51
|
Sáez-Orellana F, Godoy PA, Silva-Grecchi T, Barra KM, Fuentealba J. Modulation of the neuronal network activity by P2X receptors and their involvement in neurological disorders. Pharmacol Res 2015; 101:109-15. [PMID: 26122853 DOI: 10.1016/j.phrs.2015.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 11/16/2022]
Abstract
ATP is a key energetic molecule, fundamental to cell function, which also has an important role in the extracellular milieu as a signaling molecule, acting as a chemoattractant for immune cells and as a neuro- and gliotransmitter. The ionotropic P2X receptors are members of an ATP-gated ion channels family. These ionotropic receptors are widely expressed through the body, with 7 subunits described in mammals, which are arranged in a trimeric configuration with a central pore permeable mainly to Ca(2+) and Na(+). All 7 subunits are expressed in different brain areas, being present in neurons and glia. ATP, through these ionotropic receptors, can act as a neuromodulator, facilitating the Ca(2+)-dependent release of neurotransmitters, inducing the cross-inhibition between P2XR and GABA receptors, and exercising by this way a modulation of synaptic plasticity. Growing evidence shows that P2XR play an important role in neuronal disorders and neurodegenerative diseases, like Parkinson's and Alzheimer's disease; this role involves changes on P2XR expression levels, activation of key pathways like GSK3β, APP processing, oxidative stress and inflammatory response. This review is focused on the neuromodulatory function of P2XR on pathophysiological conditions of the brain; the recent evidence could open a window to a new therapeutic target.
Collapse
Affiliation(s)
- F Sáez-Orellana
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - P A Godoy
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - T Silva-Grecchi
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - K M Barra
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - J Fuentealba
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile; Center for Advanced Research on Biomedicine (CIAB-UdeC), University of Concepción, Chile.
| |
Collapse
|
52
|
Burnstock G. An introduction to the roles of purinergic signalling in neurodegeneration, neuroprotection and neuroregeneration. Neuropharmacology 2015; 104:4-17. [PMID: 26056033 DOI: 10.1016/j.neuropharm.2015.05.031] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022]
Abstract
Purinergic signalling appears to play important roles in neurodegeneration, neuroprotection and neuroregeneration. Initially there is a brief summary of the background of purinergic signalling, including release of purines and pyrimidines from neural and non-neural cells and their ectoenzymatic degradation, and the current characterisation of P1 (adenosine), and P2X (ion channel) and P2Y (G protein-coupled) nucleotide receptor subtypes. There is also coverage of the localization and roles of purinoceptors in the healthy central nervous system. The focus is then on the roles of purinergic signalling in trauma, ischaemia, stroke and in neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's diseases, as well as multiple sclerosis and amyotrophic lateral sclerosis. Neuroprotective mechanisms involving purinergic signalling are considered and its involvement in neuroregeneration, including the role of adult neural stem/progenitor cells. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; Department of Pharmacology and Therapeutics, The University of Melbourne, Australia.
| |
Collapse
|
53
|
Cavaliere F, Donno C, D'Ambrosi N. Purinergic signaling: a common pathway for neural and mesenchymal stem cell maintenance and differentiation. Front Cell Neurosci 2015; 9:211. [PMID: 26082684 PMCID: PMC4451364 DOI: 10.3389/fncel.2015.00211] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/16/2015] [Indexed: 01/25/2023] Open
Abstract
Extracellular ATP, related nucleotides and adenosine are among the earliest signaling molecules, operating in virtually all tissues and cells. Through their specific receptors, namely purinergic P1 for nucleosides and P2 for nucleotides, they are involved in a wide array of physiological effects ranging from neurotransmission and muscle contraction to endocrine secretion, vasodilation, immune response, and fertility. The purinergic system also participates in the proliferation and differentiation of stem cells from different niches. In particular, both mesenchymal stem cells (MSCs) and neural stem cells are endowed with several purinergic receptors and ecto-nucleotide metabolizing enzymes, and release extracellular purines that mediate autocrine and paracrine growth/proliferation, pro- or anti-apoptotic processes, differentiation-promoting effects and immunomodulatory actions. Here, we discuss the often opposing roles played by ATP and adenosine in adult neurogenesis in both physiological and pathological conditions, as well as in adipogenic and osteogenic MSC differentiation. We also focus on how purinergic ligands produced and released by transplanted stem cells can be regarded as ideal candidates to mediate the crosstalk with resident stem cell niches, promoting cell growth and survival, regulating inflammation and, therefore, contributing to local tissue homeostasis and repair.
Collapse
Affiliation(s)
- Fabio Cavaliere
- Department of Neuroscience, Achucarro Basque Center for Neuroscience, CIBERNED and University of Basque Country, Leioa Spain
| | - Claudia Donno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome Italy
| | - Nadia D'Ambrosi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome Italy
| |
Collapse
|
54
|
Abstract
Preconditioning (PC) using a preceding sublethal ischemic insult is an attractive strategy for protecting neurons by inducing ischemic tolerance in the brain. Although the underlying molecular mechanisms have been extensively studied, almost all studies have focused on neurons. Here, using a middle cerebral artery occlusion model in mice, we show that astrocytes play an essential role in the induction of brain ischemic tolerance. PC caused activation of glial cells without producing any noticeable brain damage. The spatiotemporal pattern of astrocytic, but not microglial, activation correlated well with that of ischemic tolerance. Interestingly, such activation in astrocytes lasted at least 8 weeks. Importantly, inhibiting astrocytes with fluorocitrate abolished the induction of ischemic tolerance. To investigate the underlying mechanisms, we focused on the P2X7 receptor as a key molecule in astrocyte-mediated ischemic tolerance. P2X7 receptors were dramatically upregulated in activated astrocytes. PC-induced ischemic tolerance was abolished in P2X7 receptor knock-out mice. Moreover, our results suggest that hypoxia-inducible factor-1α, a well known mediator of ischemic tolerance, is involved in P2X7 receptor-mediated ischemic tolerance. Unlike previous reports focusing on neuron-based mechanisms, our results show that astrocytes play indispensable roles in inducing ischemic tolerance, and that upregulation of P2X7 receptors in astrocytes is essential.
Collapse
|
55
|
Alzheimer’s disease shares gene expression aberrations with purinergic dysregulation of HPRT deficiency (Lesch–Nyhan disease). Neurosci Lett 2015; 590:35-9. [DOI: 10.1016/j.neulet.2015.01.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/19/2022]
|
56
|
Heine C, Sygnecka K, Scherf N, Grohmann M, Bräsigk A, Franke H. P2Y(1) receptor mediated neuronal fibre outgrowth in organotypic brain slice co-cultures. Neuropharmacology 2015; 93:252-66. [PMID: 25683778 DOI: 10.1016/j.neuropharm.2015.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/30/2015] [Accepted: 02/01/2015] [Indexed: 11/16/2022]
Abstract
Extracellular purines have multiple functional roles in development, plastic remodelling, and regeneration of the CNS by stimulating certain P2X/Y receptor (R) subtypes. In the present study we elucidated the involvement of P2YRs in neuronal fibre outgrowth in the developing nervous system. We particularly focused on the P2Y1R subtype and the dopaminergic system, respectively. For this purpose, we used organotypic slice co-cultures consisting of the ventral tegmental area/substantia nigra (VTA/SN) and the prefrontal cortex (PFC). After detecting the presence of the P2Y1R in VTA/SN, PFC, and on outgrowing fibres in the border region (e.g. on glial processes) connecting both brain slices, we could show that pharmacological modulation of the receptor influenced neuronal fibre outgrowth. Biocytin-tracing and tyrosine hydroxylase-immunolabelling together with quantitative image analysis revealed a significant increase in fibre growth in the border region of the co-cultures after treatment with ADPβS (P2Y1,12,13R agonist). The observed stimulatory potential of ADPβS was inhibited by pre-treatment with the P2X/YR antagonist PPADS. In P2Y1R knockout (P2Y1R(-/-)) mice, the ADPβS-induced stimulatory effect was absent, while growth was significantly enhanced in the co-cultures of the respective wild-type. This observation was confirmed in entorhino-hippocampal co-cultures, an example of a different projection system, expressing the P2Y1R. Using wortmannin and PD98059 we further showed that PI3K/Akt and MAPK/ERK cascades are involved in the mechanism underlying ADPβS-induced fibre growth. In conclusion, the data of this study clearly indicate that activation of the P2Y1R stimulates fibre growth and thereby emphasises the general role of this particular receptor subtype during development and regeneration.
Collapse
Affiliation(s)
- Claudia Heine
- Translational Centre for Regenerative Medicine (TRM), University of Leipzig, Philipp-Rosenthal-Straße 55, 04103 Leipzig, Germany; Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Katja Sygnecka
- Translational Centre for Regenerative Medicine (TRM), University of Leipzig, Philipp-Rosenthal-Straße 55, 04103 Leipzig, Germany; Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Nico Scherf
- Institute for Medical Informatics and Biometry (IMB), Dresden University of Technology, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Marcus Grohmann
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Annett Bräsigk
- Centre for Biotechnology and Biomedicine (BBZ), Molecular Biological-Biochemical Processing Technology, Deutscher Platz 5, 04103 Leipzig, Germany.
| | - Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany.
| |
Collapse
|
57
|
Mizuno MS, Crisma AR, Borelli P, Schäfer BT, Silveira MP, Castelucci P. Distribution of the P2X2 receptor and chemical coding in ileal enteric neurons of obese male mice ( ob/ob). World J Gastroenterol 2014; 20:13911-13919. [PMID: 25320527 PMCID: PMC4194573 DOI: 10.3748/wjg.v20.i38.13911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/29/2014] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the colocalization, density and profile of neuronal areas of enteric neurons in the ileum of male obese mice.
METHODS: The small intestinal samples of male mice in an obese group (OG) (C57BL/6J ob/ob) and a control group (CG) (+/+) were used. The tissues were analyzed using a double immunostaining technique for immunoreactivity (ir) of the P2X2 receptor, nitric oxide synthase (NOS), choline acetyl transferase (ChAT) and calretinin (Calr). Also, we investigated the density and profile of neuronal areas of the NOS-, ChAT- and Calr-ir neurons in the myenteric plexus. Myenteric neurons were labeled using an NADH-diaphorase histochemical staining method.
RESULTS: The analysis demonstrated that the P2X2 receptor was expressed in the cytoplasm and in the nuclear and cytoplasmic membranes only in the CG. Neuronal density values (neuron/cm2) decreased 31% (CG: 6579 ± 837; OG: 4556 ± 407) and 16.5% (CG: 7796 ± 528; OG: 6513 ± 610) in the NOS-ir and calretinin-ir neurons in the OG, respectively (P < 0.05). Density of ChAT-ir (CG: 6200 ± 310; OG: 8125 ± 749) neurons significantly increased 31% in the OG (P < 0.05). Neuron size studies demonstrated that NOS, ChAT, and Calr-ir neurons did not differ significantly between the CG and OG groups. The examination of NADH-diaphorase-positive myenteric neurons revealed an overall similarity between the OG and CG.
