1
|
Mou YJ, Li FM, Zhang R, Sheng R, Han R, Zhang ZL, Hu LF, Zhao YZ, Wu JC, Qin ZH. The P2X7 receptor mediates NADPH transport across the plasma membrane. Biochem Biophys Res Commun 2024; 737:150500. [PMID: 39142135 DOI: 10.1016/j.bbrc.2024.150500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Nicotinamide Adenine Dinucleotide Phosphate (NADPH) plays a vital role in regulating redox homeostasis and reductive biosynthesis. However, if exogenous NADPH can be transported across the plasma membrane has remained elusive. In this study, we present evidence supporting that NADPH can traverse the plasma membranes of cells through a mechanism mediated by the P2X7 receptor (P2X7R). Notably, we observed an augmentation of intracellular NADPH levels in cultured microglia upon exogenous NADPH supplementation in the presence of ATP. The P2X7R-mediated transmembrane transportation of NADPH was validated with P2X7R antagonists, including OX-ATP, BBG, and A-438079, or through P2X7 knockdown, which impeded NADPH transportation into cells. Conversely, overexpression of P2X7 resulted in an enhanced capacity for NADPH transport. Furthermore, transfection of hP2X7 demonstrated the ability to complement NADPH uptake in native HEK293 cells. Our findings provide evidence for the first time that NADPH is transported across the plasma membrane via a P2X7R-mediated pathway. Additionally, we propose an innovative avenue for modulating intracellular NADPH levels. This discovery holds promise for advancing our understanding of the role of NADPH in redox homeostasis and neuroinflammation.
Collapse
Affiliation(s)
- Yu-Jie Mou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Feng-Min Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rong Zhang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zhong-Ling Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, 23 Post Street, Nangang District, Harbin, HeiLongjiang 150081, China.
| | - Li-Fang Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| | - Yu-Zheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Institute of Health Science and Technology, Suzhou Gaobo Vocational College, Qingshan Road, Suzhou Science and Technology Tower, Hi-Tech Area, Suzhou, Jiangsu 215163, China.
| |
Collapse
|
2
|
Vitureira N, Rafael A, Abudara V. P2X7 receptors and pannexin1 hemichannels shape presynaptic transmission. Purinergic Signal 2024; 20:223-236. [PMID: 37713157 PMCID: PMC11189373 DOI: 10.1007/s11302-023-09965-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
Over the last decades, since the discovery of ATP as a transmitter, accumulating evidence has been reported about the role of this nucleotide and purinergic receptors, in particular P2X7 receptors, in the modulation of synaptic strength and plasticity. Purinergic signaling has emerged as a crucial player in orchestrating the molecular interaction between the components of the tripartite synapse, and much progress has been made in how this neuron-glia interaction impacts neuronal physiology under basal and pathological conditions. On the other hand, pannexin1 hemichannels, which are functionally linked to P2X7 receptors, have appeared more recently as important modulators of excitatory synaptic function and plasticity under diverse contexts. In this review, we will discuss the contribution of ATP, P2X7 receptors, and pannexin hemichannels to the modulation of presynaptic strength and its impact on motor function, sensory processing, synaptic plasticity, and neuroglial communication, with special focus on the P2X7 receptor/pannexin hemichannel interplay. We also address major hypotheses about the role of this interaction in physiological and pathological circumstances.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
3
|
Iqbal J, Bano S, Khan IA, Huang Q. A patent review of P2X7 receptor antagonists to treat inflammatory diseases (2018-present). Expert Opin Ther Pat 2024; 34:263-271. [PMID: 38828613 DOI: 10.1080/13543776.2024.2363885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION The purinergic P2X7 receptor (P2X7R) is expressed on the surface of many different types of cells, including immune cells. Targeting P2X7R with antagonists has been studied for its potential therapeutic effects in a variety of inflammatory illnesses. AREA COVERED Many chemical substances, including carboxamides, benzamides and nitrogen containing heterocyclic derivatives have demonstrated promising inhibitory potential for P2X7 receptor. The chemistry and clinical applications of P2X7R antagonists patented from 2018- present are discussed in this review. EXPERT OPINION Purinergic receptor inhibitor discovery and application has demonstrated the potential for therapeutic intervention, as demonstrated by pharmacological research. Few chemical modalities have been authorized for use in clinical settings, despite the fact that breakthroughs in crystallography and chemical biology have increased the knowledge of purinergic signaling and its consequences in disease. The many research projects and pharmaceutical movements that sustain dynamic P2X receptor programs over decades are evidence of the therapeutic values and academic persistence in purinergic study. P2X7R is an intriguing therapeutic target and possible biomarker for inflammation. Although several companies like Merck and AstraZeneca have published patents on P2X3 antagonists, the search for P2X7R antagonists has not stopped. Numerous pharmaceutical companies have disclosed different scaffolds, and some molecules are presently being studied in clinical studies.
