1
|
Stephens AD, Wilkinson T. Discovery of Therapeutic Antibodies Targeting Complex Multi-Spanning Membrane Proteins. BioDrugs 2024; 38:769-794. [PMID: 39453540 DOI: 10.1007/s40259-024-00682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning polypeptides, encompass families of proteins that are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels, transporters, enzymes, and adhesion molecules. The high specificity of monoclonal antibodies and the ability to engineer their properties offers a significant opportunity to selectively bind these target proteins, allowing direct modulation of pharmacology or enabling other mechanisms of action such as cell killing. Isolation of antibodies that bind these types of membrane proteins and exhibit the desired pharmacological function has, however, remained challenging due to technical issues in preparing membrane protein antigens suitable for enabling and driving antibody drug discovery strategies. In this article, we review progress and emerging themes in defining discovery strategies for a generation of antibodies that target these complex membrane protein antigens. We also comment on how this field may develop with the emerging implementation of computational techniques, artificial intelligence, and machine learning.
Collapse
Affiliation(s)
- Amberley D Stephens
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Trevor Wilkinson
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| |
Collapse
|
2
|
Jia W, Huang Z, Zhou L, Liou YC, Di Virgilio F, Ulrich H, Illes P, Zhang W, Huang C, Tang Y. Purinergic signalling in cancer therapeutic resistance: From mechanisms to targeting strategies. Drug Resist Updat 2023; 70:100988. [PMID: 37413937 DOI: 10.1016/j.drup.2023.100988] [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: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
Purinergic signalling, consisting of extracellular purines and purinergic receptors, modulates cell proliferation, invasion and immunological reaction during cancer progression. Here, we focus on current evidence that suggests the crucial role of purinergic signalling in mediating cancer therapeutic resistance, the major obstacle in cancer treatment. Mechanistically, purinergic signalling can modulate the tumor microenvironment (TME), epithelial-mesenchymal transition (EMT) and anti-tumor immunity, thus affecting drug sensitivity of tumor cells. Currently, some agents attempting to target purinergic signalling either in tumor cells or in tumor-associated immune cells are under preclinical or clinical investigation. Moreover, nano-based delivery technologies significantly improve the efficacy of agents targeting purinergic signalling. In this review article, we summarize the mechanisms of purinergic signalling in promoting cancer therapeutic resistance and discuss the potentials and challenges of targeting purinergic signalling in future cancer treatment.
Collapse
Affiliation(s)
- Wenhui Jia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhao Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yih-Cherng Liou
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117573, Singapore; Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117573, Singapore
| | | | - Henning Ulrich
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, Leipzig, Germany
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; Institute of TCM-Based Stress Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
3
|
Elhage A, Cuthbertson P, Sligar C, Watson D, Sluyter R. A Species-Specific Anti-Human P2X7 Monoclonal Antibody Reduces Graft-versus-Host Disease in Humanised Mice. Pharmaceutics 2023; 15:2263. [PMID: 37765233 PMCID: PMC10536354 DOI: 10.3390/pharmaceutics15092263] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a T cell-mediated inflammatory disorder that arises from allogeneic haematopoietic stem cell transplantation and is often fatal. The P2X7 receptor is an extracellular adenosine 5'-triphosphate-gated cation channel expressed on immune cells. Blockade of this receptor with small molecule inhibitors impairs GVHD in a humanised mouse model. A species-specific blocking monoclonal antibody (mAb) (clone L4) for human P2X7 is available, affording the opportunity to determine whether donor (human) P2X7 contributes to the development of GVHD in humanised mice. Using flow cytometric assays of human RPMI 8266 and murine J774 cells, this study confirmed that this mAb bound and impaired human P2X7. Furthermore, this mAb prevented the loss of human regulatory T cells (hTregs) and natural killer (hNK) T cells in vitro. NOD-scid IL2Rγnull mice were injected with 10 × 106 human peripheral blood mononuclear cells (Day 0) and an anti-hP2X7 or control mAb (100 μg i.p. per mouse, Days 0, 2, 4, 6, and 8). The anti-hP2X7 mAb increased hTregs and hNK cells at Day 21. Moreover, anti-hP2X7 mAb-treatment reduced clinical and histological GVHD in the liver and lung compared to the control treatment at disease endpoint. hTregs, hNK, and hNK T cell proportions were increased, and human T helper 17 cell proportions were decreased at endpoint. These studies indicate that blockade of human (donor) P2X7 reduces GVHD development in humanised mice, providing the first direct evidence of a role for donor P2X7 in GVHD.
Collapse
Affiliation(s)
- Amal Elhage
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Peter Cuthbertson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Chloe Sligar
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
4
|
Sluyter R, Adriouch S, Fuller SJ, Nicke A, Sophocleous RA, Watson D. Animal Models for the Investigation of P2X7 Receptors. Int J Mol Sci 2023; 24:ijms24098225. [PMID: 37175933 PMCID: PMC10179175 DOI: 10.3390/ijms24098225] [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: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The P2X7 receptor is a trimeric ligand-gated cation channel activated by extracellular adenosine 5'-triphosphate. The study of animals has greatly advanced the investigation of P2X7 and helped to establish the numerous physiological and pathophysiological roles of this receptor in human health and disease. Following a short overview of the P2X7 distribution, roles and functional properties, this article discusses how animal models have contributed to the generation of P2X7-specific antibodies and nanobodies (including biologics), recombinant receptors and radioligands to study P2X7 as well as to the pharmacokinetic testing of P2X7 antagonists. This article then outlines how mouse and rat models have been used to study P2X7. These sections include discussions on preclinical disease models, polymorphic P2X7 variants, P2X7 knockout mice (including bone marrow chimeras and conditional knockouts), P2X7 reporter mice, humanized P2X7 mice and P2X7 knockout rats. Finally, this article reviews the limited number of studies involving guinea pigs, rabbits, monkeys (rhesus macaques), dogs, cats, zebrafish, and other fish species (seabream, ayu sweetfish, rainbow trout and Japanese flounder) to study P2X7.
Collapse
Affiliation(s)
- Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Sahil Adriouch
- UniRouen, INSERM, U1234, Pathophysiology, Autoimmunity, and Immunotherapy, (PANTHER), Univ Rouen Normandie, University of Rouen, F-76000 Rouen, France
| | - Stephen J Fuller
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Nepean Hospital, Kingswood, NSW 2750, Australia
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, 80336 Munich, Germany
| | - Reece A Sophocleous
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
5
|
Li Y, Huang X, Fowler C, Lim YY, Laws SM, Faux N, Doecke JD, Trounson B, Pertile K, Rumble R, Doré V, Villemagne VL, Rowe CC, Wiley JS, Maruff P, Masters CL, Gu BJ. Identification of Leukocyte Surface P2X7 as a Biomarker Associated with Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23147867. [PMID: 35887215 PMCID: PMC9322488 DOI: 10.3390/ijms23147867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) has shown altered immune responses in the periphery. We studied P2X7 (a proinflammatory receptor and a scavenger receptor) and two integrins, CD11b and CD11c, on the surface of circulating leukocytes and analysed their associations with Aβ-PET, brain atrophy, neuropsychological assessments, and cerebrospinal fluid (CSF) biomarkers. Total 287 age-matched, sex-balanced participants were recruited in a discovery cohort and two validation cohorts through the AIBL study and studied using tri-colour flow cytometry. Our results demonstrated reduced expressions of P2X7, CD11b, and CD11c on leukocytes, particularly monocytes, in Aβ +ve cases compared with Aβ -ve controls. P2X7 and integrin downregulation was observed at pre-clinical stage of AD and stayed low throughout disease course. We further constructed a polygenic risk score (PRS) model based on 12 P2RX7 risk alleles to assess the genetic impact on P2X7 function in AIBL and ADNI cohorts. No significant association was identified between the P2RX7 gene and AD, indicating that P2X7 downregulation in AD is likely caused by environmental changes rather than genetic factors. In conclusion, the downregulation of P2X7 and integrins at pre-clinical stage of AD indicates altered pro-inflammatory responses, phagocytic functions, and migrating capabilities of circulating monocytes in early AD pathogenesis. Our study not only improves our understanding of peripheral immune involvement in early stage of AD but also provides more insights into novel biomarker development, diagnosis, and prognosis of AD.
Collapse
Affiliation(s)
- Yihan Li
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Xin Huang
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Christopher Fowler
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Yen Y. Lim
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC 3168, Australia; (Y.Y.L.); (V.D.)
| | - Simon M. Laws
- School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia;
| | - Noel Faux
- Melbourne Data Analytics Platform, Petascale Campus Initiative, The University of Melbourne, 21 Bedford St., North Melbourne, VIC 3051, Australia;
| | - James D. Doecke
- The Australian e-Health Research Centre, CSIRO, Brisbane, QLD 4029, Australia;
| | - Brett Trounson
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Kelly Pertile
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Rebecca Rumble
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Vincent Doré
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC 3168, Australia; (Y.Y.L.); (V.D.)
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Christopher C. Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
| | - James S. Wiley
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
- CogState Ltd., Melbourne, VIC 3001, Australia
| | - Colin L. Masters
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Ben J. Gu
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: ; Tel.: +61-3-9035-6317
| |
Collapse
|
6
|
Elhage A, Turner RJ, Cuthbertson P, Watson D, Sluyter R. Preparation of the Murine Anti-Human P2X7 Receptor Monoclonal Antibody (Clone L4). METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2510:77-98. [PMID: 35776321 DOI: 10.1007/978-1-0716-2384-8_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The murine anti-human P2X7 receptor monoclonal antibody (mAb) (clone L4) has been used to study the expression and function of the P2X7 receptor on primary leukocytes, keratinocytes, osteoblasts and neuronal cells, as well as various cell lines. This antibody has also been used to characterize polymorphic variants and isoforms of the P2RX7 gene and P2X7 site-directed mutations, and to identify molecules coassociated with P2X7 in the plasma membrane. This chapter describes the maintenance and cryopreservation of the L4 hybridoma cell line, as well as the generation of tissue culture supernatant containing the anti-human P2X7 mAb, and its subsequent purification by Protein A chromatography and conjugation to DyLight™ 488. Moreover, this chapter describes flow cytometric assays to assess the blocking activity and binding of the anti-human P2X7 mAb against P2X7 on human RPMI 8226 multiple myeloma cells.