CONCLUSION: Obesity may exert its effects by promoting a decrease in P2X2 receptor expression and modifications in the density of the NOS-ir, ChAT-ir and CalR-ir myenteric neurons.
Collapse
|
58
|
da Silva MV, Marosti AR, Mendes CE, Palombit K, Castelucci P. Differential effects of experimental ulcerative colitis on P2X7 receptor expression in enteric neurons. Histochem Cell Biol 2014; 143:171-84. [PMID: 25201348 DOI: 10.1007/s00418-014-1270-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2014] [Indexed: 12/17/2022]
Abstract
The digestive tracts of ulcerative colitis and Crohn's disease patients present with pathophysiological processes and intestinal necrosis. This study examined the P2X7 receptor and changes in the distal colon in enteric neurons of rats with experimental ulcerative colitis. The analysis was performed in the distal colons of rats with ulcerative colitis induced by the administration of 2,4,6-trinitrobenzene sulfonic acid (colitis group). The survival time after colitis induction was 24 h. The treated animals were compared to sham rats injected with phosphate-buffered saline and to animals with no intervention (control group). Tissues were prepared for immunohistochemical double-staining methods to examine P2X7 receptor, choline acetyltransferase (ChAT), calbindin, calretinin, anti-HuC/D (pan-neuronal) and S100β (pan-glial). The colocalization of the P2X7 receptor-immunoreactive (IR) cells was observed in the myenteric plexus with nitric oxide synthase (NOS)-, ChAT-,calbindin-, calretinin- and HuC/D-IR neurons and S100β-IR cells in the control, sham and colitis groups. The neuronal density (cell bodies/cm(2)) decreased in the myenteric plexus by 11, 18, 34, 22 and 60% in the P2X7 receptor, NOS-, ChAT-, calbindin- and calretinin-IR neurons, respectively. In addition, the densities (cell bodies/cm(2)) of HuC/D-IR neurons and S100β-IR enteric glial cells decreased by 33 and 29%, respectively. The profile areas were reduced by 6.8 and 21% in NOS- and ChAT-IR neurons, respectively. There was also a 20% increase of calbindin-IR neurons. Morphological changes were observed, such as increased neutrophils, disintegration of the intestinal epithelium and goblet cells and decreased collagen. This study demonstrated that colitis differentially affects P2X7 receptor-expressing enteric neurons based on their chemical codes and may cause changes in morphology and motility.
Collapse
Affiliation(s)
- Marcos Vinícius da Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Dr. Lineu Prestes, 2415, São Paulo, CEP 05508-900, Brazil
| | | | | | | | | |
Collapse
|
59
|
Jia S, Chen Z, Li J, Chi Y, Wang J, Li S, Luo Y, Geng B, Wang C, Cui Q, Guan Y, Yang J. FAM3A promotes vascular smooth muscle cell proliferation and migration and exacerbates neointima formation in rat artery after balloon injury. J Mol Cell Cardiol 2014; 74:173-82. [PMID: 24857820 DOI: 10.1016/j.yjmcc.2014.05.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/07/2014] [Accepted: 05/14/2014] [Indexed: 11/17/2022]
Abstract
The biological function of FAM3A, the first member of family with sequence similarity 3 (FAM3) gene family, remains largely unknown. This study aimed to determine its role in the proliferation and migration of vascular smooth muscle cells (VSMCs). Immunohistochemical staining revealed that FAM3A protein is expressed in the tunica media of rodent arteries, and its expression is reduced with an increase in prostaglandin E receptor 2 (EP2) expression after injury. In vitro, FAM3A overexpression promotes proliferation and migration of VSMCs, whereas FAM3A silencing inhibits these processes. In vivo, FAM3A overexpression results in exaggerated neointima formation of rat carotid artery after balloon injury. FAM3A activates Akt in a PI3K-dependent manner. In contrast, FAM3A induces ERK1/2 activation independent of PI3K. FAM3A protein is subcellularly located in mitochondria, where it affects ATP production and release. Activation of EP2 represses FAM3A expression, leading to impaired ATP production and release in VSMCs. FAM3A-induced activation of Akt and ERK1/2 pathways, proliferation and migration of VSMCs are inhibited by P2 receptor antagonist suramin. Furthermore, inhibition or knockdown of P2Y1 receptor inihibits FAM3A-induced proliferation and migration of VSMCs. In conclusion, FAM3A promotes proliferation and migration of VSMCs via P2Y1 receptor-mediated activation of Akt and ERK1/2 pathways. In injured vessels, FAM3A was repressed by upregulated EP2 expression, leading to the attenuation of ATP-P2Y1 receptor signaling, which is beneficial for preventing excessive proliferation and migration of VSMCs.
Collapse
MESH Headings
- Animals
- Balloon Occlusion
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cell Movement
- Cell Proliferation
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression Regulation
- Male
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/genetics
- Neointima/metabolism
- Neointima/pathology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Purinergic P2Y1/genetics
- Receptors, Purinergic P2Y1/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Shi Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Zhenzhen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Jing Li
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Yujing Chi
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - Jinyu Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Sha Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yanjin Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Bin Geng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Cheng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
60
|
Wang C, Chi Y, Li J, Miao Y, Li S, Su W, Jia S, Chen Z, Du S, Zhang X, Zhou Y, Wu W, Zhu M, Wang Z, Yang H, Xu G, Wang S, Yang J, Guan Y. FAM3A activates PI3K p110α/Akt signaling to ameliorate hepatic gluconeogenesis and lipogenesis. Hepatology 2014; 59:1779-90. [PMID: 24806753 DOI: 10.1002/hep.26945] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/18/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED FAM3A belongs to a novel cytokine-like gene family, and its physiological role remains largely unknown. In our study, we found a marked reduction of FAM3A expression in the livers of db/db and high-fat diet (HFD)-induced diabetic mice. Hepatic overexpression of FAM3A markedly attenuated hyperglycemia, insulin resistance, and fatty liver with increased Akt (pAkt) signaling and repressed gluconeogenesis and lipogenesis in the livers of those mice. In contrast, small interfering RNA (siRNA)-mediated knockdown of hepatic FAM3A resulted in hyperglycemia with reduced pAkt levels and increased gluconeogenesis and lipogenesis in the livers of C57BL/6 mice. In vitro study revealed that FAM3A was mainly localized in the mitochondria, where it increases adenosine triphosphate (ATP) production and secretion in cultured hepatocytes. FAM3A activated Akt through the p110α catalytic subunit of PI3K in an insulin-independent manner. Blockade of P2 ATP receptors or downstream phospholipase C (PLC) and IP3R and removal of medium calcium all significantly reduced FAM3A-induced increase in cytosolic free Ca(2+) levels and attenuated FAM3A-mediated PI3K/Akt activation. Moreover, FAM3A-induced Akt activation was completely abolished by the inhibition of calmodulin (CaM). CONCLUSION FAM3A plays crucial roles in the regulation of glucose and lipid metabolism in the liver, where it activates the PI3K-Akt signaling pathway by way of a Ca(2+) /CaM-dependent mechanism. Up-regulating hepatic FAM3A expression may represent an attractive means for the treatment of insulin resistance, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Wang C, Geng B, Cui Q, Guan Y, Yang J. Intracellular and extracellular adenosine triphosphate in regulation of insulin secretion from pancreatic β cells (β). J Diabetes 2014; 6:113-9. [PMID: 24134160 DOI: 10.1111/1753-0407.12098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 09/27/2013] [Accepted: 10/11/2013] [Indexed: 01/09/2023] Open
Abstract
Adenosine triphosphate (ATP) synthesis and release in mitochondria play critical roles in regulating insulin secretion in pancreatic β cells. Mitochondrial dysfunction is mainly characterized by a decrease in ATP production, which is a central event in the progression of pancreatic β cell dysfunction and diabetes. ATP has been demonstrated to regulate insulin secretion via several pathways: (i) Intracellular ATP directly closes ATP-sensitive potassium channel to open L-type calcium channel, leading to an increase in free cytosolic calcium levels and exocytosis of insulin granules; (ii) A decrease in ATP production is always associated with an increase in production of reactive oxygen species, which exerts deleterious effects on pancreatic β cell survival and insulin secretion; and (iii) ATP can be co-secreted with insulin from pancreatic β cells, and the released ATP functions as an autocrine signal to modulate insulin secretory process via P2 receptors on the cell membrane. In this review, the recent findings regarding the role and mechanism of ATP synthesis and release in regulation of insulin secretion from pancreatic β cells will be summarized and discussed.
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | | | | | | | | |
Collapse
|
62
|
Wang C, Chen Z, Li S, Zhang Y, Jia S, Li J, Chi Y, Miao Y, Guan Y, Yang J. Hepatic overexpression of ATP synthase β subunit activates PI3K/Akt pathway to ameliorate hyperglycemia of diabetic mice. Diabetes 2014; 63:947-59. [PMID: 24296716 DOI: 10.2337/db13-1096] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ATP synthase β subunit (ATPSβ) had been previously shown to play an important role in controlling ATP synthesis in pancreatic β-cells. This study aimed to investigate the role of ATPSβ in regulation of hepatic ATP content and glucose metabolism in diabetic mice. ATPSβ expression and ATP content were both reduced in the livers of type 1 and type 2 diabetic mice. Hepatic overexpression of ATPSβ elevated cellular ATP content and ameliorated hyperglycemia of streptozocin-induced diabetic mice and db/db mice. ATPSβ overexpression increased phosphorylated Akt (pAkt) levels and reduced PEPCK and G6pase expression levels in the livers. Consistently, ATPSβ overexpression repressed hepatic glucose production in db/db mice. In cultured hepatocytes, ATPSβ overexpression increased intracellular and extracellular ATP content, elevated the cytosolic free calcium level, and activated Akt independent of insulin. The ATPSβ-induced increase in cytosolic free calcium and pAkt levels was attenuated by inhibition of P2 receptors. Notably, inhibition of calmodulin (CaM) completely abolished ATPSβ-induced Akt activation in liver cells. Inhibition of P2 receptors or CaM blocked ATPSβ-induced nuclear exclusion of forkhead box O1 in liver cells. In conclusion, a decrease in hepatic ATPSβ expression in the liver, leading to the attenuation of ATP-P2 receptor-CaM-Akt pathway, may play an important role in the progression of diabetes.