Collapse
Affiliation(s)
- Jamshed Iqbal
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Sehrish Bano
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Imtiaz Ali Khan
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Qing Huang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
4
|
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
|
5
|
Hayoz S, Jia C, Hegg CC. Constitutive and evoked release of ATP in adult mouse olfactory epithelium. Open Life Sci 2024; 19:20220811. [PMID: 38250473 PMCID: PMC10795008 DOI: 10.1515/biol-2022-0811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/31/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
In adult olfactory epithelium (OE), ATP plays a role in constant cell turnover and post-injury neuroregeneration. We previously demonstrated that constitutive and ATP-evoked ATP release are present in neonatal mouse OE and underlie continuous cell turn-over and post-injury neuroregeneration, and that activation of purinergic P2X7 receptors is involved in the evoked release. We hypothesized that both releases are present in adult mouse OE. To study the putative contribution of olfactory sensory neurons to ATP release, we used olfactory sensory neuronal-like OP6 cells derived from the embryonic olfactory placode cells. Calcium imaging showed that OP6 cells and primary adult OE cell cultures express functional purinergic receptors. We monitored ATP release from OP6 cells and whole adult OE turbinates using HEK cells as biosensors and luciferin-luciferase assays. Constitutive ATP release occurs in OP6 cells and whole adult mouse OE turbinates, and P2X7 receptors mediated evoked ATP release occurs only in turbinates. The mechanisms of ATP release described in the present study might underlie the constant cell turn-over and post-injury neuroregeneration present in adult OE and thus, further studies of these mechanisms are warranted as it will improve our knowledge of OE tissue homeostasis and post-injury regeneration.
Collapse
Affiliation(s)
- Sébastien Hayoz
- Department of Physiology, University of Arizona, Tucson, Arizona 85724, USA
| | - Cuihong Jia
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee 37614, USA
| | - Colleen Cosgrove Hegg
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
6
|
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
|
7
|
Luan Y, Luan Y, Jiao Y, Liu H, Huang Z, Feng Q, Pei J, Yang Y, Ren K. Broadening Horizons: Exploring mtDAMPs as a Mechanism and Potential Intervention Target in Cardiovascular Diseases. Aging Dis 2023; 15:2395-2416. [PMID: 38270118 PMCID: PMC11567272 DOI: 10.14336/ad.2023.1130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Cardiovascular diseases (CVDs) have been recognized as the leading cause of premature mortality and morbidity worldwide despite significant advances in therapeutics. Inflammation is a key factor in CVD progression. Once stress stimulates cells, they release cellular compartments known as damage-associated molecular patterns (DAMPs). Mitochondria can release mitochondrial DAMPs (mtDAMPs) to initiate an immune response when stimulated with cellular stress. Investigating the molecular mechanisms underlying the DAMPs that regulate CVD progression is crucial for improving CVDs. Herein, we discuss the composition and mechanism of DAMPs, the significance of mtDAMPs in cellular inflammation, the presence of mtDAMPs in different types of cells, and the main signaling pathways associated with mtDAMPs. Based on this, we determined the role of DAMPs in CVDs and the effects of mtDAMP intervention on CVD progression. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of DAMPs, this review seeks to provide important theoretical foundations for developing drugs targeting CVDs.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Qi Feng
