1
|
McGown A, Renault N, Barczyk A, Nafie J, Barluzzi L, Guest D, Tizzard GJ, Coles SJ, Leach D, von Emloh D, Sutton L, Bailey K, Edmunds L, Greenland BW, Millet R, Spencer J, Dezitter X. Characterization of the Active Enantiomer and Mapping of the Stereospecific Intermolecular Pattern of a Reference P2X7 Allosteric Antagonist. ACS Pharmacol Transl Sci 2025; 8:446-459. [PMID: 39974629 PMCID: PMC11833722 DOI: 10.1021/acsptsci.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/21/2025]
Abstract
The P2X purinergic receptor 7 (P2X7) has an essential role in inflammation, innate immunity, tumor progression, neurodegenerative diseases, and several other diseases, leading subsequently to the development of P2X7 modulators. AZ11645373 is a frequently studied P2X7 antagonist tool compound but always used as a racemic mixture. Racemic AZ11645373 can be separated into its respective enantiomers by chiral chromatography, albeit in small batches, and these were stereochemically intact over two years later, by chiral high-performance liquid chromatography (HPLC) analysis. On a higher scale, significant decomposition is observed during purification. One of the enantiomers was crystallized as a palladium complex, and its (R)-configuration was determined by single-crystal X-ray diffraction, further confirmed, in solution, by vibrational circular dichroism. Biological studies demonstrated that both (S)- and (R)-forms were able to fully inhibit human P2X7, but (R)-AZ11645373 was more potent, with an IC50 of 32.9 nM. Contrary to its effect on human P2X7, (S)-AZ11645373 was ineffective on mouse P2X7, while the (R)-AZ11645373 enantiomer was a full antagonist. These results demonstrated that the antagonistic effects of racemic AZ11645373 are mainly due to its (R)-enantiomer. Site-directed mutagenesis and molecular dynamics simulations indicated that the (R)-enantiomer may form specific interactions with Phe95 and the antagonists bound to other P2X7 monomers. Phe95 is situated in the allosteric binding site at the edge of the upper vestibule and appears to be the pivotal molecular gateway between AZ11645373 allosteric binding and locking of the closed state of the P2X7 channel. All together, these structure-function relationships should be helpful for drug design of P2X7 modulators.
Collapse
Affiliation(s)
- Andrew McGown
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
- Sussex
Drug Discovery Centre, Department of Chemistry, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Nicolas Renault
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - Amélie Barczyk
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - Jordan Nafie
- Biotools,
Inc., 17546 Beeline Highway, Jupiter, Florida 33458, United States
| | - Luciano Barluzzi
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Daniel Guest
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Graham J. Tizzard
- National
Crystallography Service, School of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Simon J. Coles
- National
Crystallography Service, School of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - David Leach
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Daniel von Emloh
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Léa Sutton
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Kiera Bailey
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Lewis Edmunds
- Reach Separations
Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham NG1 1GF, U.K.
| | - Barnaby W. Greenland
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Régis Millet
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| | - John Spencer
- Department
of Chemistry, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
- Sussex
Drug Discovery Centre, Department of Chemistry, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Xavier Dezitter
- Univ.
Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for
Translational Research in Inflammation, Lille F-59000, France
| |
Collapse
|
2
|
Mutafova-Yambolieva VN. Mechanosensitive release of ATP in the urinary bladder mucosa. Purinergic Signal 2024:10.1007/s11302-024-10063-6. [PMID: 39541058 DOI: 10.1007/s11302-024-10063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The urinary bladder mucosa (urothelium and suburothelium/lamina propria) functions as a barrier between the content of the urine and the underlying bladder tissue. The bladder mucosa is also a mechanosensitive tissue that releases signaling molecules that affect functions of cells in the bladder wall interconnecting the mucosa with the detrusor muscle and the CNS. Adenosine 5'-triphosphate (ATP) is a primary mechanotransduction signal that is released from cells in the bladder mucosa in response to bladder wall distention and activates cell membrane-localized P2X and P2Y purine receptors on urothelial cells, sensory and efferent neurons, interstitial cells, and detrusor smooth muscle cells. The amounts of ATP at active receptor sites depend significantly on the amounts of extracellularly released ATP. Spontaneous and distention-induced release of ATP appear to be under differential control. This review is focused on mechanisms underlying urothelial release of ATP in response to mechanical stimulation. First, we present a brief overview of studies that report mechanosensitive ATP release in bladder cells or tissues. Then, we discuss experimental evidence for mechanosensitive release of urothelial ATP by vesicular and non-vesicular mechanisms and roles of the stretch-activated channels PIEZO channels, transient receptor potential vanilloid type 4, and pannexin 1. This is followed by brief discussion of possible involvement of calcium homeostasis modulator 1, acid-sensing channels, and connexins in the release of urothelial ATP. We conclude with brief discussion of limitations of current research and of needs for further studies to increase our understanding of mechanotransduction in the bladder wall and of purinergic regulation of bladder function.
Collapse
|
3
|
Markwardt F, Schön EC, Raycheva M, Malisetty A, Hawro Yakoob S, Berthold M, Schmalzing G. Two serial filters control P2X7 cation selectivity, Ser342 in the central pore and lateral acidic residues at the cytoplasmic interface. PNAS NEXUS 2024; 3:pgae349. [PMID: 39262850 PMCID: PMC11388005 DOI: 10.1093/pnasnexus/pgae349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024]
Abstract
The human P2X7 receptor (hP2X7R) is a homotrimeric cell surface receptor gated by extracellular ATP4- with two transmembrane helices per subunit, TM1 and TM2. A ring of three S342 residues, one from each pore-forming TM2 helix, located halfway across the membrane bilayer, functions to close and open the gate in the apo and ATP4--bound open states, respectively. The hP2X7R is selective for small inorganic cations, but can also conduct larger organic cations such as Tris+. Here, we show by voltage-clamp electrophysiology in Xenopus laevis oocytes that mutation of S342 residues to positively charged lysines decreases the selectivity for Na+ over Tris+, but maintains cation selectivity. Deep in the membrane, laterally below the S342 ring are nine acidic residues arranged as an isosceles triangle consisting of residues E14, D352, and D356 on each side, which do not move significantly during gating. When the E14K mutation is combined with lysine substitutions of D352 and/or D356, cation selectivity is lost and permeation of the small anion Cl- is allowed. Lysine substitutions of S342 together with D352 or E14 plus D356 in the acidic triangle convert the hP2X7R mutant to a fully Cl--selective ATP4--gated receptor. We conclude that the ion selectivity of wild-type hP2X7R is determined by two sequential filters in one single pathway: (i) a primary size filter, S342, in the membrane center and (ii) three cation filters lateral to the channel axis, one per subunit interface, consisting of a total of nine acidic residues at the cytoplasmic interface.