Collapse
Affiliation(s)
- Chunjiong Wang
- Department of Physiology and Pathophysiology, Key Laboratory of Molecular Cardiovascular Science, Peking (Beijing) University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Ventral tegmental area/substantia nigra and prefrontal cortex rodent organotypic brain slices as an integrated model to study the cellular changes induced by oxygen/glucose deprivation and reperfusion: effect of neuroprotective agents. Neurochem Int 2014; 66:43-54. [PMID: 24463100 DOI: 10.1016/j.neuint.2014.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/02/2013] [Accepted: 01/14/2014] [Indexed: 01/17/2023]
Abstract
Unveiling the roles of distinct cell types in brain response to insults is a partially unsolved challenge and a key issue for new neuroreparative approaches. In vivo models are not able to dissect the contribution of residential microglia and infiltrating blood-borne monocytes/macrophages, which are fundamentally undistinguishable; conversely, cultured cells lack original tissue anatomical and functional complexity, which profoundly alters reactivity. Here, we tested whether rodent organotypic co-cultures from mesencephalic ventral tegmental area/substantia nigra and prefrontal cortex (VTA/SN-PFC) represent a suitable model to study changes induced by oxygen/glucose deprivation and reperfusion (OGD/R). OGD/R induced cytotoxicity to both VTA/SN and PFC slices, with higher VTA/SN susceptibility. Neurons were highly affected, with astrocytes and oligodendrocytes undergoing very mild damage. Marked reactive astrogliosis was also evident. Notably, OGD/R triggered the activation of CD68-expressing microglia and increased expression of Ym1 and Arg1, two markers of "alternatively" activated beneficial microglia. Treatment with two well-known neuroprotective drugs, the anticonvulsant agent valproic acid and the purinergic P2-antagonist PPADS, prevented neuronal damage. Thus, VTA/SN-PFC cultures are an integrated model to investigate OGD/R-induced effects on distinct cells and easily screen neuroprotective agents. The model is particularly adequate to dissect the microglia phenotypic shift in the lack of a functional vascular compartment.
Collapse
|
64
|
Kido Y, Kawahara C, Terai Y, Ohishi A, Kobayashi S, Hayakawa M, Kamatsuka Y, Nishida K, Nagasawa K. Regulation of activity of P2X7 receptor by its splice variants in cultured mouse astrocytes. Glia 2013; 62:440-51. [PMID: 24375698 DOI: 10.1002/glia.22615] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/24/2013] [Accepted: 11/21/2013] [Indexed: 01/05/2023]
Abstract
Of purinergic receptors, P2X7 receptor (P2X7R, defined as a full-length receptor) has unique characteristics, and its activation leads to ion channel activity and pore formation, causing cell death. Previously, we demonstrated that P2X7R expressed by nonstimulated astrocyte cultures obtained from SJL-strain mice exhibits constitutive activation, implying its role in maintenance of cellular homeostasis. To obtain novel insights into its physiological roles, we examined whether constitutive activation of P2X7R is regulated by expression of its splice variants in such resting astrocytes, and whether their distinct expression profiles in different mouse strains affect activation levels of astrocytic P2X7Rs. In SJL- and ddY-mouse astrocytes, spontaneous YO-PRO-1 uptake, an indicator of pore activity of P2X7R, was detected, but the uptake by the formers was significantly greater than that by the latter. Between the two mouse strains, there was a difference in their sensitivity of YO-PRO-1 uptake to antagonists, but not in the expression levels and sequences of P2X7R and pannexin-1. Regarding expression of splice variants of P2X7R, expression of P2X7R variant-3 (P2X7R-v3) and -4 (P2X7R-v4), but not variant-2 and -k, was lower in SJL-mouse astrocytes than in ddY-mouse ones. On transfection of P2X7R-v3 and -v4 into SJL-mouse astrocytes, the pore activity was attenuated as in the case of the HEK293T cell-expression system. These findings demonstrate that basal activity of P2X7R expressed by resting astrocytes is negatively regulated by P2X7R-v3 and -v4, and that their distinct expression profiles result in the different activation levels of astrocytic P2X7Rs in different mouse strains.
Collapse
Affiliation(s)
- Yuuka Kido
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Carmo MRS, Simões AP, Fonteles AA, Souza CM, Cunha RA, Andrade GM. ATP P2Y1 receptors control cognitive deficits and neurotoxicity but not glial modifications induced by brain ischemia in mice. Eur J Neurosci 2013; 39:614-22. [PMID: 24304101 DOI: 10.1111/ejn.12435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/21/2013] [Accepted: 10/25/2013] [Indexed: 11/28/2022]
Abstract
ATP is a pleiotropic cell-to-cell signaling molecule in the brain that functions through activation of the P2 receptors (P2R), encompassing ionotropic P2XR or metabotropic P2YR. Noxious brain insults increase the extracellular levels of ATP and previous studies have implicated different P2R, namely P2Y1R, in the control of ischemic brain damage, but it remains to be defined if P2Y1R antagonists also alleviate the behavioral impairments associated with brain ischemia. Furthermore, as P2Y1R can control neuronal and glial functions, we explored if P2Y1R antagonist-mediated protection would mainly involve neuronal and/or glial processes. Adult male mice subject to permanent middle cerebral artery occlusion (pMCAO) displayed an infarcted cortical area (2,3,5-triphenyltetrazolium chloride staining), decreased neurological score with decreased working and reference memory performance (Y-maze, object recognition and aversive memory), accompanied by neuronal damage (FluoroJade C), astrogliosis (glial fibrillary acidic protein) and microgliosis (CD11b). All of these changes were attenuated by intracerebroventricular pre-treatment (10 min before pMCAO) with the generic P2R antagonist 4-[(E)-{4-formyl-5-hydroxy-6-methyl-3-[(phosphono-oxy)methyl]pyridin-2-yl}diazenyl]benzene-1,3-disulfonic acid (PPADS, 0.5-1.0 nmol/μL). In contrast, the selective P2Y1R antagonist (1R*,2S*)-4-[2-Iodo-6-(methylamino)-9H-purin-9-yl]-2-(phosphono-oxy)bicycle[3.1.0] hexane-1-methanol dihydrogen phosphate ester (MRS2500, 1.0-2.0 nmol/μL) afforded equivalent behavioral benefits but only prevented neuronal damage but not astrogliosis or microgliosis upon pMCAO. These results indicated that P2Y1R-associated neuroprotection mainly occurred through neuronal mechanisms, whereas other P2R were also involved in the control of astrocytic reactivity upon brain injury.
Collapse
Affiliation(s)
- Marta R S Carmo
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | | |
Collapse
|
66
|
Franke H, Illes P. Nucleotide signaling in astrogliosis. Neurosci Lett 2013; 565:14-22. [PMID: 24103370 DOI: 10.1016/j.neulet.2013.09.056] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 09/05/2013] [Accepted: 09/17/2013] [Indexed: 12/29/2022]
Abstract
Acute and chronic damage to the central nervous system (CNS) releases large quantities of ATP. Whereas the ATP concentration in the extracellular space is normally in the micromolar range, under these conditions it increases to millimolar levels. A number of ligand-gated cationic channels termed P2X receptors (7 mammalian subtypes), and G protein-coupled P2Y receptors (8 mammalian subtypes) are located at astrocytes, as confirmed by the measurement of the respective mRNA and protein. Activation of both the P2X7 and P2Y1,2 subtypes identified at astrocytes initiates astrogliosis isolating damaged brain areas from surrounding healthy cells and synthesizing neurotrophins and pleotrophins that participate in neuronal recovery. Astrocytes are considered as cells of high plasticity which may alter their properties in a culture medium. Therefore, recent work concentrates on investigating nucleotide effects at in situ (acute brain slices) and in vivo astrocytes. A wealth of data relates to the involvement of purinergic mechanisms in astrogliosis induced by acute CNS injury such as mechanical trauma and hypoxia/ischemia. The released ATP may act within minutes as an excitotoxic molecule; at a longer time-scale within days it causes neuroinflammation. These effects sum up as necrosis/apoptosis on the one hand and proliferation on the other. Although the role of nucleotides in chronic neurodegenerative illnesses is not quite clear, it appears that they aggravate the consequences of the primary disease. Epilepsy and neuropathic pain are also associated with the release of ATP and a pathologic glia-neuron interaction leading to astrogliosis and cell death. In view of these considerations, P2 receptor antagonists may open new therapeutic vistas in all forms of acute and chronic CNS damage.
Collapse
Affiliation(s)
- Heike Franke
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107 Leipzig, Germany
| | - Peter Illes
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, 04107 Leipzig, Germany.
| |
Collapse
|
67
|
Frégeau MO, Carrier M, Guillemette G. Mechanism of dopamine D2 receptor-induced Ca(2+) release in PC-12 cells. Cell Signal 2013; 25:2871-7. [PMID: 24055909 DOI: 10.1016/j.cellsig.2013.08.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/23/2013] [Indexed: 01/06/2023]
Abstract
Intracellular Ca(2+) levels are tightly regulated in the neuronal system. The loss of Ca(2+) homeostasis is associated with many neurological diseases and neuropsychiatric disorders such as Parkinson's, Alzheimer's, and schizophrenia. We investigated the mechanisms involved in intracellular Ca(2+) signaling in PC-12 cells. The stimulation of NGF-differentiated PC-12 cells with 3μM ATP caused an early Ca(2+) release followed by a delayed Ca(2+) release. The delayed Ca(2+) release was dependent on prior ATP priming and on dopamine secretion by PC-12 cells. Delayed Ca(2+) release was abolished in the presence of spiperone, suggesting that it is due to the activation of D2 dopamine receptors (D2R) by dopamine secreted by PC-12 cells. This was shown to be independent of PKA activation but dependent on PLC activity. An endocytosis step was required for inducing the delayed Ca(2+) release. Given the importance of calcyon in clathrin-mediated endocytosis, we verified the role of this protein in the delayed Ca(2+) release phenomenon. siRNA targeting of calcyon blocked the delayed Ca(2+) release, decreased ATP-evoked IP3R-mediated Ca(2+) release, and impaired subsequent Ca(2+) oscillations. Our results suggested that calcyon is involved in an unknown mechanism that causes a delayed IP3R-mediated Ca(2+) release in PC-12 cells. In schizophrenia, Ca(2+) dysregulation may depend on the upregulation of calcyon, which maintains elevated Ca(2+) levels as well as dopamine signaling.