- Department ofIntegrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jinyan Pei
- Quality Management Department, The Third People’s Hospital of Henan Provine, Zhengzhou, China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
8
|
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
|
9
|
Purinergic P2X7R as a potential target for pancreatic cancer. Clin Transl Oncol 2023:10.1007/s12094-023-03123-7. [PMID: 36856920 DOI: 10.1007/s12094-023-03123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023]
Abstract
Pancreatic cancer is one of the deadliest types of cancer, with a death rate nearly equal to the incidence. The P2X7 receptor (P2X7R) is a kind of extracellular adenosine triphosphate (ATP)-gated ion channel with special permeability, which exists in most tissues of human body and mediates inflammation-related signaling pathways and immune signal transduction after activation. P2X7R is also present on the surface of several tumor cells and is involved in tumor growth and progression. P2X7R expression in pancreatic cancer has also been identified in recent studies. Activation of P2X7R in pancreatic cancer can support the proliferation of pancreatic stellate cells, participate in protein interactions, and mediate ERK1/2, IL-6/STAT3, hCAP-18/LL-37, PI3K/AKT signaling pathways to promote pancreatic cancer progression. Inhibitors targeting P2X7R can inhibit the development of pancreatic cancer and are expected to be used in clinical therapy. Therefore, P2X7R is promising as a potential therapeutic target for pancreatic cancer. This article reviews the progress of research on P2X7R in pancreatic cancer.
Collapse
|
10
|
Zalpoor H, Akbari A, Nabi-Afjadi M, Forghaniesfidvajani R, Tavakol C, Barzegar Z, Iravanpour F, Hosseini M, Mousavi SR, Farrokhi MR. Hypoxia-inducible factor 1 alpha (HIF-1α) stimulated and P2X7 receptor activated by COVID-19, as a potential therapeutic target and risk factor for epilepsy. Hum Cell 2022; 35:1338-1345. [PMID: 35831562 PMCID: PMC9281298 DOI: 10.1007/s13577-022-00747-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/03/2022] [Indexed: 12/25/2022]
Abstract
Based on available evidence, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a neuroinvasive virus. According to the centers for disease control and prevention (CDC), coronavirus disease 2019 (COVID-19) may cause epilepsy. In this line, COVID-19 can stimulate hypoxia-inducible factor-1 alpha (HIF-1α) and activate P2X7 receptor. Both HIF-1α and P2X7 receptors are linked to epileptogenesis and seizures. Therefore, in the current study, we suggested that COVID-19 may have a role in epileptogenesis and seizure through HIF-1α stimulation and P2X7 receptor activation. Consequently, pharmacological targeting of these factors could be a promising therapeutic approach for such patients.
