1
|
Bayram Z, Akcabag E, Ozbey G, Nacitarhan C, Ozdem S, Turkay C, Ozdem SS. THE effect of P2X7 receptor activation on functional responses of human left internal mammary artery. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03411-1. [PMID: 39225832 DOI: 10.1007/s00210-024-03411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The Purinoreceptor 7 (P2X7R) has become a promising drug target in many cardiovascular diseases, including coronary artery disease, since prolonged activation of P2X7R could promote vascular dysfunction, atherosclerosis, and thrombosis. Thus, we aimed to study the effects of P2X7R activation on vascular relaxation responses of the human left internal mammary artery (LIMA). Sections of redundant human LIMA were cut into 3-mm wide rings,, suspended in 20-mL organ baths containing physiologic salt solution, and attached to an isometric force transducer connected to a computer-based data acquisition system. Long-term (60 min) incubation with specific P2X7R agonist Bz-ATP caused significant reductions in relaxation responses of LIMA to ATP and acetylcholine, which were reversed by selective P2X7R antagonists Brilliant Blue G or AZ11645373, whereas there were no changes in relaxation responses to endothelium-independent vasodilators isoprenaline, cAMP analog 8-Br-cAMP, and nitric oxide donor sodium nitroprusside. The impairment in relaxant responses of LIMA to endothelium-dependent vasodilators following activation of P2X7R for the long-term may contribute to postoperative LIMA vasospasm and hypertension. Modulation of P2X7R activity with selective agents may represent a new potential therapeutic approach in patients undergoing coronary artery bypass grafting surgery.
Collapse
Affiliation(s)
- Zeliha Bayram
- Turkish Medicines and Medical Devices Agency, Ankara, Turkey
| | - Esra Akcabag
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Gul Ozbey
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Cahit Nacitarhan
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Sebahat Ozdem
- Department of Medical Biochemistry, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Cengiz Turkay
- Department of Cardiovascular Surgery, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Sadi S Ozdem
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey.
| |
Collapse
|
2
|
Sakaguchi R, Takahashi N, Yoshida T, Ogawa N, Ueda Y, Hamano S, Yamaguchi K, Sawamura S, Yamamoto S, Hara Y, Kawamoto T, Suzuki R, Nakao A, Mori MX, Furukawa T, Shimizu S, Inoue R, Mori Y. Dynamic remodeling of TRPC5 channel-caveolin-1-eNOS protein assembly potentiates the positive feedback interaction between Ca 2+ and NO signals. J Biol Chem 2024; 300:107705. [PMID: 39178948 PMCID: PMC11420454 DOI: 10.1016/j.jbc.2024.107705] [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: 12/08/2023] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024] Open
Abstract
The cell signaling molecules nitric oxide (NO) and Ca2+ regulate diverse biological processes through their closely coordinated activities directed by signaling protein complexes. However, it remains unclear how dynamically the multicomponent protein assemblies behave within the signaling complexes upon the interplay between NO and Ca2+ signals. Here we demonstrate that TRPC5 channels activated by the stimulation of G-protein-coupled ATP receptors mediate Ca2+ influx, that triggers NO production from endothelial NO synthase (eNOS), inducing secondary activation of TRPC5 via cysteine S-nitrosylation and eNOS in vascular endothelial cells. Mutations in the caveolin-1-binding domains of TRPC5 disrupt its association with caveolin-1 and impair Ca2+ influx and NO production, suggesting that caveolin-1 serves primarily as the scaffold for TRPC5 and eNOS to assemble into the signal complex. Interestingly, during ATP receptor activation, eNOS is dissociated from caveolin-1 and in turn directly associates with TRPC5, which accumulates at the plasma membrane dependently on Ca2+ influx and calmodulin. This protein reassembly likely results in a relief of eNOS from the inhibitory action of caveolin-1 and an enhanced TRPC5 S-nitrosylation by eNOS localized in the proximity, thereby facilitating the secondary activation of Ca2+ influx and NO production. In isolated rat aorta, vasodilation induced by acetylcholine was significantly suppressed by the TRPC5 inhibitor AC1903. Thus, our study provides evidence that dynamic remodeling of the protein assemblies among TRPC5, eNOS, caveolin-1, and calmodulin determines the ensemble of Ca2+ mobilization and NO production in vascular endothelial cells.
Collapse
Affiliation(s)
- Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Nobuaki Takahashi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan
| | - Takashi Yoshida
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Nozomi Ogawa
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yoshifumi Ueda
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Satoshi Hamano
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kaori Yamaguchi
- Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Seishiro Sawamura
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Shinichiro Yamamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Yuji Hara
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Department of Integrative Physiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tomoya Kawamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ryosuke Suzuki
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Akito Nakao
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Masayuki X Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University, Fukuoka, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Golia MT, Gabrielli M, Verderio C. P2X 7 Receptor and Extracellular Vesicle Release. Int J Mol Sci 2023; 24:9805. [PMID: 37372953 DOI: 10.3390/ijms24129805] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/21/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Extensive evidence indicates that the activation of the P2X7 receptor (P2X7R), an ATP-gated ion channel highly expressed in immune and brain cells, is strictly associated with the release of extracellular vesicles. Through this process, P2X7R-expressing cells regulate non-classical protein secretion and transfer bioactive components to other cells, including misfolded proteins, participating in inflammatory and neurodegenerative diseases. In this review, we summarize and discuss the studies addressing the impact of P2X7R activation on extracellular vesicle release and their activities.
Collapse
Affiliation(s)
- Maria Teresa Golia
- National Research Council of Italy, Institute of Neuroscience, Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| | - Martina Gabrielli
- National Research Council of Italy, Institute of Neuroscience, Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| | - Claudia Verderio
- National Research Council of Italy, Institute of Neuroscience, Via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
| |
Collapse
|
4
|
Frolov AV, Shishkova DK, Markova VE, Sinitsky MY, Sinitskaya AV, Poddubnyak AO, Kanonykina AY, Zagorodnikov NI, Grigoriev EV, Kutikhin AG. Paracrine Effects of Conditioned Medium during Its Cross-Addition to Arterial and Venous Endothelial Cells. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
6
|
Purinergic Regulation of Endothelial Barrier Function. Int J Mol Sci 2021; 22:ijms22031207. [PMID: 33530557 PMCID: PMC7865261 DOI: 10.3390/ijms22031207] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Increased vascular permeability is a hallmark of several cardiovascular anomalies, including ischaemia/reperfusion injury and inflammation. During both ischaemia/reperfusion and inflammation, massive amounts of various nucleotides, particularly adenosine 5'-triphosphate (ATP) and adenosine, are released that can induce a plethora of signalling pathways via activation of several purinergic receptors and may affect endothelial barrier properties. The nature of the effects on endothelial barrier function may depend on the prevalence and type of purinergic receptors activated in a particular tissue. In this review, we discuss the influence of the activation of various purinergic receptors and downstream signalling pathways on vascular permeability during pathological conditions.
Collapse
|
7
|
Monaghan MLT, Bailey MA, Unwin RJ. Purinergic signalling in the kidney: In physiology and disease. Biochem Pharmacol 2020; 187:114389. [PMID: 33359067 DOI: 10.1016/j.bcp.2020.114389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Historically, the control of renal vascular and tubular function has, for the most part, concentrated on neural and endocrine regulation. However, in addition to these extrinsic factors, it is now appreciated that several complex humoral control systems exist within the kidney that can act in an autocrine and/or paracrine fashion. These paracrine systems complement neuroendocrine regulation by dynamically fine-tuning renal vascular and tubular function to buffer rapid changes in nephron perfusion and flow rate of tubular fluid. One of the most pervasive is the extracellular nucleotide/P2 receptor system, which is central to many of the intrinsic regulatory feedback loops within the kidney such as renal haemodynamic autoregulation and tubuloglomerular feedback (TGF). Although physiological actions of extracellular adenine nucleotides were reported almost 100 years ago, the conceptual framework for purinergic regulation of renal function owes much to the work of Geoffrey Burnstock. In this review, we reflect on our >20-year collaboration with Professor Burnstock and highlight the research that is still unlocking the potential of the renal purinergic system to understand and treat kidney disease.
Collapse
Affiliation(s)
- Marie-Louise T Monaghan
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom
| | - Matthew A Bailey
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom
| | - Robert J Unwin
- The Department of Renal Medicine, University College London, United Kingdom.
| |
Collapse
|
8
|
Martin-Aragon Baudel M, Espinosa-Tanguma R, Nieves-Cintron M, Navedo MF. Purinergic Signaling During Hyperglycemia in Vascular Smooth Muscle Cells. Front Endocrinol (Lausanne) 2020; 11:329. [PMID: 32528416 PMCID: PMC7256624 DOI: 10.3389/fendo.2020.00329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
The activation of purinergic receptors by nucleotides and/or nucleosides plays an important role in the control of vascular function, including modulation of vascular smooth muscle excitability, and vascular reactivity. Accordingly, purinergic receptor actions, acting as either ion channels (P2X) or G protein-coupled receptors (GCPRs) (P1, P2Y), target diverse downstream effectors, and substrates to regulate vascular smooth muscle function and vascular reactivity. Both vasorelaxant and vasoconstrictive effects have been shown to be mediated by different purinergic receptors in a vascular bed- and species-specific manner. Purinergic signaling has been shown to play a key role in altering vascular smooth muscle excitability and vascular reactivity following acute and short-term elevations in extracellular glucose (e.g., hyperglycemia). Moreover, there is evidence that vascular smooth muscle excitability and vascular reactivity is severely impaired during diabetes and that this is mediated, at least in part, by activation of purinergic receptors. Thus, purinergic receptors present themselves as important candidates mediating vascular reactivity in hyperglycemia, with potentially important clinical and therapeutic potential. In this review, we provide a narrative summarizing our current understanding of the expression, function, and signaling of purinergic receptors specifically in vascular smooth muscle cells and discuss their role in vascular complications following hyperglycemia and diabetes.