Collapse
Affiliation(s)
- Marc-Olivier Frégeau
- Faculty of Medicine and Health sciences, Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4 Canada
| | | | | |
Collapse
|
68
|
Fletcher L, Evans TM, Watts LT, Jimenez DF, Digicaylioglu M. Rapamycin treatment improves neuron viability in an in vitro model of stroke. PLoS One 2013; 8:e68281. [PMID: 23861877 PMCID: PMC3701663 DOI: 10.1371/journal.pone.0068281] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 05/28/2013] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is the leading cause of serious, long-term adult disability and is associated with sensorimotor and cognitive impairments due to neuronal degeneration. Currently, recombinant tissue plasminogen activator (rTPA) is the only FDA-approved medical therapy for treatment of patients with acute ischemic stroke. However, rTPA can only be given within 3 hours of symptom onset, and only 2% of patients are eligible. Therefore, there is an urgent need for novel neuroprotective treatment options for ischemic stroke. An emerging treatment for a diverse range of neurological disorders associated with neurodegeneration is rapamycin, a key modulator of the mammalian target of rapamycin (mTOR) pathway. The mTOR pathway is the primary regulator of the cellular response to nutrient availability, changes in energy status and stress as seen following ischemia and reperfusion. However, rapamycin’s effects on mTORC1 and mTORC2 are poorly understood in neurons. In the current study we show that rapamycin can prevent the activation of both mTORC1 and mTORC2 in cortical neurons and improve cell survival following oxygen glucose deprivation (OGD), an in vitro model of ischemic stroke. This work further supports the investigation of rapamycin as a novel neuroprotectant for ischemic stroke.
Collapse
Affiliation(s)
- Lauren Fletcher
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Teresa M. Evans
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lora Talley Watts
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - David F. Jimenez
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Murat Digicaylioglu
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
69
|
Weisman GA, Woods LT, Erb L, Seye CI. P2Y receptors in the mammalian nervous system: pharmacology, ligands and therapeutic potential. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2013; 11:722-38. [PMID: 22963441 DOI: 10.2174/187152712803581047] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 06/14/2012] [Accepted: 06/14/2012] [Indexed: 11/22/2022]
Abstract
P2Y receptors for extracellular nucleotides are coupled to activation of a variety of G proteins and stimulate diverse intracellular signaling pathways that regulate functions of cell types that comprise the central nervous system (CNS). There are 8 different subtypes of P2Y receptor expressed in cells of the CNS that are activated by a select group of nucleotide agonists. Here, the agonist selectivity of these 8 P2Y receptor subtypes is reviewed with an emphasis on synthetic agonists with high potency and resistance to degradation by extracellular nucleotidases that have potential applications as therapeutic agents. In addition, the recent identification of a wide variety of subtype-selective antagonists is discussed, since these compounds are critical for discerning cellular responses mediated by activation of individual P2Y receptor subtypes. The functional expression of P2Y receptor subtypes in cells that comprise the CNS is also reviewed and the role of each subtype in the regulation of physiological and pathophysiological responses is considered. Other topics include the role of P2Y receptors in the regulation of blood-brain barrier integrity and potential interactions between different P2Y receptor subtypes that likely impact tissue responses to extracellular nucleotides in the CNS. Overall, current research suggests that P2Y receptors in the CNS regulate repair mechanisms that are triggered by tissue damage, inflammation and disease and thus P2Y receptors represent promising targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, 540E Life Sciences Center, 1201 Rollins Road, University of Missouri, Columbia, MO 65211-7310, USA.
| | | | | | | |
Collapse
|
70
|
Choi RCY, Chu GKY, Siow NL, Yung AWY, Yung LY, Lee PSC, Lo CCW, Simon J, Dong TTX, Barnard EA, Tsim KWK. Activation of UTP-sensitive P2Y2 receptor induces the expression of cholinergic genes in cultured cortical neurons: a signaling cascade triggered by Ca2+ mobilization and extracellular regulated kinase phosphorylation. Mol Pharmacol 2013; 84:50-61. [PMID: 23592515 DOI: 10.1124/mol.112.084160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
ATP functions as an extracellular signaling molecule that is costored and coreleased with neurotransmitters at central and peripheral neuronal synapses. Stimulation by ATP upregulates the expression of synaptic genes in muscle-including the genes for nicotine acetylcholine receptor (α-, δ-, and ε-subunits) and acetylcholinesterase (AChE)-via the P2Y receptor (P2YR), but the trophic response of neurons to the activation of P2YRs is less well understood. We reported that cultured cortical neurons and the developing rat brain expressed different types of P2YRs, and among these the UTP-sensitive P2Y2R was the most abundant. P2Y2R was found to exist in membrane rafts and it colocalized with the postsynaptic protein PSD-95 in cortical neurons. Notably, agonist-dependent stimulation of P2Y2R elevated the neuronal expression of cholinergic genes encoding AChE, PRiMA (an anchor for the globular form AChE), and choline acetyltransferase, and this induction was mediated by a signaling cascade that involved Ca(2+) mobilization and extracellular regulated kinases 1/2 activation. The importance of P2Y2R action was further shown by the receptor's synergistic effect with P2Y1R in enhancing cholinergic gene expression via the robust stimulation of Ca(2+) influx. Taken together our results revealed a developmental function of P2Y2R in promoting synaptic gene expression and demonstrated the influence of costimulation of P2Y1R and P2Y2R in neurons.
Collapse
Affiliation(s)
- Roy C Y Choi
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Xu P, Rosen KM, Hedstrom K, Rey O, Guha S, Hart C, Corfas G. Nerve injury induces glial cell line-derived neurotrophic factor (GDNF) expression in Schwann cells through purinergic signaling and the PKC-PKD pathway. Glia 2013; 61:1029-40. [PMID: 23553603 DOI: 10.1002/glia.22491] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/06/2013] [Indexed: 12/22/2022]
Abstract
Upon peripheral nerve injury, specific molecular events, including increases in the expression of selected neurotrophic factors, are initiated to prepare the tissue for regeneration. However, the mechanisms underlying these events and the nature of the cells involved are poorly understood. We used the injury-induced upregulation of glial cell-derived neurotrophic factor (GDNF) expression as a tool to gain insights into these processes. We found that both myelinating and nonmyelinating Schwann cells are responsible for the dramatic increase in GDNF expression after injury. We also demonstrate that the GDNF upregulation is mediated by a signaling cascade involving activation of Schwann cell purinergic receptors, followed by protein kinase C signaling which activates protein kinase D (PKD), which leads to increased GDNF transcription. Given the potent effects of GDNF on survival and repair of injured peripheral neurons, we propose that targeting these pathways may yield therapeutic tools to treat peripheral nerve injury and neuropathies.
Collapse
Affiliation(s)
- Pin Xu
- F.M. Kirby Neurobiology Center, Children's Hospital Boston, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
72
|
Ashpole NM, Chawla AR, Martin MP, Brustovetsky T, Brustovetsky N, Hudmon A. Loss of calcium/calmodulin-dependent protein kinase II activity in cortical astrocytes decreases glutamate uptake and induces neurotoxic release of ATP. J Biol Chem 2013; 288:14599-14611. [PMID: 23543737 DOI: 10.1074/jbc.m113.466235] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The extent of calcium/calmodulin-dependent protein kinase II (CaMKII) inactivation in the brain after ischemia correlates with the extent of damage. We have previously shown that a loss of CaMKII activity in neurons is detrimental to neuronal viability by inducing excitotoxic glutamate release. In the current study we extend these findings to show that the ability of astrocytes to buffer extracellular glutamate is reduced when CaMKII is inhibited. Furthermore, CaMKII inhibition in astrocytes is associated with the rapid onset of intracellular calcium oscillations. Surprisingly, this rapid calcium influx is blocked by the N-type calcium channel antagonist, ω-conotoxin. Although the function of N-type calcium channels within astrocytes is controversial, these voltage-gated calcium channels have been linked to calcium-dependent vesicular gliotransmitter release. When extracellular glutamate and ATP levels are measured after CaMKII inhibition within our enriched astrocyte cultures, no alterations in glutamate levels are observed, whereas ATP levels in the extracellular environment significantly increase. Extracellular ATP accumulation associated with CaMKII inhibition contributes both to calcium oscillations within astrocytes and ultimately cortical neuron toxicity. Thus, a loss of CaMKII signaling within astrocytes dysregulates glutamate uptake and supports ATP release, two processes that would compromise neuronal survival after ischemic/excitotoxic insults.
Collapse
Affiliation(s)
- Nicole M Ashpole
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Aarti R Chawla
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Matthew P Martin
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Nickolay Brustovetsky
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Andy Hudmon
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| |
Collapse
|
73
|
ATP induces NO production in hippocampal neurons by P2X(7) receptor activation independent of glutamate signaling. PLoS One 2013; 8:e57626. [PMID: 23472093 PMCID: PMC3589399 DOI: 10.1371/journal.pone.0057626] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 01/27/2013] [Indexed: 12/31/2022] Open
Abstract
To assess the putative role of adenosine triphosphate (ATP) upon nitric oxide (NO) production in the hippocampus, we used as a model both rat hippocampal slices and isolated hippocampal neurons in culture, lacking glial cells. In hippocampal slices, additions of exogenous ATP or 2′(3′)-O-(4-Benzoylbenzoyl) ATP (Bz-ATP) elicited concentration-dependent NO production, which increased linearly within the first 15 min and plateaued thereafter; agonist EC50 values were 50 and 15 µM, respectively. The NO increase evoked by ATP was antagonized in a concentration-dependent manner by Coomassie brilliant blue G (BBG) or by Nω-propyl-L-arginine, suggesting the involvement of P2X7Rs and neuronal NOS, respectively. The ATP induced NO production was independent of N-methyl-D-aspartic acid (NMDA) receptor activity as effects were not alleviated by DL-2-Amino-5-phosphonopentanoic acid (APV), but antagonized by BBG. In sum, exogenous ATP elicited NO production in hippocampal neurons independently of NMDA receptor activity.
Collapse
|
74
|
P2X7 receptor activation induces reactive oxygen species formation and cell death in murine EOC13 microglia. Mediators Inflamm 2013; 2013:271813. [PMID: 23431238 PMCID: PMC3568910 DOI: 10.1155/2013/271813] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/16/2012] [Accepted: 12/04/2012] [Indexed: 12/04/2022] Open
Abstract
The P2X7 purinergic receptor is a ligand-gated cation channel expressed on leukocytes including microglia. This study aimed to determine if P2X7 activation induces the uptake of organic cations, reactive oxygen species (ROS) formation, and death in the murine microglial EOC13 cell line. Using the murine macrophage J774 cell line as a positive control, RT-PCR, immunoblotting, and immunolabelling established the presence of P2X7 in EOC13 cells. A cytofluorometric assay demonstrated that the P2X7 agonists adenosine-5′-triphosphate (ATP) and 2′(3′)-O-(4-benzoylbenzoyl) ATP induced ethidium+ or YO-PRO-12+ uptake into both cell lines. ATP induced ethidium+ uptake into EOC13 cells in a concentration-dependent manner, with an EC50 of ~130 μM. The P2X7 antagonists Brilliant Blue G, A438079, AZ10606120, and AZ11645373 inhibited ATP-induced cation uptake into EOC13 cells by 75–100%. A cytofluorometric assay demonstrated that P2X7 activation induced ROS formation in EOC13 cells, via a mechanism independent of Ca2+ influx and K+ efflux. Cytofluorometric measurements of Annexin-V binding and 7AAD uptake demonstrated that P2X7 activation induced EOC13 cell death. The ROS scavenger N-acetyl-L-cysteine impaired both P2X7-induced EOC13 ROS formation and cell death, suggesting that ROS mediate P2X7-induced EOC13 death. In conclusion, P2X7 activation induces the uptake of organic cations, ROS formation, and death in EOC13 microglia.