Collapse
Affiliation(s)
- Fritz Markwardt
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University, Magdeburger Straße 6, D-06097 Halle/Saale, Germany
| | - Eike Christian Schön
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University, Magdeburger Straße 6, D-06097 Halle/Saale, Germany
| | - Mihaela Raycheva
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074 Aachen, Germany
| | - Aparna Malisetty
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074 Aachen, Germany
| | - Sanaria Hawro Yakoob
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074 Aachen, Germany
| | - Malte Berthold
- Julius-Bernstein-Institute of Physiology, Martin-Luther-University, Magdeburger Straße 6, D-06097 Halle/Saale, Germany
| | - Günther Schmalzing
- Institute of Clinical Pharmacology, RWTH Aachen University, Wendlingweg 2, D-52074 Aachen, Germany
| |
Collapse
|
4
|
Liu X, Li Y, Huang L, Kuang Y, Wu X, Ma X, Zhao B, Lan J. Unlocking the therapeutic potential of P2X7 receptor: a comprehensive review of its role in neurodegenerative disorders. Front Pharmacol 2024; 15:1450704. [PMID: 39139642 PMCID: PMC11319138 DOI: 10.3389/fphar.2024.1450704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The P2X7 receptor (P2X7R), an ATP-gated ion channel, has emerged as a crucial player in neuroinflammation and a promising therapeutic target for neurodegenerative disorders. This review explores the current understanding of P2X7R's structure, activation, and physiological roles, focusing on its expression and function in microglial cells. The article examines the receptor's involvement in calcium signaling, microglial activation, and polarization, as well as its role in the pathogenesis of Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis. The review highlights the complex nature of P2X7R signaling, discussing its potential neuroprotective and neurotoxic effects depending on the disease stage and context. It also addresses the development of P2X7R antagonists and their progress in clinical trials, identifying key research gaps and future perspectives for P2X7R-targeted therapy development. By providing a comprehensive overview of the current state of knowledge and future directions, this review serves as a valuable resource for researchers and clinicians interested in exploring the therapeutic potential of targeting P2X7R for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoming Liu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiwen Li
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Liting Huang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yingyan Kuang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiaoxiong Wu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiangqiong Ma
- Henan Hospital of Integrated Chinese and Western Medicine, Zhengzhou, China
| | - Beibei Zhao
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jiao Lan
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
5
|
Huffer K, Tan XF, Fernández-Mariño AI, Dhingra S, Swartz KJ. Dilation of ion selectivity filters in cation channels. Trends Biochem Sci 2024; 49:417-430. [PMID: 38514273 PMCID: PMC11069442 DOI: 10.1016/j.tibs.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Ion channels establish the voltage gradient across cellular membranes by providing aqueous pathways for ions to selectively diffuse down their concentration gradients. The selectivity of any given channel for its favored ions has conventionally been viewed as a stable property, and in many cation channels, it is determined by an ion-selectivity filter within the external end of the ion-permeation pathway. In several instances, including voltage-activated K+ (Kv) channels, ATP-activated P2X receptor channels, and transient receptor potential (TRP) channels, the ion-permeation pathways have been proposed to dilate in response to persistent activation, dynamically altering ion permeation. Here, we discuss evidence for dynamic ion selectivity, examples where ion selectivity filters exhibit structural plasticity, and opportunities to fill gaps in our current understanding.
Collapse
Affiliation(s)
- Kate Huffer
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiao-Feng Tan
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ana I Fernández-Mariño
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Surbhi Dhingra
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Immanuel CN, Teng B, Dong BE, Gordon EM, Luellen C, Lopez B, Harding J, Cormier SA, Fitzpatrick EA, Schwingshackl A, Waters CM. Two-pore potassium channel TREK-1 (K2P2.1) regulates NLRP3 inflammasome activity in macrophages. Am J Physiol Lung Cell Mol Physiol 2024; 326:L367-L376. [PMID: 38252657 PMCID: PMC11281793 DOI: 10.1152/ajplung.00313.2023] [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: 10/05/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Because of the importance of potassium efflux in inflammasome activation, we investigated the role of the two-pore potassium (K2P) channel TREK-1 in macrophage inflammasome activity. Using primary alveolar macrophages (AMs) and bone marrow-derived macrophages (BMDMs) from wild-type (wt) and TREK-1-/- mice, we measured responses to inflammasome priming [using lipopolysaccharide (LPS)] and activation (LPS + ATP). We measured IL-1β, caspase-1, and NLRP3 via ELISA and Western blot. A membrane-permeable potassium indicator was used to measure potassium efflux during ATP exposure, and a fluorescence-based assay was used to assess changes in membrane potential. Inflammasome activation induced by LPS + ATP increased IL-1β secretion in wt AMs, whereas activation was significantly reduced in TREK-1-/- AMs. Priming of BMDMs using LPS was not affected by either genetic deficiency or pharmacological inhibition of TREK-1 with Spadin. Cleavage of caspase-1 following LPS + ATP treatment was significantly reduced in TREK-1-/- BMDMs. The intracellular potassium concentration in LPS-primed wt BMDMs was significantly lower compared with TREK-1-/- BMDMs or wt BMDMs treated with Spadin. Conversely, activation of TREK-1 with BL1249 caused a decrease in intracellular potassium in wt BMDMs. Treatment of LPS-primed BMDMs with ATP caused a rapid reduction in intracellular potassium levels, with the largest change observed in TREK-1-/- BMDMs. Intracellular K+ changes were associated with changes in the plasma membrane potential (Em), as evidenced by a more depolarized Em in TREK-1-/- BMDMs compared with wt, and Em hyperpolarization upon TREK-1 channel opening with BL1249. These results suggest that TREK-1 is an important regulator of NLRP3 inflammasome activation in macrophages.NEW & NOTEWORTHY Because of the importance of potassium efflux in inflammasome activation, we investigated the role of the two-pore potassium (K2P) channel TREK-1 in macrophage inflammasome activity. Using primary alveolar macrophages and bone marrow-derived macrophages from wild-type and TREK-1-/- mice, we measured responses to inflammasome priming (using LPS) and activation (LPS + ATP). Our results suggest that TREK-1 is an important regulator of NLRP3 inflammasome activation in macrophages.
Collapse
Grants
- HL131526 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Le Bonheur Children's Hospital
- 20TPA35490010 American Heart Association (AHA)
- R01 HL131526 NHLBI NIH HHS
- HL151419 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- IA-678511 American Lung Association (ALA)
- R01 HL146821 NHLBI NIH HHS
- HL146821 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL123540 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL151419 NHLBI NIH HHS
- R01 HL123540 NHLBI NIH HHS
- HHS | NIH | National Heart, Lung, and Blood Institute (NHBLI)
Collapse
Affiliation(s)
- Camille N Immanuel
- Division of Pediatric Critical Care, Department of Pediatrics, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Bin Teng
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Brittany E Dong
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Elizabeth M Gordon
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Charlean Luellen
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Benjamin Lopez
- Department of Pediatrics, University of California, Los Angeles, California, United States
| | - Jeffrey Harding
- Department of Biological Sciences, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Stephania A Cormier
- Department of Biological Sciences, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Elizabeth A Fitzpatrick
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Andreas Schwingshackl
- Department of Pediatrics, University of California, Los Angeles, California, United States
| | - Christopher M Waters
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
7
|
Zheng H, Liu Q, Zhou S, Luo H, Zhang W. Role and therapeutic targets of P2X7 receptors in neurodegenerative diseases. Front Immunol 2024; 15:1345625. [PMID: 38370420 PMCID: PMC10869479 DOI: 10.3389/fimmu.2024.1345625] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
The P2X7 receptor (P2X7R), a non-selective cation channel modulated by adenosine triphosphate (ATP), localizes to microglia, astrocytes, oligodendrocytes, and neurons in the central nervous system, with the most incredible abundance in microglia. P2X7R partake in various signaling pathways, engaging in the immune response, the release of neurotransmitters, oxidative stress, cell division, and programmed cell death. When neurodegenerative diseases result in neuronal apoptosis and necrosis, ATP activates the P2X7R. This activation induces the release of biologically active molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen species, and excitotoxic glutamate/ATP. Subsequently, this leads to neuroinflammation, which exacerbates neuronal involvement. The P2X7R is essential in the development of neurodegenerative diseases. This implies that it has potential as a drug target and could be treated using P2X7R antagonists that are able to cross the blood-brain barrier. This review will comprehensively and objectively discuss recent research breakthroughs on P2X7R genes, their structural features, functional properties, signaling pathways, and their roles in neurodegenerative diseases and possible therapies.