Collapse
Affiliation(s)
- Miguel Martin-Aragon Baudel
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
- *Correspondence: Miguel Martin-Aragon Baudel
| | - Ricardo Espinosa-Tanguma
- Departamento de Fisiologia y Biofisca, Universidad Autónoma San Luis Potosí, San Luis Potosí, Mexico
| | | | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
- Manuel F. Navedo
| |
Collapse
|
9
|
Lopez ES, Ortiz GA, Potilinski C, Croxatto JO, Gallo JE. Corneal Neovascularization: A Combined Approach of Bevacizumab and Suramin Showed Increased Antiangiogenic Effect Through Downregulation of BFGF and P2X2. Curr Eye Res 2017; 43:466-473. [DOI: 10.1080/02713683.2017.1416146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Emiliano S. Lopez
- Nanomedicine & VisionGroup; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Buenos Aires, Argentina
| | - Gustavo A. Ortiz
- Nanomedicine & VisionGroup; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Buenos Aires, Argentina
| | - Constanza Potilinski
- Nanomedicine & VisionGroup; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Buenos Aires, Argentina
| | - J. Oscar Croxatto
- Departamento de Patología Ocular, Fundación Oftalmológica Argentina “Jorge Malbran”, Buenos Aires, Argentina
| | - Juan E. Gallo
- Nanomedicine & VisionGroup; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Buenos Aires, Argentina
| |
Collapse
|
10
|
Lombardi M, Mantione ME, Baccellieri D, Ferrara D, Castellano R, Chiesa R, Alfieri O, Foglieni C. P2X7 receptor antagonism modulates IL-1β and MMP9 in human atherosclerotic vessels. Sci Rep 2017; 7:4872. [PMID: 28687781 PMCID: PMC5501842 DOI: 10.1038/s41598-017-05137-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022] Open
Abstract
In atherosclerosis, matrix metallopeptidases (MMPs) contribute to plaque rupture through weakening of the fibrous cap. Pleiotropic P2X purinoceptor 7 (P2X7), expressed in the carotid plaque (PL), is involved in interleukin 1 beta (IL-1β) release that may influence MMP9 generation, thus their possible modulation through acting on P2X7 was investigated. P2X7-related machinery was characterized and the effects of P2X7 antagonists (A740003, KN62) and MMPs inhibitors (Batimastat, Ro28-2653) were studied in ex-vivo tissue cultures of human PL’s vs. non-atherosclerotic internal mammary artery (IMA) by using molecular biology, immune-biochemical and microscopy methodologies. We highlighted atherosclerosis-related differences between PLs and IMAs molecular patterns, and their responsivity to P2X7 antagonism. High IL-1β tissue content was associated with PLs morphology and instability/vulnerability. We demonstrated that A740003, but not KN62, decreased IL-1β and MMP9 independently from NLR family pyrin domain containing 3, but in relationship with patient’s smoking status. Acting downstream P2X7 by MMPs inhibitors, diminished IL-1β mRNA without transcriptional effect at MMP9, possibly because the assumption of statin by patients. These data firstly demonstrated A740003 suitability as a specific tool to decrease inflammatory status in human vessels and might support the design of studies applying P2X7 antagonists for the local targeting and tailored therapy of atherosclerosis.
Collapse
Affiliation(s)
- Maria Lombardi
- Cardiovascular Research Area, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Maria Elena Mantione
- Cardiovascular Research Area, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Domenico Baccellieri
- Cardio-thoracic-vascular Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - David Ferrara
- Cardio-thoracic-vascular Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Renata Castellano
- Cardio-thoracic-vascular Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Roberto Chiesa
- Cardio-thoracic-vascular Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ottavio Alfieri
- Cardio-thoracic-vascular Department, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Chiara Foglieni
- Cardiovascular Research Area, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| |
Collapse
|
11
|
Ji Y, Adeola O, Strawn TL, Jeong SS, Chen R, Fay WP. Recombinant soluble apyrase APT102 inhibits thrombosis and intimal hyperplasia in vein grafts without adversely affecting hemostasis or re-endothelialization. J Thromb Haemost 2017; 15:814-825. [PMID: 28079982 PMCID: PMC5378664 DOI: 10.1111/jth.13621] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Indexed: 12/15/2022]
Abstract
Essentials New strategies are needed to inhibit thrombosis and intimal hyperplasia (IH) in vein grafts (VG). We studied effects of apyrase (APT102) on VGs and smooth muscle and endothelial cells (SMC/EC). APT102 inhibited thrombosis, SMC migration, and IH without impairing hemostasis or EC recovery. Apyrase APT102 is a single-drug approach to inhibit multiple processes that cause VG failure. SUMMARY Background Occlusion of vein grafts (VGs) after bypass surgery, owing to thrombosis and intimal hyperplasia (IH), is a major clinical problem. Apyrases are enzymes that scavenge extracellular ATP and ADP, and promote adenosine formation at sites of vascular injury, and hence have the potential to inhibit VG pathology. Objectives To examine the effects of recombinant soluble human apyrase, APT102, on platelets, smooth muscle cells (SMCs) and endothelial cells (ECs) in vitro, and on thrombosis and IH in murine VGs. Methods SMC and EC proliferation and migration were studied in vitro. Inferior vena cava segments from donor mice were grafted into carotid arteries of recipient mice. Results APT102 potently inhibited ADP-induced platelet aggregation and VG thrombosis, but it did not impair surgical hemostasis. APT102 did not directly inhibit SMC or EC proliferation, but significantly attenuated the effects of ATP on SMC and EC proliferation. APT102 significantly inhibited SMC migration, but did not inhibit EC migration, which may be mediated, at least in part, by inhibition of SMC, but not EC, migration by adenosine. At 4 weeks after surgery, there was significantly less IH in VGs of APT102-treated mice than in control VGs. APT102 significantly inhibited cell proliferation in VGs, but did not inhibit re-endothelialization. Conclusions Systemic administration of a recombinant human apyrase inhibits thrombosis and IH in VGs without increasing bleeding or compromising re-endothelialization. These results suggest that APT102 has the potential to become a novel, single-drug treatment strategy to prevent multiple pathologic processes that drive early adverse remodeling and occlusion of VGs.
Collapse
Affiliation(s)
- Y Ji
- Departments of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine and the Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - O Adeola
- Departments of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine and the Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - T L Strawn
- Departments of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine and the Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | | | - R Chen
- APT Therapeutics, St Louis, MO, USA
| | - W P Fay
- Departments of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine and the Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| |
Collapse
|
12
|
Menzies RI, Tam FW, Unwin RJ, Bailey MA. Purinergic signaling in kidney disease. Kidney Int 2016; 91:315-323. [PMID: 27780585 DOI: 10.1016/j.kint.2016.08.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/10/2016] [Accepted: 08/15/2016] [Indexed: 02/04/2023]
Abstract
Nucleotides are key subunits for nucleic acids and provide energy for intracellular metabolism. They can also be released from cells to act physiologically as extracellular messengers or pathologically as danger signals. Extracellular nucleotides stimulate membrane receptors in the P2 and P1 family. P2X are ATP-activated cation channels; P2Y and P1 are G-protein coupled receptors activated by ATP, ADP, UTP, and UDP in the case of P2 or adenosine for P1. Renal P2 receptors influence both vascular contractility and tubular function. Renal cells also express ectonucleotidases that rapidly hydrolyze extracellular nucleotides. These enzymes integrate this multireceptor purinergic-signaling complex by determining the nucleotide milieu to titrate receptor activation. Purinergic signaling also regulates immune cell function by modulating the synthesis and release of various cytokines such as IL1-β and IL-18 as part of inflammasome activation. Abnormal or excessive stimulation of this intricate paracrine system can be pro- or anti-inflammatory, and is also linked to necrosis and apoptosis. Kidney tissue injury causes a localized increase in ATP concentration, and sustained activation of P2 receptors can lead to renal glomerular, tubular, and vascular cell damage. Purinergic receptors also regulate the activity and proliferation of fibroblasts, promoting both inflammation and fibrosis in chronic disease. In this short review we summarize some of the recent findings related to purinergic signaling in the kidney. We focus predominantly on the P2X7 receptor, discussing why antagonists have so far disappointed in clinical trials and how advances in our understanding of purinergic signaling might help to reposition these compounds as potential treatments for renal disease.
Collapse
Affiliation(s)
- Robert I Menzies
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Frederick W Tam
- Imperial College Renal and Transplant Centre, Department of Medicine, Imperial College London, UK
| | - Robert J Unwin
- Cardiovascular and Metabolic Diseases Biotech Unit, AstraZeneca Gothenburg, Sweden; UCL Centre for Nephrology, University College London, London, UK.
| | - Matthew A Bailey
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
13
|
Avanzato D, Genova T, Fiorio Pla A, Bernardini M, Bianco S, Bussolati B, Mancardi D, Giraudo E, Maione F, Cassoni P, Castellano I, Munaron L. Activation of P2X7 and P2Y11 purinergic receptors inhibits migration and normalizes tumor-derived endothelial cells via cAMP signaling. Sci Rep 2016; 6:32602. [PMID: 27586846 PMCID: PMC5009337 DOI: 10.1038/srep32602] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/09/2016] [Indexed: 01/23/2023] Open
Abstract
Purinergic signaling is involved in inflammation and cancer. Extracellular ATP accumulates in tumor interstitium, reaching hundreds micromolar concentrations, but its functional role on tumor vasculature and endothelium is unknown. Here we show that high ATP doses (>20 μM) strongly inhibit migration of endothelial cells from human breast carcinoma (BTEC), but not of normal human microvascular EC. Lower doses (1–10 mm result ineffective. The anti-migratory activity is associated with cytoskeleton remodeling and is significantly prevented by hypoxia. Pharmacological and molecular evidences suggest a major role for P2X7R and P2Y11R in ATP-mediated inhibition of TEC migration: selective activation of these purinergic receptors by BzATP mimics the anti-migratory effect of ATP, which is in turn impaired by their pharmacological or molecular silencing. Downstream pathway includes calcium-dependent Adenilyl Cyclase 10 (AC10) recruitment, cAMP release and EPAC-1 activation. Notably, high ATP enhances TEC-mediated attraction of human pericytes, leading to a decrease of endothelial permeability, a hallmark of vessel normalization. Finally, we provide the first evidence of in vivo P2X7R expression in blood vessels of murine and human breast carcinoma. In conclusion, we have identified a purinergic pathway selectively acting as an antiangiogenic and normalizing signal for human tumor-derived vascular endothelium.