Collapse
|
75
|
Guo W, Zhang Z, Liu X, Burnstock G, Xiang Z, He C. Developmental expression of P2X5 receptors in the mouse prenatal central and peripheral nervous systems. Purinergic Signal 2012; 9:239-48. [PMID: 23271560 DOI: 10.1007/s11302-012-9346-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 12/04/2012] [Indexed: 12/30/2022] Open
Abstract
The functions of P2X purinoceptors (P2X1-7) in the nervous system of adults have been widely studied. However, little is known about their roles during embryonic development. Our previous work has reported an extensive expression of P2X5 receptors in the adult mouse central nervous system. In the present study, we have examined the expression pattern of P2X5 receptor mRNA and protein during prenatal development of the mouse nervous system (from embryonic day E8 to E17). P2X5 receptors appeared in the neural tube as early as E8 and were gradually confined to new-born neurons in the cortical plate and ventral horn of the spinal cord. Heavy signals for P2X5 receptors were also found in dorsal root ganglia (DRG), retina, olfactory epithelium, and nerve fibers in skeletal muscles. In conclusion, P2X5 receptors were strongly represented in the developing mouse nervous system. The transient high expression pattern of P2X5 receptors in epithelium-like structures suggests a role during early neurogenesis.
Collapse
Affiliation(s)
- Wei Guo
- Key Laboratory of Medical Neurobiology, Ministry of Health, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | |
Collapse
|
76
|
Nishida K, Kitada T, Kato J, Dohi Y, Nagasawa K. Expression of equilibrative nucleoside transporter 1 in rat circumvallate papillae. Neurosci Lett 2012; 533:104-8. [PMID: 23147119 DOI: 10.1016/j.neulet.2012.10.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 10/27/2012] [Accepted: 10/31/2012] [Indexed: 11/26/2022]
Abstract
In gustatory function, communication between four types taste buds cells plays crucial roles. ATP is one of the intercellular signaling molecules in taste buds, and the extracellular ATP fate is regulated by its cellular clearance, but there is little information on it. Therefore, we examined the expression profiles of nucleoside transporters (NTs) as a clearance system for ATP metabolite adenosine in rat circumvallate papillae (CP) by RT-PCR, real-time PCR and immunohistochemistry. Among NTs, mRNA for Ent1 was expressed by the CP, and significantly was greater in the CP as compared with non-CP. ENT1 immunoreactivity was detected in PLC-β2-positive type II (71.0±8.5%), chromogranin-A-positive type III (64.9±7.4%), and SNAP25-positive type III (77.0±10.4%) taste cells, but not in NTPDase2-positive type I ones. These results indicate that ENT1-expressing type II and III taste cells might comprise an adenosine clearance system in taste buds of the CP. ENT1 expression in taste cells is important for elucidation of complicated taste signaling.
Collapse
Affiliation(s)
- Kentaro Nishida
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, Misasagi, Kyoto 607-8414, Japan
| | | | | | | | | |
Collapse
|
77
|
Anoxic depolarization of hippocampal astrocytes: possible modulation by P2X7 receptors. Neurochem Int 2012; 62:15-22. [PMID: 23147683 DOI: 10.1016/j.neuint.2012.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 12/30/2022]
Abstract
Current responses from CA1 neurons and stratum oriens astrocytes were recorded from hippocampal brain slices by means of the whole-cell patch-clamp technique. Anoxic depolarization (AD) was induced by an oxygen/glucose-deprived (OGD) medium also containing sodium iodoacetate and antimycin, in order to block glycolysis and oxidative phosphorylation, respectively. Anoxic depolarization has been reported to be due to the sudden increase of the extracellular K(+) concentration and the accompanying explosive rise in glutamate concentration. We asked ourselves whether the release of ATP activating P2X7 receptors is also involved in the AD. Although, the AD was evoked in absolute synchrony in neurons and astrocytes, and the NMDA receptor antagonistic AP-5 depressed these responses, neither the non-selective P2 receptor antagonist PPADS, nor the highly selective P2X7 receptor antagonist A438079 interfered with the AD or its delay time in neurons/astrocytes after inducing chemical hypoxia. However, A438079, but not PPADS increased in astrocytes the slow inward current observed in a hypoxic medium. It is concluded that ATP co-released with glutamate by hypoxic stimulation has only a minor function in the present brain slice system.
Collapse
|
78
|
Nishida K, Nakatani T, Ohishi A, Okuda H, Higashi Y, Matsuo T, Fujimoto S, Nagasawa K. Mitochondrial dysfunction is involved in P2X7 receptor-mediated neuronal cell death. J Neurochem 2012; 122:1118-28. [PMID: 22774935 DOI: 10.1111/j.1471-4159.2012.07868.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
P2X7 receptor (P2X7R) is known to be a 'death receptor' in immune cells, but its functional expression in non-immune cells such as neurons is controversial. Here, we examined the involvement of P2X7R activation and mitochondrial dysfunction in ATP-induced neuronal death in cultured cortical neurons. In P2X7R- and pannexin-1-expressing neuron cultures, 5 or more mM ATP or 0.1 or more mM BzATP induced neuronal death including apoptosis, and cell death was prevented by oxATP, P2X7R-selective antagonists. ATP-treated neurons exhibited Ca(2+) entry and YO-PRO-1 uptake, the former being inhibited by oxATP and A438079, and the latter by oxATP and carbenoxolone, while P2X7R antagonism with oxATP, but not pannexin-1 blocking with carbenoxolone, prevented the ATP-induced neuronal death. The ATP treatment induced reactive oxygen species generation through activation of NADPH oxidase and activated poly(ADP-ribose) polymerase, but both of them made no or negligible contribution to the neuronal death. Rhodamine123 efflux from neuronal mitochondria was increased by the ATP-treatment and was inhibited by oxATP, and a mitochondrial permeability transition pore inhibitor, cyclosporine A, significantly decreased the ATP-induced neuronal death. In ATP-treated neurons, the cleavage of pro-caspase-3 was increased, and caspase inhibitors, Q-VD-OPh and Z-DEVD-FMK, inhibited the neuronal death. The cleavage of apoptosis-inducing factor was increased, and calpain inhibitors, MDL28170 and PD151746, inhibited the neuronal death. These findings suggested that P2X7R was functionally expressed by cortical neuron cultures, and its activation-triggered Ca(2+) entry and mitochondrial dysfunction played important roles in the ATP-induced neuronal death.
Collapse
Affiliation(s)
- Kentaro Nishida
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Franke H, Verkhratsky A, Burnstock G, Illes P. Pathophysiology of astroglial purinergic signalling. Purinergic Signal 2012; 8:629-57. [PMID: 22544529 DOI: 10.1007/s11302-012-9300-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/01/2012] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are fundamental for central nervous system (CNS) physiology and are the fulcrum of neurological diseases. Astroglial cells control development of the nervous system, regulate synaptogenesis, maturation, maintenance and plasticity of synapses and are central for nervous system homeostasis. Astroglial reactions determine progression and outcome of many neuropathologies and are critical for regeneration and remodelling of neural circuits following trauma, stroke, ischaemia or neurodegenerative disorders. They secrete multiple neurotransmitters and neurohormones to communicate with neurones, microglia and the vascular walls of capillaries. Signalling through release of ATP is the most widespread mean of communication between astrocytes and other types of neural cells. ATP serves as a fast excitatory neurotransmitter and has pronounced long-term (trophic) roles in cell proliferation, growth, and development. During pathology, ATP is released from damaged cells and acts both as a cytotoxic factor and a proinflammatory mediator, being a universal "danger" signal. In this review, we summarise contemporary knowledge on the role of purinergic receptors (P2Rs) in a variety of diseases in relation to changes of astrocytic functions and nucleotide signalling. We have focussed on the role of the ionotropic P2X and metabotropic P2YRs working alone or in concert to modify the release of neurotransmitters, to activate signalling cascades and to change the expression levels of ion channels and protein kinases. All these effects are of great importance for the initiation, progression and maintenance of astrogliosis-the conserved and ubiquitous glial defensive reaction to CNS pathologies. We highlighted specific aspects of reactive astrogliosis, especially with respect to the involvement of the P2X(7) and P2Y(1)R subtypes. Reactive astrogliosis exerts both beneficial and detrimental effects in a context-specific manner determined by distinct molecular signalling cascades. Understanding the role of purinergic signalling in astrocytes is critical to identifying new therapeutic principles to treat acute and chronic neurological diseases.
Collapse
Affiliation(s)
- Heike Franke
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstrasse 16-18, 04107, Leipzig, Germany.
| | | | | | | |
Collapse
|
80
|
Weisman GA, Ajit D, Garrad R, Peterson TS, Woods LT, Thebeau C, Camden JM, Erb L. Neuroprotective roles of the P2Y(2) receptor. Purinergic Signal 2012; 8:559-78. [PMID: 22528682 DOI: 10.1007/s11302-012-9307-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/04/2011] [Indexed: 02/07/2023] Open
Abstract
Purinergic signaling plays a unique role in the brain by integrating neuronal and glial cellular circuits. The metabotropic P1 adenosine receptors and P2Y nucleotide receptors and ionotropic P2X receptors control numerous physiological functions of neuronal and glial cells and have been implicated in a wide variety of neuropathologies. Emerging research suggests that purinergic receptor interactions between cells of the central nervous system (CNS) have relevance in the prevention and attenuation of neurodegenerative diseases resulting from chronic inflammation. CNS responses to chronic inflammation are largely dependent on interactions between different cell types (i.e., neurons and glia) and activation of signaling molecules including P2X and P2Y receptors. Whereas numerous P2 receptors contribute to functions of the CNS, the P2Y(2) receptor is believed to play an important role in neuroprotection under inflammatory conditions. While acute inflammation is necessary for tissue repair due to injury, chronic inflammation contributes to neurodegeneration in Alzheimer's disease and occurs when glial cells undergo prolonged activation resulting in extended release of proinflammatory cytokines and nucleotides. This review describes cell-specific and tissue-integrated functions of P2 receptors in the CNS with an emphasis on P2Y(2) receptor signaling pathways in neurons, glia, and endothelium and their role in neuroprotection.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO 65211-7310, USA.