Collapse
Affiliation(s)
- Huiyong Zheng
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiang Liu
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Siwei Zhou
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wenjun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Tewari M, Michalski S, Egan TM. Modulation of Microglial Function by ATP-Gated P2X7 Receptors: Studies in Rat, Mice and Human. Cells 2024; 13:161. [PMID: 38247852 PMCID: PMC10814008 DOI: 10.3390/cells13020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
P2X receptors are a family of seven ATP-gated ion channels that trigger physiological and pathophysiological responses in a variety of cells. Five of the family members are sensitive to low concentrations of extracellular ATP, while the P2X6 receptor has an unknown affinity. The last subtype, the P2X7 receptor, is unique in requiring millimolar concentrations to fully activate in humans. This low sensitivity imparts the agonist with the ability to act as a damage-associated molecular pattern that triggers the innate immune response in response to the elevated levels of extracellular ATP that accompany inflammation and tissue damage. In this review, we focus on microglia because they are the primary immune cells of the central nervous system, and they activate in response to ATP or its synthetic analog, BzATP. We start by introducing purinergic receptors and then briefly consider the roles that microglia play in neurodevelopment and disease by referencing both original works and relevant reviews. Next, we move to the role of extracellular ATP and P2X receptors in initiating and/or modulating innate immunity in the central nervous system. While most of the data that we review involve work on mice and rats, we highlight human studies of P2X7R whenever possible.
Collapse
|
9
|
Augusto-Oliveira M, Tremblay MÈ, Verkhratsky A. Receptors on Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:83-121. [PMID: 39207688 DOI: 10.1007/978-3-031-55529-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglial cells are the most receptive cells in the central nervous system (CNS), expressing several classes of receptors reflecting their immune heritage and newly acquired neural specialisation. Microglia possess, depending on the particular context, receptors to neurotransmitters and neuromodulators as well as immunocompetent receptors. This rich complement allows microglial cells to monitor the functional status of the nervous system, contribute actively to the regulation of neural activity and plasticity and homeostasis, and guard against pathogens as well as other challenges to the CNS's integrity and function.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
10
|
Yin Y, Wei L, Caseley EA, Lopez‐Charcas O, Wei Y, Li D, Muench SP, Roger S, Wang L, Jiang L. Leveraging the ATP-P2X7 receptor signalling axis to alleviate traumatic CNS damage and related complications. Med Res Rev 2023; 43:1346-1373. [PMID: 36924449 PMCID: PMC10947395 DOI: 10.1002/med.21952] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 11/11/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
The P2X7 receptor is an exceptional member of the P2X purinergic receptor family, with its activation requiring high concentrations of extracellular adenosine 5'-triphosphate (ATP) that are often associated with tissue damage and inflammation. In the central nervous system (CNS), it is highly expressed in glial cells, particularly in microglia. In this review, we discuss the role and mechanisms of the P2X7 receptor in mediating neuroinflammation and other pathogenic events in a variety of traumatic CNS damage conditions, which lead to loss of neurological and cognitive functions. We raise the perspective on the steady progress in developing CNS-penetrant P2X7 receptor-specific antagonists that leverage the ATP-P2X7 receptor signaling axis as a potential therapeutic strategy to alleviate traumatic CNS damage and related complications.
Collapse
Affiliation(s)
- Yaling Yin
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Linyu Wei
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Emily A. Caseley
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| | - Osbaldo Lopez‐Charcas
- EA4245, Transplantation, Immunology and Inflammation, Faculty of MedicineUniversity of ToursToursFrance
| | - Yingjuan Wei
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Dongliang Li
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
- Sanquan College of Xinxiang Medical UniversityXinxiangChina
| | - Steve P. Muench
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| | - Sebastian Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of MedicineUniversity of ToursToursFrance
| | - Lu Wang
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Lin‐Hua Jiang
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
11
|
Cevoli F, Arnould B, Peralta FA, Grutter T. Untangling Macropore Formation and Current Facilitation in P2X7. Int J Mol Sci 2023; 24:10896. [PMID: 37446075 DOI: 10.3390/ijms241310896] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Macropore formation and current facilitation are intriguing phenomena associated with ATP-gated P2X7 receptors (P2X7). Macropores are large pores formed in the cell membrane that allow the passage of large molecules. The precise mechanisms underlying macropore formation remain poorly understood, but recent evidence suggests two alternative pathways: a direct entry through the P2X7 pore itself, and an indirect pathway triggered by P2X7 activation involving additional proteins, such as TMEM16F channel/scramblase. On the other hand, current facilitation refers to the progressive increase in current amplitude and activation kinetics observed with prolonged or repetitive exposure to ATP. Various mechanisms, including the activation of chloride channels and intrinsic properties of P2X7, have been proposed to explain this phenomenon. In this comprehensive review, we present an in-depth overview of P2X7 current facilitation and macropore formation, highlighting new findings and proposing mechanistic models that may offer fresh insights into these untangled processes.
Collapse
Affiliation(s)
- Federico Cevoli
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Benoit Arnould
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Francisco Andrés Peralta
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- Instituto de Neurociencias, CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - Thomas Grutter
- Équipe de Chimie et Neurobiologie Moléculaire, Laboratoire de Conception et Application de Molécules Bioactives (CAMB) UMR 7199, Centre National de la Recherche Scientifique, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
- University of Strasbourg Institute for Advanced Studies (USIAS), 67000 Strasbourg, France
| |
Collapse
|
12
|
Aresta Branco MSL, Gutierrez Cruz A, Peri LE, Mutafova-Yambolieva VN. The Pannexin 1 Channel and the P2X7 Receptor Are in Complex Interplay to Regulate the Release of Soluble Ectonucleotidases in the Murine Bladder Lamina Propria. Int J Mol Sci 2023; 24:9964. [PMID: 37373111 PMCID: PMC10298213 DOI: 10.3390/ijms24129964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The bladder urothelium releases ATP into the lamina propria (LP) during filling, which can activate P2X receptors on afferent neurons and trigger the micturition reflex. Effective ATP concentrations are largely dependent on metabolism by membrane-bound and soluble ectonucleotidases (s-ENTDs), and the latter are released in the LP in a mechanosensitive manner. Pannexin 1 (PANX1) channel and P2X7 receptor (P2X7R) participate in urothelial ATP release and are physically and functionally coupled, hence we investigated whether they modulate s-ENTDs release. Using ultrasensitive HPLC-FLD, we evaluated the degradation of 1,N6-etheno-ATP (eATP, substrate) to eADP, eAMP, and e-adenosine (e-ADO) in extraluminal solutions that were in contact with the LP of mouse detrusor-free bladders during filling prior to substrate addition, as an indirect measure of s-ENDTS release. Deletion of Panx1 increased the distention-induced, but not the spontaneous, release of s-ENTDs, whereas activation of P2X7R by BzATP or high concentration of ATP in WT bladders increased both. In Panx1-/- bladders or WT bladders treated with the PANX1 inhibitory peptide 10Panx, however, BzATP had no effect on s-ENTDS release, suggesting that P2X7R activity depends on PANX1 channel opening. We concluded, therefore, that P2X7R and PANX1 are in complex interaction to regulate s-ENTDs release and maintain suitable ATP concentrations in the LP. Thus, while stretch-activated PANX1 hinders s-ENTDS release possibly to preserve effective ATP concentration at the end of bladder filling, P2X7R activation, presumably in cystitis, would facilitate s-ENTDs-mediated ATP degradation to counteract excessive bladder excitability.