Collapse
Affiliation(s)
- D Avanzato
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy
| | - T Genova
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy
| | - A Fiorio Pla
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces Centre of Excellence (NIS), University of Torino, Torino, Italy
| | - M Bernardini
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy
| | - S Bianco
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy
| | - B Bussolati
- Dept. of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - D Mancardi
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - E Giraudo
- Candiolo Cancer Research Center, Torino, Italy
| | - F Maione
- Candiolo Cancer Research Center, Torino, Italy
| | - P Cassoni
- Department of Medical Sciences, Torino, Italy
| | | | - L Munaron
- Department of Life Sciences &Systems Biology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces Centre of Excellence (NIS), University of Torino, Torino, Italy
| |
Collapse
|
14
|
Galam L, Rajan A, Failla A, Soundararajan R, Lockey RF, Kolliputi N. Deletion of P2X7 attenuates hyperoxia-induced acute lung injury via inflammasome suppression. Am J Physiol Lung Cell Mol Physiol 2016; 310:L572-81. [PMID: 26747786 DOI: 10.1152/ajplung.00417.2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence shows that hyperoxia is a serious complication of oxygen therapy in acutely ill patients that causes excessive production of free radicals leading to hyperoxia-induced acute lung injury (HALI). Our previous studies have shown that P2X7 receptor activation is required for inflammasome activation during HALI. However, the role of P2X7 in HALI is unclear. The main aim of this study was to determine the effect of P2X7 receptor gene deletion on HALI. Wild-type (WT) and P2X7 knockout (P2X7 KO) mice were exposed to 100% O2 for 72 h. P2X7 KO mice treated with hyperoxia had enhanced survival in 100% O2 compared with the WT mice. Hyperoxia-induced recruitment of inflammatory cells and elevation of IL-1β, TNF-α, monocyte chemoattractant protein-1, and IL-6 levels were attenuated in P2X7 KO mice. P2X7 deletion decreased lung edema and alveolar protein content, which are associated with enhanced alveolar fluid clearance. In addition, activation of the inflammasome was suppressed in P2X7-deficient alveolar macrophages and was associated with suppression of IL-1β release. Furthermore, P2X7-deficient alveolar macrophage in type II alveolar epithelial cells (AECs) coculture model abolished protein permeability across mouse type II AEC monolayers. Deletion of P2X7 does not lead to a decrease in epithelial sodium channel expression in cocultures of alveolar macrophages and type II AECs. Taken together, these findings show that deletion of P2X7 is a protective factor and therapeutic target for the amelioration of hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Lakshmi Galam
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Ashna Rajan
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Athena Failla
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Ramani Soundararajan
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Richard F Lockey
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
15
|
Menzies RI, Howarth AR, Unwin RJ, Tam FWK, Mullins JJ, Bailey MA. Inhibition of the purinergic P2X7 receptor improves renal perfusion in angiotensin-II-infused rats. Kidney Int 2015; 88:1079-87. [PMID: 26108066 DOI: 10.1038/ki.2015.182] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/19/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
Chronic activation of the renin-angiotensin system promotes hypertension, renal microvascular dysfunction, tissue hypoxia, and inflammation. Despite similar hypertension, an injurious response to excess angiotensin II is greater in F344 than in Lewis rats; the latter displaying renoprotection. Here we studied whether p2rx7, encoding the P2X7 receptor (P2X7R), is a candidate gene for the differential susceptibility to vascular dysfunction under high angiotensin II tone. A 14-day infusion of angiotensin II into F344 rats increased blood pressure by about 15 mm Hg without inducing fibrosis or albuminuria. In vivo pressure natriuresis was suppressed, medullary perfusion reduced by half, and the corticomedullary oxygenation gradient disrupted. Selective P2X7R antagonism restored pressure natriuresis, promoting a significant leftward shift in the intercept and increasing the slope. Sodium excretion was increased sixfold and blood pressure normalized. The specific P2X7R antagonist AZ11657312 increased renal medullary perfusion, but only in angiotensin II-treated rats. Tissue oxygenation was improved by P2X7R blockade, particularly in poorly oxygenated regions of the kidney. Thus, activation of P2X7R induces microvascular dysfunction and regional hypoxia when angiotensin II is elevated and these effects may contribute to progression of renal injury induced by chronic angiotensin II.
Collapse
Affiliation(s)
- Robert I Menzies
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK.,Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Amelia R Howarth
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Robert J Unwin
- Cardiovascular and Metabolic Diseases (iMed CVMD) R&D, AstraZeneca, Mölndal, Sweden.,UCL Centre for Nephrology, University College London, London, UK
| | - Frederick W K Tam
- Imperial College Renal and Transplant Centre, Department of Medicine, Imperial College London, London, UK
| | - John J Mullins
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Matthew A Bailey
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
Wang S, Zhao J, Wang H, Liang Y, Yang N, Huang Y. Blockage of P2X7 attenuates acute lung injury in mice by inhibiting NLRP3 inflammasome. Int Immunopharmacol 2015; 27:38-45. [PMID: 25937482 PMCID: PMC7185518 DOI: 10.1016/j.intimp.2015.04.035] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 11/19/2022]
Abstract
NLRP3 inflammasome is engaged in the inflammatory response during acute lung injury (ALI). Purinergic receptor P2X7 has been reported to be upstream of NLRP3 activation. However, the therapeutic implication of P2X7 in ALI remains to be explored. The present study used lipopolysaccharide (LPS)-induced mouse model to investigate the therapeutic potential of P2X7 blockage in ALI. Our results showed that P2X7/NLRP3 inflammasome pathway was significantly upregulated in the lungs of ALI mice as compared with control mice. P2X7 antagonist A438079 suppressed NLRP3/ASC/caspase 1 activation, production of IL-1β, IL-17A and IFN-γ and neutrophil infiltration but not the production of IL-10, resulting in a significant amelioration of lung injury. Moreover, blockage of P2X7 significantly inhibited NLRP3 inflammasome activation and IL-1β production in bone marrow derived macrophages. Similar results were obtained using another P2X7 inhibitor brilliant blue G (BBG) in vivo. Thus, pharmacological blockage of P2X7/NLRP3 pathway can be considered as a potential therapeutic strategy in patients with ALI.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Jijun Zhao
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Hongyue Wang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Yingjie Liang
- Department of Pathology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Niansheng Yang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China
| | - Yuefang Huang
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, China.
| |
Collapse
|
17
|
Abstract
There are nineteen different receptor proteins for adenosine, adenine and uridine nucleotides, and nucleotide sugars, belonging to three families of G protein-coupled adenosine and P2Y receptors, and ionotropic P2X receptors. The majority are functionally expressed in blood vessels, as purinergic receptors in perivascular nerves, smooth muscle and endothelial cells, and roles in regulation of vascular contractility, immune function and growth have been identified. The endogenous ligands for purine receptors, ATP, ADP, UTP, UDP and adenosine, can be released from different cell types within the vasculature, as well as from circulating blood cells, including erythrocytes and platelets. Many purine receptors can be activated by two or more of the endogenous ligands. Further complexity arises because of interconversion between ligands, notably adenosine formation from the metabolism of ATP, leading to complex integrated responses through activation of different subtypes of purine receptors. The enzymes responsible for this conversion, ectonucleotidases, are present on the surface of smooth muscle and endothelial cells, and may be coreleased with neurotransmitters from nerves. What selectivity there is for the actions of purines/pyrimidines comes from differential expression of their receptors within the vasculature. P2X1 receptors mediate the vasocontractile actions of ATP released as a neurotransmitter with noradrenaline (NA) from sympathetic perivascular nerves, and are located on the vascular smooth muscle adjacent to the nerve varicosities, the sites of neurotransmitter release. The relative contribution of ATP and NA as functional cotransmitters varies with species, type and size of blood vessel, neuronal firing pattern, the tone/pressure of the blood vessel, and in ageing and disease. ATP is also a neurotransmitter in non-adrenergic non-cholinergic perivascular nerves and mediates vasorelaxation via smooth muscle P2Y-like receptors. ATP and adenosine can act as neuromodulators, with the most robust evidence being for prejunctional inhibition of neurotransmission via A1 adenosine receptors, but also prejunctional excitation and inhibition of neurotransmission via P2X and P2Y receptors, respectively. P2Y2, P2Y4 and P2Y6 receptors expressed on the vascular smooth muscle are coupled to vasocontraction, and may have a role in pathophysiological conditions, when purines are released from damaged cells, or when there is damage to the protective barrier that is the endothelium. Adenosine is released during hypoxia to increase blood flow via vasodilator A2A and A2B receptors expressed on the endothelium and smooth muscle. ATP is released from endothelial cells during hypoxia and shear stress and can act at P2Y and P2X4 receptors expressed on the endothelium to increase local blood flow. Activation of endothelial purine receptors leads to the release of nitric oxide, hyperpolarising factors and prostacyclin, which inhibits platelet aggregation and thus ensures patent blood flow. Vascular purine receptors also regulate endothelial and smooth muscle growth, and inflammation, and thus are involved in the underlying processes of a number of cardiovascular diseases.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | - William R Dunn
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
18
|
Burnstock G, Pelleg A. Cardiac purinergic signalling in health and disease. Purinergic Signal 2015; 11:1-46. [PMID: 25527177 PMCID: PMC4336308 DOI: 10.1007/s11302-014-9436-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 01/09/2023] Open
Abstract
This review is a historical account about purinergic signalling in the heart, for readers to see how ideas and understanding have changed as new experimental results were published. Initially, the focus is on the nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory nerves, as well as in intracardiac neurons. Control of the heart by centers in the brain and vagal cardiovascular reflexes involving purines are also discussed. The actions of adenine nucleotides and nucleosides on cardiomyocytes, atrioventricular and sinoatrial nodes, cardiac fibroblasts, and coronary blood vessels are described. Cardiac release and degradation of ATP are also described. Finally, the involvement of purinergic signalling and its therapeutic potential in cardiac pathophysiology is reviewed, including acute and chronic heart failure, ischemia, infarction, arrhythmias, cardiomyopathy, syncope, hypertrophy, coronary artery disease, angina, diabetic cardiomyopathy, as well as heart transplantation and coronary bypass grafts.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | |
Collapse
|
19
|
Shear stress modulates endothelial KLF2 through activation of P2X4. Purinergic Signal 2015; 11:139-53. [PMID: 25563726 DOI: 10.1007/s11302-014-9442-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial cells that are in direct contact with blood flow are exposed to fluid shear stress and regulate vascular homeostasis. Studies report endothelial cells to release ATP in response to shear stress that in turn modulates cellular functions via P2 receptors with P2X4 mediating shear stress-induced calcium signaling and vasodilation. A recent study shows that a loss-of-function polymorphism in the human P2X4 resulting in a Tyr315>Cys variant is associated with increased pulse pressure and impaired endothelial vasodilation. Although the importance of shear stress-induced Krüppel-like factor 2 (KLF2) expression in atheroprotection is well studied, whether ATP regulates KLF2 remains unanswered and is the objective of this study. Using an in vitro model, we show that in human umbilical vein endothelial cells (HUVECs), apyrase decreased shear stress-induced KLF2, KLF4, and NOS3 expression but not that of NFE2L2. Exposure of HUVECs either to shear stress or ATPγS under static conditions increased KLF2 in a P2X4-dependent manner as was evident with both the receptor antagonist and siRNA knockdown. Furthermore, transient transfection of static cultures of human endothelial cells with the Tyr315>Cys mutant P2X4 construct blocked ATP-induced KLF2 expression. Also, P2X4 mediated the shear stress-induced phosphorylation of extracellular regulated kinase-5, a known regulator of KLF2. This study demonstrates a major physiological finding that the shear-induced effects on endothelial KLF2 axis are in part dependent on ATP release and P2X4, a previously unidentified mechanism.
