Schang
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Université Paris Cité, UMR 1153 CRESS, Paris, France.
| | | |
Collapse
|
22
|
Boccazzi M, Raffaele S, Zanettin T, Abbracchio MP, Fumagalli M. Altered Purinergic Signaling in Neurodevelopmental Disorders: Focus on P2 Receptors. Biomolecules 2023; 13:biom13050856. [PMID: 37238724 DOI: 10.3390/biom13050856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
With the umbrella term 'neurodevelopmental disorders' (NDDs) we refer to a plethora of congenital pathological conditions generally connected with cognitive, social behavior, and sensory/motor alterations. Among the possible causes, gestational and perinatal insults have been demonstrated to interfere with the physiological processes necessary for the proper development of fetal brain cytoarchitecture and functionality. In recent years, several genetic disorders caused by mutations in key enzymes involved in purine metabolism have been associated with autism-like behavioral outcomes. Further analysis revealed dysregulated purine and pyrimidine levels in the biofluids of subjects with other NDDs. Moreover, the pharmacological blockade of specific purinergic pathways reversed the cognitive and behavioral defects caused by maternal immune activation, a validated and now extensively used rodent model for NDDs. Furthermore, Fragile X and Rett syndrome transgenic animal models as well as models of premature birth, have been successfully utilized to investigate purinergic signaling as a potential pharmacological target for these diseases. In this review, we examine results on the role of the P2 receptor signaling in the etiopathogenesis of NDDs. On this basis, we discuss how this evidence could be exploited to develop more receptor-specific ligands for future therapeutic interventions and novel prognostic markers for the early detection of these conditions.
Collapse
Affiliation(s)
- Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Stefano Raffaele
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Thomas Zanettin
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
23
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
24
|
Kari S, Kandhavelu J, Murugesan A, Thiyagarajan R, Kidambi S, Kandhavelu M. Mitochondrial complex III bypass complex I to induce ROS in GPR17 signaling activation in GBM. Biomed Pharmacother 2023; 162:114678. [PMID: 37054539 DOI: 10.1016/j.biopha.2023.114678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/15/2023] Open
Abstract
Guanine nucleotide binding protein (G protein) coupled receptor 17 (GPR17) plays crucial role in Glioblastoma multiforme (GBM) cell signaling and is primarily associated with reactive oxidative species (ROS) production and cell death. However, the underlying mechanisms by which GPR17 regulates ROS level and mitochondrial electron transport chain (ETC) complexes are still unknown. Here, we investigate the novel link between the GPR17 receptor and ETC complex I and III in regulating level of intracellular ROS (ROSi) in GBM using pharmacological inhibitors and gene expression profiling. Incubation of 1321N1 GBM cells with ETC I inhibitor and GPR17 agonist decreased the ROS level, while treatment with GPR17 antagonist increased the ROS level. Also, inhibition of ETC III and activation of GPR17 increased the ROS level whereas opposite function was observed with antagonist interaction. The similar functional role was also observed in multiple GBM cells, LN229 and SNB19, where ROS level increased in the presence of Complex III inhibitor. The level of ROS varies in Complex I inhibitor and GPR17 antagonist treatment conditions suggesting that ETC I function differs depending on the GBM cell line. RNAseq analysis revealed that ∼ 500 genes were commonly expressed in both SNB19 and LN229, in which 25 genes are involved in ROS pathway. Furthermore, 33 dysregulated genes were observed to be involved in mitochondria function and 36 genes of complex I-V involved in ROS pathway. Further analysis revealed that induction of GPR17 leads to loss of function of NADH dehydrogenase genes involved in ETC I, while cytochrome b and Ubiquinol Cytochrome c Reductase family genes in ETC III. Overall, our findings suggest that mitochondrial ETC III bypass ETC I to increase ROSi in GPR17 signaling activation in GBM and could provide new opportunities for developing targeted therapy for GBM.
Collapse
Affiliation(s)
- Sana Kari
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, ArvoYlpönkatu 34, 33520 Tampere, Finland; Science Center, Tampere University Hospital, ArvoYlpönkatu 34, 33520 Tampere, Finland
| | - Jeyalakshmi Kandhavelu
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, ArvoYlpönkatu 34, 33520 Tampere, Finland; Science Center, Tampere University Hospital, ArvoYlpönkatu 34, 33520 Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, ArvoYlpönkatu 34, 33520 Tampere, Finland; Science Center, Tampere University Hospital, ArvoYlpönkatu 34, 33520 Tampere, Finland; Department of Biotechnology, Lady Doak College, Thallakulam, Madurai 625002, India
| | - Ramesh Thiyagarajan
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Kingdom of Saudi Arabia
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, ArvoYlpönkatu 34, 33520 Tampere, Finland; Science Center, Tampere University Hospital, ArvoYlpönkatu 34, 33520 Tampere, Finland.
| |
Collapse
|
25
|
Nicholas RA. Commentary on 'Identification and characterization of select oxysterols as ligands for GPR17'. Br J Pharmacol 2023; 180:1186-1187. [PMID: 36669769 DOI: 10.1111/bph.16026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/22/2023] Open
Affiliation(s)
- Robert A Nicholas
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
26
|
Chapman TW, Olveda GE, Bame X, Pereira E, Hill RA. Oligodendrocyte death initiates synchronous remyelination to restore cortical myelin patterns in mice. Nat Neurosci 2023; 26:555-569. [PMID: 36928635 PMCID: PMC10208560 DOI: 10.1038/s41593-023-01271-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/09/2023] [Indexed: 03/18/2023]
Abstract
Myelin degeneration occurs in neurodegenerative diseases and aging. In these conditions, resident oligodendrocyte progenitor cells (OPCs) differentiate into oligodendrocytes that carry out myelin repair. To investigate the cellular dynamics underlying these events, we developed a noninflammatory demyelination model that combines intravital two-photon imaging with a single-cell ablation technique called two-photon apoptotic targeted ablation (2Phatal). Oligodendrocyte 2Phatal in both sexes results in a myelin degeneration cascade that triggers rapid forms of synchronous remyelination on defined axons. This remyelination is driven by oligodendrocytes differentiated from a subset of morphologically distinct, highly branched OPCs. Moreover, remyelination efficiency depends on the initial myelin patterns, as well as the age of the organism. In summary, using 2Phatal, we show a form of rapid synchronous remyelination, mediated by a distinct subset of OPCs, capable of restoring the original myelin patterning in adulthood but not aging.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Genaro E Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Xhoela Bame
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Elizabeth Pereira
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
27
|
