Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Thromb Thrombolysis ; 22(3): 213-20, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17111196

ABSTRACT

BACKGROUND: P2Y12 is the major platelet receptor that mediates ADP-induced aggregation. P2Y12 is also expressed by vascular cells. The factors that regulate P2Y12 expression have not been determined. Since nicotine (NIC) has effects on platelet activation and vascular function, and because nicotinic and purinerigic receptors may interact, we determined whether nicotine altered P2Y12 expression. METHODS: Four cell lines (human coronary artery endothelial cells, HCAEC; human umbilical vein endothelial cells, HUVEC; human aortic smooth muscle cells, HASMC; and human megakaryoblastic cells, MEG-01) were cultured in the absence or presence of nicotine. Immunoblotting for P2Y12, P2Y2, and actin was performed. RESULTS: Nicotine, at concentrations of 0.1-1.0 microM, induced P2Y12 (but not P2Y2) expression in all the four cell lines. HASMC exhibited the greatest induction with a sixfold mean increase in P2Y12 expression in response to 0.25 microM nicotine. The induction was inhibited by nicotinic acetylcholine receptor antagonists. Healthy smokers were observed to have higher P2Y12 expression in platelet lysates compared to non-smokers. CONCLUSION: Nicotine induces the expression of P2Y12 in vascular cells and megakaryoblasts, and is mediated by nicotinic acetylcholine receptors. Smokers exhibit higher platelet P2Y12, possibly mediated via nicotine. These results may contribute to a better understanding of the effects of cigarette smoking on platelet activation and the vessel wall. CONDENSED ABSTRACT: The factors that regulate the expression of P2Y12, the platelet ADP receptor, have not been determined. Four cell lines (human coronary artery endothelial cells, HCAEC; human umbilical vein endothelial cells, HUVEC; human aortic smooth muscle cells, HASMC; and human megakaryoblastic cells, MEG-01) were cultured in the absence or presence of nicotine. Nicotine, at concentrations of 0.1-1.0 microM, induced P2Y12 expression in all the four cell lines. HASMC exhibited the greatest induction with a sixfold mean increase in P2Y12 expression in response to 0.25 microM nicotine. The induction was inhibited by nicotinic acetylcholine receptor antagonists. Healthy smokers were observed to have higher P2Y12 expression in platelet lysates compared to non-smokers. These results may contribute to a better understanding of the effects of cigarette smoking on platelet activation and the vessel wall.


Subject(s)
Endothelial Cells/metabolism , Megakaryocytes/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Purinergic P2/metabolism , Adult , Female , Humans , Male , Middle Aged , Platelet Activation/drug effects , Receptors, Purinergic P2Y12 , Smoking/adverse effects , Tumor Cells, Cultured , Up-Regulation/drug effects
2.
Biol Trace Elem Res ; 109(3): 267-80, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16632895

ABSTRACT

Glutamate is removed mainly by astrocytes from the extracellular fluid via high-affinity astroglial Na+ -dependent excitatory amino acid transporters, glutamate/aspartate transporter (GLAST), and glutamate transporter-1 (GLT-1). Mercuric chloride (HgCl2) is a highly toxic compound that inhibits glutamate uptake in astrocytes, resulting in excessive extracellular glutamate accumulation, leading to excitotoxicity and neuronal cell death. The mechanisms associated with the inhibitory effects of HgCl2 on glutamate uptake are unknown. This study examines the effects of HgCl2 on the transport of 3H-D-aspartate, a nonmetabolizable glutamate analog, using Chinese hamster ovary cells (CHO) transfected with two glutamate transporter subtypes, GLAST (EAAT1) and GLT-1 (EAAT2), as a model system. Additionally, studies were undertaken to determine the effects of HgCl2 on mRNA and protein levels of these transporters. The results indicate that (1) HgCl2 leads to significant (p < 0.001) inhibition of glutamate uptake via both transporters, but is a more potent inhibitor of glutamate transport via GLAST and (2) the effect of HgCl2 on inhibition of glutamate uptake in transfected CHO cells is not associated with changes in transporter protein levels despite a significant decrease in mRNA expression; thus, (3) HgCl2 inhibition is most likely related to its direct binding to the functional thiol groups of the transporters and interference with their uptake function.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamates/metabolism , Mercuric Chloride/pharmacology , Animals , Biological Transport/drug effects , CHO Cells , Cell Line , Cricetinae , D-Aspartic Acid/analogs & derivatives , D-Aspartic Acid/metabolism , Excitatory Amino Acid Transporter 2/genetics , Glutamate Plasma Membrane Transport Proteins/genetics , In Vitro Techniques , Mercuric Chloride/metabolism , Mutation , RNA, Messenger/metabolism , Sulfhydryl Compounds/metabolism , Time Factors , Transfection
3.
Biol Trace Elem Res ; 105(1-3): 71-86, 2005.
Article in English | MEDLINE | ID: mdl-16034155