Collapse
|
20
|
Menzies RI, Unwin RJ, Bailey MA. Renal P2 receptors and hypertension. Acta Physiol (Oxf) 2015; 213:232-41. [PMID: 25345692 DOI: 10.1111/apha.12412] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/23/2014] [Accepted: 10/17/2014] [Indexed: 12/11/2022]
Abstract
The regulation of extracellular fluid volume is a key component of blood pressure homeostasis. Long-term blood pressure is stabilized by the acute pressure natriuresis response by which changes in renal perfusion pressure evoke corresponding changes in renal sodium excretion. A wealth of experimental evidence suggests that a defect in the pressure natriuresis response contributes to the development and maintenance of hypertension. The mechanisms underlying the relationship between renal perfusion pressure and sodium excretion are incompletely understood. Increased blood flow through the vasa recta increases renal interstitial hydrostatic pressure, thereby reducing the driving force for transepithelial sodium reabsorption. Paracrine signalling also contributes to the overall natriuretic response by inhibiting tubular sodium reabsorption in several nephron segments. In this brief review, we discuss the role of purinergic signalling in the renal control of blood pressure. ATP is released from renal tubule and vascular cells in response to increased flow and can activate P2 receptor subtypes expressed in both epithelial and vascular endothelial/smooth muscle cells. In concert, these effects integrate the vascular and tubular responses to increased perfusion pressure and targeting P2 receptors, particularly P2X7, may prove beneficial for treatment of hypertension.
Collapse
Affiliation(s)
- R. I. Menzies
- University/British Heart Foundation; Centre for Cardiovascular Science; The University of Edinburgh; Edinburgh UK
- MRC Institute for Genetics and Molecular Medicine; The University of Edinburgh; Edinburgh UK
| | - R. J. Unwin
- UCL Centre for Nephrology; University College London; London UK
| | - M. A. Bailey
- University/British Heart Foundation; Centre for Cardiovascular Science; The University of Edinburgh; Edinburgh UK
| |
Collapse
|
21
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
22
|
Menzies RI, Unwin RJ, Dash RK, Beard DA, Cowley AW, Carlson BE, Mullins JJ, Bailey MA. Effect of P2X4 and P2X7 receptor antagonism on the pressure diuresis relationship in rats. Front Physiol 2013; 4:305. [PMID: 24187541 PMCID: PMC3807716 DOI: 10.3389/fphys.2013.00305] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 10/03/2013] [Indexed: 12/31/2022] Open
Abstract
Reduced glomerular filtration, hypertension and renal microvascular injury are hallmarks of chronic kidney disease, which has a global prevalence of ~10%. We have shown previously that the Fischer (F344) rat has lower GFR than the Lewis rat, and is more susceptible to renal injury induced by hypertension. In the early stages this injury is limited to the pre-glomerular vasculature. We hypothesized that poor renal hemodynamic function and vulnerability to vascular injury are causally linked and genetically determined. In the present study, normotensive F344 rats had a blunted pressure diuresis relationship, compared with Lewis rats. A kidney microarray was then interrogated using the Endeavour enrichment tool to rank candidate genes for impaired blood pressure control. Two novel candidate genes, P2rx7 and P2rx4, were identified, having a 7− and 3− fold increased expression in F344 rats. Immunohistochemistry localized P2X4 and P2X7 receptor expression to the endothelium of the pre-glomerular vasculature. Expression of both receptors was also found in the renal tubule; however there was no difference in expression profile between strains. Brilliant Blue G (BBG), a relatively selective P2X7 antagonist suitable for use in vivo, was administered to both rat strains. In Lewis rats, BBG had no effect on blood pressure, but increased renal vascular resistance, consistent with inhibition of some basal vasodilatory tone. In F344 rats BBG caused a significant reduction in blood pressure and a decrease in renal vascular resistance, suggesting that P2X7 receptor activation may enhance vasoconstrictor tone in this rat strain. BBG also reduced the pressure diuresis threshold in F344 rats, but did not alter its slope. These preliminary findings suggest a physiological and potential pathophysiological role for P2X7 in controlling renal and/or systemic vascular function, which could in turn affect susceptibility to hypertension-related kidney damage.
Collapse
Affiliation(s)
- Robert I Menzies
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh Edinburgh, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Zamboulis DE, Senior JM, Clegg PD, Gallagher JA, Carter SD, Milner PI. Distribution of purinergic P2X receptors in the equine digit, cervical spinal cord and dorsal root ganglia. Purinergic Signal 2013; 9:383-93. [PMID: 23381684 PMCID: PMC3757141 DOI: 10.1007/s11302-013-9356-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 01/23/2013] [Indexed: 01/04/2023] Open
Abstract
Purinergic pathways are considered important in pain transmission, and P2X receptors are a key part of this system which has received little attention in the horse. The aim of this study was to identify and characterise the distribution of P2X receptor subtypes in the equine digit and associated vasculature and nervous tissue, including peripheral nerves, dorsal root ganglia and cervical spinal cord, using PCR, Western blot analysis and immunohistochemistry. mRNA signal for most of the tested P2X receptor subunits (P2X1-5, 7) was detected in all sampled equine tissues, whereas P2X6 receptor subunit was predominantly expressed in the dorsal root ganglia and spinal cord. Western blot analysis validated the specificity of P2X1-3, 7 antibodies, and these were used in immunohistochemistry studies. P2X1-3, 7 receptor subunits were found in smooth muscle cells in the palmar digital artery and vein with the exception of the P2X3 subunit that was present only in the vein. However, endothelial cells in the palmar digital artery and vein were positive only for P2X2 and P2X3 receptor subunits. Neurons and nerve fibres in the peripheral and central nervous system were positive for P2X1-3 receptor subunits, whereas glial cells were positive for P2X7 and P2X1 and 2 receptor subunits. This previously unreported distribution of P2X subtypes may suggest important tissue specific roles in physiological and pathological processes.
Collapse
Affiliation(s)
- D. E. Zamboulis
- />Faculty of Health and Life Sciences, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Chester High Road, Neston, Cheshire CH64 7TE UK
| | - J. M. Senior
- />Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Leahurst Campus, Neston, CH64 7TE UK
| | - P. D. Clegg
- />Faculty of Health and Life Sciences, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Chester High Road, Neston, Cheshire CH64 7TE UK
| | - J. A. Gallagher
- />Faculty of Health and Life Sciences, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L69 3GA UK
| | - S. D. Carter
- />Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE UK
| | - P. I. Milner
- />Faculty of Health and Life Sciences, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Chester High Road, Neston, Cheshire CH64 7TE UK
| |
Collapse
|
24
|
Mishra A. New insights of P2X7 receptor signaling pathway in alveolar functions. J Biomed Sci 2013; 20:26. [PMID: 23634990 PMCID: PMC3652760 DOI: 10.1186/1423-0127-20-26] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/29/2013] [Indexed: 12/20/2022] Open
Abstract
Purinergic P2X7 receptor (P2X7R), an ATP-gated cation channel, is unique among all other family members because of its ability to respond to various stimuli and to modulate pro-inflammatory signaling. The activation of P2X7R in immune cells is absolutely required for mature interleukin -1beta (IL-1beta) and IL-18 production and release. Lung alveoli are lined by the structural alveolar epithelial type I (AEC I) and alveolar epithelial type II cells (AEC II). AEC I plays important roles in alveolar barrier protection and fluid homeostasis whereas AEC II synthesizes and secrete surfactant and prevents alveoli from collapse. Earlier studies indicated that purinergic P2X7 receptors were specifically expressed in AEC I. However, their implication in alveolar functions has not been explored. This paper reviews two important signaling pathways of P2X7 receptors in surfactant homeostatsis and Acute Lung Injury (ALI). Thus, P2X7R resides at the critical nexus of alveolar pathophysiology.