Collapse
Affiliation(s)
| | | | | | - Violeta N. Mutafova-Yambolieva
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV 89557, USA; (M.S.L.A.B.); (A.G.C.); (L.E.P.)
| |
Collapse
|
13
|
Tang Y, Rubini P, Yin HY, Illes P. Acupuncture for Counteracting P2X4 and P2X7 Receptor Involvement in Neuroinflammation. PURINERGIC SIGNALING IN NEURODEVELOPMENT, NEUROINFLAMMATION AND NEURODEGENERATION 2023:359-374. [DOI: 10.1007/978-3-031-26945-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Abstract
Within the family of purinergic receptors, the P2X1 receptor is a ligand-gated ion channel that plays a role in urogenital, immune and cardiovascular function. Specifically, the P2X1 receptor has been implicated in controlling smooth muscle contractions of the vas deferens and therefore has emerged as an exciting drug target for male contraception. In addition, the P2X1 receptor contributes to smooth muscle contractions of the bladder and is a target to treat bladder dysfunction. Finally, platelets and neutrophils have populations of P2X1 receptors that could be targeted for thrombosis and inflammatory conditions. Drugs that specifically target the P2X1 receptor have been challenging to develop, and only recently have small molecule antagonists of the P2X1 receptor been available. However, these ligands need further biological validation for appropriate selectivity and drug-like properties before they will be suitable for use in preclinical models of disease. Although the atomic structure of the P2X1 receptor has yet to be determined, the recent discovery of several other P2X receptor structures and improvements in the field of structural biology suggests that this is now a distinct possibility. Such efforts may significantly improve drug discovery efforts at the P2X1 receptor.
Collapse
|
15
|
Demeules M, Scarpitta A, Hardet R, Gondé H, Abad C, Blandin M, Menzel S, Duan Y, Rissiek B, Magnus T, Mann AM, Koch-Nolte F, Adriouch S. Evaluation of nanobody-based biologics targeting purinergic checkpoints in tumor models in vivo. Front Immunol 2022; 13:1012534. [PMID: 36341324 PMCID: PMC9626963 DOI: 10.3389/fimmu.2022.1012534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Adenosine triphosphate (ATP) represents a danger signal that accumulates in injured tissues, in inflammatory sites, and in the tumor microenvironment. ATP promotes tumor growth but also anti-tumor immune responses notably via the P2X7 receptor. ATP can also be catabolized by CD39 and CD73 ecto-enzymes into immunosuppressive adenosine. P2X7, CD39 and CD73 have attracted much interest in cancer as targets offering the potential to unleash anti-tumor immune responses. These membrane proteins represent novel purinergic checkpoints that can be targeted by small drugs or biologics. Here, we investigated nanobody-based biologics targeting mainly P2X7, but also CD73, alone or in combination therapies. Blocking P2X7 inhibited tumor growth and improved survival of mice in cancer models that express P2X7. P2X7-potentiation by a nanobody-based biologic was not effective alone to control tumor growth but enhanced tumor control and immune responses when used in combination with oxaliplatin chemotherapy. We also evaluated a bi-specific nanobody-based biologic that targets PD-L1 and CD73. This novel nanobody-based biologic exerted a potent anti-tumor effect, promoting tumor rejection and improving survival of mice in two tumor models. Hence, this study highlights the importance of purinergic checkpoints in tumor control and open new avenues for nanobody-based biologics that may be further exploited in the treatment of cancer.
Collapse
Affiliation(s)
- Mélanie Demeules
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Allan Scarpitta
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Romain Hardet
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Henri Gondé
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Catalina Abad
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Marine Blandin
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Bonn, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yinghui Duan
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Marei Mann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sahil Adriouch
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie Univ, Rouen, France
- *Correspondence: Sahil Adriouch,
| |
Collapse
|
16
|
Sattler C, Benndorf K. Enlightening activation gating in P2X receptors. Purinergic Signal 2022; 18:177-191. [PMID: 35188598 PMCID: PMC9123132 DOI: 10.1007/s11302-022-09850-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
P2X receptors are trimeric nonselective cation channels gated by ATP. They assemble from seven distinct subunit isoforms as either homo- or heteromeric complexes and contain three extracellularly located binding sites for ATP. P2X receptors are expressed in nearly all tissues and are there involved in physiological processes like synaptic transmission, pain, and inflammation. Thus, they are a challenging pharmacological target. The determination of crystal and cryo-EM structures of several isoforms in the last decade in closed, open, and desensitized states has provided a firm basis for interpreting the huge amount of functional and biochemical data. Electrophysiological characterization in conjugation with optical approaches has generated significant insights into structure–function relationships of P2X receptors. This review focuses on novel optical and related approaches to better understand the conformational changes underlying the activation of these receptors.
Collapse
Affiliation(s)
- Christian Sattler
- Institut Für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany.
| | - Klaus Benndorf
- Institut Für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany.
| |
Collapse
|
17
|
Dunning K, Peverini L, Grutter T. Using Symmetrical Organic Cation Solutions to Study P2X7 Ion Permeation. Methods Mol Biol 2022; 2510:239-252. [PMID: 35776328 DOI: 10.1007/978-1-0716-2384-8_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
P2X7 receptors are ATP-gated ion channels permeable to metal cations, such as Na+, K+, and Ca2+. They also exhibit permeability to various large molecular weight species, reaching up to 900 Da, in a process known as macropore formation, which is a unique functional hallmark across the P2X family. While well-documented in a range of different cell types, the molecular mechanism underlying this phenomenon is poorly understood, and has been clouded through the use of electrophysiological methodology prone to artifacts as a result of significant changes in ionic concentrations in asymmetric conditions. In this chapter, we discuss the permeation properties of P2X7, the related methodological challenges and the use of symmetrical organic cation solutions as a useful technique for probing P2X7 permeation.
Collapse
Affiliation(s)
- Kate Dunning
- Centre National de la Recherche Scientifique, University of Strasbourg, CAMB UMR 7199, Strasbourg, France
| | - Laurie Peverini
- Channel-Receptors Unit, Institut Pasteur, UMR 3571, Paris, France
| | - Thomas Grutter
- Centre National de la Recherche Scientifique, University of Strasbourg, CAMB UMR 7199, Strasbourg, France.
- University of Strasbourg Institute for Advanced Studies (USIAS), Strasbourg, France.
- Equipe de Chimie et Neurobiologie Moléculaire, Faculté de Pharmacie, UMR 7199 CNRS/Université de Strasbourg, Illkirch, France.
| |
Collapse
|
18
|
Airway Exposure to Polyethyleneimine Nanoparticles Induces Type 2 Immunity by a Mechanism Involving Oxidative Stress and ATP Release. Int J Mol Sci 2021; 22:ijms22169071. [PMID: 34445774 PMCID: PMC8396525 DOI: 10.3390/ijms22169071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Polyethyleneimine (PEI) induced immune responses were investigated in human bronchial epithelial (hBE) cells and mice. PEI rapidly induced ATP release from hBE cells and pretreatment with glutathione (GSH) blocked the response. PEI activated two conductive pathways, VDAC-1 and pannexin 1, which completely accounted for ATP efflux across the plasma membrane. Moreover, PEI increased intracellular Ca2+ concentration ([Ca2+]i), which was reduced by the pannexin 1 inhibitor, 10Panx (50 μM), the VDAC-1 inhibitor, DIDS (100 μM), and was nearly abolished by pretreatment with GSH (5 mM). The increase in [Ca2+]i involved Ca2+ uptake through two pathways, one blocked by oxidized ATP (oATP, 300 μM) and another that was blocked by the TRPV-1 antagonist A784168 (100 nM). PEI stimulation also increased IL-33 mRNA expression and protein secretion. In vivo experiments showed that acute (4.5 h) PEI exposure stimulated secretion of Th2 cytokines (IL-5 and IL-13) into bronchoalveolar lavage (BAL) fluid. Conjugation of PEI with ovalbumin also induced eosinophil recruitment and secretion of IL-5 and IL-13 into BAL fluid, which was inhibited in IL-33 receptor (ST2) deficient mice. In conclusion, PEI-induced oxidative stress stimulated type 2 immune responses by activating ATP-dependent Ca2+ uptake leading to IL-33 secretion, similar to allergens derived from Alternaria.