ABSTRACT

Thimerosal, also known as thimersal, Merthrolate, or sodiumethyl-mercurithiosalicylate, is an organic mercurial compound that is used in a variety of commercial as well as biomedical applications. As a preservative, it is used in a number of vaccines and pharmaceutical products. Its active ingredient is ethylmercury. Both inorganic and organic mercurials are known to interfere with glutamate homeostasis. Brain glutamate is removed mainly by astrocytes from the extracellular fluid via high-affinity astroglial Na+-dependent excitatory amino acid transporters, glutamate/ aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1). The effects of thimerosal on glutamate homeostasis have yet to be determined. As a first step in this process, we examined the effects of thimerosal on the transport of [3H]-d-aspartate, a nonmetabolizable glutamate analog, in Chinese hamster ovary (CHO) cells transfected with two glutamate transporter subtypes, GLAST (EAAT1) and GLT-1 (EAAT2). Additionally, studies were undertaken to determine the effects of thimerosal on mRNA and protein levels of these transporters. The results indicate that thimerosal treatment caused significant but selective changes in both glutamate transporter mRNA and protein expression in CHO cells. Thimerosal-mediated inhibition of glutamate transport in the CHO-K1 cell line DdB7 was more pronounced in the GLT-1-transfected cells compared with the GLAST- transfected cells. These studies suggest that thimerosal accumulation in the central nervous system might contribute to dysregulation of glutamate homeostasis.


Subject(s)
Amino Acid Transport System X-AG/genetics , Ethylmercury Compounds/pharmacology , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/pharmacokinetics , Thimerosal/pharmacology , Animals , Biological Transport , Blotting, Northern , Blotting, Western , Brain/metabolism , CHO Cells , Cell Line , Central Nervous System/drug effects , Cricetinae , DNA, Complementary/metabolism , In Vitro Techniques , Preservatives, Pharmaceutical/pharmacology , RNA, Messenger/metabolism , Time Factors , Transfection
4.
Brain Res Mol Brain Res ; 137(1-2): 11-22, 2005 Jun 13.
Article in English | MEDLINE | ID: mdl-15950756

ABSTRACT

Excessive free radical formation has been implicated as one of the causative factors in neurotoxic damage associated with variety of metals, including methylmercury (MeHg). Although the mechanism(s) associated with MeHg-dependent neurotoxicity remains far from clear, overwhelming data give credence to a mediatory role for astrocytes, a major cell type that preferentially accumulates MeHg. To extend our recent findings of MeHg-induced increase in ROS formation (G. Shanker, J.L. Aschner, T. Syversen et al., Free radical formation in cerebral cortical astrocytes in culture induced by methylmercury, Mol. Brain Res. 128 (2004) 48-57), the present studies were designed to assess the effect of modulating intracellular glutathione (GSH) content, on ROS generation, in the absence and presence of MeHg. Intracellular GSH was reduced by treatment with 100 microM buthionine-L-sulfoxane (BSO) for 24 h, and increased by treatment with 1 mM l-2-oxothiazolidine-4-carboxylic acid (OTC) for 24 h. Additionally, the effects of the selective antioxidants, catalase (1000 U/ml for 1 h), an H2O2 scavenger, and n-propyl gallate (100 microM for 1 h), a superoxide radical (*O2-) and possibly hydroxyl radical (*OH) scavenger on MeHg-induced ROS formation were examined. After these treatments, astrocytes were exposed to +/-10 microM MeHg for 30 min, following which the fluorescent probes, CM-H2DCFA and CM-H2XRos were added; 20 min later, laser scanning confocal microscopy (LSCM) images were obtained. Exposure of astrocytes for 24 h to 100 microM BSO, a GSH synthesis inhibitor, led to a significant increase in mitochondrial ROS (i.e., *O2-, *NO, and ONOO-) formation, as assessed with CM-H2XRos mitotracker red dye. Similarly, BSO increased ROS formation in various intracellular organelles, as assessed with CM-H2DCFDA. BSO in combination with MeHg increased fluorescence levels in astrocytes to levels above those noted with BSO or MeHg alone, but this effect was statistically indistinguishable from either of these groups (BSO or MeHg). Pretreatment of astrocytes for 24 h with 1 mM OTC abolished the MeHg-induced increase in ROS. Results similar to those obtained with OTC were observed with the free radical scavenger, n-propyl gallate (n-PG). The latter had no significant effects on astrocytic fluorescence when administered alone. This *O2- and possibly *OH radical scavenger significantly attenuated MeHg-induced ROS formation. Catalase, an H2O2 scavenger, was less effective in reducing MeHg-induced ROS formation. Taken together, these studies point to the important protective effect of adequate intracellular GSH content as well as antioxidants against MeHg-triggered oxidative stress in primary astrocyte cultures.