Collapse
Affiliation(s)
- Amarjit Mishra
- National Institute of Health, 10 Center Dr, Bldg No, 10, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
|
26
|
Moccia F, Berra-Romani R, Tanzi F. Update on vascular endothelial Ca 2+ signalling: A tale of ion channels, pumps and transporters. World J Biol Chem 2012; 3:127-58. [PMID: 22905291 PMCID: PMC3421132 DOI: 10.4331/wjbc.v3.i7.127] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 02/05/2023] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca2+ signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca2+ levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca2+ signals, ranging from brief, localized Ca2+ pulses to prolonged Ca2+ oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca2+ signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca2+ releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca2+ removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca2+ machinery in vascular ECs under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesco Moccia
- Francesco Moccia, Franco Tanzi, Department of Biology and Biotechnologies "Lazzaro Spallanzani", Laboratory of Physiology, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | | | | |
Collapse
|
27
|
Klein K, Aeschlimann A, Jordan S, Gay R, Gay S, Sprott H. ATP induced brain-derived neurotrophic factor expression and release from osteoarthritis synovial fibroblasts is mediated by purinergic receptor P2X4. PLoS One 2012; 7:e36693. [PMID: 22715356 PMCID: PMC3360754 DOI: 10.1371/journal.pone.0036693] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 04/10/2012] [Indexed: 11/26/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a neuromodulator involved in nociceptive hypersensitivity in the central nervous system, is also expressed in synoviocytes of osteoarthritis (OA) and rheumatoid arthritis (RA) patients. We investigated the role of P2 purinoreceptors in the induction of BDNF expression in synovial fibroblasts (SF) of OA and RA patients. Cultured SF from patients with symptomatic knee OA and RA were stimulated with purinoreceptor agonists ATP, ADP, or UTP. The expression of BDNF mRNA was measured by quantitative TaqMan PCR. BDNF release into cell culture supernatants was monitored by ELISA. P2X4 expression in synovial tissue was detected by immunohistochemistry. Endogenous P2X4 expression was decreased by siRNA transfection before ATP stimulation. Kinase pathways were blocked before ATP stimulation. BDNF mRNA expression levels in OASF were increased 2 h and 5 h after ATP stimulation. Mean BDNF levels in cell culture supernatants of unstimulated OASF and RASF were 19 (±9) and 67 (±49) pg/ml, respectively. BDNF levels in SF supernatants were only elevated 5 h after ATP stimulation. BDNF mRNA expression in OASF was induced both by P2X receptor agonists ATP and ADP, but not by UTP, an agonist of P2Y purinergic receptors. The ATP-induced BDNF mRNA expression in OASF was decreased by siRNA-mediated reduction of endogenous P2X4 levels compared to scrambled controls. Inhibition of p38, but not p44/42 signalling reduced the ATP-mediated BDNF mRNA induction. Here we show a functional role of the purinergic receptor P2X4 and p38 kinase in the ATP-induced expression and release of the neurotrophin BDNF in SF.
Collapse
Affiliation(s)
- Kerstin Klein
- Center of Experimental Rheumatology, Division of Rheumatology and Institute of Physical Medicine, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | | | - Suzana Jordan
- Center of Experimental Rheumatology, Division of Rheumatology and Institute of Physical Medicine, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Renate Gay
- Center of Experimental Rheumatology, Division of Rheumatology and Institute of Physical Medicine, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Steffen Gay
- Center of Experimental Rheumatology, Division of Rheumatology and Institute of Physical Medicine, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Haiko Sprott
- Center of Experimental Rheumatology, Division of Rheumatology and Institute of Physical Medicine, University Hospital Zurich, Zurich, Switzerland
- Zurich Center of Integrative Human Physiology (ZIHP), Zurich, Switzerland
- * E-mail:
| |
Collapse
|
28
|
Kaczmarek-Hájek K, Lörinczi E, Hausmann R, Nicke A. Molecular and functional properties of P2X receptors--recent progress and persisting challenges. Purinergic Signal 2012; 8:375-417. [PMID: 22547202 PMCID: PMC3360091 DOI: 10.1007/s11302-012-9314-7] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 10/18/2011] [Indexed: 12/16/2022] Open
Abstract
ATP-gated P2X receptors are trimeric ion channels that assemble as homo- or heteromers from seven cloned subunits. Transcripts and/or proteins of P2X subunits have been found in most, if not all, mammalian tissues and are being discovered in an increasing number of non-vertebrates. Both the first crystal structure of a P2X receptor and the generation of knockout (KO) mice for five of the seven cloned subtypes greatly advanced our understanding of their molecular and physiological function and their validation as drug targets. This review summarizes the current understanding of the structure and function of P2X receptors and gives an update on recent developments in the search for P2X subtype-selective ligands. It also provides an overview about the current knowledge of the regulation and modulation of P2X receptors on the cellular level and finally on their physiological roles as inferred from studies on KO mice.
Collapse
Affiliation(s)
- Karina Kaczmarek-Hájek
- Max Planck Institute for Experimental Medicine, Hermann Rein Str. 3, 37075, Göttingen, Germany
| | | | | | | |
Collapse
|
29
|
Gündüz D, Aslam M, Krieger U, Becker L, Grebe M, Arshad M, Sedding DG, Härtel FV, Abdallah Y, Piper HM, Voss RK, Noll T. Opposing effects of ATP and adenosine on barrier function of rat coronary microvasculature. J Mol Cell Cardiol 2012; 52:962-70. [PMID: 22266063 DOI: 10.1016/j.yjmcc.2012.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 12/07/2011] [Accepted: 01/05/2012] [Indexed: 11/25/2022]
Abstract
ATP can differentially affect the micro- and macrovascular endothelial barrier. It has been shown that it can both increase and/or decrease macromolecule permeability of microvascular endothelial cells and microvessels, in vivo. We hypothesised that the barrier stabilising effect is mediated by ATP itself via P2 receptors, while barrier-disrupting effect is mediated by its metabolite adenosine via adenosine receptors. The effects of ATP, ADP, AMP and adenosine on barrier function were studied in cultured rat coronary microvascular endothelial monolayers (RCEC) in vitro, as well as in rat mesentery vessels, and in rat hearts in vivo. ATP and ADP showed a biphasic effect on permeability of RCEC monolayers with a reduction followed by a later increase in albumin permeability. The permeability decreasing effect of ATP was enhanced by ecto-nucleotidase inhibitor ARL67156 while permeability increasing effect was enhanced by apyrase, an extracellular ecto-nucleotidase. Moreover, the permeability increasing effect was abrogated by adenosine receptor antagonists, 8-phenyltheophylline (8-PT) and DMPX. Adenosine and adenosine receptor agonists 5'-(N-ethylcarboxamido)-adenosine (NECA), CGS21680, and R-PIA enhanced albumin permeability which was antagonised by 8-PT, A(1), and A(2) but not by A(3) receptor antagonists. Likewise, immunofluorescence microscopy of VE-cadherin and actin showed that NECA induces a disturbance of intercellular junctions. Pre-incubation of ATP antagonised the effects of NECA on permeability, actin cytoskeleton and intercellular junctions. Similar effects of the applied substances were observed in rat mesentery artery by determining the vascular leakage using intravital microscopy as well as in rat hearts by assessing myocardial water contents in vivo. In conclusion, the study demonstrates that in RCEC, ATP, ADP, and its metabolite adenosine play opposing roles on endothelial barrier function.
Collapse
Affiliation(s)
- Dursun Gündüz
- Zentrum für Innere Medizin, Abteilung Kardiologie/Angiologie, Universitätsklinikum Giessen und Marburg, Geissen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Stokes L, Scurrah K, Ellis JA, Cromer BA, Skarratt KK, Gu BJ, Harrap SB, Wiley JS. A Loss-of-Function Polymorphism in the Human P2X4 Receptor Is Associated With Increased Pulse Pressure. Hypertension 2011; 58:1086-92. [DOI: 10.1161/hypertensionaha.111.176180] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The P2X4 receptor is involved in endothelium-dependent changes in large arterial tone in response to shear stress and is, therefore, potentially relevant to arterial compliance and pulse pressure. Four identified nonsynonymous polymorphisms in
P2RX4
were reproduced in recombinantly expressed human P2X4. Electrophysiological studies showed that one of these, the Tyr315>Cys mutation (rs28360472), significantly reduced the peak amplitude of the ATP-induced inward current to 10.9% of wild-type P2X4 receptors in transfected HEK-293 cells (10 μmol/L of ATP; n=4–8 cells;
P
<0.001). Concentration-response curves for ATP and the partial agonist BzATP demonstrate that the 315Cys-P2X4 mutant had an increased EC
50
value for both ligands. Mutation of Tyr315>Cys likely disrupts the agonist binding site of P2X4 receptors, a finding supported by molecular modeling based on the zebrafish P2X4 receptor crystal structure. We tested inheritance of rs28360472 encoding the Tyr315>Cys mutation in
P2RX4
against pulse pressure in 2874 subjects from the Victorian Family Heart Study. The minor allele frequency was 0.014 (1.4%). In a variance components analysis we found significant association with pulse pressure (
P
=0.023 for total association) where 1 minor allele increased pulse pressure by 2.84 mm Hg (95% CI: 0.41–5.27). This increase in pulse pressure associated with inheritance of 315Cys-P2X4 receptors might reflect reduced large arterial compliance as a result of impaired endothelium-dependent vasodilation in large arteries.