Wang X, Baskaran L, Chan M, Boisvert W, Hausenloy DJ. Targeting leukotriene biosynthesis to prevent atherosclerotic cardiovascular disease. CONDITIONING MEDICINE 2023; 6:33-41. [PMID: 38800614 PMCID: PMC11126214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death and disability worldwide. As such, new treatments are needed to prevent the onset and progression of atherosclerosis to improve outcomes in patients with coronary, cerebrovascular, and peripheral arterial disease. In this regard, inflammation is known to be a critical driver of atherosclerosis formation and progression, thus it is a viable target for vascular protection in patients at risk of developing ASCVD. Leukotrienes, key pro-inflammatory lipid mediators derived from arachidonic acid, are associated with atheroma inflammation and progression. Genetic mutations in key components of the leukotriene synthesis pathway, such as 5-lipoxygenase (5-LO) and 5-lipoxygenase-activating protein (FLAP), are associated with an increased risk of cardiovascular disease, and pharmacological inhibition of 5-LO and FLAP has been reported to prevent atheroma formation in pre-clinical and early clinical studies. In this article, we provide an overview of these studies and highlight the therapeutic potential of targeting leukotriene synthesis to prevent atheroma inflammation and progression and improve outcomes in patients at risk of ASCVD.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | | | - Mark Chan
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| | - William Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Yong Loo Lin Medical School, National University of Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
| |
Collapse
|
28
|
Konda Mani S, Thiyagarajan R, Yli-Harja O, Kandhavelu M, Murugesan A. Structural analysis of human G-protein-coupled receptor 17 ligand binding sites. J Cell Biochem 2023; 124:533-544. [PMID: 36791278 DOI: 10.1002/jcb.30388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/17/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023]
Abstract
The human G protein coupled membrane receptor (GPR17), the sensor of brain damage, is identified as a biomarker for many neurological diseases. In human brain tissue, GPR17 exist in two isoforms, long and short. While cryo-electron microscopy technology has provided the structure of the long isoform of GPR17 with Gi complex, the structure of the short isoform and its activation mechanism remains unclear. Recently, we theoretically modeled the structure of the short isoform of GPR17 with Gi signaling protein and identified novel ligands. In the present work, we demonstrated the presence of two distinct ligand binding sites in the short isoform of GPR17. The molecular docking of GPR17 with endogenous (UDP) and synthetic ligands (T0510.3657, MDL29950) found the presence of two distinct binding pockets. Our observations revealed that endogenous ligand UDP can bind stronger in two different binding pockets as evidenced by glide and autodock vina scores, whereas the other two ligand's binding with GPR17 has less docking score. The analysis of receptor-UDP interactions shows complexes' stability in the lipid environment by 100 ns atomic molecular dynamics simulations. The amino acid residues VAL83, ARG87, and PHE111 constitute ligand binding site 1, whereas site 2 constitutes ASN67, ARG129, and LYS232. Root mean square fluctuation analysis showed the residues 83, 87, and 232 with higher fluctuations during molecular dynamics simulation in both binding pockets. Our findings imply that the residues of GPR17's two binding sites are crucial, and their interaction with UDP reveals the protein's hidden signaling and communication properties. Furthermore, this finding may assist in the development of targeted therapies for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Saravanan Konda Mani
- Department of Biotechnology, Bharath Institute of Higher Education & Research, Chennai, Tamilnadu, India
| | - Ramesh Thiyagarajan
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Olli Yli-Harja
- Computaional Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute for Systems Biology, Seattle, Washington, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMeditech and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Akshaya Murugesan
- BioMeditech and Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Department of Biotechnology, Lady Doak College, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
29
|
Korbecki J, Rębacz-Maron E, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis. Cancers (Basel) 2023; 15:cancers15030946. [PMID: 36765904 PMCID: PMC9913267 DOI: 10.3390/cancers15030946] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive gliomas. New and more effective therapeutic approaches are being sought based on studies of the various mechanisms of GBM tumorigenesis, including the synthesis and metabolism of arachidonic acid (ARA), an omega-6 polyunsaturated fatty acid (PUFA). PubMed, GEPIA, and the transcriptomics analysis carried out by Seifert et al. were used in writing this paper. In this paper, we discuss in detail the biosynthesis of this acid in GBM tumors, with a special focus on certain enzymes: fatty acid desaturase (FADS)1, FADS2, and elongation of long-chain fatty acids family member 5 (ELOVL5). We also discuss ARA metabolism, particularly its release from cell membrane phospholipids by phospholipase A2 (cPLA2, iPLA2, and sPLA2) and its processing by cyclooxygenases (COX-1 and COX-2), lipoxygenases (5-LOX, 12-LOX, 15-LOX-1, and 15-LOX-2), and cytochrome P450. Next, we discuss the significance of lipid mediators synthesized from ARA in GBM cancer processes, including prostaglandins (PGE2, PGD2, and 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2)), thromboxane A2 (TxA2), oxo-eicosatetraenoic acids, leukotrienes (LTB4, LTC4, LTD4, and LTE4), lipoxins, and many others. These lipid mediators can increase the proliferation of GBM cancer cells, cause angiogenesis, inhibit the anti-tumor response of the immune system, and be responsible for resistance to treatment.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ewa Rębacz-Maron
- Department of Ecology and Anthropology, Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Correspondence: ; Tel.: +48-914-661-515
| |
Collapse
|
30
|
Harrington AW, Liu C, Phillips N, Nepomuceno D, Kuei C, Chang J, Chen W, Sutton SW, O'Malley D, Pham L, Yao X, Sun S, Bonaventure P. Identification and characterization of select oxysterols as ligands for GPR17. Br J Pharmacol 2023; 180:401-421. [PMID: 36214386 DOI: 10.1111/bph.15969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE G-protein coupled receptor 17 (GPR17) is an orphan receptor involved in the process of myelination, due to its ability to inhibit the maturation of oligodendrocyte progenitor cells (OPCs) into myelinating oligodendrocytes. Despite multiple claims that the biological ligand has been identified, it remains an orphan receptor. EXPERIMENTAL APPROACH Seventy-seven oxysterols were screened in a cell-free [35 S]GTPγS binding assay using membranes from cells expressing GPR17. The positive hits were characterized using adenosine 3',5' cyclic monophosphate (cAMP), inositol monophosphate (IP1) and calcium mobilization assays, with results confirmed in rat primary oligodendrocytes. Rat and pig brain extracts were separated by high-performance liquid chromatography (HPLC) and endogenous activator(s) were identified in receptor activation assays. Gene expression studies of GPR17, and CYP46A1 (cytochrome P450 family 46 subfamily A member 1) enzymes responsible for the conversion of cholesterol into specific oxysterols, were performed using quantitative real-time PCR. KEY RESULTS Five oxysterols were able to stimulate GPR17 activity, including the brain cholesterol, 24(S)-hydroxycholesterol (24S-HC). A specific brain fraction from rat and pig extracts containing 24S-HC activates GPR17 in vitro. Expression of Gpr17 during mouse brain development correlates with the expression of Cyp46a1 and the levels of 24S-HC itself. Other active oxysterols have low brain concentrations below effective ranges. CONCLUSIONS AND IMPLICATIONS Oxysterols, including but not limited to 24S-HC, could be physiological activators for GPR17 and thus potentially regulate OPC differentiation and myelination through activation of the receptor.