Collapse
Affiliation(s)
- Amal Elhage
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Ross J Turner
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, NSW, Australia
| | - Peter Cuthbertson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Debbie Watson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Ronald Sluyter
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia.
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
7
|
Janho dit Hreich S, Benzaquen J, Hofman P, Vouret-Craviari V. The Purinergic Landscape of Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14081926. [PMID: 35454832 PMCID: PMC9025794 DOI: 10.3390/cancers14081926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Lung cancer is the most common cancer worldwide. Despite recent therapeutic advances, including targeted therapies and immune checkpoint inhibitors, the disease progresses in almost all advanced lung cancers and in up to 50% of early-stage cancers. The purpose of this review is to discuss whether purinergic checkpoints (CD39, CD73, P2RX7, and ADORs), which shape the immune response in the tumor microenvironment, may represent novel therapeutic targets to combat progression of non-small cell lung cancer by enhancing the antitumor immune response.
Collapse
Affiliation(s)
- Serena Janho dit Hreich
- Institute of Research on Cancer and Aging (IRCAN, CNRS, INSERM), FHU OncoAge, Université Côte d’Azur, 06108 Nice, France; (S.J.d.H.); (J.B.)
| | - Jonathan Benzaquen
- Institute of Research on Cancer and Aging (IRCAN, CNRS, INSERM), FHU OncoAge, Université Côte d’Azur, 06108 Nice, France; (S.J.d.H.); (J.B.)
| | - Paul Hofman
- CHU Nice, Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d’Azur, 06000 Nice, France;
- Institute of Research on Cancer and Aging (IRCAN, CNRS, INSERM, Team 4), Université Côte d’Azur, 06100 Nice, France
- CHU Nice, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Université Côte d’Azur, 06000 Nice, France
| | - Valérie Vouret-Craviari
- Institute of Research on Cancer and Aging (IRCAN, CNRS, INSERM), FHU OncoAge, Université Côte d’Azur, 06108 Nice, France; (S.J.d.H.); (J.B.)
- Correspondence: ; Tel.: +33-492-031-223
| |
Collapse
|
8
|
Wong J, Gu BJ, Teoh H, Krupa M, Monif M, Slee M, Wiley JS. Flow Cytometry Identifies an Early Stage of Platelet Apoptosis Produced by Agonists of the P2X1 and P2X7 Receptors. Platelets 2022; 33:621-631. [PMID: 35042433 DOI: 10.1080/09537104.2021.1981844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Platelets express P2X1 receptors and our data also show the expression of P2X7 receptors. We studied the role of both receptors in platelet apoptosis by incubation of PRP with P2X agonists, then centrifuged to remove viable platelets, and analyzed the supernatant by flow cytometry to identify a sparse platelet-derived population that stained with MitoTracker dyes and CD41. BzATP, a potent agonist of P2X receptors, and ABT737, an activator of intrinsic apoptosis, produced altered platelets that stained moderately for annexin V and corresponded to an early stage apoptotic platelet (ESAP). Over a range of BzATP concentrations, we observed a dose-dependent formation of ESAPs between 5 and 500 uM BzATP, together with a variable formation of ESAPs at nanomolar ATP or BzATP (50-200 nM). Production of ESAPs occurred with αβ-meATP, while responses with either BzATP or αβ-meATP showed desensitization at a higher agonist concentration. Formation of ESAPs by either 100 nM or 0.5 mM BzATP was inhibited by preincubation of platelets with latrunculin A, an inhibitor of the actin cytoskeleton that prevents apoptosis. ESAP production was totally inhibited by preincubation of platelets with methyl-beta-cyclodextrin, which removes cholesterol from lipid rafts. Our data show that both P2X1 and P2X7 receptors are localized in platelet lipid rafts where P2X-agonists act to produce early stage apoptotic platelets.
Collapse
Affiliation(s)
- Joelyn Wong
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ben J Gu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Harry Teoh
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - Malgorzata Krupa
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - Mastura Monif
- Department of Neurology, Royal Melbourne Hospital, Parkville, Australia.,Department of Neuroscience, Monash University, Clayton, Australia
| | - Mark Slee
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - James S Wiley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Haematology Department, Box Hill Hospital, Australia
| |
Collapse
|
9
|
Hurtado-Navarro L, Baroja-Mazo A, Pelegrín P. Characterization of P2X7 Receptors in Human Blood Cells. Methods Mol Biol 2022; 2510:279-290. [PMID: 35776331 DOI: 10.1007/978-1-0716-2384-8_15] [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] [Indexed: 06/15/2023]
Abstract
P2X7 receptors are expressed in different circulating immune cells. Depending on the cell in which they are expressed, their function could vary from activation of the leucine-rich repeat receptor pyrin domain containing 3 (NLRP3) inflammasome or the shedding of different surface receptors. The P2X7 receptor has been involved in the evolution of several inflammatory diseases, and thus its characterization in blood samples is important to investigate its role in human diseases. In this chapter, we present different methods to detect the presence and the activation of P2X7 receptors in whole blood samples. These methods could be used to evaluate the degree of P2X7 receptor activation in blood samples from patients suffering different chronic inflammatory diseases.
Collapse
Affiliation(s)
- Laura Hurtado-Navarro
- Biomedical Research Institute of Murcia (IMIB), Clinical University Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Alberto Baroja-Mazo
- Biomedical Research Institute of Murcia (IMIB), Clinical University Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Pablo Pelegrín
- Department of Biochemistry and Molecular Biology B and Immunology, University of Murcia, Murcia, Spain.
| |
Collapse
|
10
|
Stähler T, Danquah W, Demeules M, Gondé H, Hardet R, Haag F, Adriouch S, Koch-Nolte F, Menzel S. Development of Antibody and Nanobody Tools for P2X7. Methods Mol Biol 2022; 2510:99-127. [PMID: 35776322 DOI: 10.1007/978-1-0716-2384-8_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Antibodies that recognize the ATP-gated P2X7 ion channel are etablished research tools. Nanobodies correspond to the antigen-binding variable immunoglobulin domain (VHH) of heavy chain antibodies that naturally occur in camelids. Nanobodies display better solubility than the variable domains (VH) of conventional antibodies. Therefore, it is much easier to construct bivalent and multivalent fusion proteins with nanobodies than with VH domains or with paired VH-VL domains. Moreover, nanobodies can bind functional crevices that are poorly accessbile to conventional VH-VL domains. This makes nanobodies particulary well suited as functional modulators. Here we provide protocols to raise antibodies and nanobodies against mouse and human P2X7 using cDNA-immunization. This approach evokes antibodies and nanobodies that recognize the P2X7 ion channel in native confirmation, some of which inhibit or potentiate gating of P2X7 by extracellular ATP. Furthermore, we developed protocols for producing P2X7-specific nanobodies and antibodies in vivo using rAAV vectors (AAVnano). This approach can be used either to durably inhibit or potentiate P2X7 function in vivo, or to deplete P2X7-expressing cells.
Collapse
Affiliation(s)
- Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Welbeck Danquah
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Demeules
- UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, aNd immunoTHERapies (PANTHER), Normandie University, 76000 Rouen, France
| | - Henri Gondé
- UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, aNd immunoTHERapies (PANTHER), Normandie University, 76000 Rouen, France
| | - Romain Hardet
- UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, aNd immunoTHERapies (PANTHER), Normandie University, 76000 Rouen, France
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sahil Adriouch
- UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, aNd immunoTHERapies (PANTHER), Normandie University, 76000 Rouen, France
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Ren W, Rubini P, Tang Y, Engel T, Illes P. Inherent P2X7 Receptors Regulate Macrophage Functions during Inflammatory Diseases. Int J Mol Sci 2021; 23:ijms23010232. [PMID: 35008658 PMCID: PMC8745241 DOI: 10.3390/ijms23010232] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages are mononuclear phagocytes which derive either from blood-borne monocytes or reside as resident macrophages in peripheral (Kupffer cells of the liver, marginal zone macrophages of the spleen, alveolar macrophages of the lung) and central tissue (microglia). They occur as M1 (pro-inflammatory; classic) or M2 (anti-inflammatory; alternatively activated) phenotypes. Macrophages possess P2X7 receptors (Rs) which respond to high concentrations of extracellular ATP under pathological conditions by allowing the non-selective fluxes of cations (Na+, Ca2+, K+). Activation of P2X7Rs by still higher concentrations of ATP, especially after repetitive agonist application, leads to the opening of membrane pores permeable to ~900 Da molecules. For this effect an interaction of the P2X7R with a range of other membrane channels (e.g., P2X4R, transient receptor potential A1 [TRPA1], pannexin-1 hemichannel, ANO6 chloride channel) is required. Macrophage-localized P2X7Rs have to be co-activated with the lipopolysaccharide-sensitive toll-like receptor 4 (TLR4) in order to induce the formation of the inflammasome 3 (NLRP3), which then activates the pro-interleukin-1β (pro-IL-1β)-degrading caspase-1 to lead to IL-1β release. Moreover, inflammatory diseases (e.g., rheumatoid arthritis, Crohn’s disease, sepsis, etc.) are generated downstream of the P2X7R-induced upregulation of intracellular second messengers (e.g., phospholipase A2, p38 mitogen-activated kinase, and rho G proteins). In conclusion, P2X7Rs at macrophages appear to be important targets to preserve immune homeostasis with possible therapeutic consequences.
Collapse
Affiliation(s)
- Wenjing Ren
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of TCM, Chengdu 610075, China; (W.R.); (P.R.); (Y.T.)
- School of Acupunct3ure and Tuina, Chengdu University of TCM, Chengdu 610075, China
| | - Patrizia Rubini
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of TCM, Chengdu 610075, China; (W.R.); (P.R.); (Y.T.)
- School of Acupunct3ure and Tuina, Chengdu University of TCM, Chengdu 610075, China
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of TCM, Chengdu 610075, China; (W.R.); (P.R.); (Y.T.)