Subject(s)
Antioxidants/pharmacology , Astrocytes/drug effects , Free Radicals/metabolism , Glutathione/drug effects , Methylmercury Compounds/antagonists & inhibitors , Oxidative Stress/drug effects , Animals , Animals, Newborn , Antioxidants/therapeutic use , Astrocytes/metabolism , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Fluorescent Dyes , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Glutathione/metabolism , Indicators and Reagents , Mercury Poisoning, Nervous System/metabolism , Methylmercury Compounds/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Pyrrolidonecarboxylic Acid , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacology , Thiazolidines
5.
Brain Res Mol Brain Res ; 128(1): 48-57, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15337317

ABSTRACT

Oxidative stress has been implicated in neurotoxic damage associated with various metals, including methylmercury (MeHg). Although the mechanism(s) of MeHg-induced neurotoxicity remains unclear, evidence supports a mediatory role for astrocytes, a cell type that preferentially accumulates MeHg. Using scanning confocal microscopy (LSCM), the present study was undertaken to examine the role of astrocytes as the site of reactive oxygen species (ROS). Three redox-sensitive fluorescent probes were used for ROS analysis, (a) CM-H2DCFDA (chloromethyl derivative of dichlorodihydrofluorescein diacetate), a probe for intracellular hydrogen peroxide (H2O2); (b) hydroethidine (HETH), a probe for superoxide anion (*O2-), and (c) CM-H2XRos (chloromethyl derivative of dihydro X-rosamine), and a probe that is selective for mitochondrial reactive oxygen intermediates. Astrocytes were treated with 10 microM MeHg for 30 min, following which the various fluorescent probes were added; 20 min later LSCM images were collected. Astrocytes loaded with CM-H2DCFDA and HE demonstrated a significant MeHg-induced increase in fluorescence intensity indicative of increased intracellular H2O2 and *O2-, respectively. Similar results were obtained with the mitotracker dye, CM-H2XRos. Additionally, exposure of astrocytes for 24 h to 100 microM buthionine-L-sulfoxane (BSO), a glutathione (GSH) synthesis inhibitor, caused a significant increase in ROS formation. Furthermore, BSO pretreatment significantly enhanced the MeHg-induced formation of *O2-, indicating an important role for GSH in the maintenance of optimal cellular redox status. Time-course experiments performed in the simultaneous presence of CM-H2XRos and CM-H2DCFDA demonstrated that the MeHg-induced CM-H2XRos fluorescence changes preceded those of CM-H2DCFDA, suggesting that the mitochondria represent an early primary site for ROS formation. Taken together, these studies illustrate that MeHg induces the generation of astrocyte-derived ROS and support a role for astrocytic ROS in MeHg-associated neurotoxic damage.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Cortex/cytology , Methylmercury Compounds/pharmacology , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Cerebral Cortex/metabolism , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/metabolism , Microscopy, Confocal , Mitochondria/metabolism , Oxidants/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
6.
Neurotoxicology ; 25(3): 399-406, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15019302

ABSTRACT

Cytosolic phospholipase A2 (cPLA2) plays an important role in the stimulus-dependent hydrolysis of sn-2 ester bond from membrane phospholipids, releasing arachidonic acid (AA), which along with its metabolites is involved in a number of regulatory functions. The present study examined the effect of methylmercury (MeHg; 0, 2.5, 5.0 microM) on cPLA2 activation in primary hippocampal neurons by assessing the release of 3H-AA. A significant increase in AA release was observed in cultures treated with 5 microM MeHg (10, 30, 60 and 120 min). This effect was due to neuronal cPLA2 activation, since it was completely abolished by arachidonyl trifluoromethyl ketone (AACOCF3), a specific inhibitor of cPLA2. Additional studies confirmed, by means of western blot analysis, that MeHg (5.0 and 10 microM; 16h) potently increases neuronal cPLA2 protein expression. These results suggest that cPLA2-stimulated hydrolysis and release of AA are potential mediators of MeHg-induced neurotoxicity.