Collapse
Affiliation(s)
| | - Changlu Liu
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Naomi Phillips
- Janssen Research & Development, LLC, San Diego, California, USA
| | | | - Chester Kuei
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Joseph Chang
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Weixuan Chen
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Steven W Sutton
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Daniel O'Malley
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Ly Pham
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Xiang Yao
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Siquan Sun
- Janssen Research & Development, LLC, San Diego, California, USA
| | | |
Collapse
|
31
|
Murugesan A, Nguyen P, Ramesh T, Yli-Harja O, Kandhavelu M, Saravanan KM. Molecular modeling and dynamics studies of the synthetic small molecule agonists with GPR17 and P2Y1 receptor. J Biomol Struct Dyn 2022; 40:12908-12916. [PMID: 34542380 DOI: 10.1080/07391102.2021.1977707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The human Guanine Protein coupled membrane Receptor 17 (hGPR17), an orphan receptor that activates uracil nucleotides and cysteinyl leukotrienes is considered as a crucial target for the neurodegenerative diseases. Yet, the detailed molecular interaction of potential synthetic ligands of GPR17 needs to be characterized. Here, we have studied a comparative analysis on the interaction specificity of GPR17-ligands with hGPR17 and human purinergic G protein-coupled receptor (hP2Y1) receptors. Previously, we have simulated the interaction stability of synthetic ligands such as T0510.3657, AC1MLNKK, and MDL29951 with hGPR17 and hP2Y1 receptor in the lipid environment. In the present work, we have comparatively studied the protein-ligand interaction of hGPR17-T0510.3657 and P2Y1-MRS2500. Sequence analysis and structural superimposition of hGPR17 and hP2Y1 receptor revealed the similarities in the structural arrangement with the local backbone root mean square deviation (RMSD) value of 1.16 Å and global backbone RMSD value of 5.30 Å. The comparative receptor-ligand interaction analysis between hGPR17 and hP2Y1 receptor exposed the distinct binding sites in terms of geometrical properties. Further, the molecular docking of T0510.3657 with the hP2Y1 receptor have shown non-specific interaction. The experimental validation also revealed that Gi-coupled activation of GPR17 by specific ligands leads to the adenylyl cyclase inhibition, while there is no inhibition upon hP2Y1 activation. Overall, the above findings suggest that T0510.3657-GPR17 binding specificity could be further explored for the treatment of numerous neuronal diseases. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, India
| | - Phung Nguyen
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj, Kingdom of Saudi Arabia
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute for Systems Biology, Seattle, WA, USA
| | | | - Konda Mani Saravanan
- Scigen Research and Innovation Pvt Ltd, Periyar Technology Business Incubator, Thanjavur, Tamil Nadu, India
| |
Collapse
|
32
|
Daniele S, Saporiti S, Capaldi S, Pietrobono D, Russo L, Guerrini U, Laurenzi T, Ataie Kachoie E, Palazzolo L, Russo V, Abbracchio MP, Eberini I, Trincavelli ML. Functional Heterodimerization between the G Protein-Coupled Receptor GPR17 and the Chemokine Receptors 2 and 4: New Evidence. Int J Mol Sci 2022; 24:261. [PMID: 36613703 PMCID: PMC9820414 DOI: 10.3390/ijms24010261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
GPR17, a G protein-coupled receptor, is a pivotal regulator of myelination. Its endogenous ligands trigger receptor desensitization and downregulation allowing oligodendrocyte terminal maturation. In addition to its endogenous agonists, GPR17 could be promiscuously activated by pro-inflammatory oxysterols and chemokines released at demyelinating lesions. Herein, the chemokine receptors CXCR2 and CXCR4 were selected to perform both in silico modelling and in vitro experiments to establish their structural and functional interactions with GPR17. The relative propensity of GPR17 and CXCR2 or CXCR4 to form homo- and hetero-dimers was assessed by homology modelling and molecular dynamics (MD) simulations, and co-immunoprecipitation and immunoenzymatic assay. The interaction between chemokine receptors and GPR17 was investigated by determining receptor-mediated modulation of intracellular cyclic adenosine monophosphate (cAMP). Our data show the GPR17 association with CXCR2 or CXCR4 and the negative regulation of these interactions by CXCR agonists or antagonists. Moreover, GPR17 and CXCR2 heterodimers can functionally influence each other. In contrast, CXCR4 can influence GPR17 functionality, but not vice versa. According to MD simulations, all the dimers reached conformational stability and negative formation energy, confirming the experimental observations. The cross-talk between these receptors could play a role in the development of the neuroinflammatory milieu associated with demyelinating events.
Collapse
Affiliation(s)
- Simona Daniele
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Saporiti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Stefano Capaldi
- Dipartimento di Biotecnologie, Università degli Studi di Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Deborah Pietrobono
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Lara Russo
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Tommaso Laurenzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Elham Ataie Kachoie
- Dipartimento di Biotecnologie, Università degli Studi di Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Vincenzo Russo
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology Scientific, Institute San Raffaele, 20132 Milan, Italy
| | - Maria Pia Abbracchio
- Laboratorio di Farmacologia Molecolare e Cellulare Della Trasmissione Purinergica, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari & Data Science Research Center (DSRC), Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | | |
Collapse
|
33
|
Ye F, Wong T, Chen G, Zhang Z, Zhang B, Gan S, Gao W, Li J, Wu Z, Pan X, Du Y. Cryo-EM structure of G-protein-coupled receptor GPR17 in complex with inhibitory G protein. MedComm (Beijing) 2022; 3:e159. [PMID: 36105372 PMCID: PMC9464062 DOI: 10.1002/mco2.159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/11/2022] Open
Abstract
GPR17 is a class A orphan G protein-coupled receptor (GPCR) expressed in neurons and oligodendrocyte progenitors of the central nervous system (CNS). The signalling of GPR17 occurs through the heterotrimeric Gi, but its activation mechanism is unclear. Here, we employed cryo-electron microscopy (cryo-EM) technology to elucidate the structure of activated GPR17-Gi complex. The 3.02 Å resolution structure, together with mutagenesis studies, revealed that the extracellular loop2 of GPR17 occupied the orthosteric binding pocket to promote its self-activation. The active GPR17 carried several typical microswitches like other class A GPCRs. Moreover, the Gi interacted with the key residues of transmembrane helix 3 (TM3), the amphipathic helix 8 (Helix8), and intracellular loops 3 (ICL3) in GPR17 to engage in the receptor core. In summary, our results highlight the activation mechanism of GPR17 from the structural basis. Elucidating the structural and activation mechanism of GPR17 may facilitate the pharmacological intervention for acute/chronic CNS injury.