- School of Acupunct3ure and Tuina, Chengdu University of TCM, Chengdu 610075, China
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Peter Illes
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of TCM, Chengdu 610075, China; (W.R.); (P.R.); (Y.T.)
- School of Acupunct3ure and Tuina, Chengdu University of TCM, Chengdu 610075, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107 Leipzig, Germany
- Correspondence:
| |
Collapse
|
12
|
Huang Z, Tan S. P2X7 Receptor as a Potential Target for Major Depressive Disorder. Curr Drug Targets 2021; 22:1108-1120. [PMID: 33494675 DOI: 10.2174/1389450122666210120141908] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 11/22/2022]
Abstract
Major depressive disorder (MDD) is a common mental disorder. Although the genetic, biochemical, and psychological factors have been related to the development of MDD, it is generally believed that a series of pathological changes in the brain caused by chronic stress is the main cause of MDD. However, the specific mechanisms underlying chronic stress-induced MDD are largely undermined. Recent investigations have found that increased pro-inflammatory cytokines and changes in the inflammatory pathway in the microglia cells in the brain are the potential pathophysiological mechanism of MDD. P2X7 receptor (P2X7R) and its mediated signaling pathway play a key role in microglia activation. The present review aimed to present and discuss the accumulating data on the role of P2X7R in MDD. Firstly, we summarized the research progress in the correlation between P2X7R and MDD. Subsequently, we presented the P2X7R mediated microglia activation in MDD and the role of P2X7R in increased blood-brain barrier (BBB) permeability caused by chronic stress. Lastly, we also discussed the potential mechanism underlying-P2X7R expression changes after chronic stress. In conclusion, P2X7R is a key molecule regulating the activation of microglia. Chronic stress activates microglia in the hippocampus by secreting interleukin- 1β (IL-1β) and other inflammatory cytokines, and increasing the BBB permeability, thus promoting the occurrence and development of MDD, which indicated that P2X7R might be a promising therapeutic target for MDD.
Collapse
Affiliation(s)
- Zeyi Huang
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 421001, Hunan, China
| | - Sijie Tan
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 421001, Hunan, China
| |
Collapse
|
13
|
Therapeutic Antibodies Targeting Potassium Ion Channels. Handb Exp Pharmacol 2021; 267:507-545. [PMID: 33963460 DOI: 10.1007/164_2021_464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Monoclonal antibodies combine specificity and high affinity binding with excellent pharmacokinetic properties and are rapidly being developed for a wide range of drug targets including clinically important potassium ion channels. Nonetheless, while therapeutic antibodies come with great promise, K+ channels represent particularly difficult targets for biologics development for a variety of reasons that include their dynamic structures and relatively small extracellular loops, their high degree of sequence conservation (leading to immune tolerance), and their generally low-level expression in vivo. The process is made all the more difficult when large numbers of antibody candidates must be screened for a given target, or when lead candidates fail to cross-react with orthologous channels in animal disease models due to their highly selective binding properties. While the number of antibodies targeting potassium channels in preclinical or clinical development is still modest, significant advances in the areas of protein expression and antibody screening are converging to open the field to an avalanche of new drugs. Here, the opportunities and constraints associated with the discovery of antibodies against K+ channels are discussed, with an emphasis on novel technologies that are opening the field to exciting new possibilities for biologics development.
Collapse
|
14
|
Mini-TrpRS is essential for IFNγ-induced monocyte-derived giant cell formation. Cytokine 2021; 142:155486. [PMID: 33721618 DOI: 10.1016/j.cyto.2021.155486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/07/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022]
Abstract
Truncated tryptophanyl-tRNA synthetase (mini-TrpRS), like any other aminoacyl-tRNA synthetases, canonically functions as a protein synthesis enzyme. Here we provide evidence for an additional signaling role of mini-TrpRS in the formation of monocyte-derived multinuclear giant cells (MGCs). Interferon-gamma (IFNγ) readily induced monocyte aggregation leading to MGC formation with paralleled marked upregulation of mini-TrpRS. Small interfering (si)RNA, targeting mini-TrpRS in the presence of IFNγ prevented monocyte aggregation. Moreover, blockade of mini-TrpRS, either by siRNA, or the cognate amino acid and decoy substrate D-Tryptophan to prevent mini-TrpRS signaling, resulted in a marked reduction in expression of the purinergic receptor P2X 7 (P2RX7) in monocytes activated by IFNγ. Our findings identify mini-TrpRS as a critical signaling molecule in a mechanism by which IFNγ initiates monocyte-derived giant cell formation.
Collapse
|
15
|
P2X7 receptor and the NLRP3 inflammasome: Partners in crime. Biochem Pharmacol 2020; 187:114385. [PMID: 33359010 DOI: 10.1016/j.bcp.2020.114385] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
Adenosine triphosphate (ATP) is a molecule that on one hand plays a central role in cellular energetics and which on the other is a ubiquitous signaling molecule when released into the extracellular media. Extracellular ATP accumulates in inflammatory environments where it acts as a damage-associated molecular pattern and activates the purinergic P2X receptor 7 (P2X7) in immune cells. P2X7 receptor activation induces the formation of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3 (NLRP3) inflammasome and the activation of the inflammatory caspase-1. Caspase-1 causes an inflammatory type of cell death called pyroptosis through the release of pro-inflammatory cytokines and intracellular content. Consequently, intense research efforts have been devoted to the design of novel anti-inflammatory therapies, focusing in particular on the P2X7 receptor and the NLRP3 pathway and the introduction of new blocking molecules in early phase clinical trials.
Collapse
|
16
|
Barczyk A, Bauderlique‐Le Roy H, Jouy N, Renault N, Hottin A, Millet R, Vouret‐Craviari V, Adriouch S, Idziorek T, Dezitter X. Flow cytometry: An accurate tool for screening
P2RX7
modulators. Cytometry A 2020. [DOI: 10.1002/cyto.a.24287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Amélie Barczyk
- Univ. Lille, Inserm, CHU Lille, U1286 – Infinite – Institute for Translational Research in Inflammation Lille France
| | - Hélène Bauderlique‐Le Roy
- Univ. Lille, UMS 2014‐US 41 PLBS BICel, Flow Cytometry Core Facility, Institut Pasteur de Lille Lille cedex France
| | - Nathalie Jouy
- Univ. Lille, UMS 2014‐US 41 PLBS BICel, Flow Cytometry Core Facility, IRCL, 1 place de Verdun Lille cedex France
| | - Nicolas Renault
- Univ. Lille, Inserm, CHU Lille, U1286 – Infinite – Institute for Translational Research in Inflammation Lille France
| | - Audrey Hottin
- Univ. Lille, Inserm, CHU Lille, U1286 – Infinite – Institute for Translational Research in Inflammation Lille France
| | - Régis Millet
- Univ. Lille, Inserm, CHU Lille, U1286 – Infinite – Institute for Translational Research in Inflammation Lille France
| | - Valérie Vouret‐Craviari
- University Cote d'Azur, Institute for Research on Cancer and Aging, IRCAN U1081 UMR CNRS 7284 Nice France
| | - Sahil Adriouch
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies (PANTHER) Rouen France
| | - Thierry Idziorek
- Univ. Lille, UMS 2014‐US 41 PLBS BICel, Flow Cytometry Core Facility, IRCL, 1 place de Verdun Lille cedex France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut de Recherche contre le Cancer de Lille, UMR9020 – UMR‐S 1277 ‐ Canther – Cancer Heterogeneity, Plasticity and Resistance to Therapies Lille France
| | - Xavier Dezitter
- Univ. Lille, Inserm, CHU Lille, U1286 – Infinite – Institute for Translational Research in Inflammation Lille France
| |
Collapse
|
17
|
Di Virgilio F, Vultaggio-Poma V, Sarti AC. P2X receptors in cancer growth and progression. Biochem Pharmacol 2020; 187:114350. [PMID: 33253643 DOI: 10.1016/j.bcp.2020.114350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that ion channels have a crucial role in tumors, either as promoters of cancer cell growth, or modulators of immune cell functions, or both. Among ion channels, P2X receptors have a special status because they are gated by ATP, a common and abundant component of the tumor microenvironment. Furthermore, one P2X receptor, i.e. P2X7, may also function as a conduit for ATP release, thus fuelling the increased extracellular ATP level in the tumor interstitium. These findings show that P2X receptors and extracellular ATP are indissoluble partners and key regulators of tumor growth, and suggest the exploitation of the extracellular ATP-P2X partnership to develop innovative therapeutic approaches to cancer.
Collapse
|
18
|
Alarcón-Vila C, Baroja-Mazo A, de Torre-Minguela C, Martínez CM, Martínez-García JJ, Martínez-Banaclocha H, García-Palenciano C, Pelegrin P. CD14 release induced by P2X7 receptor restricts inflammation and increases survival during sepsis. eLife 2020; 9:60849. [PMID: 33135636 PMCID: PMC7690950 DOI: 10.7554/elife.60849] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022] Open
Abstract
P2X7 receptor activation induces the release of different cellular proteins, such as CD14, a glycosylphosphatidylinositol (GPI)-anchored protein to the plasma membrane important for LPS signaling via TLR4. Circulating CD14 has been found at elevated levels in sepsis, but the exact mechanism of CD14 release in sepsis has not been established. Here, we show for first time that P2X7 receptor induces the release of CD14 in extracellular vesicles, resulting in a net reduction in macrophage plasma membrane CD14 that functionally affects LPS, but not monophosphoryl lipid A, pro-inflammatory cytokine production. Also, we found that during a murine model of sepsis, P2X7 receptor activity is important for maintaining elevated levels of CD14 in biological fluids and a decrease in its activity results in higher bacterial load and exacerbated organ damage, ultimately leading to premature deaths. Our data reveal that P2X7 is a key receptor for helping to clear sepsis because it maintains elevated concentrations of circulating CD14 during infection. When the immune system detects an infection, it often launches an inflammatory response to fight off the disease. This defense mechanism is activated by a cascade of signaling molecules that can aggravate inflammation, causing it to damage the body’s own tissues and organs. This life-threatening reaction is referred to as sepsis, and kills around 11 million people each year. New approaches are therefore needed to help alleviate the damage caused by this condition. The inflammatory response is often triggered by proteins called receptors, which sit on the surface of immune cells. When these receptors are activated, they induce cells to secrete proteins that travel around the body and activate immune cells that can eliminate the infection. In 2016, a group of researchers showed that a receptor called P2X7 stimulates the release of a signaling molecule called CD14. Patients with sepsis often have elevated amounts of CD14 in their bloodstream. Yet, it remained unclear what causes this rise in CD14 and what role this molecule plays in the development of sepsis. Now, Alarcón-Vila et al. – including some of the researchers involved in the 2016 study – have investigated the role of P2X7 in mice undergoing sepsis. This was done by puncturing the mice’s intestines, causing bacteria to leak out and initiate an over-active immune response. Alarcón-Vila et al. found that mice lacking the P2X7 receptor had less CD14 and struggled to eliminate the bacterial infection from their system. This increase in bacteria caused excessive damage to the mice’s organs, ultimately leading to premature death. These findings suggest that P2X7 plays an important role in preventing the onset of sepsis by helping maintain high levels of CD14 following infection. This result could help to identify new therapies that reduce the mortality rates of septic infections.