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Weisman GA, Camden JM, Peterson TS, Ajit D, Woods LT, Erb L. P2 receptors for extracellular nucleotides in the central nervous system: role of P2X7 and P2Y₂ receptor interactions in neuroinflammation. Mol Neurobiol 2012; 46:96-113. [PMID: 22467178 DOI: 10.1007/s12035-012-8263-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/21/2012] [Indexed: 12/16/2022]
Abstract
Extracellular nucleotides induce cellular responses in the central nervous system (CNS) through the activation of ionotropic P2X and metabotropic P2Y nucleotide receptors. Activation of these receptors regulates a wide range of physiological and pathological processes. In this review, we present an overview of the current literature regarding P2X and P2Y receptors in the CNS with a focus on the contribution of P2X7 and P2Y(2) receptor-mediated responses to neuroinflammatory and neuroprotective mechanisms.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO 65211-7310, USA.
| | | | | | | | | | | |
Collapse
|
82
|
Nerve growth factor and brain-derived neurotrophic factor but not granulocyte colony-stimulating factor, nimodipine and dizocilpine, require ATP for neuroprotective activity after oxygen-glucose deprivation of primary neurons. Brain Res 2012; 1448:20-6. [PMID: 22386494 DOI: 10.1016/j.brainres.2012.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/31/2012] [Accepted: 02/07/2012] [Indexed: 11/21/2022]
Abstract
In previous work, we have demonstrated by radiolabeling, mass spectrometry and site-directed mutagenesis that nerve growth factor (NGF) as well as brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are capable of ATP-binding and that this binding appears to be essential for their neuroprotective activity. In this study, we attempted to shed some light on the question whether ATP is a general prerequisite for neuroprotection. Therefore, we used the non-ATP-binding granulocyte colony-stimulating factor (GCSF), the calcium antagonist nimodipine and the NMDA antagonist dizocilpine to find out whether they need ATP for neuroprotection comparable to NGF and BDNF. However, ATP was not necessary for the neuroprotective effects of GCSF, nimodipine and dizocilpine on primary cultures of rat cortical neurons damaged by oxygen-glucose deprivation whereas neuroprotection was demonstrable for NGF and BDNF only when ATP was present in the culture medium at a concentration higher than ca. 0.4nmol/l. In circular dichroism studies ATP caused changes of the secondary structure of NGF but not of GCSF. Taken together, we suggest that ATP is not a general prerequisite for neuroprotectivity but some growth factors like NGF and BDNF can stimulate their receptors only if they have bound ATP.
Collapse
|
83
|
Purinergic signaling in human pluripotent stem cells is regulated by the housekeeping gene encoding hypoxanthine guanine phosphoribosyltransferase. Proc Natl Acad Sci U S A 2012; 109:3377-82. [PMID: 22331909 DOI: 10.1073/pnas.1118067109] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Lesch-Nyhan disease (LND) is an X-linked genetic disorder caused by mutations of the hypoxanthine guanine phosphoribosyltransferase (HPRT) purine biosynthesis gene and characterized by aberrant purine metabolism, deficient basal ganglia dopamine levels, dystonia, and severe neurobehavioral manifestations, including compulsive self-injurious behavior. Although available evidence has identified important roles for purinergic signaling in brain development, the mechanisms linking HPRT deficiency, purinergic pathways, and neural dysfunction of LND are poorly understood. In these studies aimed at characterizing purinergic signaling in HPRT deficiency, we used a lentivirus vector stably expressing an shRNA targeted to the HPRT gene to produce HPRT-deficient human CVB induced pluripotent stem cells and human HUES11 embryonic stem cells. Both CVB and HUES11 cells show >99% HPRT knockdown and demonstrate markedly decreased expression of the purinergic P2Y1 receptor mRNA. In CVB cells, P2Y1 mRNA and protein down-regulation by HPRT knockdown is refractory to activation by the P2Y1 receptor agonist ATP and shows aberrant purinergic signaling, as reflected by marked deficiency of the transcription factor pCREB and constitutive activation of the MAP kinases phospho-ERK1/2. Moreover, HPRT-knockdown CVB cells also demonstrate marked reduction of phosphorylated β-catenin. These results indicate that the housekeeping gene HPRT regulates purinergic signaling in pluripotent human stem cells, and that this regulation occurs at least partly through aberrant P2Y1-mediated expression and signaling. We propose that such mechanisms may play a role in the neuropathology of HPRT-deficiency LND and may point to potential molecular targets for modulation of this intractable neurological phenotype.
Collapse
|
84
|
Fausther M, Lecka J, Soliman E, Kauffenstein G, Pelletier J, Sheung N, Dranoff JA, Sévigny J. Coexpression of ecto-5'-nucleotidase/CD73 with specific NTPDases differentially regulates adenosine formation in the rat liver. Am J Physiol Gastrointest Liver Physiol 2012; 302:G447-59. [PMID: 22135310 PMCID: PMC3287391 DOI: 10.1152/ajpgi.00165.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ectonucleotidases modulate purinergic signaling by hydrolyzing ATP to adenosine. Here we characterized the impact of the cellular distribution of hepatic ectonucleotidases, namely nucleoside triphosphate diphosphohydrolase (NTPDase)1/CD39, NTPDase2/CD39L1, NTPDase8, and ecto-5'-nucleotidase/CD73, and of their specific biochemical properties, on the levels of P1 and P2 receptor agonists, with an emphasis on adenosine-producing CD73. Immunostaining and enzyme histochemistry showed that the distribution of CD73 (protein and AMPase activity) overlaps partially with those of NTPDase1, -2, and -8 (protein levels and ATPase and ADPase activities) in normal rat liver. CD73 is expressed in fibroblastic cells located underneath vascular endothelial cells and smooth muscle cells, which both express NTPDase1, in portal spaces in a distinct fibroblast population next to NTPDase2-positive portal fibroblasts, and in bile canaliculi, together with NTPDase8. In fibrotic rat livers, CD73 protein expression and activity are redistributed but still overlap with the NTPDases mentioned. The ability of the observed combinations of ectonucleotidases to generate adenosine over time was evaluated by reverse-phase HPLC with the recombinant rat enzymes at high "inflammatory" (500 μM) and low "physiological" (1 μM) ATP concentrations. Overall, ATP was rapidly converted to adenosine by the NTPDase1+CD73 combination, but not by the NTPDase2+CD73 combination. In the presence of NTPDase8 and CD73, ATP was sequentially dephosphorylated to the CD73 inhibitor ADP, and then to AMP, thus resulting in a delayed formation of adenosine. In conclusion, the specific cellular cocompartmentalization of CD73 with hepatic NTPDases is not redundant and may lead to the differential activation of P1 and P2 receptors, under normal and fibrotic conditions.
Collapse
Affiliation(s)
- Michel Fausther
- 1Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL), Québec and Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada; ,2Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and ,3Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Joanna Lecka
- 1Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL), Québec and Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada;
| | - Elwy Soliman
- 2Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and
| | - Gilles Kauffenstein
- 1Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL), Québec and Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada;
| | - Julie Pelletier
- 1Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL), Québec and Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada;
| | - Nina Sheung
- 2Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and
| | - Jonathan A. Dranoff
- 2Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and ,3Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jean Sévigny
- 1Centre de recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec (pavillon CHUL), Québec and Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada;
| |
Collapse
|
85
|
del Puerto A, Díaz-Hernández JI, Tapia M, Gomez-Villafuertes R, Benitez MJ, Zhang J, Miras-Portugal MT, Wandosell F, Díaz-Hernández M, Garrido JJ. Adenylate cyclase 5 coordinates the action of ADP, P2Y1, P2Y13 and ATP-gated P2X7 receptors on axonal elongation. J Cell Sci 2012; 125:176-88. [PMID: 22250198 DOI: 10.1242/jcs.091736] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In adult brains, ionotropic or metabotropic purinergic receptors are widely expressed in neurons and glial cells. They play an essential role in inflammation and neurotransmission in response to purines secreted to the extracellular medium. Recent studies have demonstrated a role for purinergic receptors in proliferation and differentiation of neural stem cells although little is known about their role in regulating the initial neuronal development and axon elongation. The objective of our study was to investigate the role of some different types of purinergic receptors, P2Y1, P2Y13 and P2X7, which are activated by ADP or ATP. To study the role and crosstalk of P2Y1, P2Y13 and P2X7 purinergic receptors in axonal elongation, we treated neurons with specific agonists and antagonists, and we nucleofected neurons with expression or shRNA plasmids. ADP and P2Y1-GFP expression improved axonal elongation; conversely, P2Y13 and ATP-gated P2X7 receptors halted axonal elongation. Signaling through each of these receptor types was coordinated by adenylate cyclase 5. In neurons nucleofected with a cAMP FRET biosensor (ICUE3), addition of ADP or Blue Brilliant G, a P2X7 antagonist, increased cAMP levels in the distal region of the axon. Adenylate cyclase 5 inhibition or suppression impaired these cAMP increments. In conclusion, our results demonstrate a crosstalk between two metabotropic and one ionotropic purinergic receptor that regulates cAMP levels through adenylate cyclase 5 and modulates axonal elongation triggered by neurotropic factors and the PI3K-Akt-GSK3 pathway.
Collapse
Affiliation(s)
- Ana del Puerto
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Stanojević I, Drakulić D, Petrović S, Milošević M, Jovanović N, Horvat A. Kinetic characterization of ecto-nucleoside triphosphate diphosphohydrolases in brain nerve terminals during rat postnatal development. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY A 2011. [DOI: 10.1134/s0036024411130292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
87
|
Glaser T, Cappellari AR, Pillat MM, Iser IC, Wink MR, Battastini AMO, Ulrich H. Perspectives of purinergic signaling in stem cell differentiation and tissue regeneration. Purinergic Signal 2011; 8:523-37. [PMID: 22143354 DOI: 10.1007/s11302-011-9282-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/09/2011] [Indexed: 12/20/2022] Open
Abstract
Replacement of lost or dysfunctional tissues by stem cells has recently raised many investigations on therapeutic applications. Purinergic signaling has been shown to regulate proliferation, differentiation, cell death, and successful engraftment of stem cells originated from diverse origins. Adenosine triphosphate release occurs in a controlled way by exocytosis, transporters, and lysosomes or in large amounts from damaged cells, which is then subsequently degraded into adenosine. Paracrine and autocrine mechanisms induced by immune responses present critical factors for the success of stem cell therapy. While P1 receptors generally exert beneficial effects including anti-inflammatory activity, P2 receptor-mediated actions depend on the subtype of stimulated receptors and localization of tissue repair. Pro-inflammatory actions and excitatory tissue damages mainly result from P2X7 receptor activation, while other purinergic receptor subtypes participate in proliferation and differentiation, thereby providing adequate niches for stem cell engraftment and novel mechanisms for cell therapy and endogenous tissue repair. Therapeutic applications based on regulation of purinergic signaling are foreseen for kidney and heart muscle regeneration, Clara-like cell replacement for pulmonary and bronchial epithelial cells as well as for induction of neurogenesis in case of neurodegenerative diseases.