Collapse
Affiliation(s)
- Fang Ye
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
- The Chinese University of Hong KongShenzhen Futian Biomedical Innovation R&D CenterShenzhenGuangdongChina
| | - Thian‐Sze Wong
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Geng Chen
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Zhiyi Zhang
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Binghao Zhang
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Wei Gao
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Jiancheng Li
- Instrumental Analysis CenterShenzhen UniversityShenzhenGuangdongChina
| | - Zhangsong Wu
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Xin Pan
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Yang Du
- Kobilka Institute of Innovative Drug DiscoveryShenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhenGuangdongChina
- The Chinese University of Hong KongShenzhen Futian Biomedical Innovation R&D CenterShenzhenGuangdongChina
| |
Collapse
|
34
|
Collins HY, Emery B. Fishing for a balance: Toward promyelinating drug discovery. Cell Chem Biol 2022; 29:1467-1469. [PMID: 36270232 DOI: 10.1016/j.chembiol.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this issue, Häberlein et al. demonstrate a role for GPR17 in regulating zebrafish oligodendrocyte differentiation. Zebrafish expressing a humanized GPR17 respond to modulators, which are inactive against the endogenous zebrafish receptor. These findings highlight the potential for humanized zebrafish as an in vivo platform for targeted remyelination drug screens.
Collapse
Affiliation(s)
- Hannah Y Collins
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR 97239, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ben Emery
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
35
|
Säfholm J, Abma W, Bankova LG, Boyce JA, Al-Ameri M, Orre AC, Wheelock CE, Dahlén SE, Adner M. Cysteinyl-maresin 3 inhibits IL-13 induced airway hyperresponsiveness through alternative activation of the CysLT 1 receptor. Eur J Pharmacol 2022; 934:175257. [PMID: 36116518 DOI: 10.1016/j.ejphar.2022.175257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Cysteinyl-maresins, also known as maresin-conjugates in tissue regeneration (MCTRs), are recently discovered lipid mediators proposed to reduce airway inflammation. OBJECTIVE To investigate the influence of MCTRs on IL-13-induced airway hyperresponsiveness in isolated human and mice airways. METHODS Before responsiveness to contractile agonists were assessed in myographs, human small bronchi were cultured for 2 days and mouse tracheas were cultured for 1-4 days. During the culture procedure airways were exposed to interleukin (IL)-13 in the presence or absence of MCTRs. Signalling mechanisms were explored using pharmacologic agonists and antagonists, and genetically modified mice. RESULTS IL-13 treatment increased contractions to histamine, carbachol and leukotriene D4 (LTD4) in human small bronchi, and to 5-hydroxytryptamine (5-HT) in mouse trachea. In both preparations, co-incubation of the explanted tissues with MCTR3 reduced the IL-13 induced enhancement of contractions. In mouse trachea, this inhibitory effect of MCTR3 was blocked by three different CysLT1 receptor antagonists (montelukast, zafirlukast and pobilukast) during IL-13 exposure. Likewise, MCTR3 failed to reduce the IL-13-induced 5-HT responsiveness in mice deficient of the CysLT1 receptor. However, co-incubation with the classical CysLT1 receptor agonist LTD4 did not alter the IL-13-induced 5-HT hyperreactivity. CONCLUSIONS MCTR3, but not LTD4, decreased the IL-13-induced airway hyperresponsiveness by activation of the CysLT1 receptor. The distinct actions of the two lipid mediators on the CysLT1 receptor suggest an alternative signalling pathway appearing under inflammatory conditions, where this new action of MCTR3 implicates potential to inhibit airway hyperresponsiveness in asthma.
Collapse
Affiliation(s)
- Jesper Säfholm
- Institute of Environmental Medicine, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden; Centre for Allergy Research, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden
| | - Willem Abma
- Institute of Environmental Medicine, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden; Centre for Allergy Research, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden
| | - Lora G Bankova
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joshua A Boyce
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mamdoh Al-Ameri
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden; Department of Cardiothoracic Surgery and Anesthesiology, Karolinska University Hospital, Stockholm, Sweden
| | - Ann-Charlotte Orre
- Department of Cardiothoracic Surgery and Anesthesiology, Karolinska University Hospital, Stockholm, Sweden
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Institute of Environmental Medicine, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden; Centre for Allergy Research, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Adner
- Institute of Environmental Medicine, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden; Centre for Allergy Research, Karolinska Institutet, Biomedicum 5B, Solnavägen 9, SE-171 65, Solna, Sweden.
| |
Collapse
|
36
|
Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15091039. [PMID: 36145259 PMCID: PMC9505853 DOI: 10.3390/ph15091039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing environmental distress is associated with a growing asthma incidence; no treatments are available but montelukast (MTK)—an antagonist of the cysteinyl leukotrienes receptor 1—is widely used in the management of symptoms among adults and children. Recently, new molecular targets have been identified and MTK has been proposed for repurposing in other therapeutic applications, with several ongoing clinical trials. The proposed applications include neuroinflammation control, which could be explored in some neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases (AD and PD). However, this drug has been associated with an increasing number of reported neuropsychiatric adverse drug reactions (ADRs). Besides, and despite being on the market since 1998, MTK metabolism is still poorly understood and the mechanisms underlying neuropsychiatric ADRs remain unknown. We review the role of MTK as a modulator of leukotriene pathways and systematize the current knowledge about MTK metabolism. Known toxic effects of MTK are discussed, and repurposing applications are presented comprehensively, with a focus on AD and PD.
Collapse
Affiliation(s)
- Cátia F. Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Maria Matilde Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Gonçalo C. Justino
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
37
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
38
|
Montelukast, an Antagonist of Cysteinyl Leukotriene Signaling, Impairs Burn Wound Healing. Plast Reconstr Surg 2022; 150:92e-104e. [PMID: 35536768 DOI: 10.1097/prs.0000000000009228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Burns are severe injuries often associated with impaired wound healing. Impaired healing is caused by multiple factors, including dysregulated inflammatory responses at the wound site. Interestingly, montelukast, an antagonist for cysteinyl leukotrienes and U.S. Food and Drug Administration approved for treatment of asthma and allergy, was previously shown to enhance healing in excision wounds and to modulate local inflammation. METHODS In this study, the authors examined the effect of montelukast on wound healing in a mouse model of scald burn injury. Burn wound tissues isolated from montelukast- and vehicle-treated mice at various times after burn injury were analyzed for wound areas ( n = 34 to 36), reepithelialization ( n = 14), inflammation ( n = 8 to 9), and immune cell infiltration ( n = 3 to 6) and proliferation ( n = 7 to 8). RESULTS In contrast to previously described beneficial effects in excision wounds, this study shows that montelukast delays burn wound healing by impairing the proliferation of keratinocytes and endothelial cells. This occurs largely independently of inflammatory responses at the wound site, suggesting that montelukast impairs specifically the proliferative phase of wound healing in burns. Wound healing rates in mice in which leukotrienes are not produced were not affected by montelukast. CONCLUSION Montelukast delays wound healing mainly by reducing the proliferation of local cells after burn injury. CLINICAL RELEVANCE STATEMENT Although additional and clinical studies are necessary, our study suggests that burn patients who are on montelukast may exhibit delayed healing, necessitating extra observation.