Collapse
Affiliation(s)
- Cristina Alarcón-Vila
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Alberto Baroja-Mazo
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Carlos de Torre-Minguela
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Carlos M Martínez
- Plataforma de Patología, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Murcia, Spain
| | - Juan J Martínez-García
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Helios Martínez-Banaclocha
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Pablo Pelegrin
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| |
Collapse
|
19
|
Human P2X7 Receptor Causes Cycle Arrest in RPMI-8226 Myeloma Cells to Alter the Interaction with Osteoblasts and Osteoclasts. Cells 2020; 9:cells9112341. [PMID: 33105696 PMCID: PMC7690412 DOI: 10.3390/cells9112341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma is a malignant expansion of plasma cells and aggressively affects bone health. We show that P2X7 receptor altered myeloma growth, which affects primary bone cells in vitro. Expression on six human myeloma cell lines confirmed the heterogeneity associated with P2X7 receptor. Pharmacology with 2′(3′)-O-(4-benzoylbenzoyl) adenosine 5′-triphosphate (BzATP) as agonist showed dose-dependent membranal pores on RPMI-8226 (p = 0.0027) and blockade with P2X7 receptor antagonists. Ca2+ influx with increasing doses of BzATP (p = 0.0040) was also inhibited with antagonists. Chronic P2X7 receptor activation reduced RPMI-8226 viability (p = 0.0208). No apoptosis or RPMI-8226 death was observed by annexin V/propidium iodide (PI) labeling and caspase-3 cleavage, respectively. However, bromodeoxyuridine (BrdU) labelling showed an accumulation of RPMI-8226 in the S phase of cell cycle progression (61.5%, p = 0.0114) with significant decline in G0/G1 (5.2%, p = 0.0086) and G2/M (23.5%, p = 0.0015) phases. As myeloma pathology depends on a positive and proximal interaction with bone, we show that P2X7 receptor on RPMI-8226 inhibited the myeloma-induced suppression on mineralization (p = 0.0286) and reversed the excessive osteoclastic resorption. Our results demonstrate a view of how myeloma cell growth is halted by P2X7 receptor and the consequences on myeloma–osteoblast and myeloma–osteoclast interaction in vitro.
Collapse
|
20
|
Lara R, Adinolfi E, Harwood CA, Philpott M, Barden JA, Di Virgilio F, McNulty S. P2X7 in Cancer: From Molecular Mechanisms to Therapeutics. Front Pharmacol 2020; 11:793. [PMID: 32581786 PMCID: PMC7287489 DOI: 10.3389/fphar.2020.00793] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
P2X7 is a transmembrane receptor expressed in multiple cell types including neurons, dendritic cells, macrophages, monocytes, B and T cells where it can drive a wide range of physiological responses from pain transduction to immune response. Upon activation by its main ligand, extracellular ATP, P2X7 can form a nonselective channel for cations to enter the cell. Prolonged activation of P2X7, via high levels of extracellular ATP over an extended time period can lead to the formation of a macropore, leading to depolarization of the plasma membrane and ultimately to cell death. Thus, dependent on its activation state, P2X7 can either drive cell survival and proliferation, or induce cell death. In cancer, P2X7 has been shown to have a broad range of functions, including playing key roles in the development and spread of tumor cells. It is therefore unsurprising that P2X7 has been reported to be upregulated in several malignancies. Critically, ATP is present at high extracellular concentrations in the tumor microenvironment (TME) compared to levels observed in normal tissues. These high levels of ATP should present a survival challenge for cancer cells, potentially leading to constitutive receptor activation, prolonged macropore formation and ultimately to cell death. Therefore, to deliver the proven advantages for P2X7 in driving tumor survival and metastatic potential, the P2X7 macropore must be tightly controlled while retaining other functions. Studies have shown that commonly expressed P2X7 splice variants, distinct SNPs and post-translational receptor modifications can impair the capacity of P2X7 to open the macropore. These receptor modifications and potentially others may ultimately protect cancer cells from the negative consequences associated with constitutive activation of P2X7. Significantly, the effects of both P2X7 agonists and antagonists in preclinical tumor models of cancer demonstrate the potential for agents modifying P2X7 function, to provide innovative cancer therapies. This review summarizes recent advances in understanding of the structure and functions of P2X7 and how these impact P2X7 roles in cancer progression. We also review potential therapeutic approaches directed against P2X7.
Collapse
Affiliation(s)
- Romain Lara
- Biosceptre (UK) Limited, Cambridge, United Kingdom
| | - Elena Adinolfi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mike Philpott
- Centre for Cutaneous Research, Blizard Institute, Bart's & The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
21
|
Baeza-Raja B, Goodyear A, Liu X, Lam K, Yamamoto L, Li Y, Dodson GS, Takeuchi T, Kisseleva T, Brenner DA, Dabbagh K. Pharmacological inhibition of P2RX7 ameliorates liver injury by reducing inflammation and fibrosis. PLoS One 2020; 15:e0234038. [PMID: 32492075 PMCID: PMC7269334 DOI: 10.1371/journal.pone.0234038] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular adenosine triphosphate (eATP) released by damaged cells, and its purinergic receptors, comprise a crucial signaling network after injury. Purinergic receptor P2X7 (P2RX7), a major driver of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and IL-1β processing, has been shown to play a role in liver injury in murine diet- and chemically-induced liver injury models. It is unclear, however, whether P2RX7 plays a role in non-alcoholic steatohepatitis (NASH) and which cell type is the main target of P2RX7 pharmacological inhibition. Here, we report that P2RX7 is expressed by infiltrating monocytes and resident Kupffer cells in livers from NASH-affected individuals. Using primary isolated human cells, we demonstrate that P2RX7 expression in CD14+ monocytes and Kupffer cells primarily mediates IL-1β release. In addition, we show that pharmacological inhibition of P2RX7 in monocytes and Kupffer cells, blocks IL-1β release, reducing hepatocyte caspase 3/7 activity, IL-1β-mediated CCL2 and CCL5 chemokine gene expression and secretion, and hepatic stellate cell (HSC) procollagen secretion. Consequently, in a chemically-induced nonhuman primate model of liver fibrosis, treatment with a P2RX7 inhibitor improved histological characteristics of NASH, protecting from liver inflammation and fibrosis. Taken together, these findings underscore the critical role of P2RX7 in the pathogenesis of NASH and implicate P2RX7 as a promising therapeutic target for the management of this disease.
Collapse
Affiliation(s)
- Bernat Baeza-Raja
- Second Genome Inc., South San Francisco, California, United States of America
| | - Andrew Goodyear
- Second Genome Inc., South San Francisco, California, United States of America
| | - Xiao Liu
- Department of Surgery, University of California San Diego, La Jolla, California, United States of America
| | - Kevin Lam
- Department of Surgery, University of California San Diego, La Jolla, California, United States of America
| | - Lynn Yamamoto
- Second Genome Inc., South San Francisco, California, United States of America
| | - Yingwu Li
- Second Genome Inc., South San Francisco, California, United States of America
| | - G. Steven Dodson
- Second Genome Inc., South San Francisco, California, United States of America
| | - Toshi Takeuchi
- Second Genome Inc., South San Francisco, California, United States of America
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, California, United States of America
| | - David A. Brenner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Karim Dabbagh
- Second Genome Inc., South San Francisco, California, United States of America
| |
Collapse
|
22
|
Generating therapeutic monoclonal antibodies to complex multi-spanning membrane targets: Overcoming the antigen challenge and enabling discovery strategies. Methods 2020; 180:111-126. [PMID: 32422249 DOI: 10.1016/j.ymeth.2020.05.006] [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: 02/24/2020] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning helices, encompass families of proteins which are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels and transporters. Although these proteins have typically been targeted by small molecule drugs and peptides, the high specificity of monoclonal antibodies offers a significant opportunity to selectively modulate these target proteins. However, it remains the case that isolation of antibodies with desired pharmacological function(s) has proven difficult due to technical challenges in preparing membrane protein antigens suitable to support antibody drug discovery. In this review recent progress in defining strategies for generation of membrane protein antigens is outlined. We also highlight antibody isolation strategies which have generated antibodies which bind the membrane protein and modulate the protein function.