Collapse
|
19
|
P2X7 Receptors and TMEM16 Channels Are Functionally Coupled with Implications for Macropore Formation and Current Facilitation. Int J Mol Sci 2021; 22:ijms22126542. [PMID: 34207150 PMCID: PMC8234106 DOI: 10.3390/ijms22126542] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 02/03/2023] Open
Abstract
P2X7 receptors (P2X7) are cationic channels involved in many diseases. Following their activation by extracellular ATP, distinct signaling pathways are triggered, which lead to various physiological responses such as the secretion of pro-inflammatory cytokines or the modulation of cell death. P2X7 also exhibit unique behaviors, such as “macropore” formation, which corresponds to enhanced large molecule cell membrane permeability and current facilitation, which is caused by prolonged activation. These two phenomena have often been confounded but, thus far, no clear mechanisms have been resolved. Here, by combining different approaches including whole-cell and single-channel recordings, pharmacological and biochemical assays, CRISPR/Cas9 technology and cell imaging, we provide evidence that current facilitation and macropore formation involve functional complexes comprised of P2X7 and TMEM16, a family of Ca2+-activated ion channel/scramblases. We found that current facilitation results in an increase of functional complex-embedded P2X7 open probability, a result that is recapitulated by plasma membrane cholesterol depletion. We further show that macropore formation entails two distinct large molecule permeation components, one of which requires functional complexes featuring TMEM16F subtype, the other likely being direct permeation through the P2X7 pore itself. Such functional complexes can be considered to represent a regulatory hub that may orchestrate distinct P2X7 functionalities.
Collapse
|
20
|
Collado-Díaz V, Martinez-Cuesta MÁ, Blanch-Ruiz MA, Sánchez-López A, García-Martínez P, Peris JE, Usach I, Ivorra MD, Lacetera A, Martín-Santamaría S, Esplugues JV, Alvarez A. Abacavir Increases Purinergic P2X7 Receptor Activation by ATP: Does a Pro-inflammatory Synergism Underlie Its Cardiovascular Toxicity? Front Pharmacol 2021; 12:613449. [PMID: 33867979 PMCID: PMC8045785 DOI: 10.3389/fphar.2021.613449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/15/2021] [Indexed: 11/15/2022] Open
Abstract
The cardiovascular toxicity of Abacavir is related to its purinergic structure. Purinergic P2X7-receptors (P2X7R), characterized by activation by high concentrations of ATP and with high plasticity, seem implicated. We appraise the nature of the interplay between Abacavir and P2X7R in generating vascular inflammation. The effects of Abacavir on leukocyte-endothelium interactions were compared with those of its metabolite carbovir triphosphate (CBV-TP) or ATP in the presence of apyrase (ATP-ase) or A804598 (P2X7R-antagonist). CBV-TP and ATP levels were evaluated by HPLC, while binding of Abacavir, CBV-TP and ATP to P2X7R was assessed by radioligand and docking studies. Hypersensitivity studies explored a potential allosteric action of Abacavir. Clinical concentrations of Abacavir (20 µmol/L) induced leukocyte-endothelial cell interactions by specifically activating P2X7R, but the drug did not show affinity for the P2X7R ATP-binding site (site 1). CBV-TP levels were undetectable in Abacavir-treated cells, while those of ATP were unaltered. The effects of Abacavir were Apyrase-dependent, implying dependence on endogenous ATP. Exogenous ATP induced a profile of proinflammatory actions similar to Abacavir, but was not entirely P2X7R-dependent. Docking calculations suggested ATP-binding to sites 1 and 2, and Abacavir-binding only to allosteric site 2. A combination of concentrations of Abacavir (1 µmol/L) and ATP (0.1 µmol/L) that had no effect when administered separately induced leukocyte-endothelium interactions mediated by P2X7R and involving Connexin43 channels. Therefore, Abacavir acts as a positive allosteric modulator of P2X7R, turning low concentrations of endogenous ATP themselves incapable of stimulating P2X7R into a functional proinflammatory agonist of the receptor.
Collapse
Affiliation(s)
- Víctor Collado-Díaz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Maria Ángeles Martinez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,CIBERehd, Valencia, Spain
| | | | - Ainhoa Sánchez-López
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | - José E Peris
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
| | - Iris Usach
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Valencia, Valencia, Spain
| | - Maria Dolores Ivorra
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Alessandra Lacetera
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sonsoles Martín-Santamaría
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,CIBERehd, Valencia, Spain.,FISABIO- Fundación Hospital Universitario Dr. Peset, Valencia, Spain
| | - Angeles Alvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,CIBERehd, Valencia, Spain
| |
Collapse
|
21
|
Kanellopoulos JM, Almeida-da-Silva CLC, Rüütel Boudinot S, Ojcius DM. Structural and Functional Features of the P2X4 Receptor: An Immunological Perspective. Front Immunol 2021; 12:645834. [PMID: 33897694 PMCID: PMC8059410 DOI: 10.3389/fimmu.2021.645834] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular nucleotides are important mediators of activation, triggering various responses through plasma membrane P2 and P1 receptors. P2 receptors are further subdivided into ionotropic P2X receptors and G protein-coupled P2Y receptors. P2X4 is an ATP-gated cation channel broadly expressed in most tissues of the body. Within the P2X family, P2X4 has a unique subcellular distribution, being preferentially localized in lysosomes. In these organelles, high ATP concentrations do not trigger P2X4 because of the low pH. However, when the pH increases to 7.4, P2X4 can be stimulated by intra-lysosomal ATP, which is in its active, tetra-anionic form. Elucidation of P2X4, P2X3 and P2X7 structures has shed some light on the functional differences between these purinergic receptors. The potential interaction between P2X4 and P2X7 has been extensively studied. Despite intensive effort, it has not been possible yet to determine whether P2X4 and P2X7 interact as heterotrimers or homotrimers at the plasma membrane. However, several publications have shown that functional interactions between P2X4 and P2X7 do occur. Importantly, these studies indicate that P2X4 potentiates P2X7-dependent activation of inflammasomes, leading to increased release of IL-1β and IL-18. The role of P2X4 in various diseases could be beneficial or deleterious even though the pathophysiological mechanisms involved are still poorly defined. However, in diseases whose physiopathology involves activation of the NLRP3 inflammasome, P2X4 was found to exacerbate severity of disease. The recent production of monoclonal antibodies specific for the human and mouse P2X4, some of which are endowed with agonist or antagonist properties, raises the possibility that they could be used therapeutically. Analysis of single nucleotide polymorphisms of the human P2RX4 gene has uncovered the association of P2RX4 gene variants with susceptibility to several human diseases.