Collapse
|
39
|
Wang S, Wang Y, Zou S. A Glance at the Molecules That Regulate Oligodendrocyte Myelination. Curr Issues Mol Biol 2022; 44:2194-2216. [PMID: 35678678 PMCID: PMC9164040 DOI: 10.3390/cimb44050149] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Oligodendrocyte (OL) myelination is a critical process for the neuronal axon function in the central nervous system. After demyelination occurs because of pathophysiology, remyelination makes repairs similar to myelination. Proliferation and differentiation are the two main stages in OL myelination, and most factors commonly play converse roles in these two stages, except for a few factors and signaling pathways, such as OLIG2 (Oligodendrocyte transcription factor 2). Moreover, some OL maturation gene mutations induce hypomyelination or hypermyelination without an obvious function in proliferation and differentiation. Herein, three types of factors regulating myelination are reviewed in sequence.
Collapse
Affiliation(s)
- Shunqi Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Yingxing Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
| | - Suqi Zou
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
40
|
Leukotriene Signaling as a Target in α-Synucleinopathies. Biomolecules 2022; 12:biom12030346. [PMID: 35327537 PMCID: PMC8944962 DOI: 10.3390/biom12030346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/12/2022] [Accepted: 02/12/2022] [Indexed: 01/04/2023] Open
Abstract
Parkinson’s disease (PD) and dementia with Lewy bodies (DLB) are two common types of α-synucleinopathies and represent a high unmet medical need. Despite diverging clinical manifestations, both neurodegenerative diseases share several facets of their complex pathophysiology. Apart from α-synuclein aggregation, an impairment of mitochondrial functions, defective protein clearance systems and excessive inflammatory responses are consistently observed in the brains of PD as well as DLB patients. Leukotrienes are lipid mediators of inflammatory signaling traditionally known for their role in asthma. However, recent research advances highlight a possible contribution of leukotrienes, along with their rate-limiting synthesis enzyme 5-lipoxygenase, in the pathogenesis of central nervous system disorders. This review provides an overview of in vitro as well as in vivo studies, in summary suggesting that dysregulated leukotriene signaling is involved in the pathological processes underlying PD and DLB. In addition, we discuss how the leukotriene signaling pathway could serve as a future drug target for the therapy of PD and DLB.
Collapse
|
41
|
Sokolowska M, Rovati GE, Diamant Z, Untersmayr E, Schwarze J, Lukasik Z, Sava F, Angelina A, Palomares O, Akdis C, O'Mahony L, Jesenak M, Pfaar O, Torres MJ, Sanak M, Dahlén S, Woszczek G. Effects of non-steroidal anti-inflammatory drugs and other eicosanoid pathway modifiers on antiviral and allergic responses: EAACI task force on eicosanoids consensus report in times of COVID-19. Allergy 2022; 77:2337-2354. [PMID: 35174512 PMCID: PMC9111413 DOI: 10.1111/all.15258] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 11/29/2022]
Abstract
Non‐steroidal anti‐inflammatory drugs (NSAIDs) and other eicosanoid pathway modifiers are among the most ubiquitously used medications in the general population. Their broad anti‐inflammatory, antipyretic, and analgesic effects are applied against symptoms of respiratory infections, including SARS‐CoV‐2, as well as in other acute and chronic inflammatory diseases that often coexist with allergy and asthma. However, the current pandemic of COVID‐19 also revealed the gaps in our understanding of their mechanism of action, selectivity, and interactions not only during viral infections and inflammation, but also in asthma exacerbations, uncontrolled allergic inflammation, and NSAIDs‐exacerbated respiratory disease (NERD). In this context, the consensus report summarizes currently available knowledge, novel discoveries, and controversies regarding the use of NSAIDs in COVID‐19, and the role of NSAIDs in asthma and viral asthma exacerbations. We also describe here novel mechanisms of action of leukotriene receptor antagonists (LTRAs), outline how to predict responses to LTRA therapy and discuss a potential role of LTRA therapy in COVID‐19 treatment. Moreover, we discuss interactions of novel T2 biologicals and other eicosanoid pathway modifiers on the horizon, such as prostaglandin D2 antagonists and cannabinoids, with eicosanoid pathways, in context of viral infections and exacerbations of asthma and allergic diseases. Finally, we identify and summarize the major knowledge gaps and unmet needs in current eicosanoid research.
Collapse
Affiliation(s)
- Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne ‐ Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - G Enrico Rovati
- Department of Pharmaceutical Sciences Section of Pharmacology and Biosciences University of Milan Milano Italy
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology Skane University Hospital Lund Sweden
- Department Microbiology Immunology and Transplantation Ku Leuven, Catholic University of Leuven Belgium
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Jürgen Schwarze
- Child Life and Health and Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Zuzanna Lukasik
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- VIB Center for Inflammation Research Ghent University Ghent Belgium
| | - Florentina Sava
- London North Genomic Laboratory Hub Great Ormond Street Hospital for Children NHS Foundation Trust London UK
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University Madrid Spain
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne ‐ Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Liam O'Mahony
- Departments of Medicine and Microbiology APC Microbiome IrelandUniversity College Cork Cork Ireland
| | - Milos Jesenak
- Department of Pulmonology and Phthisiology Department of Allergology and Clinical Immunology Department of Pediatrics Jessenius Faculty of Medicine in Martin Comenius University in BratislavaUniversity Teaching Hospital in Martin Slovakia
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - María José Torres
- Allergy Unit Málaga Regional University Hospital‐IBIMA‐UMA Málaga Spain
| | - Marek Sanak
- Department of Medicine Jagiellonian University Medical College Krakow Poland
| | - Sven‐Erik Dahlén
- Institute of Environmental Medicine and the Centre for Allergy Research, Karolinska Institute, and the Department of Respiratory Medicine Karolinska University Hospital Stockholm Sweden
| | - Grzegorz Woszczek
- Asthma UK Centre in Allergic Mechanisms of Asthma School of Immunology and Microbial Sciences King's College London London UK
| |
Collapse
|
42
|
Xiao Y, Zhang Y, Gao YH, Zhao ZH, He J, Gao R, Guo YX, Wang LB, Li X. A targeted extracellular vesicles loaded with montelukast in the treatment of demyelinating diseases. Biochem Biophys Res Commun 2022; 594:31-37. [DOI: 10.1016/j.bbrc.2022.01.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022]
|
43
|
Lin KN, Zhang K, Zhao W, Huang SY, Li H. Insulin-like Growth Factor 1 Promotes Cell Proliferation by Downregulation of G-Protein-Coupled Receptor 17 Expression via PI3K/Akt/FoxO1 Signaling in SK-N-SH Cells. Int J Mol Sci 2022; 23:ijms23031513. [PMID: 35163437 PMCID: PMC8835821 DOI: 10.3390/ijms23031513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) not only regulates neuronal function and development but also is neuroprotective in the setting of acute ischemic stroke. G-protein-coupled receptor 17 (GPR17) expression in brain tissue serves as an indicator of brain damage. As whether IGF-1 regulates GPR17 expression remains unknown, the aim of this study is to investigate how IGF-1 regulates GPR17 expression in vitro. Human neuroblastoma SK-N-SH cells were used. Lentivirus-mediated short hairpin RNA (shRNA) was constructed to mediate the silencing of FoxO1, while adenoviral vectors were used for its overexpression. Verification of the relevant signaling cascade was performed using a FoxO1 inhibitor (AS1842856), a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002), and a GPR17 antagonist (cangrelor). Cell proliferation was analyzed using EdU staining; immunofluorescence staining was used to detect the expression and subcellular localization of FoxO1. Chromatin immunoprecipitation was used to analyze the binding of FoxO1 to the GPR17 promoter in SK-N-SH cells. The expression of FoxO1, GPR17, and protein kinase B (also known as Akt) mRNA and protein as well as the levels of FoxO1 and Akt phosphorylation were investigated in this study. IGF-1 was found to downregulate FoxO1 and GPR17 expression in SK-N-SH cells while promoting cell viability and proliferation. Inhibition of FoxO1 and antagonism of GPR17 were found to play a role similar to that of IGF-1. Silencing of FoxO1 by lentivirus-mediated shRNA resulted in the downregulation of FoxO1 and GPR17 expression. The overexpression of FoxO1 via adenoviral vectors resulted in the upregulation of FoxO1 and GPR17 expression. Blocking of PI3K signaling by LY294002 inhibited the effect of IGF-1 on GPR17 suppression. Results from chromatin immunoprecipitation revealed that IGF-1 promotes FoxO1 nuclear export and reduces FoxO1 binding to the GPR17 promoter in SK-N-SH cells. Here, we conclude that IGF-1 enhances cell viability and proliferation in SK-N-SH cells via the promotion of FoxO1 nuclear export and reduction of FoxO1 binding to the GPR17 promoter via PI3K/Akt signaling. Our findings suggest that the enhancement of IGF-1 signaling to antagonize GPR17 serves as a potential therapeutic strategy in the management of acute ischemic stroke.