Subject(s)
Arachidonic Acid/metabolism , Cytosol/drug effects , Methylmercury Compounds/pharmacology , Neurons/drug effects , Phospholipases A/biosynthesis , Animals , Cells, Cultured , Cytosol/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/metabolism , Phospholipases A/genetics , Phospholipases A2 , Rats
7.
Biol Trace Elem Res ; 95(1): 1-10, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14555794

ABSTRACT

Methylmercury (MeHg) is a potent neurotoxicant. Any source of environmental mercury represents a potential risk for human MeHg poisoning, because the methylation of inorganic mercury to MeHg in waterways results ultimately in its accumulation in the sea food chain, which represents the most prevalent source for human consumption. A small amount of MeHg accumulates in the central nervous system (CNS), particularly in astrocytes. Astrocytic swelling, excitatory amino acid (EAA) release and uptake inhibition, as well as EAA transporter expression inhibition are known sequelae of MeHg exposure. Herein, we review the effect of MeHg on additional transport systems (for cystine and cysteine) as well as arachidonic acid (AA) release and cytosolic phospholipase A2 (cPLA2) regulation and attempt to integrate the effects of MeHg in astrocytes within a mechanistic hypothesis that explains the inability of these cells to maintain control of the proper milieu of the extracellular fluid and, in turn, leads to neuronal demise.


Subject(s)
Astrocytes/drug effects , Astrocytes/pathology , Mercury Poisoning, Nervous System/metabolism , Methylmercury Compounds/toxicity , Animals , Arachidonic Acid/biosynthesis , Astrocytes/metabolism , Cysteine/antagonists & inhibitors , Cysteine/metabolism , Cystine/antagonists & inhibitors , Cystine/metabolism , Enzyme Activation/drug effects , Glutathione/metabolism , Humans , Mercury Poisoning, Nervous System/pathology , Phospholipases A/metabolism , Phospholipases A2
8.
Brain Res Mol Brain Res ; 110(1): 85-91, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12573536

ABSTRACT

Excessive generation of reactive oxygen species (ROS) has been suggested as a causal factor in various neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease [Brain Res. 830 (1999) 10-15; Biochem. J. 310 (1995) 83-90; Free Radic. Biol. Med. 27 (1999) 612-616]. The present work examined the role of ROS in the neurotoxicity of methylmercury (MeHg). ROS formation in primary astrocytic cultures of neonatal rat cerebral cortex was monitored by 2',7'-dichlorodihydrofluorescein diacetate (H(2)DCF-DA) fluorescence. MeHg, at 10 and 20 microM caused a significant increase in ROS formation (10 microM, P<0.01; 20 microM, P<0.001). Additional studies established the effectiveness of antioxidants/free radical scavengers in attenuating the MeHg-stimulated ROS formation in the following rank-order: (1) Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid), a non-thiol containing antioxidant, (2) n-propyl gallate (PG), a free radical scavenger, (3) superoxide dismutase (SOD), an antioxidant enzyme that dismutates superoxide anion radical, (4) alpha-phenyl-tert-butyl nitrone (PBN), a lipophilic hydroxyl radical spin trapping agent. A significant inhibition of MeHg-induced ROS generation was also noted in astrocytes preincubated (3 h) with arachidonyl trifluoromethyl ketone (AACOCF(3,) 20 microM, P<0.05), a specific inhibitor of cytosolic phospholipase A(2) (cPLA(2)). Conversely, pretreatment (24 h) with 100 microM buthionine-L-sulfoxamine [BSO, a glutathione (GSH) synthesis inhibitor], significantly increased (P<0.05) ROS formation in MeHg treated astrocytes compared to controls. Combined, these studies invoke ROS as potent mediators of MeHg cytotoxicity and support the hypothesis that excessive ROS generation, at least in part, plays an important role in MeHg-induced neurotoxicity.


Subject(s)
Antioxidants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Chromans/pharmacology , Methylmercury Compounds/toxicity , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Arachidonic Acids/pharmacology , Astrocytes/cytology , Cells, Cultured , Cyclic N-Oxides , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Neuroprotective Agents/pharmacology , Nitrogen Oxides/pharmacology , Propyl Gallate/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/pharmacology
9.
Brain Res Mol Brain Res ; 106(1-2): 1-11, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12393259