Collapse
|
23
|
He S, Moutaoufik MT, Islam S, Persad A, Wu A, Aly KA, Fonge H, Babu M, Cayabyab FS. HERG channel and cancer: A mechanistic review of carcinogenic processes and therapeutic potential. Biochim Biophys Acta Rev Cancer 2020; 1873:188355. [PMID: 32135169 DOI: 10.1016/j.bbcan.2020.188355] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/21/2022]
Abstract
The human ether-à-go-go related gene (HERG) encodes the alpha subunit of Kv11.1, which is a voltage-gated K+ channel protein mainly expressed in heart and brain tissue. HERG plays critical role in cardiac repolarization, and mutations in HERG can cause long QT syndrome. More recently, evidence has emerged that HERG channels are aberrantly expressed in many kinds of cancer cells and play important roles in cancer progression. HERG could therefore be a potential biomarker for cancer and a possible molecular target for anticancer drug design. HERG affects a number of cellular processes, including cell proliferation, apoptosis, angiogenesis and migration, any of which could be affected by dysregulation of HERG. This review provides an overview of available information on HERG channel as it relates to cancer, with focus on the mechanism by which HERG influences cancer progression. Molecular docking attempts suggest two possible protein-protein interactions of HERG with the ß1-integrin receptor and the transcription factor STAT-1 as novel HERG-directed therapeutic targeting which avoids possible cardiotoxicity. The role of epigenetics in regulating HERG channel expression and activity in cancer will also be discussed. Finally, given its inherent extracellular accessibility as an ion channel, we discuss regulatory roles of this molecule in cancer physiology and therapeutic potential. Future research should be directed to explore the possibilities of therapeutic interventions targeting HERG channels while minding possible complications.
Collapse
Affiliation(s)
- Siyi He
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | | | - Saadul Islam
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Amit Persad
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Adam Wu
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Khaled A Aly
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W8, Canada; Department of Medical Imaging, Royal University Hospital, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Francisco S Cayabyab
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
24
|
Pegoraro A, Bortolotti D, Marci R, Caselli E, Falzoni S, De Marchi E, Di Virgilio F, Rizzo R, Adinolfi E. The P2X7 Receptor 489C>T Gain of Function Polymorphism Favors HHV-6A Infection and Associates With Female Idiopathic Infertility. Front Pharmacol 2020; 11:96. [PMID: 32153407 PMCID: PMC7046806 DOI: 10.3389/fphar.2020.00096] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 01/16/2023] Open
Abstract
The P2X7 receptor (P2X7R) is an ATP-gated ion channel known for its proinflammatory activity. Despite its participation in host defense against pathogens, the role played in viral infections, notably those caused by herpes viruses, has been seldom studied. Here we investigated the effect of P2X7R expression on human herpes virus 6 A (HHV-6A) infection of P2X7R-expressing HEK293 cells. We show that functional P2X7R increases while its blockade decreases viral load. Interestingly, HHV-6A infection was enhanced in HEK293 cells transfected with P2X7R cDNA bearing the gain of function 489C>T SNP (rs208294, replacing a histidine for tyrosine at position 155). The P2X7R 489C>T polymorphism correlated with HHV-6A infection also in a cohort of 50 women affected with idiopathic infertility, a condition previously shown to correlate with HHV-6A infection. None of the infertile women infected by HHV-6A was homozygote for 489CC genotype, while on the contrary HHV-6A infection significantly associated with the presence of the rs208294 allele. Levels of soluble human leukocyte antigen G (sHLA-G), a factor promoting embryo implant, measured in uterine flushings negatively correlated with the 489TT genotype and HHV-6A infection, while proinflammatory cytokines interleukins 1α (IL-1α), 1β (IL-1β), and 8 (IL-8) positively correlated with both the 489T allele presence and viral infection. Taken together these data point to the P2X7R as a new therapeutic target to prevent HHV-6A infection and the associated infertility.
Collapse
Affiliation(s)
- Anna Pegoraro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Daria Bortolotti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberto Marci
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Obstetrics and Gynaecology, School of Medicine, University of Geneve, Geneve, Switzerland
| | | | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Roberta Rizzo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
25
|
Skarratt KK, Gu BJ, Lovelace MD, Milligan CJ, Stokes L, Glover R, Petrou S, Wiley JS, Fuller SJ. A P2RX7 single nucleotide polymorphism haplotype promotes exon 7 and 8 skipping and disrupts receptor function. FASEB J 2020; 34:3884-3901. [PMID: 32003498 DOI: 10.1096/fj.201901198rr] [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: 05/14/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
P2X7 is an ATP-gated membrane ion channel that is expressed by multiple cell types. Brief exposure to ATP induces the opening of a nonselective cation channel; while repeated or prolonged exposure induces formation of a transmembrane pore. This process may be partially regulated by alternative splicing of full-length P2RX7A pre-mRNA, producing isoforms that delete or retain functional domains. Here, we report cloning and expression of a novel P2RX7 splice variant, P2RX7L, that is, characterized by skipping of exons 7 and 8. In HEK 293 cells, expression of P2RX7L produces a protein isoform, P2X7L, that forms a heteromer with P2X7A. A haplotype defined by six single nucleotide polymorphisms (SNPs) (rs208307, rs208306, rs36144485, rs208308, rs208309, and rs373655596) promotes allele-specific alternative splicing, increasing mRNA levels of P2RX7L and another isoform, P2RX7E, which in addition has a truncated C-terminus. Skipping of exons 7 and 8 is predicted to delete critical amino acids in the ATP-binding site. P2X7L-transfected HEK 293 cells have phagocytic but not channel, pore, or membrane-blebbing function, and double-transfected P2X7L and P2X7A cells have reduced pore function. Heteromeric receptor complexes of P2X7A and P2X7L are predicted to have reduced numbers of ATP-binding sites, which potentially alters receptor function compared to homomeric P2X7A complexes.
Collapse
Affiliation(s)
- Kristen K Skarratt
- Department of Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
| | - Ben J Gu
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Michael D Lovelace
- Department of Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
| | - Carol J Milligan
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Leanne Stokes
- Department of Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia.,School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Rachel Glover
- Department of Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - James S Wiley
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Fuller
- Department of Medicine, Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
| |
Collapse
|
26
|
Li M, Luo S, Zhang Y, Jia L, Yang C, Peng X, Zhao R. Production, characterization, and application of a monoclonal antibody specific for the extracellular domain of human P2X7R. Appl Microbiol Biotechnol 2020; 104:2017-2028. [PMID: 31930453 DOI: 10.1007/s00253-019-10340-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/14/2019] [Accepted: 12/27/2019] [Indexed: 12/17/2022]
Abstract
This paper focuses on the production of a high-affinity monoclonal antibody (mAb) that can efficiently detect and block purinergic ligand-gated ion channel 7 receptor (P2X7R). To achieve this goal, the extracellular domain of human P2X7R, P2X7R-ECD, was used as an immunogen for BALB/c mice, inducing them to produce spleen lymphocytes that were subsequently fused with myeloma cells. Screening of the resultant hybridoma clones resulted in the selection of one stable positive clone that produced a qualified mAb, named 4B3A4. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis demonstrated that the purity of the purified 4B3A4 mAb was above 85%, with prominent bands corresponding to molecular weights of 55 kDa (heavy chain) and 25 kDa (light chain), and the BCA assay showed that the concentration of the purified 4B3A4 mAb was 0.3 mg/mL. Western blot analysis revealed that the 4B3A4 mAb could specifically recognize and bind both P2X7R-ECD and the full-length P2X7R protein. Laser scanning confocal microscopy (LSCM) revealed that the 4B3A4 mAb specifically bound to P2X7R on the membrane of human peripheral blood mononuclear cells (PBMCs). P2X7R expression was significantly different between healthy individuals and people with certain cancers as determined by flow cytometry (FCM). In addition, the 4B3A4 mAb significantly reduced ATP-stimulated Ca2+ entry and YO-PRO-1 uptake, which indicated that the 4B3A4 mAb effectively blocked P2X7R activity. These data indicate that the 4B3A4 mAb can be further used as not only an antibody to detect cell surface P2X7R but also as a therapeutic antibody to target P2X7R-related signaling pathways.
Collapse
Affiliation(s)
- Mingxuan Li
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Shuping Luo
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yunfang Zhang
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Lina Jia
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Chuanyu Yang
- Department of Blood Transfusion, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xiaoxiang Peng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Ronglan Zhao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, Shandong, China.
| |
Collapse
|
27
|
Ito Y, Kitakawa M, Koshikawa S, Watanabe H, Sueki H. CD14 and CD16 expression in noninfectious granulomatous skin diseases. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2020. [DOI: 10.1002/cia2.12091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yuta Ito
- Department of Dermatology Showa University School of Medicine Tokyo Japan
| | - Maki Kitakawa
- Department of Dermatology Showa University School of Medicine Tokyo Japan
| | - Sachiko Koshikawa
- Department of Dermatology Showa University School of Medicine Tokyo Japan
| | - Hideaki Watanabe
- Department of Dermatology Showa University School of Medicine Tokyo Japan
| | - Hirohiko Sueki
- Department of Dermatology Showa University School of Medicine Tokyo Japan
| |
Collapse
|
28
|
Di Virgilio F, Jiang LH, Roger S, Falzoni S, Sarti AC, Vultaggio-Poma V, Chiozzi P, Adinolfi E. Structure, function and techniques of investigation of the P2X7 receptor (P2X7R) in mammalian cells. Methods Enzymol 2019; 629:115-150. [PMID: 31727237 DOI: 10.1016/bs.mie.2019.07.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The P2X7 receptor [P2X7R or P2RX7 in National Center for Biotechnology Information (NCBI) gene nomenclature] is a member of the P2X receptor (P2XR) subfamily of P2 receptors (P2Rs). The P2X7R is an extracellular ATP-gated ion channel with peculiar permeability properties expressed by most cell types, mainly in the immune system, where it has a leading role in cytokine release, oxygen radical generation, T lymphocyte differentiation and proliferation. A role in cancer cell growth and tumor progression has also been demonstrated. These features make the P2X7R an appealing target for drug development in inflammation and cancer. The functional P2X7R, recently (partially) crystallized and 3-D solved, is formed by the assembly of three identical subunits (homotrimer). The P2X7R is preferentially permeable to small cations (Ca2+, Na+, K+), and in most (but not all) cell types also to large positively charged molecules of molecular mass up to 900Da. Permeability to negatively charged species of comparable molecular mass (e.g., Lucifer yellow) is debated. Several highly selective P2X7R pharmacological blockers have been developed over the years, thus providing powerful tools for P2X7R studies. Biophysical properties and coupling to several different physiological responses make the P2X7R amenable to investigation by electrophysiology and cell biology techniques, which allow its identification and characterization in many different cell types and tissues. A careful description of the physiological features of the P2X7R is a prerequisite for an effective therapeutic development. Here we describe the most common techniques to asses P2X7R functions, including patch-clamp, intracellular calcium measurements, and membrane permeabilization to large fluorescent dyes in a selection of different cell types. In addition, we also describe common toxicity assays used to verify the effects of P2X7R stimulation on cell viability.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- EA4245 Transplantation, Immunology and Inflammation, University of Tours, Tours, France
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Valentina Vultaggio-Poma
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Chiozzi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
29
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
30
|
P2X7 receptor induces mitochondrial failure in monocytes and compromises NLRP3 inflammasome activation during sepsis. Nat Commun 2019; 10:2711. [PMID: 31221993 PMCID: PMC6586640 DOI: 10.1038/s41467-019-10626-x] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is characterized by a systemic inflammatory response followed by immunosuppression of the host. Metabolic defects and mitochondrial failure are common in immunocompromised patients with sepsis. The NLRP3 inflammasome is important for establishing an inflammatory response after activation by the purinergic P2X7 receptor. Here, we study a cohort of individuals with intra-abdominal origin sepsis and show that patient monocytes have impaired NLRP3 activation by the P2X7 receptor. Furthermore, most sepsis-related deaths are among patients whose NLRP3 activation is profoundly altered. In monocytes from sepsis patients, the P2X7 receptor is associated with mitochondrial dysfunction. Furthermore, activation of the P2X7 receptor results in mitochondrial damage, which in turn inhibits NLRP3 activation by HIF-1α. We show that mortality increases in a mouse model of sepsis when the P2X7 receptor is activated in vivo. These data reveal a molecular mechanism initiated by the P2X7 receptor that contributes to NLRP3 impairment during infection. Systemic sepsis is a potentially life-threatening illness and immunocompromised individuals are especially vulnerable. Here, using a cohort of patients with intra-abdominal origin sepsis, the authors show an important role for the NLRP3 inflammasome in establishing a host response, and NLRP3 dysfunction is a common feature of sepsis mortality.