Collapse
Affiliation(s)
- Ka-Na Lin
- Center for Brain Science & Clinical Research Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Kan Zhang
- Department of Anesthesiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
| | - Wei Zhao
- Department of Pharmacy, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (W.Z.); (S.-Y.H.)
| | - Shi-Ying Huang
- Department of Pharmacy, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (W.Z.); (S.-Y.H.)
| | - Hao Li
- Center for Brain Science & Clinical Research Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
- Department of Pharmacy, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (W.Z.); (S.-Y.H.)
- Correspondence:
| |
Collapse
|
44
|
Intestinal Gpr17 deficiency improves glucose metabolism by promoting GLP-1 secretion. Cell Rep 2022; 38:110179. [PMID: 34986353 PMCID: PMC8972502 DOI: 10.1016/j.celrep.2021.110179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) in intestinal enteroendocrine cells (EECs) respond to nutritional, neural, and microbial cues and modulate the release of gut hormones. Here we show that Gpr17, an orphan GPCR, is co-expressed in glucagon-like peptide-1 (GLP-1)-expressing EECs in human and rodent intestinal epithelium. Acute genetic ablation of Gpr17 in intestinal epithelium improves glucose tolerance and glucose-stimulated insulin secretion (GSIS). Importantly, inducible knockout (iKO) mice and Gpr17 null intestinal organoids respond to glucose or lipid ingestion with increased secretion of GLP-1, but not the other incretin glucose-dependent insulinotropic polypeptide (GIP). In an in vitro EEC model, overexpression or agonism of Gpr17 reduces voltage-gated calcium currents and decreases cyclic AMP (cAMP) production, and these are two critical factors regulating GLP-1 secretion. Together, our work shows that intestinal Gpr17 signaling functions as an inhibitory pathway for GLP-1 secretion in EECs, suggesting intestinal GPR17 is a potential target for diabetes and obesity intervention. Yan et al. locate GPR17 expression in the enteroendocrine cells of human and rodent intestinal epithelium. They find that GPR17 signaling inhibits intracellular rise of cAMP and calcium and that loss of intestinal Gpr17 in rodents leads to better glucose tolerance via increased hormone secretion in response to nutrient ingestion.
Collapse
|
45
|
Liu W, Yang Y, Zeng Z, Tian Y, Wu Q, Zhou M, Fu ZF, Zhao L. G protein-coupled receptor 17 restricts rabies virus replication via BAK-mediated apoptosis. Vet Microbiol 2021; 265:109326. [PMID: 34979406 DOI: 10.1016/j.vetmic.2021.109326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023]
Abstract
Rabies, caused by rabies virus (RABV), is an ancient zoonotic disease that significantly affects human and animal health throughout the world. RABV causes acute encephalitis in mammals with a high fatality rate in developing countries. G protein-coupled receptor 17 (GPR17) is a vital gene in the central nervous system (CNS) that plays important roles in demyelinating diseases and ischemia brain. However, it is still unclear whether GPR17 participates in the regulation of RABV infection. Here, we found that upregulation or activation of GPR17 can reduce the virus titer; conversely, the inactivation or silence of GPR17 led to increased RABV replication in N2a cells. The recombinant RABV expressing GPR17 (rRABV-GPR17) showed reduced replication capacity compared to the parent virus rRABV. Moreover, overexpression of GPR17 can attenuate RABV pathogenicity in mice. Further study demonstrated that GPR17 suppressed RABV replication via BAK-mediated apoptosis. Our findings uncover an unappreciated role of GPR17 in suppressing RABV infection, where GPR17 mediates cell apoptosis to limit RABV replication and may be an attractive candidate for new therapeutic interventions in the treatment of rabies.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaping Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zonghui Zeng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuling Tian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiong Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
46
|
The oligodendrocyte-enriched orphan G protein-coupled receptor Gpr62 is dispensable for central nervous system myelination. Neural Dev 2021; 16:6. [PMID: 34844642 PMCID: PMC8630896 DOI: 10.1186/s13064-021-00156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myelination is a highly regulated process in the vertebrate central nervous system (CNS) whereby oligodendrocytes wrap axons with multiple layers of insulating myelin in order to allow rapid electrical conduction. Establishing the proper pattern of myelin in neural circuits requires communicative axo-glial interactions, however, the molecular interactions that occur between oligodendrocytes and axons during developmental myelination and myelin maintenance remain to be fully elucidated. Our previous work identified G protein-coupled receptor 62 (Gpr62), an uncharacterized orphan g-protein coupled receptor, as being selectively expressed by mature oligodendrocytes within the CNS, suggesting a potential role in myelination or axoglial interactions. However, no studies to date have assessed the functional requirement for Gpr62 in oligodendrocyte development or CNS myelination. METHODS To address this, we generated a knockout mouse strain lacking the Gpr62 gene. We assessed CNS myelination during both postnatal development and adulthood using immunohistochemistry, electron microscopy and western blot. In addition, we utilized AAV-mediated expression of a tagged Gpr62 in oligodendrocytes to determine the subcellular localization of the protein in vivo. RESULTS We find that virally expressed Gpr62 protein is selectively expressed on the adaxonal myelin layer, suggestive of a potential role for Gpr62 in axo-myelinic signaling. Nevertheless, Gpr62 knockout mice display normal oligodendrocyte numbers and apparently normal myelination within the CNS during both postnatal development and adulthood. CONCLUSIONS We conclude that in spite of being well-placed to mediate neuronal-oligodendrocyte communications, Gpr62 is overall dispensable for CNS myelination.