Collapse
Affiliation(s)
- Leanne Stokes
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Katrina Scurrah
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Justine A. Ellis
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Brett A. Cromer
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Kristen K. Skarratt
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Ben J. Gu
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - Stephen B. Harrap
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| | - James S. Wiley
- From the Sydney Medical School Nepean (L.S., K.K.S., B.J.G., J.S.W.), University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia; Centre for Molecular, Environmental, Genetic and Analytic Epidemiology (K.S.), Florey Neurosciences Institute (B.A.C., B.J.G., J.S.W.), and Department of Physiology (J.A.E., S.B.H.), University of Melbourne, Melbourne, Victoria, Australia; Murdoch Childrens Research Institute (J.A.E.), Parkville, Victoria, Australia; Health Innovations Research Institute
| |
Collapse
|
31
|
Wu T, Dai M, Shi XR, Jiang ZG, Nuttall AL. Functional expression of P2X4 receptor in capillary endothelial cells of the cochlear spiral ligament and its role in regulating the capillary diameter. Am J Physiol Heart Circ Physiol 2011; 301:H69-78. [PMID: 21460192 DOI: 10.1152/ajpheart.01035.2010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cochlear lateral wall generates the endocochlear potential (EP), which creates a driving force for the hair cell transduction current and is essential for normal hearing. Blood flow at the cochlear lateral wall is critically important for maintaining the EP. The vulnerability of the EP to hypoxia suggests that the blood flow in the cochlear lateral wall is dynamically and precisely regulated to meet the changing metabolic needs of the cochlear lateral wall. It has been reported that ATP, an important extracellular signaling molecule, plays an essential role in regulating cochlear blood flow. However, the cellular mechanism underlying ATP-induced regional blood flow changes has not been investigated. In the current study, we demonstrate that 1) the P2X4 receptor is expressed in endothelial cells (ECs) of spiral ligament (SL) capillaries. 2) ATP elicits a characteristic current through P2X4 on ECs in a dose-dependent manner (EC(50) = 0.16 mM). The ATP current has a reversal potential at ∼0 mV; is inhibited by 5-(3-bromophenyl)-1,3-dihydro-2H-benzofuro[3,2-e]-1,4-diazepin-2-one (5-BDBD), LaCl(3), pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid) tetrasodium salt hydrate (PPADS), and extracellular acidosis; and is less sensitive to α,β-methyleneadenosine 5'-triphosphate (α,β-MeATP) and 2'- and 3'-O-(4-benzoyl-benzoyl) adenosine 5'-triphosphate (BzATP). 3) ATP elicits a transient increase of intracellular Ca(2+) in ECs. 4) In accordance with the above in vitro findings, perilymphatic ATP (1 mM) caused dilation in SL capillaries in vivo by 11.5%. N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME), a nonselective inhibitor of nitric oxide synthase, or 5-BDBD, the specific P2X4 inhibitor, significantly blocked the dilation. These findings support our hypothesis that extracellular ATP regulates cochlear lateral blood flow through P2X4 activation in ECs.
Collapse
Affiliation(s)
- T Wu
- Oregon Hearing Research Center, NRC04, Oregon Health & Science Univ., 3181 S.W. Sam Jackson Park Rd., Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
32
|
|
33
|
|
34
|
Faria M, Magalhães-Cardoso T, Lafuente-de-Carvalho JM, Correia-de Sá P. Decreased ecto-NTPDase1/CD39 activity leads to desensitization of P2 purinoceptors regulating tonus of corpora cavernosa in impotent men with endothelial dysfunction. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 27:761-8. [PMID: 18600538 DOI: 10.1080/15257770802145744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Vascular responses to adenine nucleotides in human corpora cavernosa from men with vasculogenic erectile dysfunction were investigated. We also evaluated the catabolism of extracellular adenine nucleotides to probe its relevance to vascular hemodynamics in impotent men. Human corpora cavernosa have high NTPDase1/CD39 activity, converting ATP directly into AMP, without significant ADP formation. Extracellular ATP hydrolysis is slower in impotent patients. Adenine nucleotides have dual roles on phenylephrine-contracted strips of corpora cavernosa operated by P2X-contractant and P2Y-relaxant receptors. Prolonged exposure to endogenous ATP related to decreased NTPDase1/CD39 activity leads to P2-purinoceptor desensitization in impotent men. Shutting down ATP signaling in vasculogenic impotent men may represent a defense mechanism for preventing purinergic overstimulation.
Collapse
Affiliation(s)
- M Faria
- Laboratório de Farmacologia e Neurobiologia/UMIB, Instituto de Ciências Biomédicas Abel Salazar-Universidade do Porto (ICBAS-UP), Porto, Poturgal
| | | | | | | |
Collapse
|
35
|
Palomino-Doza J, Rahman TJ, Avery PJ, Mayosi BM, Farrall M, Watkins H, Edwards CRW, Keavney B. Ambulatory blood pressure is associated with polymorphic variation in P2X receptor genes. Hypertension 2008; 52:980-5. [PMID: 18852390 DOI: 10.1161/hypertensionaha.108.113282] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The P2X receptor gene family encodes a series of proteins that function as ATP-gated nonselective ion channels. P2X receptor channels are involved in transducing aldosterone-mediated signaling in the distal renal tubule and are potential candidate genes for blood pressure regulation. We performed a family based quantitative genetic association study in 248 families ascertained through a proband with hypertension to investigate the relationship between common genetic variation in the P2X4, P2X6, and P2X7 genes and ambulatory blood pressure. We genotyped 28 single nucleotide polymorphisms, which together captured the common genetic variability in the 3 genes. We corrected our results for multiple comparisons specifying a false discovery rate of 5%. We found significant evidence of association between the single nucleotide polymorphism rs591874 in the first intron of the P2X7 gene and blood pressure. The strongest association was found for nighttime diastolic blood pressure (P=0.0032), although association was present for both systolic and diastolic blood pressures measured by an observer during the day and at night. Genotypes were associated with a 0.2 SD ( approximately 2.5 mm Hg) difference in night diastolic blood pressure per allele and accounted for approximately 1% of the total variability in this measurement. Other suggestive associations were found, but these were nonsignificant after correction for multiple testing. These genetic data suggest that drugs affecting P2X receptor signaling may have promise as clinical antihypertensive agents.
Collapse
Affiliation(s)
- Julian Palomino-Doza
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Seiffert K, Ding W, Wagner JA, Granstein RD. ATPgammaS enhances the production of inflammatory mediators by a human dermal endothelial cell line via purinergic receptor signaling. J Invest Dermatol 2006; 126:1017-27. [PMID: 16410784 DOI: 10.1038/sj.jid.5700135] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenosine 5'-triphosphate (ATP) affects multiple intra- and extracellular processes, including vascular tone and immune responses. Microvascular endothelial cells (EC) play a central role in inflammation by recruitment of inflammatory cells from blood to tissues. We hypothesized that ATP (secreted by neurons and/or released after perturbation of cutaneous cells) may influence secretion of inflammatory messengers by dermal microvascular EC through actions on purinergic P2 receptors. Addition of the hydrolysis-resistant ATP analogue, adenosine 5'-O-(3-thiotriphosphate) (ATPgammaS), to subconfluent cultures of the human microvascular endothelial cell-1 (HMEC-1) cell line led to a dose- and time-dependent increase in release of IL-6, IL-8, monocyte chemoattractant protein-1, and growth-regulated oncogene alpha. Both ATPgammaS-induced release and basal production of these proteins were significantly inhibited by the purinergic antagonists pyridoxal-5'-phosphate-6-azophenyl-2',5'-disulfonic acid (PPADS), pyridoxal-5'-phosphate-6(2'-naphthylazo-6-nitro-4',8'-disulfonate), and suramin. ATPgammaS increased expression of intercellular adhesion molecule-1 (ICAM-1), whereas suramin and PPADS decreased both ATPgammaS-induced and basal ICAM-1 expression. Using PCR, we found that HMEC-1 strongly express mRNA for the P2X(4), P2X(5), P2X(7), P2Y(2), and P2Y(11) receptors and weakly express mRNA for P2X(1) and P2X(3) receptors. Purinergic nucleotides may mediate acute inflammation in the skin and thus contribute to physiological and pathophysiological inflammation. For example, ATP may contribute to both the vasodilation and the inflammation associated with rosacea.
Collapse
Affiliation(s)
- Kristina Seiffert
- Department of Dermatology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021 USA
| | | | | | | |
Collapse
|
37
|
Abstract
The concept of a purinergic signaling system, using purine nucleotides and nucleosides as extracellular messengers, was first proposed over 30 years ago. After a brief introduction and update of purinoceptor subtypes, this article focuses on the diverse pathophysiological roles of purines and pyrimidines as signaling molecules. These molecules mediate short-term (acute) signaling functions in neurotransmission, mechanosensory transduction, secretion and vasodilatation, and long-term (chronic) signaling functions in cell proliferation, differentiation, and death involved in development and regeneration. Plasticity of purinoceptor expression in pathological conditions is frequently observed, including an increase in the purinergic component of autonomic cotransmission. Recent advances in therapies using purinergic-related drugs in a wide range of pathological conditions will be addressed with speculation on future developments in the field.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, Royal Free and University College Medical School, London NW3 2PF, UK.
| |
Collapse
|
38
|
|
39
|
Judkins CP, Sobey CG, Dang TT, Miller AA, Dusting GJ, Drummond GR. NADPH-induced contractions of mouse aorta do not involve NADPH oxidase: a role for P2X receptors. J Pharmacol Exp Ther 2006; 317:644-50. [PMID: 16407465 DOI: 10.1124/jpet.105.096610] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Reactive oxygen species elicit vascular effects ranging from acute dilatation because of hydrogen peroxide-mediated opening of K(+) channels to contraction arising from superoxide-dependent inactivation of endothelium-derived nitric oxide. Given that NADPH oxidases are major sources of superoxide in the vascular wall, this study examined the effects of exogenous NADPH, a substrate of these enzymes, on superoxide generation and isometric tone in mouse isolated aortic rings. NADPH caused concentration-dependent increases in superoxide generation (measured by lucigenin-enhanced chemiluminescence) and vascular tone (isometric tension recordings). However, surprisingly, whereas oxidized NADP(+) was unable to support superoxide production, it was equally as effective as reduced NADPH at stimulating vasocontraction. In addition, an NADPH oxidase inhibitor, diphenyleneiodonium, markedly attenuated NADPH-induced superoxide production, yet had no effect on vasocontractions to NADPH. In contrast, a broad specificity P2X receptor antagonist, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid, as well as the P2X1 selective antagonist, NF023, markedly attenuated both endothelium-dependent and -independent vasocontractions to NADPH, as did the P2X-desensitizing agent alpha,beta-methylene-ATP. Importantly, alpha,beta-methylene-ATP had no effect on superoxide production induced by NADPH. In conclusion, these findings suggest little role for NADPH oxidase-derived superoxide in the contractile effects of NADPH in the mouse aorta. Rather, NADPH seems to act as an agonist at two distinct P2X receptor populations; one located on the endothelium and the other on smooth muscle layer, both of which ultimately lead to contraction.