Collapse
Affiliation(s)
- Jean M Kanellopoulos
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | | | - Sirje Rüütel Boudinot
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, United States
| |
Collapse
|
22
|
Sagar S, Kapoor H, Chaudhary N, Roy SS. Cellular and mitochondrial calcium communication in obstructive lung disorders. Mitochondrion 2021; 58:184-199. [PMID: 33766748 DOI: 10.1016/j.mito.2021.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+) signalling is well known to dictate cellular functioning and fate. In recent years, the accumulation of Ca2+ in the mitochondria has emerged as an important factor in Chronic Respiratory Diseases (CRD) such as Asthma and Chronic Obstructive Pulmonary Disease (COPD). Various reports underline an aberrant increase in the intracellular Ca2+, leading to mitochondrial ROS generation, and further activation of the apoptotic pathway in these diseases. Mitochondria contribute to Ca2+ buffering which in turn regulates mitochondrial metabolism and ATP production. Disruption of this Ca2+ balance leads to impaired cellular processes like apoptosis or necrosis and thus contributes to the pathophysiology of airway diseases. This review highlights the key role of cytoplasmic and mitochondrial Ca2+ signalling in regulating CRD, such as asthma and COPD. A better understanding of the dysregulation of mitochondrial Ca2+ homeostasis in these diseases could provide cues for the development of advanced therapeutic interventions in these diseases.
Collapse
Affiliation(s)
- Shakti Sagar
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Himanshi Kapoor
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Nisha Chaudhary
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Soumya Sinha Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
23
|
Ferrari D, la Sala A, Milani D, Celeghini C, Casciano F. Purinergic Signaling in Controlling Macrophage and T Cell Functions During Atherosclerosis Development. Front Immunol 2021; 11:617804. [PMID: 33664731 PMCID: PMC7921745 DOI: 10.3389/fimmu.2020.617804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a hardening and narrowing of arteries causing a reduction of blood flow. It is a leading cause of death in industrialized countries as it causes heart attacks, strokes, and peripheral vascular disease. Pathogenesis of the atherosclerotic lesion (atheroma) relies on the accumulation of cholesterol-containing low-density lipoproteins (LDL) and on changes of artery endothelium that becomes adhesive for monocytes and lymphocytes. Immunomediated inflammatory response stimulated by lipoprotein oxidation, cytokine secretion and release of pro-inflammatory mediators, worsens the pathological context by amplifying tissue damage to the arterial lining and increasing flow-limiting stenosis. Formation of thrombi upon rupture of the endothelium and the fibrous cup may also occur, triggering thrombosis often threatening the patient’s life. Purinergic signaling, i.e., cell responses induced by stimulation of P2 and P1 membrane receptors for the extracellular nucleotides (ATP, ADP, UTP, and UDP) and nucleosides (adenosine), has been implicated in modulating the immunological response in atherosclerotic cardiovascular disease. In this review we will describe advancements in the understanding of purinergic modulation of the two main immune cells involved in atherogenesis, i.e., monocytes/macrophages and T lymphocytes, highlighting modulation of pro- and anti-atherosclerotic mediated responses of purinergic signaling in these cells and providing new insights to point out their potential clinical significance.
Collapse
Affiliation(s)
- Davide Ferrari
- Department of Life Science and Biotechnology, Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
| | - Andrea la Sala
- Certification Unit, Health Directorate, Bambino Gesù Pediatric Hospital, IRCCS, Rome, Italy
| | - Daniela Milani
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Claudio Celeghini
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
24
|
Zhu X, Li Q, Song W, Peng X, Zhao R. P2X7 receptor: a critical regulator and potential target for breast cancer. J Mol Med (Berl) 2021; 99:349-358. [PMID: 33486566 DOI: 10.1007/s00109-021-02041-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is currently the most common cancer and the leading cause of cancer death among women worldwide. Advanced breast cancer is prone to metastasis, and there is currently no drug to cure metastatic breast cancer. The purinergic ligand-gated ion channel 7 receptor is an ATP-gated nonselective cation channel receptor and is involved in signal transduction, growth regulation, cytokine secretion, and tumor cell development. Recent studies have shown that upregulation of the P2X7 receptor in breast cancer can mediate AKT signaling pathways, Ca2 þ-activated SK3 potassium channels, and EMT and regulate the secretion of small extracellular vesicles to promote breast cancer invasion and migration, which are affected by factors such as hypoxia and ATP. In addition, studies have shown that microRNAs can bind to the 3' untranslated region of the P2X7 receptor, which affects the occurrence and development of breast cancer by upregulating and downregulating P2X7 receptor expression. Studies have shown that new P2X7 receptor inhibitors, such as emodin and Uncaria tomentosa, can inhibit P2X7 receptor-mediated breast cancer invasion and are expected to be used clinically. This article reviews the research progress on the relationship between the P2X7 receptor and breast cancer to provide new ideas and a basis for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaodi Zhu
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China
| | - Qianqian Li
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China
| | - Wei Song
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China
| | - Xiaoxiang Peng
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China.
| | - Ronglan Zhao
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China.
| |
Collapse
|
25
|
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
|
26
|
Martínez-Cuesta MÁ, Blanch-Ruiz MA, Ortega-Luna R, Sánchez-López A, Álvarez Á. Structural and Functional Basis for Understanding the Biological Significance of P2X7 Receptor. Int J Mol Sci 2020; 21:ijms21228454. [PMID: 33182829 PMCID: PMC7696479 DOI: 10.3390/ijms21228454] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022] Open
Abstract
The P2X7 receptor (P2X7R) possesses a unique structure associated to an as yet not fully understood mechanism of action that facilitates cell permeability to large ionic molecules through the receptor itself and/or nearby membrane proteins. High extracellular adenosine triphosphate (ATP) levels—inexistent in physiological conditions—are required for the receptor to be triggered and contribute to its role in cell damage signaling. The inconsistent data on its activation pathways and the few studies performed in natively expressed human P2X7R have led us to review the structure, activation pathways, and specific cellular location of P2X7R in order to analyze its biological relevance. The ATP-gated P2X7R is a homo-trimeric receptor channel that is occasionally hetero-trimeric and highly polymorphic, with at least nine human splice variants. It is localized predominantly in the cellular membrane and has a characteristic plasticity due to an extended C-termini, which confers it the capacity of interacting with membrane structural compounds and/or intracellular signaling messengers to mediate flexible transduction pathways. Diverse drugs and a few endogenous molecules have been highlighted as extracellular allosteric modulators of P2X7R. Therefore, studies in human cells that constitutively express P2X7R need to investigate the precise endogenous mediator located nearby the activation/modulation domains of the receptor. Such research could help us understand the possible physiological ATP-mediated P2X7R homeostasis signaling.
Collapse
Affiliation(s)
- María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (A.S.-L.)
- CIBERehd, Valencia, Spain
- Correspondence: (M.Á.M.-C.); (Á.Á.); Tel.: +34-963983716 (M.Á.M.-C.); +34-963864898 (Á.Á.)
| | - María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (A.S.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (A.S.-L.)
| | - Ainhoa Sánchez-López
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (A.S.-L.)
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (A.S.-L.)