Collapse
|
31
|
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, Lehen'kyi V. Monoclonal Antibodies Targeting Ion Channels and Their Therapeutic Potential. Front Pharmacol 2019; 10:606. [PMID: 31231216 PMCID: PMC6561378 DOI: 10.3389/fphar.2019.00606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) represent a rapidly growing pharmaceutical class of protein drugs that becomes an important part of the precision therapy. mAbs are characterized by their high specificity and affinity for the target antigen, which is mostly present on the cell surface. Ion channels are a large family of transmembrane proteins that control ion transport across the cell membrane. They are involved in almost all biological processes in both health and disease and are widely considered as prospective targets. However, no antibody-based drug targeting ion channel has been developed so far that has progressed to clinical use. Thus, we provide a comprehensive review of the elaborated mAbs against ion channels, describe their mechanisms of action, and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Aline Hantute-Ghesquier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France
| | - V'yacheslav Lehen'kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channel Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, Villeneuve d'Ascq, France.,FONDATION ARC, Villejuif, France
| |
Collapse
|
32
|
Benzaquen J, Heeke S, Janho Dit Hreich S, Douguet L, Marquette CH, Hofman P, Vouret-Craviari V. Alternative splicing of P2RX7 pre-messenger RNA in health and diseases: Myth or reality? Biomed J 2019; 42:141-154. [PMID: 31466708 PMCID: PMC6717933 DOI: 10.1016/j.bj.2019.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
Alternative splicing (AS) tremendously increases the use of genetic information by generating protein isoforms that differ in protein-protein interactions, catalytic activity and/or subcellular localization. This review is not dedicated to AS in general, but rather we focus our attention on AS of P2RX7 pre-mRNA. Whereas P2RX7 mRNA is expressed by virtually all eukaryotic mammalian cells, the expression of this channel receptor is restrained to certain cells. When expressed at the cell membrane, P2RX7 controls downstream events including release of inflammatory molecules, phagocytosis, cell proliferation and death and metabolic events. Therefore, P2RX7 is an important actor of health and diseases. In this review, we summarize the general mechanisms leading to AS. Further, we recapitulate our current knowledge concerning the functional regions in P2RX7, identified at the genetic or exonic levels, and how AS may affect the expression of these regions. Finally, the potential of P2RX7 splice variants to control the fate of cancer cells is discussed.
Collapse
Affiliation(s)
- Jonathan Benzaquen
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; FHU OncoAge, Nice, France
| | - Simon Heeke
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, Nice, France; FHU OncoAge, Nice, France
| | | | | | - Charles Hugo Marquette
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; FHU OncoAge, Nice, France; University of Cote d'Azur, CHU de Nice, Department of Pulmonary Medicine, FHU OncoAge, Nice, France
| | - Paul Hofman
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, Nice, France; Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, Nice, France; FHU OncoAge, Nice, France
| | | |
Collapse
|
33
|
Ion Channel Targeting with Antibodies and Antibody Fragments for Cancer Diagnosis. Antibodies (Basel) 2019; 8:antib8020033. [PMID: 31544839 PMCID: PMC6640718 DOI: 10.3390/antib8020033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
The antibody era has greatly impacted cancer management in recent decades. Indeed, antibodies are currently applied for both cancer diagnosis and therapy. For example, monoclonal antibodies are the main constituents of several in vitro diagnostics, which are applied at many levels of cancer diagnosis. Moreover, the great improvement provided by in vivo imaging, especially for early-stage cancer diagnosis, has traced the path for the development of a complete new class of antibodies, i.e., engineered antibody fragments. The latter embody the optimal characteristics (e.g., low renal retention, rapid clearance, and small size) which make them ideal for in vivo applications. Furthermore, the present review focuses on reviewing the main applications of antibodies and antibody fragments for solid cancer diagnosis, both in vitro and in vivo. Furthermore, we review the scientific evidence showing that ion channels represent an almost unexplored class of ideal targets for both in vitro and in vivo diagnostic purposes. In particular, we review the applications, in solid cancers, of monoclonal antibodies and engineered antibody fragments targeting the voltage-dependent ion channel Kv 11.1, also known as hERG1.
Collapse
|
34
|
Therapeutic Monoclonal Antibodies to Complex Membrane Protein Targets: Antigen Generation and Antibody Discovery Strategies. BioDrugs 2019; 32:339-355. [PMID: 29934752 DOI: 10.1007/s40259-018-0289-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell surface membrane proteins comprise a wide array of structurally and functionally diverse proteins involved in a variety of important physiological and homeostatic processes. Complex integral membrane proteins, which are embedded in the lipid bilayer by multiple transmembrane-spanning helices, are represented by families of proteins that are important target classes for drug discovery. Such protein families include G-protein-coupled receptors, ion channels and transporters. Although these targets have typically been the domain of small-molecule drugs, the exquisite specificity of monoclonal antibodies offers a significant opportunity to selectively modulate these target proteins. Nevertheless, the isolation of antibodies with desired pharmacological functions has proved difficult because of technical challenges in preparing membrane protein antigens for antibody drug discovery. In this review, we describe recent progress in defining strategies for the generation of membrane protein antigens. We also describe antibody-isolation strategies that identify antibodies that bind the membrane protein and modulate protein function.
Collapse
|
35
|
Wulff H, Christophersen P, Colussi P, Chandy KG, Yarov-Yarovoy V. Antibodies and venom peptides: new modalities for ion channels. Nat Rev Drug Discov 2019; 18:339-357. [PMID: 30728472 PMCID: PMC6499689 DOI: 10.1038/s41573-019-0013-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ion channels play fundamental roles in both excitable and non-excitable tissues and therefore constitute attractive drug targets for myriad neurological, cardiovascular and metabolic diseases as well as for cancer and immunomodulation. However, achieving selectivity for specific ion channel subtypes with small-molecule drugs has been challenging, and there currently is a growing trend to target ion channels with biologics. One approach is to improve the pharmacokinetics of existing or novel venom-derived peptides. In parallel, after initial studies with polyclonal antibodies demonstrated the technical feasibility of inhibiting channel function with antibodies, multiple preclinical programmes are now using the full spectrum of available technologies to generate conventional monoclonal and engineered antibodies or nanobodies against extracellular loops of ion channels. After a summary of the current state of ion channel drug discovery, this Review discusses recent developments using the purinergic receptor channel P2X purinoceptor 7 (P2X7), the voltage-gated potassium channel KV1.3 and the voltage-gated sodium channel NaV1.7 as examples of targeting ion channels with biologics.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | | | | | - K George Chandy
- Molecular Physiology Laboratory, Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Vladimir Yarov-Yarovoy
- Department of Physiology & Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
36
|
Antibodies binding the head domain of P2X4 inhibit channel function and reverse neuropathic pain. Pain 2019; 160:1989-2003. [DOI: 10.1097/j.pain.0000000000001587] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Koch-Nolte F, Eichhoff A, Pinto-Espinoza C, Schwarz N, Schäfer T, Menzel S, Haag F, Demeules M, Gondé H, Adriouch S. Novel biologics targeting the P2X7 ion channel. Curr Opin Pharmacol 2019; 47:110-118. [PMID: 30986625 DOI: 10.1016/j.coph.2019.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 01/05/2023]
Abstract
Targeting the P2X7 ion channel, a danger sensor for extracellular nucleotides, improves outcomes in models of inflammation, cancer, and brain-diseases. Antibodies and nanobodies have been developed that antagonize or potentiate gating of P2X7. Their potential advantages over small-molecule drugs include high specificity, lower off-target effects, and tunable in vivo half-life. Genetic fusion of P2X7-specific biologics to binding modules may enable targeting of specific cell subsets. Besides directly modulating P2X7 function, antibodies can also initiate specific depletion of P2X7-expressing cells. Adeno-associated viral vectors (AAV) can be used to express P2X7-specific antibodies in vivo to achieve long-lasting biological effects. Furthermore, if successfully targeted to P2X7-expressing cells, AAVs may enable modulation of the function of P2X7-expressing immune cells via encoded transgenic RNA or proteins.