Collapse
Affiliation(s)
- Courtney P Judkins
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | |
Collapse
|
40
|
Yamamoto K, Sokabe T, Matsumoto T, Yoshimura K, Shibata M, Ohura N, Fukuda T, Sato T, Sekine K, Kato S, Isshiki M, Fujita T, Kobayashi M, Kawamura K, Masuda H, Kamiya A, Ando J. Impaired flow-dependent control of vascular tone and remodeling in P2X4-deficient mice. Nat Med 2005; 12:133-7. [PMID: 16327800 DOI: 10.1038/nm1338] [Citation(s) in RCA: 254] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 11/03/2005] [Indexed: 12/31/2022]
Abstract
The structure and function of blood vessels adapt to environmental changes such as physical development and exercise. This phenomenon is based on the ability of the endothelial cells to sense and respond to blood flow; however, the underlying mechanisms remain unclear. Here we show that the ATP-gated P2X4 ion channel, expressed on endothelial cells and encoded by P2rx4 in mice, has a key role in the response of endothelial cells to changes in blood flow. P2rx4(-/-) mice do not have normal endothelial cell responses to flow, such as influx of Ca(2+) and subsequent production of the potent vasodilator nitric oxide (NO). Additionally, vessel dilation induced by acute increases in blood flow is markedly suppressed in P2rx4(-/-) mice. Furthermore, P2rx4(-/-) mice have higher blood pressure and excrete smaller amounts of NO products in their urine than do wild-type mice. Moreover, no adaptive vascular remodeling, that is, a decrease in vessel size in response to a chronic decrease in blood flow, was observed in P2rx4(-/-) mice. Thus, endothelial P2X4 channels are crucial to flow-sensitive mechanisms that regulate blood pressure and vascular remodeling.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Animals
- Blood Pressure
- Blood Vessels/pathology
- Blotting, Northern
- Calcium/metabolism
- Carotid Arteries/pathology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Transfer Techniques
- Green Fluorescent Proteins/metabolism
- Immunohistochemistry
- Mesenteric Arteries/pathology
- Mice
- Mice, Transgenic
- Microscopy, Fluorescence
- Models, Biological
- NG-Nitroarginine Methyl Ester/pharmacology
- Nitric Oxide/metabolism
- Polymerase Chain Reaction
- Receptors, Purinergic P2/genetics
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2X4
- Regional Blood Flow
- Time Factors
Collapse
Affiliation(s)
- Kimiko Yamamoto
- Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Slater M, Barden JA. Differentiating keratoacanthoma from squamous cell carcinoma by the use of apoptotic and cell adhesion markers. Histopathology 2005; 47:170-8. [PMID: 16045778 DOI: 10.1111/j.1365-2559.2005.02155.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Keratoacanthomas (KA) are well-differentiated squamoproliferative skin lesions that grow rapidly and regress spontaneously. In contrast, squamous cell carcinomas (SCC) can have variable differentiation, inexorably progress and on occasion metastasize. Distinguishing between KA and SCC using haematoxylin and eosin-stained sections from an initial biopsy can often be difficult. There is also some debate as to whether KA is simply a variety of well-differentiated SCC or a distinct entity. METHODS AND RESULTS Initial biopsy sections from 25 cases of SCC and 20 of KA were labelled with markers for both the initiation (the cytolytic receptor P2X7) and end-stage (terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling) of apoptosis, telomerase-associated protein (TP1) and the cell adhesion protein E-cadherin. As this was a retrospective study, the clinical outcome of each case was known. This resulted in a unique labelling pattern of each marker for SCC and KA, allowing a differential diagnosis between the two conditions. The simplest marker to use for this purpose was anti-P2X7. Sections from five cases that were initially very difficult to diagnose were correctly identified as SCC using this method. CONCLUSIONS These results support the view that KA has a different pathogenesis and biochemistry from that of SCC, and is a distinct entity. Anti-P2X7 labelling, using routine immunohistochemical techniques, provides a method for differentially diagnosing these conditions.
Collapse
Affiliation(s)
- M Slater
- School of Biomedical Sciences, Department of Anatomy and Histology, The University of Sydney, Sydney, NSW 2006, Australia.
| | | |
Collapse
|
42
|
Kolosova IA, Mirzapoiazova T, Adyshev D, Usatyuk P, Romer LH, Jacobson JR, Natarajan V, Pearse DB, Garcia JGN, Verin AD. Signaling pathways involved in adenosine triphosphate-induced endothelial cell barrier enhancement. Circ Res 2005; 97:115-24. [PMID: 15994434 DOI: 10.1161/01.res.0000175561.55761.69] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Endothelial barrier dysfunction caused by inflammatory agonists is a frequent underlying cause of vascular leak and edema. Novel strategies to preserve barrier integrity could have profound clinical impact. Adenosine triphosphate (ATP) released from endothelial cells by shear stress and injury has been shown to protect the endothelial barrier in some settings. We have demonstrated that ATP and its nonhydrolyzed analogues enhanced barrier properties of cultured endothelial cell monolayers and caused remodeling of cell-cell junctions. Increases in cytosolic Ca2+ and Erk activation caused by ATP were irrelevant to barrier enhancement. Experiments using biochemical inhibitors or siRNA indicated that G proteins (specifically Galphaq and Galphai2), protein kinase A (PKA), and the PKA substrate vasodilator-stimulated phosphoprotein were involved in ATP-induced barrier enhancement. ATP treatment decreased phosphorylation of myosin light chain and specifically activated myosin-associated phosphatase. Depletion of Galphaq with siRNA prevented ATP-induced activation of myosin phosphatase. We conclude that the mechanisms of ATP-induced barrier enhancement are independent of intracellular Ca2+, but involve activation of myosin phosphatase via a novel G-protein-coupled mechanism and PKA.
Collapse
Affiliation(s)
- Irina A Kolosova
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Adenosine triphosphate (ATP) is now established as a principle vaso-active mediator in the vasculature. Its actions on arteries are complex, and are mediated by the P2X and P2Y receptor families. It is generally accepted that ATP induces a bi-phasic response in arteries, inducing contraction via the P2X and P2Y receptors on the smooth muscle cells, and vasodilation via the actions of P2Y receptors located on the endothelium. However, a number of recent studies have placed P2X1 receptors on the endothelium of some arteries. The use of a specific P2X1 receptor ligand, alpha, beta methylene ATP has demonstrated that P2X1 receptors also have a bi-functional role. The actions of ATP on P2X1 receptors is therefore dependant on its location, inducing contraction when located on the smooth muscle cells, and dilation when expressed on the endothelium, comparable to that of P2Y receptors.
Collapse
Affiliation(s)
- L S Harrington
- Unit of Critical Care, NHLI, Imperial College, London, UK.
| | | |
Collapse
|
44
|
Lemon G, Brockhausen J, Li GH, Gibson WG, Bennett MR. Calcium mobilization and spontaneous transient outward current characteristics upon agonist activation of P2Y2 receptors in smooth muscle cells. Biophys J 2005; 88:1507-23. [PMID: 15556987 PMCID: PMC1305209 DOI: 10.1529/biophysj.104.043976] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2004] [Accepted: 08/12/2004] [Indexed: 11/18/2022] Open
Abstract
A quantitative model is provided that links the process of metabotropic receptor activation and sequestration to the generation of inositol 1,4,5-trisphosphate, the subsequent release of calcium from the central sarcoplasmic reticulum, and the consequent release of calcium from subsarcolemma sarcoplasmic reticulum that acts on large-conductance potassium channels to generate spontaneous transient outward currents (STOCs). This model is applied to the case of STOC generation in vascular A7r5 smooth muscle cells that have been transfected with a chimera of the P2Y(2) metabotropic receptor and green fluorescent protein (P2Y(2)-GFP) and exposed to the P2Y(2) receptor agonist uridine 5'-triphosphate. The extent of P2Y(2)-GFP sequestration from the membrane on exposure to uridine 5'-triphosphate, the ensuing changes in cytosolic calcium concentration, as well as the interval between STOCs that are subsequently generated, are used to determine parameter values in the model. With these values, the model gives a good quantitative prediction of the dynamic changes in STOC amplitude observed upon activation of metabotropic P2Y(2) receptors in the vascular smooth muscle cell line.
Collapse
MESH Headings
- Animals
- Biological Transport, Active/drug effects
- Biological Transport, Active/physiology
- Calcium/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Cell Line
- Computer Simulation
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Models, Biological
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Purinergic P2 Receptor Agonists
- Receptors, Purinergic P2/metabolism
- Receptors, Purinergic P2Y2
- Uridine Triphosphate/pharmacology
Collapse
Affiliation(s)
- G Lemon
- The School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|
45
|
Sylte MJ, Kuckleburg CJ, Inzana TJ, Bertics PJ, Czuprynski CJ. Stimulation of P2X receptors enhances lipooligosaccharide-mediated apoptosis of endothelial cells. J Leukoc Biol 2005; 77:958-65. [PMID: 15728716 DOI: 10.1189/jlb.1004597] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Exposure of endothelial cells to lipid A-containing molecules, such as lipopolysaccharide (LPS) or lipooligosaccharide (LOS), causes the release of purinergic compounds [e.g., adenosine 5'-triphosphate (ATP)] and can lead to apoptosis. The P2X family of purinergic receptors (e.g., P2X(7)) has been reported to modulate LPS signaling events and to participate in apoptosis. We investigated the role that P2X receptors play in the apoptosis that follows exposure of bovine endothelial cells to Haemophilus somnus LOS. Addition of P2X inhibitors, such as periodate-oxidized ATP (oATP) or pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid tetrasodium, significantly reduced LOS-induced apoptosis. Incubation of endothelial cells with apyrase, which degrades ATP, diminished LOS-induced apoptosis of endothelial cells. Concomitant addition of P2X agonists [e.g., 2',3'-(4-benzoyl)-benzoyl ATP or ATP] to LOS-treated endothelial cells significantly enhanced caspase-3 activation. The P2X antagonist oATP significantly blocked caspase-8 but not caspase-9 activation in LOS-treated endothelial cells. Together, these data indicate that stimulation of P2X receptors enhances LOS-induced apoptosis of endothelial cells, possibly as a result of endogenous release of ATP, which results in caspase-8 activation.