- CIBERehd, Valencia, Spain
- Correspondence: (M.Á.M.-C.); (Á.Á.); Tel.: +34-963983716 (M.Á.M.-C.); +34-963864898 (Á.Á.)
| |
Collapse
|
27
|
Demeules M, Scarpitta A, Abad C, Gondé H, Hardet R, Pinto-Espinoza C, Eichhoff AM, Schäfer W, Haag F, Koch-Nolte F, Adriouch S. Evaluation of P2X7 Receptor Function in Tumor Contexts Using rAAV Vector and Nanobodies (AAVnano). Front Oncol 2020; 10:1699. [PMID: 33042812 PMCID: PMC7518291 DOI: 10.3389/fonc.2020.01699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine triphosphate (ATP) represents a danger signal that accumulates in injured tissues, in inflammatory sites, and in the tumor microenvironment. Extracellular ATP is known to signal through plasma membrane receptors of the P2Y and P2X families. Among the P2X receptors, P2X7 has attracted increasing interest in the field of inflammation as well as in cancer. P2X7 is expressed by immune cells and by most malignant tumor cells where it plays a crucial yet complex role that remains to be clarified. P2X7 activity has been associated with production and release of pro-inflammatory cytokines, modulation of the activity and survival of immune cells, and the stimulation of proliferation and migratory properties of tumor cells. Hence, P2X7 plays an intricate role in the tumor microenvironment combining beneficial and detrimental effects that need to be further investigated. For this, we developed a novel methodology termed AAVnano based on the use of Adeno-associated viral vectors (AAV) encoding nanobodies targeting P2X7. We discuss here the advantages of this tool to study the different functions of P2X7 in cancer and other pathophysiological contexts.
Collapse
Affiliation(s)
- Mélanie Demeules
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Allan Scarpitta
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Catalina Abad
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Henri Gondé
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Romain Hardet
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | | | - Anna Marei Eichhoff
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Schäfer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sahil Adriouch
- Normandie University, UNIROUEN, INSERM, U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| |
Collapse
|
28
|
P2X7 Receptors Amplify CNS Damage in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21175996. [PMID: 32825423 PMCID: PMC7504621 DOI: 10.3390/ijms21175996] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
ATP is a (co)transmitter and signaling molecule in the CNS. It acts at a multitude of ligand-gated cationic channels termed P2X to induce rapid depolarization of the cell membrane. Within this receptor-channel family, the P2X7 receptor (R) allows the transmembrane fluxes of Na+, Ca2+, and K+, but also allows the slow permeation of larger organic molecules. This is supposed to cause necrosis by excessive Ca2+ influx, as well as depletion of intracellular ions and metabolites. Cell death may also occur by apoptosis due to the activation of the caspase enzymatic cascade. Because P2X7Rs are localized in the CNS preferentially on microglia, but also at a lower density on neuroglia (astrocytes, oligodendrocytes) the stimulation of this receptor leads to the release of neurodegeneration-inducing bioactive molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen and nitrogen molecules, and the excitotoxic glutamate/ATP. Various neurodegenerative reactions of the brain/spinal cord following acute harmful events (mechanical CNS damage, ischemia, status epilepticus) or chronic neurodegenerative diseases (neuropathic pain, Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis) lead to a massive release of ATP via the leaky plasma membrane of neural tissue. This causes cellular damage superimposed on the original consequences of neurodegeneration. Hence, blood-brain-barrier permeable pharmacological antagonists of P2X7Rs with excellent bioavailability are possible therapeutic agents for these diseases. The aim of this review article is to summarize our present state of knowledge on the involvement of P2X7R-mediated events in neurodegenerative illnesses endangering especially the life quality and duration of the aged human population.
Collapse
|
29
|
McCarthy AE, Yoshioka C, Mansoor SE. Full-Length P2X 7 Structures Reveal How Palmitoylation Prevents Channel Desensitization. Cell 2019; 179:659-670.e13. [PMID: 31587896 DOI: 10.1016/j.cell.2019.09.017] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/30/2019] [Accepted: 09/13/2019] [Indexed: 12/24/2022]
Abstract
P2X receptors are trimeric, non-selective cation channels activated by extracellular ATP. The P2X7 receptor subtype is a pharmacological target because of involvement in apoptotic, inflammatory, and tumor progression pathways. It is the most structurally and functionally distinct P2X subtype, containing a unique cytoplasmic domain critical for the receptor to initiate apoptosis and not undergo desensitization. However, lack of structural information about the cytoplasmic domain has hindered understanding of the molecular mechanisms underlying these processes. We report cryoelectron microscopy structures of full-length rat P2X7 receptor in apo and ATP-bound states. These structures reveal how one cytoplasmic element, the C-cys anchor, prevents desensitization by anchoring the pore-lining helix to the membrane with palmitoyl groups. They show a second cytoplasmic element with a unique fold, the cytoplasmic ballast, which unexpectedly contains a zinc ion complex and a guanosine nucleotide binding site. Our structures provide first insights into the architecture and function of a P2X receptor cytoplasmic domain.
Collapse
Affiliation(s)
- Alanna E McCarthy
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA; Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Craig Yoshioka
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA
| | - Steven E Mansoor
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA; Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
30
|
Homerin G, Jawhara S, Dezitter X, Baudelet D, Dufrénoy P, Rigo B, Millet R, Furman C, Ragé G, Lipka E, Farce A, Renault N, Sendid B, Charlet R, Leroy J, Phanithavong M, Richeval C, Wiart JF, Allorge D, Adriouch S, Vouret-Craviari V, Ghinet A. Pyroglutamide-Based P2X7 Receptor Antagonists Targeting Inflammatory Bowel Disease. J Med Chem 2019; 63:2074-2094. [PMID: 31525963 DOI: 10.1021/acs.jmedchem.9b00584] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This report deals with the design, the synthesis, and the pharmacological evaluation of pyroglutamide-based P2X7 antagonists. A dozen were shown to possess improved properties, among which inhibition of YO-PRO-1/TO-PRO-3 uptake and IL1β release upon BzATP activation of the receptor and dampening signs of DSS-induced colitis on mice, in comparison with reference antagonist GSK1370319A. Docking study and biological evaluation of synthesized compounds has highlighted new SAR, and low toxicity profiles of pyroglutamides herein described are clues for the finding of a usable h-P2X7 antagonist drug. Such a drug would raise the hope for a cure to many P2X7-dependent pathologies, including inflammatory, neurological, and immune diseases.
Collapse
Affiliation(s)
- Germain Homerin
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Samir Jawhara
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Xavier Dezitter
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Davy Baudelet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Pierrick Dufrénoy
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Benoît Rigo
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France
| | - Régis Millet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Christophe Furman
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Guillaume Ragé
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Emmanuelle Lipka
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Laboratoire de Chimie Analytique, Faculté des Sciences Pharmaceutiques et Biologiques de Lille, F-59006 Lille Cedex, France
| | - Amaury Farce
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Nicolas Renault
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Institut de Chimie Pharmaceutique Albert Lespagnol, IFR114, 3 rue du Pr Laguesse, F-59006 Lille, France
| | - Boualem Sendid
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Rogatien Charlet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Jordan Leroy
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France
| | - Mélodie Phanithavong
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Camille Richeval
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Jean-François Wiart
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Delphine Allorge
- Laboratoire de Toxicologie & Génopathies, CHRU de Lille, Centre de Biologie Pathologie, Blvd du Pr. J. Leclercq, CS 70001, F-59037 Lille, France
| | - Sahil Adriouch
- INSERM U905, F-76183 Rouen, France.,Institute for Research and Innovation in Biomedicine, Normandie University, F-76183 Rouen, France
| | - Valérie Vouret-Craviari
- Institute for Research on Cancer and Aging (IRCAN), F-06100 Nice, France.,University of Nice Cote d'Azur (UCA), F-06100 Nice, France
| | - Alina Ghinet
- CHRU de Lille, Faculté de Médecine-Pôle Recherche, Inserm U995, LIRIC, Université de Lille, Place Verdun, F-59045 Lille Cedex, France.,Yncréa Hauts-de-France, UCLille, Laboratoire de Pharmacochimie, Hautes Etudes d'Ingénieur (HEI), 13 rue de Toul, F-59046 Lille, France.,Faculty of Chemistry, "Al. I. Cuza" University of Iasi, Blvd Carol I, nr. 11, 700506 Iasi, Romania
| |
Collapse
|
31
|
Kanellopoulos JM, Delarasse C. Pleiotropic Roles of P2X7 in the Central Nervous System. Front Cell Neurosci 2019; 13:401. [PMID: 31551714 PMCID: PMC6738027 DOI: 10.3389/fncel.2019.00401] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022] Open
Abstract
The purinergic receptor P2X7 is expressed in neural and immune cells known to be involved in neurological diseases. Its ligand, ATP, is a signaling molecule that can act as a neurotransmitter in physiological conditions or as a danger signal when released in high amount by damaged/dying cells or activated glial cells. Thus, ATP is a danger-associated molecular pattern. Binding of ATP by P2X7 leads to the activation of different biochemical pathways, depending on the physiological or pathological environment. The aim of this review is to discuss various functions of P2X7 in the immune and central nervous systems. We present evidence that P2X7 may have a detrimental or beneficial role in the nervous system, in the context of neurological pathologies: epilepsy, Alzheimer’s disease, multiple sclerosis, amyotrophic lateral sclerosis, age-related macular degeneration and cerebral artery occlusion.