Collapse
|
47
|
Braune M, Scherf N, Heine C, Sygnecka K, Pillaiyar T, Parravicini C, Heimrich B, Abbracchio MP, Müller CE, Franke H. Involvement of GPR17 in Neuronal Fibre Outgrowth. Int J Mol Sci 2021; 22:ijms222111683. [PMID: 34769111 PMCID: PMC8584086 DOI: 10.3390/ijms222111683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
Characterization of new pharmacological targets is a promising approach in research of neurorepair mechanisms. The G protein-coupled receptor 17 (GPR17) has recently been proposed as an interesting pharmacological target, e.g., in neuroregenerative processes. Using the well-established ex vivo model of organotypic slice co-cultures of the mesocortical dopaminergic system (prefrontal cortex (PFC) and substantia nigra/ventral tegmental area (SN/VTA) complex), the influence of GPR17 ligands on neurite outgrowth from SN/VTA to the PFC was investigated. The growth-promoting effects of Montelukast (MTK; GPR17- and cysteinyl-leukotriene receptor antagonist), the glial cell line-derived neurotrophic factor (GDNF) and of two potent, selective GPR17 agonists (PSB-16484 and PSB-16282) were characterized. Treatment with MTK resulted in a significant increase in mean neurite density, comparable with the effects of GDNF. The combination of MTK and GPR17 agonist PSB-16484 significantly inhibited neuronal growth. qPCR studies revealed an MTK-induced elevated mRNA-expression of genes relevant for neuronal growth. Immunofluorescence labelling showed a marked expression of GPR17 on NG2-positive glia. Western blot and RT-qPCR analysis of untreated cultures suggest a time-dependent, injury-induced stimulation of GPR17. In conclusion, MTK was identified as a stimulator of neurite fibre outgrowth, mediating its effects through GPR17, highlighting GPR17 as an interesting therapeutic target in neuronal regeneration.
Collapse
Affiliation(s)
- Max Braune
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany; (M.B.); (C.H.); (K.S.)
| | - Nico Scherf
- Methods and Development Group Neural Data Analysis and Statistical Computing, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103 Leipzig, Germany;
| | - Claudia Heine
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany; (M.B.); (C.H.); (K.S.)
| | - Katja Sygnecka
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany; (M.B.); (C.H.); (K.S.)
| | - Thanigaimalai Pillaiyar
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; (T.P.); (C.E.M.)
| | - Chiara Parravicini
- Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (C.P.); (M.P.A.)
| | - Bernd Heimrich
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Albertstr. 23, 79104 Freiburg, Germany;
| | - Maria P. Abbracchio
- Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (C.P.); (M.P.A.)
| | - Christa E. Müller
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; (T.P.); (C.E.M.)
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany; (M.B.); (C.H.); (K.S.)
- Correspondence: ; Tel.: +49-(0)341-9724602; Fax: +49-(0)341-9724609
| |
Collapse
|
48
|
Trost A, Motloch K, Koller A, Bruckner D, Runge C, Schroedl F, Bogner B, Kaser-Eichberger A, Strohmaier C, Ladek AM, Preishuber-Pfluegl J, Brunner SM, Aigner L, Reitsamer HA. Inhibition of the cysteinyl leukotriene pathways increases survival of RGCs and reduces microglial activation in ocular hypertension. Exp Eye Res 2021; 213:108806. [PMID: 34715090 DOI: 10.1016/j.exer.2021.108806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/05/2021] [Accepted: 10/21/2021] [Indexed: 10/20/2022]
Abstract
Glaucoma is the second leading cause of blindness worldwide. This multifactorial, neurodegenerative group of diseases is characterized by the progressive loss of retinal ganglion cells (RGCs) and their axons, leading to irreversible visual impairment and blindness. There is a huge unmet and urging need for the development of new and translatable strategies and treatment options to prevent this progressive loss of RGC. Accumulating evidence points towards a critical role of neuroinflammation, in particular microglial cells, in the pathogenesis of glaucoma. Leukotrienes are mediators of neuroinflammation and are involved in many neurodegenerative diseases. Therefore, we tested the leukotriene receptors CysLT1R/GPR17-selective antagonist Montelukast (MTK) for its efficacy to modulate the reactive state of microglia in order to ameliorate RGCs loss in experimental glaucoma. Ocular hypertension (OHT) was induced unilaterally by injection of 8 μm magnetic microbead (MB) into the anterior chamber of female Brown Norway rats. The contralateral, untreated eye served as control. Successful induction of OHT was verified by daily IOP measurement using a TonoLab rebound tonometer. Simultaneously to OHT induction, one group received daily MTK treatment and the control group vehicle solution by oral gavage. Animals were sacrificed 13-15 days after MB injection. Retina and optic nerves (ON) of OHT and contralateral eyes were analyzed by immunofluorescence with specific markers for RGCs (Brn3a), microglial cells/macrophages (Iba1 and CD68), and cysteinyl leukotriene pathway receptors (CysLT1R and GPR17). Protein labeling was documented by confocal microscopy and analyzed with ImageJ plugins. Further, mRNA expression of genes of the inflammatory and leukotriene pathway was analyzed in retinal tissue. MTK treatment resulted in a short-term IOP reduction at day 2, which dissipated by day 5 of OHT induction in MTK treated animals. Furthermore, MTK treatment resulted in a decreased activation of Iba1+ microglial cells in the retina and ON, and in a significantly increased RGC survival in OHT eyes. Within the retina, GPR17 and CysLT1R expression was demonstrated in single RCGs and in microglial cells respectively. Further, increased mRNA expression of pro-inflammatory genes was detected in OHT induced retinas. In the ON, OHT induction increased the number of GPR17+ cells, showing a trend of reduction following MTK treatment. This study shows for the first time a significantly increased RGC survival in an acute OHT model following treatment with the leukotriene receptor antagonist MTK. These results strongly suggest a neuroprotective effect of MTK and a potential new therapeutic strategy for glaucoma treatment.