Collapse
Affiliation(s)
- Hamidreza Zalpoor
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
| | - Abdullatif Akbari
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Chanour Tavakol
- Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohreh Barzegar
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farideh Iravanpour
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshid Hosseini
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Reza Mousavi
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farrokhi
- Department of Neurosurgery, Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. .,Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
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
|
12
|
Mansoor SE. How Structural Biology Has Directly Impacted Our Understanding of P2X Receptor Function and Gating. Methods Mol Biol 2022; 2510:1-29. [PMID: 35776317 DOI: 10.1007/978-1-0716-2384-8_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
P2X receptors are ATP-gated ion channels expressed in a wide variety of eukaryotic cells. They play key roles in diverse processes such as platelet activation, smooth muscle contraction, synaptic transmission, nociception, cell proliferation, and inflammation making this receptor family an important pharmacological target. Structures of P2X receptors solved by X-ray crystallography have been instrumental in helping to define mechanisms of molecular P2X receptor function. In 2009, the first X-ray structure of the P2X4 receptor subtype confirmed a trimeric stoichiometry and revealed the overall architecture of the functional ion channel. Subsequent X-ray structures have provided the molecular details to define the orthosteric ATP binding pocket, the orthosteric antagonist binding pocket, an allosteric antagonist binding pocket, and the pore architecture in each of the major conformational states of the receptor gating cycle. Moreover, the unique gating mechanism by which P2X receptor subtypes desensitize at differing rates, referred to as the helical recoil model of receptor desensitization, was discovered directly from X-ray structures of the P2X3 receptor. However, structures of P2X receptors solved by X-ray crystallography have only been able to provide limited information on the cytoplasmic domain of this receptor family, as this domain was always truncated to varying degrees in order to facilitate crystallization. Because the P2X7 receptor subtype has a significantly larger cytoplasmic domain that has been shown to be necessary for its ability to initiate apoptosis, an absence of structural information on the P2X7 receptor cytoplasmic domain has limited our understanding of its complex signaling pathways as well as its unusual ability to remain open without undergoing desensitization. This absence of cytoplasmic structural information for P2X7 receptors was recently overcome when the first full-length P2X7 receptor structures were solved by single-particle cryogenic electron microscopy. These structures finally provide insight into the large and unique P2X7 receptor cytoplasmic domain and revealed two novel structural elements and several surprising findings: first, a cytoplasmic structural element called the cytoplasmic ballast was identified that contains a dinuclear zinc ion complex and a high affinity guanosine nucleotide binding site and second, a palmitoylated membrane proximal structural element called the C-cys anchor was identified which prevents P2X7 receptor desensitization. This chapter will highlight the major structural and functional aspects of P2X receptors discovered through structural biology, with a key emphasis on the most recent cryogenic electron microscopy structures of the full-length, wild-type P2X7 receptor.
Collapse
Affiliation(s)
- Steven E Mansoor
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA.
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
13
|
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
|
14
|
D’Amico R, Fusco R, Siracusa R, Impellizzeri D, Peritore AF, Gugliandolo E, Interdonato L, Sforza AM, Crupi R, Cuzzocrea S, Genovese T, Cordaro M, Di Paola R. Inhibition of P2X7 Purinergic Receptor Ameliorates Fibromyalgia Syndrome by Suppressing NLRP3 Pathway. Int J Mol Sci 2021; 22:ijms22126471. [PMID: 34208781 PMCID: PMC8234677 DOI: 10.3390/ijms22126471] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Fibromyalgia is a chronic condition characterized by persistent widespread pain that significantly reduces quality of life in patients. The purinergic P2X7 receptor (P2X7R) seems to be involved in different pain states and neuroinflammation. The purpose of this study is to investigate the positive effects of P2X7R inhibition by the antagonist Brilliant Blue G (BBG) in a rat model of reserpine-induced fibromyalgia. Sprague-Dawley male rats were injected with 1 mg/kg of reserpine for three consecutive days. Later, animals were administered BBG (50 mg/kg) intraperitoneally for seven days. Reserpine injections induced a significant increase in pain pro-inflammatory mediators as well as a significant increase in neuroinflammation. Chronic pain, in turn, led to depressive-like symptoms and reduced neurogenesis. Blockage of P2X7R by BBG administrations is able to attenuate the behavioral deficits, pain mediators and microglial activation induced by reserpine injection. Additionally, BBG prevents NLRP3 inflammasome activation and consequently the release of active interleukin (IL)-1 and IL-18, involved in the activation of nociceptors. In conclusion, these results suggest that inhibition of P2X7R should be further investigated to develop a potential approach for the management of fibromyalgia.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (E.G.); (R.C.)
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Andrea Maria Sforza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (E.G.); (R.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
- Correspondence: (S.C.); (T.G.); Tel.: +39-090-676-5208 (S.C. & T.G.)