Collapse
Affiliation(s)
| | - Cécile Delarasse
- Inserm, Sorbonne Université, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
32
|
Zheng XB, Zhang YL, Li Q, Liu YG, Wang XD, Yang BL, Zhu GC, Zhou CF, Gao Y, Liu ZX. Effects of 1,8-cineole on neuropathic pain mediated by P2X2 receptor in the spinal cord dorsal horn. Sci Rep 2019; 9:7909. [PMID: 31133659 PMCID: PMC6536508 DOI: 10.1038/s41598-019-44282-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
As an intractable health threat, neuropathic pain is now a key problem in clinical therapy, which can be caused by lesions affecting the peripheral nervous systems. 1,8-cineole is a natural monoterpene cyclic ether present in eucalyptus and has been reported to exhibit anti-inflammatory and antioxidant effects. Research has shown that 1,8-cineole inhibits P2X3 receptor-mediated neuropathic pains in dorsal root ganglion. The P2X2 and P2X3 receptors participate in the transmission of algesia and nociception information by primary sensory neurons. In the present study, We thus investigated in the spinal cord dorsal horn whether 1,8-cineole inhibits the expression of P2X2 receptor-mediated neuropathic pain. This study used rats in five random groups: group of chronic constriction injury(CCI) with dimethysulfoxide control (CCI + DMSO); group of CCI; sham group(Sham); group of CCI treated with a low dose 1,8-cineole (CCI + 50 mg/kg); group of CCI with a high dose (CCI + 100 mg/kg). We observed the effects of 1,8-cineole on thermal withdrawal latency (TWL) and mechanical withdrawal threshold (MWT). We examined P2X2 receptors mRNA change in rat spinal cord dorsal horn by In situ nucleic acid hybridization(ISH) and Quantitative realtime polymerase chain reaction (qRT-PCR) methods. Western Blotting and Immunohistochemical staining methods were used to observe P2X2 receptor protein expressions in the rat spinal cord dorsal horn. It demonstrated that oral administration of 1,8-cineole inhibits over-expression of P2X2 receptor protein and mRNA in the spinal cord and dorsal horn in the CCI rats. And the study explored new methods for the prevention and treatment of neuropathic pain.
Collapse
Affiliation(s)
- Xiao-Bo Zheng
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Jiangxi Health Vocational College, Nanchang, 330052, Jiangxi, People's Republic of China
| | - Ya-Ling Zhang
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Qing Li
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yi-Guo Liu
- Grade 2018, Medical School of Tongji University, Shanghai, 310000, People's Republic of China
| | - Xiang-Dong Wang
- Jiangxi Health Vocational College, Nanchang, 330052, Jiangxi, People's Republic of China
| | - Bao-Lin Yang
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Gao-Chun Zhu
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Cong-Fa Zhou
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yun Gao
- Department of physiology, Basic Medical School, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Zeng-Xu Liu
- Department of Anatomy, Medical School of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
33
|
Ugur M, Ugur Ö. A Mechanism-Based Approach to P2X7 Receptor Action. Mol Pharmacol 2019; 95:442-450. [PMID: 30737253 DOI: 10.1124/mol.118.115022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/30/2019] [Indexed: 01/08/2023] Open
Abstract
The ligand-gated ion channel P2X7 receptor attracts special attention due to its widespread presence as well as its unusual responses. Besides relatively well-understood mechanisms such as intracellular Ca2+ increase and K+ depletion, the P2X7 receptor activates other peculiar responses whose mechanisms are not fully understood. The best known among these is the permeabilization of the cell membrane to large molecules. This permeabilization has been explained by the activation of a nonselective permeation pathway by the P2X7 receptor, a phenomenon called "pore formation." However, with the emergence of new data, it became apparent that large molecules enter the cell directly through the pore of the ion channel, similar to the smaller ions. This explanation seems to be true for cationic large molecules. On the other hand, there is still convincing evidence indicating that the P2X7 receptor activates a separate pathway that permeates anionic large molecules in some cell types. Furthermore, there exist functional data suggesting that the P2X7 receptor may also activate other intracellular signaling molecules or other ion channels. Interestingly and contrary to what is expected from a ligand-gated channel, these activations occur in a seemingly direct manner. Somewhat overshadowed by the pore formation hypothesis, these action mechanisms may lead to a better understanding of not only the P2X7 receptor itself but also some important physiologic functions such as the release of anionic autocoids/neurotransmitters in the central nervous system. This review discusses, assesses, and draws attention to the data concerning these neglected but potentially important points in the P2X7 receptor field.
Collapse
Affiliation(s)
- Mehmet Ugur
- Department of Biophysics (M.U.) and Department of Pharmacology (O.U.), Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Özlem Ugur
- Department of Biophysics (M.U.) and Department of Pharmacology (O.U.), Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
34
|
Coughlin Q, Hopper AT, Blanco MJ, Tirunagaru V, Robichaud AJ, Doller D. Allosteric Modalities for Membrane-Bound Receptors: Insights from Drug Hunting for Brain Diseases. J Med Chem 2019; 62:5979-6002. [PMID: 30721063 DOI: 10.1021/acs.jmedchem.8b01651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Medicinal chemists are accountable for embedding the appropriate drug target profile into the molecular architecture of a clinical candidate. An accurate characterization of the functional effects following binding of a drug to its biological target is a fundamental step in the discovery of new medicines, informing the translation of preclinical efficacy and safety observations into human trials. Membrane-bound proteins, particularly ion channels and G protein-coupled receptors (GPCRs), are biological targets prone to allosteric modulation. Investigations using allosteric drug candidates and chemical tools suggest that their functional effects may be tailored with a high degree of translational alignment, making them molecular tools to correct pathophysiological functional tone and enable personalized medicine when a causative target-to-disease link is known. We present select examples of functional molecular fine-tuning of allosterism and discuss consequences relevant to drug design.
Collapse
|
35
|
Illes P, Rubini P, Huang L, Tang Y. The P2X7 receptor: a new therapeutic target in Alzheimer’s disease. Expert Opin Ther Targets 2019; 23:165-176. [DOI: 10.1080/14728222.2019.1575811] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Patrizia Rubini
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Lumei Huang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| |
Collapse
|