Collapse
Affiliation(s)
- Andrea Trost
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria.
| | - Karolina Motloch
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Andreas Koller
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Daniela Bruckner
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Christian Runge
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Falk Schroedl
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria; Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Barbara Bogner
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria; Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Clemens Strohmaier
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria; Department of Ophthalmology and Optometry, Johannes Kepler University, Linz, Austria
| | - Anja-Maria Ladek
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Julia Preishuber-Pfluegl
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Susanne Maria Brunner
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Austria
| | - Herbert Anton Reitsamer
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, 5020, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Austria
| |
Collapse
|
49
|
Xiong LY, Ouk M, Wu CY, Rabin JS, Lanctôt KL, Herrmann N, Black SE, Edwards JD, Swardfager W. Leukotriene receptor antagonist use and cognitive decline in normal cognition, mild cognitive impairment, and Alzheimer's dementia. Alzheimers Res Ther 2021; 13:147. [PMID: 34479635 PMCID: PMC8418104 DOI: 10.1186/s13195-021-00892-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/24/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Leukotriene receptor antagonists (LTRAs) alleviate Alzheimer's disease (AD) pathology and improve cognition in animal models; however, clinical evidence is limited. This study aimed to explore the associations between the use of LTRAs (montelukast or zafirlukast) and cognitive performance in people with normal cognition, mild cognitive impairment (MCI), or AD dementia. We hypothesized that LTRA use would be associated with better cognitive performance over time. METHODS This longitudinal observational study used data from the National Alzheimer's Coordinating Center. Within groups of participants with normal cognition, MCI, or AD dementia, LTRA users were matched 1:3 to non-users using propensity score matching. Cognitive domains including immediate and delayed memory (Wechsler Memory Scale Revised-Logical Memory IA and IIA), psychomotor processing speed (Digit Symbol Substitution Test), and language (animal naming, vegetable naming, and Boston Naming Test) were compared between users and non-users in mixed-effects linear or Poisson regression models. RESULTS In AD dementia, LTRA use was associated with a slower decline in psychomotor processing speed, as measured by the Digit Symbol Substitution Test (Β = 1.466 [0.253, 2.678] symbols/year, n = 442), and language, as measured by animal naming (Β = 0.541 [0.215, 0.866] animals/year, n = 566), vegetable naming (B = 0.309 [0.056, 0.561] vegetables/year, n = 565), and the Boston Naming Test (B = 0.529 [0.005, 1.053] items/year, n = 561). Effect sizes were small but persisted after controlling for a 10% false discovery rate. LTRA use was not associated with changes in memory performance in AD, nor was it associated with changes in cognitive performance in people with normal cognition or MCI. In a post hoc analysis, LTRA use was associated with a slower decline in clinical progression in MCI (B = -0.200 [-0.380, -0.019] points/year, n = 800) and AD dementia (B = -0.321 [-0.597, -0.046] points/year, n = 604) as measured by CDR Sum of Boxes. CONCLUSIONS The use of LTRAs was associated with preserved function in non-amnestic cognitive domains in AD dementia. The role of leukotrienes and their receptors in cognitive decline warrants further investigation and the leukotriene pathway may represent a target for AD treatment.
Collapse
Affiliation(s)
- Lisa Y Xiong
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Room 4207, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Michael Ouk
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
| | - Che-Yuan Wu
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Room 4207, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Jennifer S Rabin
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, Canada
| | - Krista L Lanctôt
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Room 4207, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- KITE UHN Toronto Rehabilitation Institute, Toronto, Canada
| | - Nathan Herrmann
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sandra E Black
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- KITE UHN Toronto Rehabilitation Institute, Toronto, Canada
- Canadian Partnership for Stroke Recovery, Toronto, Canada
| | - Jodi D Edwards
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
- ICES, Ottawa, Canada
| | - Walter Swardfager
- Dr. Sandra Black Centre for Brain Resilience & Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada.
- Department of Pharmacology & Toxicology, University of Toronto, Room 4207, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- KITE UHN Toronto Rehabilitation Institute, Toronto, Canada.
- Canadian Partnership for Stroke Recovery, Toronto, Canada.
| |
Collapse
|
50
|
Doan P, Nguyen P, Murugesan A, Subramanian K, Konda Mani S, Kalimuthu V, Abraham BG, Stringer BW, Balamuthu K, Yli-Harja O, Kandhavelu M. Targeting Orphan G Protein-Coupled Receptor 17 with T0 Ligand Impairs Glioblastoma Growth. Cancers (Basel) 2021; 13:cancers13153773. [PMID: 34359676 PMCID: PMC8345100 DOI: 10.3390/cancers13153773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/10/2021] [Accepted: 07/22/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM), or glioblastoma chemotherapy, has one of the poorest improvements across all types of cancers. Despite the different rationales explored in targeted therapy for taming the GBM aggressiveness, its phenotypic plasticity, drug toxicity, and adaptive resistance mechanisms pose many challenges in finding an effective cure. Our manuscript reports the expression and prognostic role of orphan receptor GPR17 in glioma, the molecular mechanism of action of the novel ligand of GPR17, and provides evidence how the T0 agonist promotes glioblastoma cell death through modulation of the MAPK/ERK, PI3K–Akt, STAT, and NF-κB pathways. The highlights are as follows: GPR17 expression is associated with greater survival for both low-grade glioma (LGG) and GBM; GA-T0, a potent GPR17 receptor agonist, causes significant GBM cell death and apoptosis; GPR17 signaling promotes cell cycle arrest at the G1 phase in GBM cells; key genes are modulated in the signaling pathways that inhibit GBM cell proliferation; and GA-T0 crosses the blood–brain barrier and reduces tumor volume. Abstract Glioblastoma, an invasive high-grade brain cancer, exhibits numerous treatment challenges. Amongst the current therapies, targeting functional receptors and active signaling pathways were found to be a potential approach for treating GBM. We exploited the role of endogenous expression of GPR17, a G protein-coupled receptor (GPCR), with agonist GA-T0 in the survival and treatment of GBM. RNA sequencing was performed to understand the association of GPR17 expression with LGG and GBM. RT-PCR and immunoblotting were performed to confirm the endogenous expression of GPR17 mRNA and its encoded protein. Biological functions of GPR17 in the GBM cells was assessed by in vitro analysis. HPLC and histopathology in wild mice and an acute-toxicity analysis in a patient-derived xenograft model were performed to understand the clinical implication of GA-T0 targeting GPR17. We observed the upregulation of GPR17 in association with improved survival of LGG and GBM, confirming it as a predictive biomarker. GA-T0-stimulated GPR17 leads to the inhibition of cyclic AMP and calcium flux. GPR17 signaling activation enhances cytotoxicity against GBM cells and, in patient tissue-derived mesenchymal subtype GBM cells, induces apoptosis and prevents proliferation by stoppage of the cell cycle at the G1 phase. Modulation of the key genes involved in DNA damage, cell cycle arrest, and in several signaling pathways, including MAPK/ERK, PI3K–Akt, STAT, and NF-κB, prevents tumor regression. In vivo activation of GPR17 by GA-T0 reduces the tumor volume, uncovering the potential of GA-T0–GPR17 as a targeted therapy for GBM treatment. Conclusion: Our analysis suggests that GA-T0 targeting the GPR17 receptor presents a novel therapy for treating glioblastoma.
Collapse
Affiliation(s)
- Phuong Doan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | - Phung Nguyen
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
- Department of Biotechnology, Lady Doak College, Thallakulam, Madurai 625002, India
| | - Kumar Subramanian
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | | | - Vignesh Kalimuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (K.B.)
| | - Bobin George Abraham
- Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland;
| | - Brett W. Stringer
- College of Medicine and Public Health, Flinders University, Sturt Rd., Bedford Park, SA 5042, Australia;
| | - Kadalmani Balamuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (K.B.)
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland;
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
- Correspondence: ; Tel.: +358-504721724
| |
Collapse
|