ABSTRACT

Cytosolic phospholipase A(2) (cPLA(2)) stimulates the hydrolysis of sn-2 ester bond in membrane phospholipids releasing arachidonic acid (AA) and lysophospholipids. The present study examined the effect of methylmercury (MeHg) on cPLA(2) activation and AA release in primary cultures of neonatal rat cerebral astrocytes. Astrocytes were preloaded overnight at 37 degrees C with 3H-AA to metabolically label phospholipids. The effect of MeHg on the activation of cPLA(2) was measured by the release of 3H-AA from astrocytes over 120 min. MeHg (5 microM) caused a significant increase in AA release at 10, 30, 60, and 120 min, whereas 2.5 microM MeHg significantly increased AA release only at 120 min. MeHg-induced increase in 3H-AA release was due to cPLA(2) activation, since arachidonyl trifluoromethyl ketone (AACOCF(3)), a selective inhibitor of cPLA(2), completely abolished MeHg's effect. Consistent with these observations, MeHg (5.0 and 10.0 microM) increased cPLA(2) mRNA (6 h) and cPLA(2) protein expression (5.0 and 10.0 microM; 24 h). The time-course of these effects suggests an immediate direct or indirect effect of MeHg on cPLA(2) activation and 3H-AA release as well as a long-term effect involving the induction of cPLA(2). Thin layer chromatographic analysis of 3H-AA-labeled phospholipids showed that MeHg-stimulated astrocyte 3H-AA release was not due to increased incorporation of 3H-AA into the putative substrates of cPLA(2). These results invoke cPLA(2) as a putative target for MeHg toxicity, and support the notion that cPLA(2)-stimulated hydrolysis and release of AA play a critical role in MeHg-induced neurotoxicity.


Subject(s)
Arachidonic Acids/metabolism , Astrocytes/metabolism , Methylmercury Compounds/pharmacology , Phospholipases A/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Phospholipases A/antagonists & inhibitors , Phospholipases A/genetics , Phospholipases A2 , Phospholipids/chemistry , Phospholipids/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stearic Acids/metabolism , Time Factors
10.
Neurotoxicology ; 23(2): 165-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12224757

ABSTRACT

The present study focused on central nervous system (CNS) transport kinetics of manganese phosphate and manganese sulfate; these findings were correlated with the transport kinetics of manganese chloride (MnCl2), a model Mn compound that has been previously studied. A series of studies was performed to address the transport of Mn salts in confluent cultured endothelial cells. The initial rate of uptake (5 min) of Mn salts (chloride, sulfate, and phosphate) in rat brain endothelial (RBE4) cell cultures is salt-dependent, with the highest rates of uptake for Mn chloride and Mn sulfate (as reflected by the greatest displacement of 54Mn compared with control). Mn phosphate had a lower rate of uptake than the other two Mn salts. These data show that brain endothelial cells efficiently transport Mn sulfate.


Subject(s)
Brain/cytology , Brain/metabolism , Chlorides/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Manganese Compounds/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Sulfates/pharmacokinetics , Animals , Cells, Cultured , Rats
11.
Neurotoxicology ; 23(6): 755-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12520765

ABSTRACT

Methylmercury (MeHg) is a highly neurotoxic, environmentally ubiquitous chemical that exerts its toxic effects by largely unknown mechanisms. Maintenance of optimal intracellular concentrations of glutathione (GSH) is vital for cellular defenses against damage from free radicals. Since astrocytes play an essential role in providing GSH precursors to neurons, studies were directed at the effect of MeHg on cystine transport in both cell types. Astrocytes accumulated cystine via three independent transporters, referred to as system XAG-, system XC-, and gamma-glutamyltranspeptidase (GGT). In contrast, neurons accumulated cystine exclusively via system XC- and GGT. MeHg potently inhibited cystine uptake in astrocytes (but not in neurons), and this effect could be fully accounted for by inhibition of the system XAG- transporter. The transport of glutamate in astrocytes is also inhibited by reactive oxygen species (ROS). Accordingly, additional studies examined the ability of thiol reducing or oxidizing agents to inhibit the astrocytic transport of 3H-D-aspartate, a glutamate analog. The antioxidant catalase significantly attenuated MeHg-induced inhibition of astrocytic 3H-aspartate uptake. Combinedly, these studies suggest that inhibition of cystine uptake and decreased astrocytic GSH levels and efflux reduce the availability of precursors for GSH synthesis in neurons. In addition, MeHg-induced generation of H2O2 plays a role in the inhibition of astrocytic glutamate transport. These effects likely increase neuronal vulnerability to MeHg-induced oxidative stress, and excess N-methyl D-aspartate (NMDA) receptor activation leading to neuronal demise.


Subject(s)
Astrocytes/drug effects , Cell Communication/drug effects , Methylmercury Compounds/toxicity , Neurons/drug effects , Animals , Astrocytes/metabolism , Cell Communication/physiology , Humans , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...