Collapse
Affiliation(s)
- Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Anna Eichhoff
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), 76000, Rouen, France
| | | | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Schäfer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mélanie Demeules
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), 76000, Rouen, France
| | - Henri Gondé
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), 76000, Rouen, France
| | - Sahil Adriouch
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular diseases and regenerative THERapies (PANTHER), 76000, Rouen, France.
| |
Collapse
|
38
|
Vijayamahantesh, Vijayalaxmi. Tinkering with targeting nucleotide signaling for control of intracellular Leishmania parasites. Cytokine 2019; 119:129-143. [PMID: 30909149 DOI: 10.1016/j.cyto.2019.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
Abstract
Nucleotides are one of the most primitive extracellular signalling molecules across all phyla and regulate a multitude of responses. The biological effects of extracellular nucleotides/sides are mediated via the specific purinergic receptors present on the cell surface. In mammalian system, adenine nucleotides are the predominant nucleotides found in the extracellular milieu and mediate a constellation of physiological functions. In the context of host-pathogen interaction, extracellular ATP is recognized as a danger signal and potentiates the release of pro-inflammatory mediators from activated immune cells, on the other hand, its breakdown product adenosine exerts potential anti-inflammatory and immunosuppressive actions. Therefore, it is increasingly apparent that the interplay between extracellular ATP/adenosine ratios has a significant role in coordinating the regulation of the immune system in health and diseases. Several pathogens express ectonucleotidases on their surface and exploit the purinergic signalling as one of the mechanisms to modulate the host immune response. Leishmania pathogens are one of the most successful intracellular pathogens which survive within host macrophages and manipulate protective Th1 response into disease promoting Th2 response. In this review, we discuss the regulation of extracellular ATP and adenosine levels, the role of ATP/adenosine counter signalling in regulating the inflammation and immune responses during infection and how Leishmania parasites exploit the purinergic signalling to manipulate host response. We also discuss the challenges and opportunities in targeting purinergic signalling and the future prospects.
Collapse
Affiliation(s)
- Vijayamahantesh
- Department of Biochemistry, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| | - Vijayalaxmi
- Department of Zoology, Karnatak University, Dharwad, Karnataka, India
| |
Collapse
|
39
|
Colley CS, England E, Linley JE, Wilkinson TCI. Screening Strategies for the Discovery of Ion Channel Monoclonal Antibodies. ACTA ACUST UNITED AC 2018; 82:e44. [DOI: 10.1002/cpph.44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Caroline S. Colley
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - Elizabeth England
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - John E. Linley
- Neuroscience, IMED Biotech Unit, AstraZeneca; Cambridge United Kingdom
| | | |
Collapse
|
40
|
ATP in the tumour microenvironment drives expression of nfP2X 7, a key mediator of cancer cell survival. Oncogene 2018; 38:194-208. [PMID: 30087439 PMCID: PMC6328436 DOI: 10.1038/s41388-018-0426-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/29/2022]
Abstract
The ATP-gated receptor P2X7 is expressed in multiple malignant tumours including neuroblastoma, melanoma, prostate, lung and breast. P2X7 has a significant role in mediating diverse cell responses, which upon dysregulation are associated with tumour initiation and development. The rapid, ATP-mediated activation of P2X7 induces a fast-inward cation current in cells. However, prolonged ATP-mediated activation of P2X7 leads to formation of a pore that increases membrane permeability and eventually causes cell death. This presents a potential paradox, as the tumour microenvironment contains extracellular ATP at levels sufficient to activate the P2X7 pore and trigger cell death. However, P2X7 expression is associated with enhanced cancer cell survival, proliferation and metastatic potential. At least one distinct conformational form of P2X7, termed non-pore functional P2X7 (nfP2X7), has been described, which is not able to form a functional pore. We demonstrate for the first time in this study that exposure to a high ATP concentration, equivalent to those measured in the tumour microenvironment, drives nfP2X7 expression and also that nfP2X7 is essential for tumour cell survival. We show that monoclonal antibodies raised against a P2X7 amino acid sequence (200–216), whose conformation is distinct from that of wild-type (WT) P2X7, bind specifically to nfP2X7 expressed on the surface of tumour cells. We also show that nfP2X7 is broadly expressed in patient-derived tumour sections from a wide range of cancers. Therefore, antibodies raised against E200 provide tools that can differentiate between forms of the P2X7 receptor that have a key role in cancer.
Collapse
|
41
|
Amores-Iniesta J, Barberà-Cremades M, Martínez CM, Pons JA, Revilla-Nuin B, Martínez-Alarcón L, Di Virgilio F, Parrilla P, Baroja-Mazo A, Pelegrín P. Extracellular ATP Activates the NLRP3 Inflammasome and Is an Early Danger Signal of Skin Allograft Rejection. Cell Rep 2018; 21:3414-3426. [PMID: 29262323 PMCID: PMC5746605 DOI: 10.1016/j.celrep.2017.11.079] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 09/20/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022] Open
Abstract
Immune cells are equipped with a number of receptors that recognize sterile injury and pathogens. We find that host immune cells release ATP as an inflammatory signal in response to allogeneic transplantation. ATP then acts via a feedback mechanism on the P2X7 channel to activate the NLRP3 inflammasome and subsequently process and release interleukin (IL)-18. This process is a necessary stage in the deleterious Th1 response against allotransplantation via interferon-γ production. Lack of IL-18 resulted in a decrease in graft-infiltrating CD8 cells but an increase in regulatory T cells. In human liver transplant patients undergoing progressive immunosuppressive drug withdrawal, we found that patients experiencing acute rejection had higher levels of the P2X7 receptor in circulating inflammatory monocytes compared to tolerant patients. These data suggest that the pharmacological inhibition of the P2X7 receptor or the NLRP3 inflammasome will aid in inducing transplant tolerance without complete immunoparalysis.
Collapse
Affiliation(s)
- Joaquín Amores-Iniesta
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Maria Barberà-Cremades
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Carlos M Martínez
- Experimental Pathology Unit, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - José A Pons
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Beatriz Revilla-Nuin
- Genomic Unit, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Laura Martínez-Alarcón
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Pascual Parrilla
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Alberto Baroja-Mazo
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Pablo Pelegrín
- Experimental Surgery Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain.
| |
Collapse
|
42
|
Stortelers C, Pinto-Espinoza C, Van Hoorick D, Koch-Nolte F. Modulating ion channel function with antibodies and nanobodies. Curr Opin Immunol 2018; 52:18-26. [DOI: 10.1016/j.coi.2018.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/21/2022]
|
43
|
Di Virgilio F, Schmalzing G, Markwardt F. The Elusive P2X7 Macropore. Trends Cell Biol 2018; 28:392-404. [PMID: 29439897 DOI: 10.1016/j.tcb.2018.01.005] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 12/27/2022]
Abstract
ATP, which is released under pathological conditions and is considered a damage-associated molecular pattern (DAMP), activates P2X7 receptors (P2X7Rs), trimeric plasma membrane ion channels selective for small cations. P2X7Rs are partners in NOD-like receptor containing a pyrin (NLRP3) inflammasome activation and promoters of tumor cell growth. P2X7R overstimulation triggers the ATP-dependent opening of a nonselective plasma membrane pore, known as a 'macropore', which allows fluxes of large hydrophilic molecules. The pathophysiological functions of P2X7R are thought to be dependent on activation of this conductance pathway, yet its molecular identity is unknown. Recent reports show that P2X7R permeability to organic solutes is an early and intrinsic property of the channel itself. A better understanding of P2X7R-dependent changes in plasma membrane permeability will allow a rationale development of novel anti-inflammatory and anticancer drugs.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Günther Schmalzing
- Department of Pharmacology and Toxicology, University of Aachen, Aachen, Germany
| | - Fritz Markwardt
- Institute for Physiology, Martin Luther University, Halle/Saale, Germany
| |
Collapse
|
44
|
Ou A, Gu BJ, Wiley JS. The scavenger activity of the human P2X7 receptor differs from P2X7 pore function by insensitivity to antagonists, genetic variation and sodium concentration: Relevance to inflammatory brain diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1051-1059. [PMID: 29329985 DOI: 10.1016/j.bbadis.2018.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 01/08/2023]
Abstract
Activation of P2X7 receptors is widely recognised to initiate proinflammatory responses. However P2X7 also has a dual function as a scavenger receptor which is active in the absence of ATP and plasma proteins and may be important in central nervous system (CNS) diseases. Here, we investigated both P2X7 pore formation and its phagocytic function in fresh human monocytes (as a model of microglia) by measuring ATP-induced ethidium dye uptake and fluorescent bead uptake respectively. This was studied in monocytes expressing various polymorphic variants as well as in the presence of different P2X7 antagonists and ionic media. P2X7-mediated phagocytosis was found to account for about half of Latrunculin (or Cytochalasin D)-sensitive bead engulfment by fresh human monocytes. Monocytes harbouring P2X7 Ala348Thr or Glu496Ala polymorphic variants showed increase or loss of ethidium uptake respectively, but these changes in pore formation did not always correspond to the changes in phagocytosis of YG beads. Unlike pore function, P2X7-mediated phagocytosis was not affected by three potent selective P2X7 antagonists and remained identical in Na+ and K+ media. Taken together, our results show that P2X7 is a scavenger receptor with important function in the CNS but its phagocytic function has features distinct from its pore function. Both P2X7 pore formation and P2X7-mediated phagocytosis should be considered in the design of new P2X7 antagonists for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Amber Ou
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ben J Gu
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - James S Wiley
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
45
|
Dreisig K, Kristensen NP, Dommer MW, Jørgensen NR, Kornum BR. N-terminal tagging of human P2X7 receptor disturbs calcium influx and dye uptake. Purinergic Signal 2018; 14:83-90. [PMID: 29290027 DOI: 10.1007/s11302-017-9598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 12/13/2017] [Indexed: 11/26/2022] Open
Abstract
The P2X7 receptor is a frequently studied member of the purinergic receptor family signalling via channel opening and membrane pore formation. Fluorescent imaging is an important molecular method for studying cellular receptor expression and localization. Fusion of receptors to fluorescent proteins might cause major functional changes and requires careful functional evaluation such as has been done for the rat P2X7 receptor. This study examines fusion constructs of the human P2X7 receptor. We assessed surface expression, channel opening with calcium influx, and pore formation using YO-PRO-1 dye uptake in response to BzATP stimulation in transfected cells. We found that tagging at the N-terminal of the human P2X7 receptor with the enhanced green fluorescent protein (eGFP) disturbed channel opening and pore formation despite intact surface expression. A triple hemagglutinin (3HA) fused to the N-terminal also disrupted pore formation but not channel opening showing that even a small tag alters the normal function of the receptor. Together, this suggests that in contrast to what has been observed for the rat P2X7 receptor, the human P2X7 receptor contains N-terminal motifs important for signalling that prevent the construction of a functionally active fusion protein.