Collapse
Affiliation(s)
- Talita Glaser
- Departamento de Bioquímica , Instituto de Química, Universidade São Paulo, Av. Prof. Lineu Prestes, 748-Bloco 8S/Room 0858, CEP: 05508-900, São Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
88
|
Shirakawa T, Nakano K, Hachiya N, Kato N, Kaneko K. The involvement of P2X1 receptor in pyramidal cell degeneration in the rat hippocampus after trimethyltin administration. Neurosci Res 2011; 71:396-404. [DOI: 10.1016/j.neures.2011.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 07/20/2011] [Accepted: 08/10/2011] [Indexed: 12/14/2022]
|
89
|
Neuropeptide Y and extracellular signal-regulated kinase mediate injury-induced neuroregeneration in mouse olfactory epithelium. Mol Cell Neurosci 2011; 49:158-70. [PMID: 22154958 DOI: 10.1016/j.mcn.2011.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/02/2011] [Accepted: 11/17/2011] [Indexed: 12/29/2022] Open
Abstract
In the olfactory epithelium (OE), injury induces ATP release, and subsequent activation of P2 purinergic receptors by ATP promotes neuroregeneration by increasing basal progenitor cell proliferation. The molecular mechanisms underlying ATP-induced increases in OE neuroregeneration have not been established. In the present study, the roles of neuroproliferative factors neuropeptide Y (NPY) and fibroblast growth factor 2 (FGF2), and p44/42 extracellular signal-regulated kinase (ERK) on ATP-mediated increases of neuroregeneration in the OE were investigated. ATP increased basal progenitor cell proliferation in the OE via activation of P2 purinergic receptors in vitro and in vivo as monitored by incorporation of 5'-ethynyl-2'-deoxyuridine, a thymidine analog, into DNA, and proliferating cell nuclear antigen (PCNA) protein levels. ATP induced p44/42 ERK activation in globose basal cells (GBCs) but not horizontal basal cells (HBCs). ATP differentially regulated p44/42 ERK over time in the OE both in vitro and in vivo with transient inhibition (5-15 min) followed by activation (30 min-1 h) of p44/42 ERK. In addition, ATP indirectly activated p44/42 ERK in the OE via ATP-induced NPY release and subsequent activation of NPY Y1 receptors in the basal cells. There were no synergistic effects of ATP and NPY or FGF2 on OE neuroregeneration. These data clearly have implications for the pharmacological modulation of neuroregeneration in the olfactory epithelium.
Collapse
|
90
|
Illes P, Verkhratsky A, Burnstock G, Franke H. P2X receptors and their roles in astroglia in the central and peripheral nervous system. Neuroscientist 2011; 18:422-38. [PMID: 22013151 DOI: 10.1177/1073858411418524] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Astrocytes are a class of neural cells that control homeostasis at all levels of the central and peripheral nervous system. There is a bidirectional neuron-glia interaction via a number of extracellular signaling molecules, glutamate and ATP being the most widespread. ATP activates ionotropic P2X and metabotropic P2Y receptors, which operate in both neurons and astrocytes. Morphological, biochemical, and functional evidence indicates the expression of astroglial P2X(1/5) heteromeric and P2X(7) homomeric receptors, which mediate physiological and pathophysiological responses. Activation of P2X(1/5) receptors triggers rapid increase of intracellular Na(+) that initiates immediate cellular reactions, such as the depression of the glutamate transporter to keep high glutamate concentrations in the synaptic cleft, the activation of the local lactate shuttle to supply energy substrate to pre- and postsynaptic neuronal structures, and the reversal of the Na(+)/Ca(2+) exchange resulting in additional Ca(2+) entry. The consequences of P2X(7) receptor activation are mostly but not exclusively mediated by the entry of Ca(2+) and result in reorganization of the cytoskeleton, inflammation, apoptosis/necrosis, and proliferation, usually at a prolonged time scale. Thus, astroglia detect by P2X(1/5) and P2X(7) receptors both physiological concentrations of ATP secreted from presynaptic nerve terminals and also much higher concentrations of ATP attained under pathological conditions.
Collapse
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Germany.
| | | | | | | |
Collapse
|
91
|
The role of purinergic receptors in retinal function and disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 664:385-91. [PMID: 20238039 DOI: 10.1007/978-1-4419-1399-9_44] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Extracellular ATP acts as a neurotransmitter in the central and peripheral nervous systems. In this review, the role of purinergic receptors in neuronal signaling and bi-directional glial-neuronal communication in the retina will be considered. There is growing evidence that a range of P2X and P2Y receptors are expressed on most classes of retinal neurons and that activation of P2 receptors modulates retinal function. Furthermore, neuronal control of glial function is achieved through neuronal release of ATP and activation of P2Y receptors expressed by Müller cells. Altered purinergic signaling in Müller cells has been implicated in gliotic changes in the diseased retina and furthermore, elevations in extracellular ATP may lead to apoptosis of retinal neurons.
Collapse
|
92
|
Ferenz KB, Rose K, König S, Krieglstein J. ATP-NGF-complex, but not NGF, is the neuroprotective ligand. Neurochem Int 2011; 59:989-95. [PMID: 21930174 DOI: 10.1016/j.neuint.2011.08.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 08/25/2011] [Accepted: 08/26/2011] [Indexed: 12/12/2022]
Abstract
We have shown previously that nerve growth factor (NGF) requires only low nanomolar ATP concentrations in the cell culture medium to protect cortical rat neurons (CRN) from cellular damage induced by staurosporine (STS). We have also demonstrated before that NGF and other growth factors form stable non-covalent complexes with ATP. Here we demonstrated that 8N(1)ATP-NGF, but not NGF, protected CRN against damage. The photo-reactive ATP derivative 8N(3)ATP was incubated with NGF and was trapped in its position by UV irradiation forming a covalent bond. The cross-link with a molar ratio of 1:1 (8N(1)ATP:NGF) was confirmed by mass spectrometry. Circular dichroism experiments revealed that 8N(1)ATP altered the secondary structure of NGF in the same way as ATP did. Covalently bound 8N(1)ATP-NGF was shown to be stable in the presence of the ATP-hydrolyzing enzyme alkaline phosphatase while the non-covalent ATP-NGF-complex dissociated with the removal of free ATP from the solution. 8N(1)ATP-NGF protected CRN against damage by STS independently of free ATP in the culture medium. These results suggest that the ATP-NGF-complex, but not NGF, is the active ligand.
Collapse
Affiliation(s)
- Katja Bettina Ferenz
- Institut für Pharmazeutische und Medizinische Chemie, Fachbereich Chemie und Pharmazie, Germany
| | | | | | | |
Collapse
|
93
|
Interaction of purinergic receptors with GPCRs, ion channels, tyrosine kinase and steroid hormone receptors orchestrates cell function. Purinergic Signal 2011; 8:91-103. [PMID: 21887492 DOI: 10.1007/s11302-011-9260-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 08/16/2011] [Indexed: 01/26/2023] Open
Abstract
Extracellular purines and pyrimidines have emerged as key regulators of a wide range of physiological and pathophysiological cellular processes acting through P1 and P2 cell surface receptors. Increasing evidence suggests that purinergic receptors can interact with and/or modulate the activity of other classes of receptors and ion channels. This review will focus on the interactions of purinergic receptors with other GPCRs, ion channels, receptor tyrosine kinases, and steroid hormone receptors. Also, the signal transduction pathways regulated by these complexes and their new functional properties are discussed.
Collapse
|
94
|
Burnstock G, Krügel U, Abbracchio MP, Illes P. Purinergic signalling: from normal behaviour to pathological brain function. Prog Neurobiol 2011; 95:229-74. [PMID: 21907261 DOI: 10.1016/j.pneurobio.2011.08.006] [Citation(s) in RCA: 315] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 08/12/2011] [Accepted: 08/15/2011] [Indexed: 02/07/2023]
Abstract
Purinergic neurotransmission, involving release of ATP as an efferent neurotransmitter was first proposed in 1972. Later, ATP was recognised as a cotransmitter in peripheral nerves and more recently as a cotransmitter with glutamate, noradrenaline, GABA, acetylcholine and dopamine in the CNS. Both ATP, together with some of its enzymatic breakdown products (ADP and adenosine) and uracil nucleotides are now recognised to act via P2X ion channels and P1 and P2Y G protein-coupled receptors, which are widely expressed in the brain. They mediate both fast signalling in neurotransmission and neuromodulation and long-term (trophic) signalling in cell proliferation, differentiation and death. Purinergic signalling is prominent in neurone-glial cell interactions. In this review we discuss first the evidence implicating purinergic signalling in normal behaviour, including learning and memory, sleep and arousal, locomotor activity and exploration, feeding behaviour and mood and motivation. Then we turn to the involvement of P1 and P2 receptors in pathological brain function; firstly in trauma, ischemia and stroke, then in neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's, as well as multiple sclerosis and amyotrophic lateral sclerosis. Finally, the role of purinergic signalling in neuropsychiatric diseases (including schizophrenia), epilepsy, migraine, cognitive impairment and neuropathic pain will be considered.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK.
| | | | | | | |
Collapse
|
95
|
Ohtani M, Ohura K, Oka T. Involvement of P2X receptors in the regulation of insulin secretion, proliferation and survival in mouse pancreatic β-cells. Cell Physiol Biochem 2011; 28:355-66. [PMID: 21865744 DOI: 10.1159/000331752] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2011] [Indexed: 12/18/2022] Open
Abstract
In order to clarify the functional role of ionotropic purinergic (P2X) receptors in pancreatic β-cells, we examined the effect of several P2 receptor agonists and antagonists on insulin secretion by mouse pancreatic islets, mouse Beta-TC6 cell proliferation and survival of dispersed islet cells in culture. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed the expression of mRNAs of P2X(4) receptor in mouse islets and P2X(1), P2X(2), P2X(3), P2X(4), P2X(5) and P2X(7) receptors in Beta-TC6 cells. The presence of P2X(4) receptor proteins in islets and Beta-TC6 cells was confirmed by immunofluorescent staining and Western blot analysis. We have previously found that the functional P2Y(1) receptor but not P2Y(2) and P2Y(4) receptors was present in islets. In this study we found that a nonspecific P2 receptor agonist, ATP (1 μM) stimulated insulin secretion by islets in the presence of high glucose (20 mM) in culture. The effect of ATP was partially inhibited by a P2 receptor antagonist PPADS as well as a P2Y(1) receptor antagonist MRS2179. In addition, a P2X(4) receptor potentiator ivermectin per se augmented glucose-induced insulin secretion and slightly potentiated the effect of ATP. These results suggested the involvement of P2Y(1)and P2X receptors. We also found that ATP inhibited proliferation of Beta-TC6 cells in a concentration-dependent manner during 72 h culture. The inhibitory effect of ATP was completely reversed by PPADS and partially by treating cells with small interfering RNA targeted for P2X(4) receptor mRNA. Furthermore, we found that the phosphorylation of the extracellular signal-regulated kinase 1 and 2 (ERK1/2) was suppressed by treatment with ATP in Beta-TC6 cells. In addition, we found that ATP reduced the cell viability and DNA synthesis of islet cells in culture. The effect of ATP on the cell viability was blocked by PPADS or MRS2179. These results suggested that P2X receptors as well as the P2Y(1) receptor played a role in the modulation of insulin secretion, proliferation and cell viability in mouse pancreatic β-cells.