Collapse
Affiliation(s)
- Matt J Sylte
- Department of Pathobiological Sciences, School of Veterinary Medicine, Madison, WI 63706, USA
| | | | | | | | | |
Collapse
|
46
|
Harrington LS, Mitchell JA. Novel role for P2X receptor activation in endothelium-dependent vasodilation. Br J Pharmacol 2004; 143:611-7. [PMID: 15466440 PMCID: PMC1575439 DOI: 10.1038/sj.bjp.0706004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1 ATP is an important vasoactive mediator, which acts via two receptor classes: P2X and P2Y. Activation of P2X receptors has traditionally been associated with the well-characterised vasoconstrictor properties of ATP. 2 In the current study, we have shown that the P2X(1 & 3) receptor ligand, alpha, beta methylene ATP, induces vasodilation of rat isolated mesenteric arteries and that P2X1 receptors are abundantly expressed in the endothelium of these vessels. 3 Second-order rat mesenteric arteries were mounted in myographs and vasomotor responses recorded. Both ATP and alpha, beta methylene ATP induced a constriction followed by a vasodilation. The dilator effects of either ATP or alpha, beta methylene ATP were slower in onset than those induced by acetylcholine. By contrast, the traditional vasodilator P2Y ligand, ADP, induced vasodilation without contraction. 4 Vasodilation induced by alpha, beta methylene ATP was endothelial dependent, but was not affected by treatment of the vessels with L-NAME plus indomethacin alone. Dilation was, however, partially inhibited by the combination of apamin plus charybdotoxin and blocked by treating vessels with all four drugs. 5 Using confocal microscopy, P2X1 receptor immunoreactivity was localised to the endothelial, smooth muscle and adventitial layers of mesenteric vessels. P2X1 protein migrated as a primary band at around 50-60 kDa in vascular tissue. 6 These results show for the first time that P2X1 receptors are expressed on the endothelium and that a selective ligand of this receptor results in vasoconstriction followed by vasodilation. These observations have important implications for our understanding of the role of purines in biological responses.
Collapse
Affiliation(s)
- Louise S Harrington
- Unit of Critical Care Medicine, Royal Brompton Hospital, The National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, London, SW3 6LY
| | - Jane A Mitchell
- Unit of Critical Care Medicine, Royal Brompton Hospital, The National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, London, SW3 6LY
- Author for correspondence:
| |
Collapse
|
47
|
Glass CA, Bates DO. The role of endothelial cell Ca2+store release in the regulation of microvascular permeabilityin vivo. Exp Physiol 2004; 89:343-51. [PMID: 15123553 DOI: 10.1113/expphysiol.2003.026948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Microvascular permeability is regulated by changes in intracellular calcium concentration. The mechanism by which this increase in calcium determines permeability under normal conditions and during stimulation with agonists remains to be elucidated. In order to determine whether calcium release from intracellular stores could contribute towards the regulation of vascular permeability, hydraulic conductivity (Lp) was measured in frog mesenteric microvessels during stimulation of the endothelial cells of these vessels with agonists that release calcium from the intracellular stores. ATP (which acts through activation of inositol 1,4,5-trisphosphate (IP3) receptors) increased Lp in the absence of calcium influx across the plasma membrane 2.3 +/- 0.3 fold (mean +/- s.e.m., P < 0.01, n = 8), which was less than the increase in the presence of calcium influx (3.1 +/- 1.1 fold). Caffeine (which acts through activation of ryanodine receptors) also increased Lp in the absence of calcium influx across the plasma membrane 3.8 +/- 1.0 fold (P < 0.01, n = 9), but by at least as much as it does in the presence of calcium influx (2.8 +/- 0.5 fold). It is surprising that there was a strong positive correlation between the size of the response during store release and the baseline permeability (r = 0.91 for ATP, r = 0.75 for caffeine). This suggests that the filling state of the stores may regulate the baseline permeability of the microvessels.
Collapse
Affiliation(s)
- C A Glass
- Microvascular Research Laboratories, Department of Physiology, Preclinical Veterinary School, Southwell Street, University of Bristol, Bristol BS2 8EJ, UK.
| | | |
Collapse
|
48
|
Burnstock G, Knight GE. Cellular Distribution and Functions of P2 Receptor Subtypes in Different Systems. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 240:31-304. [PMID: 15548415 DOI: 10.1016/s0074-7696(04)40002-3] [Citation(s) in RCA: 584] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review is aimed at providing readers with a comprehensive reference article about the distribution and function of P2 receptors in all the organs, tissues, and cells in the body. Each section provides an account of the early history of purinergic signaling in the organ?cell up to 1994, then summarizes subsequent evidence for the presence of P2X and P2Y receptor subtype mRNA and proteins as well as functional data, all fully referenced. A section is included describing the plasticity of expression of P2 receptors during development and aging as well as in various pathophysiological conditions. Finally, there is some discussion of possible future developments in the purinergic signaling field.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Institute, Royal Free and University College Medical School, London NW3 2PF, United Kingdom
| | | |
Collapse
|
49
|
Schwiebert EM, Zsembery A. Extracellular ATP as a signaling molecule for epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1615:7-32. [PMID: 12948585 DOI: 10.1016/s0005-2736(03)00210-4] [Citation(s) in RCA: 346] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The charge of this invited review is to present a convincing case for the fact that cells release their ATP for physiological reasons. Many of our "purinergic" colleagues as well as ourselves have experienced resistance to this concept, because it is teleologically counter-intuitive. This review serves to integrate the three main tenets of extracellular ATP signaling: ATP release from cells, ATP receptors on cells, and ATP receptor-driven signaling within cells to affect cell or tissue physiology. First principles will be discussed in the Introduction concerning extracellular ATP signaling. All possible cellular mechanisms of ATP release will then be presented. Use of nucleotide and nucleoside scavengers as well as broad-specificity purinergic receptor antagonists will be presented as a method of detecting endogenous ATP release affecting a biological endpoint. Innovative methods of detecting released ATP by adapting luciferase detection reagents or by using "biosensors" will be presented. Because our laboratory has been primarily interested in epithelial cell physiology and pathophysiology for several years, the role of extracellular ATP in regulation of epithelial cell function will be the focus of this review. For ATP release to be physiologically relevant, receptors for ATP are required at the cell surface. The families of P2Y G protein-coupled receptors and ATP-gated P2X receptor channels will be introduced. Particular attention will be paid to P2X receptor channels that mediate the fast actions of extracellular ATP signaling, much like neurotransmitter-gated channels versus metabotropic heptahelical neurotransmitter receptors that couple to G proteins. Finally, fascinating biological paradigms in which extracellular ATP signaling has been implicated will be highlighted. It is the goal of this review to convert and attract new scientists into the exploding field of extracellular nucleotide signaling and to convince the reader that extracellular ATP is indeed a signaling molecule.
Collapse
Affiliation(s)
- Erik M Schwiebert
- Department of Physiology and Biophysics, University of Alabama at Birmingham, 35294-0005, USA.
| | | |
Collapse
|
50
|
Dutta R, Singh U, Li TB, Fornage M, Teng BB. Hepatic gene expression profiling reveals perturbed calcium signaling in a mouse model lacking both LDL receptor and Apobec1 genes. Atherosclerosis 2003; 169:51-62. [PMID: 12860250 DOI: 10.1016/s0021-9150(03)00133-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a complex disease that gene and environment interaction influences the progression of atherosclerotic lesion development. Our laboratory used mice lacking both the low density lipoprotein (LDL) receptor and Apobec1 genes (LDLR-/-Apobec1-/-, designated LDb) to investigate gene-gene interaction and the influence of an environmental factor (high-fat diet) on gene networks. LDb mice (males and females) at 5 months of age were fed a chow or high fat diet for 3-month. The mice on a chow diet had elevated plasma cholesterol and triglyceride levels and developed atherosclerosis. Feeding a high-fat diet accelerated the development of lesions >1.5-fold. We performed microarray analysis of the expression of 12442 murine genes in the livers of these animals, which identified 54 genes in males and 77 genes in females were significantly perturbed by the high-fat diet. Moreover, most of these genes (>70%) were upregulated. The results suggested that glycolysis, fat transport, and steroid hormone biosynthesis pathways were upregulated, probably to compensate for the high fat intake. Furthermore, a batch of stress-responsive genes was upregulated. The study also shows a dynamic cellular communication network including T cells, neutrophils, and monocytes/macrophages, which related to inflammatory and immune/complement responses. Importantly, this study discovered that many genes involved in calcium signaling and bone formation were up regulated. Alizarin Red S staining was used to detect calcium deposits in the region of atherosclerotic lesions. Real-time quantitative RT-PCR and Western blot analyses provided verification of the gene expression levels. In conclusion, this study demonstrated the global differential gene expression profiles, which are influenced by feeding a high fat diet to LDb mice. The results of the study provide new insights into the significance of calcification in atherogenesis.
Collapse
Affiliation(s)
- Ranjan Dutta
- Research Center for Human Genetics, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 2121 W. Holcombe Blvd, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|