Collapse
Affiliation(s)
- Karin Dreisig
- Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
| | | | | | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
- OPEN, Odense Patient Data Explorative Network Odense University Hospital/Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Birgitte Rahbek Kornum
- Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark.
- Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
46
|
Danquah W, Meyer-Schwesinger C, Rissiek B, Pinto C, Serracant-Prat A, Amadi M, Iacenda D, Knop JH, Hammel A, Bergmann P, Schwarz N, Assunção J, Rotthier W, Haag F, Tolosa E, Bannas P, Boué-Grabot E, Magnus T, Laeremans T, Stortelers C, Koch-Nolte F. Nanobodies that block gating of the P2X7 ion channel ameliorate inflammation. Sci Transl Med 2017; 8:366ra162. [PMID: 27881823 DOI: 10.1126/scitranslmed.aaf8463] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 04/11/2016] [Accepted: 10/27/2016] [Indexed: 12/17/2022]
Abstract
Ion channels are desirable therapeutic targets, yet ion channel-directed drugs with high selectivity and few side effects are still needed. Unlike small-molecule inhibitors, antibodies are highly selective for target antigens but mostly fail to antagonize ion channel functions. Nanobodies-small, single-domain antibody fragments-may overcome these problems. P2X7 is a ligand-gated ion channel that, upon sensing adenosine 5'-triphosphate released by damaged cells, initiates a proinflammatory signaling cascade, including release of cytokines, such as interleukin-1β (IL-1β). To further explore its function, we generated and characterized nanobodies against mouse P2X7 that effectively blocked (13A7) or potentiated (14D5) gating of the channel. Systemic injection of nanobody 13A7 in mice blocked P2X7 on T cells and macrophages in vivo and ameliorated experimental glomerulonephritis and allergic contact dermatitis. We also generated nanobody Dano1, which specifically inhibited human P2X7. In endotoxin-treated human blood, Dano1 was 1000 times more potent in preventing IL-1β release than small-molecule P2X7 antagonists currently in clinical development. Our results show that nanobody technology can generate potent, specific therapeutics against ion channels, confirm P2X7 as a therapeutic target for inflammatory disorders, and characterize a potent new drug candidate that targets P2X7.
Collapse
Affiliation(s)
- Welbeck Danquah
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Catherine Meyer-Schwesinger
- Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Björn Rissiek
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Carolina Pinto
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Arnau Serracant-Prat
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Miriam Amadi
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Domenica Iacenda
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Jan-Hendrik Knop
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Anna Hammel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Philine Bergmann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Université de Bordeaux, Institut des Maladies Neurodégénératives, CNRS UMR 5293, Bordeaux 33076, France
| | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Joana Assunção
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | - Wendy Rotthier
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Peter Bannas
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eric Boué-Grabot
- Université de Bordeaux, Institut des Maladies Neurodégénératives, CNRS UMR 5293, Bordeaux 33076, France
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Toon Laeremans
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | | | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| |
Collapse
|
47
|
Discovery of functional monoclonal antibodies targeting G-protein-coupled receptors and ion channels. Biochem Soc Trans 2017; 44:831-7. [PMID: 27284048 DOI: 10.1042/bst20160028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Indexed: 11/17/2022]
Abstract
The development of recombinant antibody therapeutics is a significant area of growth in the pharmaceutical industry with almost 50 approved monoclonal antibodies on the market in the US and Europe. Despite this growth, however, certain classes of important molecular targets have remained intractable to therapeutic antibodies due to complexity of the target molecules. These complex target molecules include G-protein-coupled receptors and ion channels which represent a large potential target class for therapeutic intervention with monoclonal antibodies. Although these targets have typically been addressed by small molecule approaches, the exquisite specificity of antibodies provides a significant opportunity to provide selective modulation of these target proteins. Given this opportunity, substantial effort has been applied to address the technical challenges of targeting these complex membrane proteins with monoclonal antibodies. In this review recent progress made in the strategies for discovery of functional monoclonal antibodies for these challenging membrane protein targets is addressed.
Collapse
|
48
|
Sadovnick AD, Gu BJ, Traboulsee AL, Bernales CQ, Encarnacion M, Yee IM, Criscuoli MG, Huang X, Ou A, Milligan CJ, Petrou S, Wiley JS, Vilariño-Güell C. Purinergic receptors P2RX4 and P2RX7 in familial multiple sclerosis. Hum Mutat 2017; 38:736-744. [PMID: 28326637 DOI: 10.1002/humu.23218] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/08/2017] [Accepted: 03/16/2017] [Indexed: 12/11/2022]
Abstract
Genetic variants in the purinergic receptors P2RX4 and P2RX7 have been shown to affect susceptibility to multiple sclerosis (MS). In this study, we set out to evaluate whether rare coding variants of major effect could also be identified in these purinergic receptors. Sequencing analysis of P2RX4 and P2RX7 in 193 MS patients and 100 controls led to the identification of a rare three variant haplotype (P2RX7 rs140915863:C>T [p.T205M], P2RX7 rs201921967:A>G [p.N361S], and P2RX4 rs765866317:G>A [p.G135S]) segregating with disease in a multi-incident family with six family members diagnosed with MS (logarithm of odds = 3.07). Functional analysis of this haplotype in HEK293 cells revealed impaired P2X7 surface expression (P < 0.01), resulting in over 95% inhibition of adenosine triphosphate (ATP)-induced pore function (P < 0.001) and a marked reduction in phagocytic ability (P < 0.05). In addition, transfected cells showed 40% increased peak ATP-induced inward current (P < 0.01), and a greater Ca2+ response to the P2X4 135S variant compared with wild type (P < 0.0001). Our study nominates rare genetic variants in P2RX4 and P2RX7 as major genetic contributors to disease, further supporting a role for these purinergic receptors in MS and the disruption of transmembrane cation channels leading to impairment of phagocytosis as the pathological mechanisms of disease.
Collapse
Affiliation(s)
- A Dessa Sadovnick
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Division of Neurology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Ben J Gu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony L Traboulsee
- Division of Neurology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Cecily Q Bernales
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Mary Encarnacion
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Irene M Yee
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Maria G Criscuoli
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Xin Huang
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Amber Ou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Carol J Milligan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - James S Wiley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | | |
Collapse
|
49
|
Localization of the gate and selectivity filter of the full-length P2X7 receptor. Proc Natl Acad Sci U S A 2017; 114:E2156-E2165. [PMID: 28235784 DOI: 10.1073/pnas.1610414114] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The P2X7 receptor (P2X7R) belongs to the P2X family of ATP-gated cation channels. P2X7Rs are expressed in epithelial cells, leukocytes, and microglia, and they play important roles in immunological and inflammatory processes. P2X7Rs are obligate homotrimers, with each subunit having two transmembrane helices, TM1 and TM2. Structural and functional data regarding the P2X2 and P2X4 receptors indicate that the central trihelical TM2 bundle forms the intrinsic transmembrane channel of P2X receptors. Here, we studied the accessibility of single cysteines substituted along the pre-TM2 and TM2 helix (residues 327-357) of the P2X7R using as readouts (i) the covalent maleimide fluorescence accessibility of the surface-bound P2X7R and (ii) covalent modulation of macroscopic and single-channel currents using extracellularly and intracellularly applied methanethiosulfonate (MTS) reagents. We found that the channel opening extends from the pre-TM2 region through the outer half of the trihelical TM2 channel. Covalently adducted MTS ethylammonium+ (MTSEA+) strongly increased the probability that the channel was open by delaying channel closing of seven of eight responsive human P2X7R (hP2X7R) mutants. Structural modeling, as supported by experimental probing, suggested that resulting intraluminal hydrogen bonding interactions stabilize the open-channel state. The additional decrease in single-channel conductance by MTSEA+ in five of seven positions identified Y336, S339, L341C, Y343, and G345 as the narrowest part of the channel lumen. The gate and ion-selectivity filter of the P2X7R could be colocalized at and around residue S342. None of our results provided any evidence for dilation of the hP2X7R channel on sustained stimulation with ATP4.
Collapse
|
50
|
Abstract
The P2X7 receptor is a trimeric ion channel gated by extracellular adenosine 5'-triphosphate. The receptor is present on an increasing number of different cells types including stem, blood, glial, neural, ocular, bone, dental, exocrine, endothelial, muscle, renal and skin cells. The P2X7 receptor induces various downstream events in a cell-specific manner, including inflammatory molecule release, cell proliferation and death, metabolic events, and phagocytosis. As such this receptor plays important roles in heath and disease. Increasing knowledge about the P2X7 receptor has been gained from studies of, but not limited to, protein chemistry including cloning, site-directed mutagenesis, crystal structures and atomic modeling, as well as from studies of primary tissues and transgenic mice. This chapter focuses on the P2X7 receptor itself. This includes the P2RX7 gene and its products including splice and polymorphic variants. This chapter also reviews modulators of P2X7 receptor activation and inhibition, as well as the transcriptional regulation of the P2RX7 gene via its promoter and enhancer regions, and by microRNA and long-coding RNA. Furthermore, this chapter discusses the post-translational modification of the P2X7 receptor by N-linked glycosylation, adenosine 5'-diphosphate ribosylation and palmitoylation. Finally, this chapter reviews interaction partners of the P2X7 receptor, and its cellular localisation and trafficking within cells.
Collapse
Affiliation(s)
- Ronald Sluyter
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia. .,Centre for Medical and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia. .,Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.
| |
Collapse
|