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
- Correspondence: (S.C.); (T.G.); Tel.: +39-090-676-5208 (S.C. & T.G.)
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, via Consolare Valeria, 98125 Messina, Italy;
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.D.); (R.F.); (R.S.); (D.I.); (A.F.P.); (L.I.); or (A.M.S.); (R.D.P.)
| |
Collapse
|
15
|
Wang A, Shi X, Yu R, Qiao B, Yang R, Xu C. The P2X 7 Receptor Is Involved in Diabetic Neuropathic Pain Hypersensitivity Mediated by TRPV1 in the Rat Dorsal Root Ganglion. Front Mol Neurosci 2021; 14:663649. [PMID: 34163328 PMCID: PMC8215290 DOI: 10.3389/fnmol.2021.663649] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022] Open
Abstract
The purinergic 2X7 (P2X7) receptor expressed in satellite glial cells (SGCs) is involved in the inflammatory response, and transient receptor potential vanilloid 1 (TRPV1) participates in the process of neurogenic inflammation, such as that in diabetic neuropathic pain (DNP) and peripheral neuralgia. The main purpose of this study was to explore the role of the P2X7 receptor in DNP hypersensitivity mediated by TRPV1 in the rat and its possible mechanism. A rat model of type 2 diabetes mellitus-related neuropathic pain (NPP) named the DNP rat model was established in this study. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of DNP rats were increased after intrathecal injection of the P2X7 receptor antagonist A438079, and the mRNA and protein levels of TRPV1 in the dorsal root ganglion (DRG) were decreased in DNP rats treated with A438079 compared to untreated DNP rats; in addition, A438079 also decreased the phosphorylation of p38 and extracellular signal-regulated kinase 1/2 (ERK1/2) in the DNP group. Based on these results, the P2X7 receptor might be involved in DNP mediated by TRPV1.
Collapse
Affiliation(s)
- Anhui Wang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
| | - Xiangchao Shi
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Ruoyang Yu
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Bao Qiao
- Medical Department, Queen Mary School, Nanchang University, Nanchang, China
| | - Runan Yang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, China
| |
Collapse
|
16
|
Biswas D, Ambalavanan P, Ravins M, Anand A, Sharma A, Lim KXZ, Tan RYM, Lim HY, Sol A, Bachrach G, Angeli V, Hanski E. LL-37-mediated activation of host receptors is critical for defense against group A streptococcal infection. Cell Rep 2021; 34:108766. [PMID: 33657368 DOI: 10.1016/j.celrep.2021.108766] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 12/03/2020] [Accepted: 01/27/2021] [Indexed: 12/21/2022] Open
Abstract
Group A Streptococcus (GAS) causes diverse human diseases, including life-threatening soft-tissue infections. It is accepted that the human antimicrobial peptide LL-37 protects the host by killing GAS. Here, we show that GAS extracellular protease ScpC N-terminally cleaves LL-37 into two fragments of 8 and 29 amino acids, preserving its bactericidal activity. At sub-bactericidal concentrations, the cleavage inhibits LL-37-mediated neutrophil chemotaxis, shortens neutrophil lifespan, and eliminates P2X7 and EGF receptors' activation. Mutations at the LL-37 cleavage site protect the peptide from ScpC-mediated splitting, maintaining all its functions. The mouse LL-37 ortholog CRAMP is neither cleaved by ScpC nor does it activate P2X7 or EGF receptors. Treating wild-type or CRAMP-null mice with sub-bactericidal concentrations of the non-cleavable LL-37 analogs promotes GAS clearance that is abolished by the administration of either P2X7 or EGF receptor antagonists. We demonstrate that LL-37-mediated activation of host receptors is critical for defense against GAS soft-tissue infections.