Collapse
Affiliation(s)
- Masahiro Ohtani
- Department of Pharmacology, Osaka Dental University, Hirakata, Japan
| | | | | |
Collapse
|
96
|
Ortega F, Pérez-Sen R, Delicado EG, Teresa Miras-Portugal M. ERK1/2 activation is involved in the neuroprotective action of P2Y13 and P2X7 receptors against glutamate excitotoxicity in cerebellar granule neurons. Neuropharmacology 2011; 61:1210-21. [PMID: 21798274 DOI: 10.1016/j.neuropharm.2011.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 07/05/2011] [Accepted: 07/08/2011] [Indexed: 10/17/2022]
Abstract
Cerebellar granule neurons express several types of nucleotide receptors, with the metabotropic P2Y(13) and the ionotropic P2X7 being the most relevant in this model. In the present study we investigated the role of P2Y(13) and P2X7 nucleotide receptors in ERK1/2 signalling. The nucleotidic agonists 2MeSADP (2-methylthioadenosine-5'-diphosphate) for P2Y(13) and BzATP (2'(3')-O-(4-benzoylbenzoyl)adenosine-5'-triphosphate) for P2X7 receptors were coupled to ERK1/2 activation in granule neurons, being able to increase around two-fold the levels of ERK1/2 phosphorylation. These effects were sensitive to the inhibitory action of the antagonists MRS-2211 and A-438079, specific for P2Y(13) and P2X7 receptors, respectively. Although both receptor subtypes shared the same pattern of transient ERK1/2 phosphorylation, they differed in the intracellular cascades they triggered, being PI3K-dependent for P2Y(13) and calcium/calmodulin kinase II (CaMKII)-dependent for P2X7. These two different ERK-mediated pathways were involved in the neuroprotective effects displayed by both P2Y(13) and P2X7 receptors against apoptosis induced by an excitotoxic concentration of glutamate, in a similar manner to the neurotrophin, BDNF. In addition, P2Y(13) and P2X7 receptor agonists were also able to phosphorylate and activate the ERK-dependent target CREB, which could be involved in their neuroprotective effect. These results indicate that nucleotide receptors share with trophic factors the same survival routes in neurons, such as the ERK signalling route, and therefore, can contribute to the maintenance of granule neurons in conditions in which survival is being compromised.
Collapse
Affiliation(s)
- Felipe Ortega
- Department of Biochemistry, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain.
| | | | | | | |
Collapse
|
97
|
Rico EP, Rosemberg DB, Langoni ADS, Souto AA, Dias RD, Bogo MR, Bonan CD, Souza DO. Chronic ethanol treatment alters purine nucleotide hydrolysis and nucleotidase gene expression pattern in zebrafish brain. Neurotoxicology 2011; 32:871-8. [PMID: 21704070 DOI: 10.1016/j.neuro.2011.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 04/12/2011] [Accepted: 05/23/2011] [Indexed: 12/20/2022]
Abstract
Ethanol is a widely consumed drug that acts on the central nervous system (CNS), modifying several signal transduction pathways activated by hormones and neurotransmitters. The zebrafish is an experimental model for the study of human diseases and the use of this species in biochemical and behavioral studies on alcoholism and alcohol-dependence has increased recently. However, there are no data concerning the effects of chronic ethanol exposure on the purinergic system, where extracellular nucleotides act as signaling molecules. Purinergic signaling is controlled by a group of enzymes named ectonucleotidases, which include NTPDases and ecto-5'-nucleotidase already characterized in zebrafish brain. The aim of this study was to evaluate nucleotide hydrolysis by NTPDases and ecto-5'-nucleotidase after long-term ethanol exposure. Additionally, the gene expression patterns of NTPDases1-3 and 5'-nucleotidase were determined. Animals were exposed to 0.5% ethanol for 7, 14, and 28 days. There were no significant changes in ATP and GTP hydrolysis after all treatments. However, a decrease in ADP (46% and 34%) and GDP (48% and 36%) hydrolysis was verified after 7 and 14 days, respectively. After 7 and 14 days of ethanol exposure, a significant decrease in AMP hydrolysis (48% and 36%) was also observed, whereas GMP hydrolysis was inhibited only after 7 days (46%). NTPDase2_mv and NTPDase3 mRNA transcript levels decreased after 7 and 14 days, respectively. In contrast, ethanol increased NTPDase1, NTPDase2_mq, and NTPDase3 transcript levels after 28 days of exposure. NTPDase2_mg and 5'-nucleotidase gene expression was not altered. Therefore, the ectonucleotidase pathway may be a target of chronic ethanol toxicity and the regulation of purinergic system could play a key role in the neurochemical mechanisms underlying the effects of ethanol on the CNS.
Collapse
Affiliation(s)
- Eduardo Pacheco Rico
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Blockade of P2 nucleotide receptors after spinal cord injury reduced the gliotic response and spared tissue. J Mol Neurosci 2011; 46:167-76. [PMID: 21647706 DOI: 10.1007/s12031-011-9567-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/23/2011] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) triggers a sequel of events commonly associated with cell death and dysfunction of glias and neurons surrounding the lesion. Although astrogliosis and glial scar formation have been involved in both damage and repair processes after SCI, their role remains controversial. Our goal was to investigate the effects of the P2 receptors antagonists, PPADS and suramin, in the establishment of the reactive gliosis and the formation of the glial scar. Molecular biology, immunohistochemistry, spared tissue, and locomotor behavioral studies were used to evaluate astrogliosis, in adult female Sprague-Dawley rats treated with P2 antagonists after moderate injury with the NYU impactor device. Semi-quantitative RT-PCR confirmed the presence of P2Y(1,) P2Y(2,) P2Y(4,) P2Y(6,) P2Y(12), and P2X(2) receptors in the adult spinal cord. Immunohistochemistry studies confirmed a significant decrease in GFAP-labeled cells at the injury epicenter as well as a decrease in spared tissue after treatment with the antagonists. Functional open field testing revealed no significant locomotor score differences between treated and control animals. Our work is consistent with studies suggesting that astrogliosis is an important event after SCI that limits tissue damage and lesion spreading.
Collapse
|
99
|
Okuda H, Higashi Y, Nishida K, Fujimoto S, Nagasawa K. Contribution of P2X7 receptors to adenosine uptake by cultured mouse astrocytes. Glia 2011; 58:1757-65. [PMID: 20645413 DOI: 10.1002/glia.21046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Nucleotides and nucleosides play important roles by maintaining brain homeostasis, and their extracellular concentrations are mainly regulated by ectonucleotidases and nucleoside transporters expressed by astrocytes. Extracellularly applied NAD(+) prevents astrocyte death caused by excessive activation of poly(ADP-ribose) polymerase-1, of which the molecular mechanism has not been fully elucidated. Recently, exogenous NAD(+) was reported to enter astrocytes via the P2X7 receptor (P2X7R)-associated channel/pore. In this study, we examined whether the intact form of NAD(+) is incorporated into astrocytes. A large portion of extracellularly added NAD(+) was degraded into metabolites such as AMP and adenosine in the extracellular space. The uptake of adenine ring-labeled [(14)C]NAD(+), but not nicotinamide moiety-labeled [(3)H]NAD(+), showed time- and temperature-dependency, and was significantly enhanced on addition of apyrase, and was reduced by 8-Br-cADPR and ARL67156, inhibitors of CD38 and ectoapyrase, respectively, and P2X7R knockdown, suggesting that the detected uptake of [(14)C]NAD(+) resulted from [(14)C]adenosine acting as a metabolite of [(14)C]NAD(+). Pharmacological and genetic inhibition of P2X7R with brilliant blue G, KN-62, oxATP, and siRNA transfection resulted in a decrease of [(3)H]adenosine uptake, and the uptake was also reduced by low concentration of carbenoxolone and pannexin1 selective peptide blocker (10)panx. Taken together, these results indicate that exogenous NAD(+) is degraded by ectonucleotidases and that adenosine, as its metabolite, is taken up into astrocytes via the P2X7R-associated channel/pore.
Collapse
Affiliation(s)
- Hiroto Okuda
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, Japan
| | | | | | | | | |
Collapse
|
100
|
Hracskó Z, Baranyi M, Csölle C, Gölöncsér F, Madarász E, Kittel A, Sperlágh B. Lack of neuroprotection in the absence of P2X7 receptors in toxin-induced animal models of Parkinson's disease. Mol Neurodegener 2011; 6:28. [PMID: 21542899 PMCID: PMC3113297 DOI: 10.1186/1750-1326-6-28] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 05/04/2011] [Indexed: 01/06/2023] Open
Abstract
Background Previous studies indicate a role of P2X7 receptors in processes that lead to neuronal death. The main objective of our study was to examine whether genetic deletion or pharmacological blockade of P2X7 receptors influenced dopaminergic cell death in various models of Parkinson's disease (PD). Results mRNA encoding P2X7 and P2X4 receptors was up-regulated after treatment of PC12 cells with 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP). P2X7 antagonists protected against MPTP and rotenone induced toxicity in the LDH assay, but failed to protect after rotenone treatment in the MTT assay in PC12 cells and in primary midbrain culture. In vivo MPTP and in vitro rotenone pretreatments increased the mRNA expression of P2X7 receptors in the striatum and substantia nigra of wild-type mice. Basal mRNA expression of P2X4 receptors was higher in P2X7 knockout mice and was further up-regulated by MPTP treatment. Genetic deletion or pharmacological inhibition of P2X7 receptors did not change survival rate or depletion of striatal endogenous dopamine (DA) content after in vivo MPTP or in vitro rotenone treatment. However, depletion of norepinephrine was significant after MPTP treatment only in P2X7 knockout mice. The basal ATP content was higher in the substantia nigra of wild-type mice, but the ADP level was lower. Rotenone treatment elicited a similar reduction in ATP content in the substantia nigra of both genotypes, whereas reduction of ATP was more pronounced after rotenone treatment in striatal slices of P2X7 deficient mice. Although the endogenous amino acid content remained unchanged, the level of the endocannabinoid, 2-AG, was elevated by rotenone in the striatum of wild-type mice, an effect that was absent in mice deficient in P2X7 receptors. Conclusions We conclude that P2X7 receptor deficiency or inhibition does not support the survival of dopaminergic neurons in an in vivo or in vitro models of PD.
Collapse
Affiliation(s)
- Zsuzsanna Hracskó
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1083 Budapest, Szigony u, 43, Hungary.
| | | | | | | | | | | | | |
Collapse
|