Collapse
Affiliation(s)
- Debabrata Biswas
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.
| | - Poornima Ambalavanan
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Aparna Anand
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Abhinay Sharma
- Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Kimberly Xuan Zhen Lim
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Rachel Ying Min Tan
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Hwee Ying Lim
- Department of Microbiology and Immunology, National University of Singapore, LSI Immunology Programme, National University of Singapore, Singapore, Singapore
| | - Asaf Sol
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | - Veronique Angeli
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore; Department of Microbiology and Immunology, National University of Singapore, LSI Immunology Programme, National University of Singapore, Singapore, Singapore
| | - Emanuel Hanski
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), MMID Phase II, Campus for Research Excellence and Technological Enterprise (CREATE), and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore; Department of Microbiology and Molecular Genetics, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel.
| |
Collapse
|
17
|
Zhao YF, Tang Y, Illes P. Astrocytic and Oligodendrocytic P2X7 Receptors Determine Neuronal Functions in the CNS. Front Mol Neurosci 2021; 14:641570. [PMID: 33642994 PMCID: PMC7906075 DOI: 10.3389/fnmol.2021.641570] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
P2X7 receptors are members of the ATP-gated cationic channel family with a preferential localization at the microglial cells, the resident macrophages of the brain. However, these receptors are also present at neuroglia (astrocytes, oligodendrocytes) although at a considerably lower density. They mediate necrosis/apoptosis by the release of pro-inflammatory cytokines/chemokines, reactive oxygen species (ROS) as well as the excitotoxic (glio)transmitters glutamate and ATP. Besides mediating cell damage i.e., superimposed upon chronic neurodegenerative processes in Alzheimer’s Disease, Parkinson’s Disease, multiple sclerosis, and amyotrophic lateral sclerosis, they may also participate in neuroglial signaling to neurons under conditions of high ATP concentrations during any other form of neuroinflammation/neurodegeneration. It is a pertinent open question whether P2X7Rs are localized on neurons, or whether only neuroglia/microglia possess this receptor-type causing indirect effects by releasing the above-mentioned signaling molecules. We suggest as based on molecular biology and functional evidence that neurons are devoid of P2X7Rs although the existence of neuronal P2X7Rs cannot be excluded with absolute certainty.
Collapse
Affiliation(s)
- Ya-Fei Zhao
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Tang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,International Collaborative Center on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peter Illes
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,International Collaborative Center on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
18
|
Abstract
The P2X7 receptor for extracellular ATP is a well-established mediator of tumoral development and progression both in solid cancers and hematological malignancies. The human P2X7 gene is highly polymorphic, and several splice variants of the receptor have been identified in time. P2X7 single-nucleotide polymorphisms (SNPs) have been broadly analyzed by studies relating them to pathologies as different as infectious, inflammatory, nervous, and bone diseases, among which cancer is included. Moreover, in the last years, an increasing number of reports concentrated on P2X7 splice variants’ different roles and their implications in pathological conditions, including oncogenesis. Here, we give an overview of established and recent literature demonstrating a role for human P2X7 gene products in oncological conditions, mainly focusing on current data emerging on P2X7 isoform B and nfP2X7. We explored the role of these and other genetic variants of P2X7 in cancer insurgence, dissemination, and progression, as well as the effect of chemotherapy on isoforms expression. The described literature strongly suggests that P2X7 variants are potential new biomarkers and therapeutical targets in oncological conditions and that their study in carcinogenesis deserves to be further pursued.
Collapse
|
19
|
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
|
20
|
Zhang WJ, Luo C, Pu FQ, Zhu JF, Zhu Z. The role and pharmacological characteristics of ATP-gated ionotropic receptor P2X in cancer pain. Pharmacol Res 2020; 161:105106. [DOI: 10.1016/j.phrs.2020.105106] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
|
21
|
Maldifassi MC, Momboisse F, Guerra MJ, Vielma AH, Maripillán J, Báez-Matus X, Flores-Muñoz C, Cádiz B, Schmachtenberg O, Martínez AD, Cárdenas AM. The interplay between α7 nicotinic acetylcholine receptors, pannexin-1 channels and P2X7 receptors elicit exocytosis in chromaffin cells. J Neurochem 2020; 157:1789-1808. [PMID: 32931038 DOI: 10.1111/jnc.15186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 08/18/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Pannexin-1 (Panx1) forms plasma membrane channels that allow the exchange of small molecules between the intracellular and extracellular compartments, and are involved in diverse physiological and pathological responses in the nervous system. However, the signaling mechanisms that induce their opening still remain elusive. Here, we propose a new mechanism for Panx1 channel activation through a functional crosstalk with the highly Ca2+ permeable α7 nicotinic acetylcholine receptor (nAChR). Consistent with this hypothesis, we found that activation of α7 nAChRs induces Panx1-mediated dye uptake and ATP release in the neuroblastoma cell line SH-SY5Y-α7. Using membrane permeant Ca2+ chelators, total internal reflection fluorescence microscopy in SH-SY5Y-α7 cells expressing a membrane-tethered GCAMP3, and Src kinase inhibitors, we further demonstrated that Panx1 channel opening depends on Ca2+ signals localized in submembrane areas, as well as on Src kinases. In turn, Panx1 channels amplify cytosolic Ca2+ signals induced by the activation of α7 nAChRs, by a mechanism that seems to involve ATP release and P2X7 receptor activation, as hydrolysis of extracellular ATP with apyrase or blockage of P2X7 receptors with oxidized ATP significantly reduces the α7 nAChR-Ca2+ signal. The physiological relevance of this crosstalk was also demonstrated in neuroendocrine chromaffin cells, wherein Panx1 channels and P2X7 receptors contribute to the exocytotic release of catecholamines triggered by α7 nAChRs, as measured by amperometry. Together these findings point to a functional coupling between α7 nAChRs, Panx1 channels and P2X7 receptors with physiological relevance in neurosecretion.
Collapse
Affiliation(s)
- María C Maldifassi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | - María J Guerra
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alex H Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jaime Maripillán
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Universidad de Valparaíso, Chile
| | - Bárbara Cádiz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias Biológicas, Universidad de Valparaíso, Chile
| | - Oliver Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
22
|
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
|
23
|
Kopp R, Krautloher A, Ramírez-Fernández A, Nicke A. P2X7 Interactions and Signaling - Making Head or Tail of It. Front Mol Neurosci 2019; 12:183. [PMID: 31440138 PMCID: PMC6693442 DOI: 10.3389/fnmol.2019.00183] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022] Open
Abstract
Extracellular adenine nucleotides play important roles in cell-cell communication and tissue homeostasis. High concentrations of extracellular ATP released by dying cells are sensed as a danger signal by the P2X7 receptor, a non-specific cation channel. Studies in P2X7 knockout mice and numerous disease models have demonstrated an important role of this receptor in inflammatory processes. P2X7 activation has been shown to induce a variety of cellular responses that are not usually associated with ion channel function, for example changes in the plasma membrane composition and morphology, ectodomain shedding, activation of lipases, kinases, and transcription factors, as well as cytokine release and apoptosis. In contrast to all other P2X family members, the P2X7 receptor contains a long intracellular C-terminus that constitutes 40% of the whole protein and is considered essential for most of these effects. So far, over 50 different proteins have been identified to physically interact with the P2X7 receptor. However, few of these interactions have been confirmed in independent studies and for the majority of these proteins, the interaction domains and the physiological consequences of the interactions are only poorly described. Also, while the structure of the P2X7 extracellular domain has recently been resolved, information about the organization and structure of its C-terminal tail remains elusive. After shortly describing the structure and assembly of the P2X7 receptor, this review gives an update of the identified or proposed interaction domains within the P2X7 C-terminus, describes signaling pathways in which this receptor has been involved, and provides an overlook of the identified interaction partners.
Collapse
Affiliation(s)
- Robin Kopp
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna Krautloher
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Antonio Ramírez-Fernández
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|