Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 336: 50-56, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33147512

ABSTRACT

Nerve agent exposure can cause debilitating neurological damage even with treatment. Currently accepted treatments involve attenuating the cholinergic crisis and seizure onset but do not focus directly on neuroprotection. Hence, there is a need for improved treatments to reduce neurological deficits. It is important to understand the pathophysiology of nerve agent mediated injury in order to identify effective treatment targets. Nerve agent-induced seizures are believed to be the main contributor to the neuropathology. Recently seizures have been shown to cause vascular changes that may actually attenuate neurological damage. This study evaluated the effect of soman-induced convulsive seizures on the relationship between CNS oxygen consumption and supply. To simultaneously assess changes in oxygenation and perfusion, rats were implanted with permanently fixed fiber-optic tissue oxygen sensing probes in the motor cortex and imaged with continuous arterial spin labelling MRI to measure cerebral blood flow. Baseline tissue oxygen tension (ptO2) and cerebral blood flow (CBF) were measured in isoflurane anaesthetized rats at least one day prior to soman or saline exposure. Rats were pretreated with HI-6 dimethansulfonate and atropine methyl nitrate (125 mg/kg and 20 mg/kg; intraperitoneal) followed by a convulsive dose of soman (90 µg/kg; subcutaneous) or equal volume of saline. Three additional treatments of HI-6/AMN were administered to improve survival. At 1.5 -hs after exposure, ptO2 and cerebral blood flow measurements were conducted. There was a significant decrease in CBF 1.5 -hs following soman exposure but no change in ptO2 was found. When we correlated ptO2 and CBF, for a given ptO2, there was lower CBF following soman exposure. This may indicate metabolism is inhibited, possibly because of mitochondrial impairment, therefore reducing oxygen demand. These data show hypoperfusion in brain following soman exposure which would be expected to contribute to soman-related neuropathology.


Subject(s)
Cerebrovascular Circulation , Chemical Warfare Agents , Energy Metabolism , Motor Cortex/blood supply , Oxygen Consumption , Oxygen/blood , Seizures/chemically induced , Soman , Animals , Disease Models, Animal , Male , Rats, Sprague-Dawley , Seizures/blood , Seizures/physiopathology , Time Factors
2.
Sci Rep ; 10(1): 13007, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32747689

ABSTRACT

Organophosphorus compounds, such as chemical warfare nerve agents and pesticides, are known to cause neurological damage. This study measured nerve agent-related neuropathology and determined whether quantitative T2 MRI could be used as a biomarker of neurodegeneration. Quantitative T2 MRI was performed using a 9.4 T MRI on rats prior to and following soman exposure. T2 images were taken at least 24 h prior, 1 h and 18-24 h after soman exposure. Rats were pre- and post-treated with HI-6 dimethanesulfonate and atropine methyl nitrate. A multicomponent T2 acquisition and analysis was performed. Brains were stained with Fluoro-Jade C to assess neurodegeneration. Rats exposed to soman developed behavioral expression of electrographic seizures. At 18-24 h after soman exposure, significant increases in T2, a possible marker of edema, were found in multiple regions. The largest changes were in the piriform cortex (before: 47.7 ± 1.4 ms; 18-24 h: 82.3 ± 13.4 ms). Fluoro-Jade C staining showed significant neurodegeneration 18-24 h post exposure. The piriform cortex had the strongest correlation between the change in relaxation rate and percent neurodegeneration (r = 0.96, p < 0.001). These findings indicate there is regionally specific neurodegeneration 24 h after exposure to soman. The high correlation between T2 relaxivity and histopathology supports the use of T2 as a marker of injury.


Subject(s)
Chemical Warfare Agents/toxicity , Magnetic Resonance Imaging/methods , Soman/toxicity , Animals , Male , Models, Animal , Piriform Cortex/drug effects , Rats , Rats, Sprague-Dawley , Seizures/chemically induced
3.
Neurotoxicology ; 65: 28-37, 2018 03.
Article in English | MEDLINE | ID: mdl-29378300

ABSTRACT

Nerve agents (NAs) are potent organophosphorus (OP) compounds with applications in chemical warfare. OP compounds act by inhibiting acetylcholinesterase (AChE). Soman (O-pinacolyl methylphosphonofluoridate) is one of the most potent NAs. It is well known that small doses of NAs can be lethal, and that even non-lethal exposure leads to long-term mental debilitation/neurological damage. However, the neuropathology following exposure to sub-lethal nerve agents is not well understood. In this study, we examined changes in tissue oxygenation (pO2) in the cortex and hippocampus after a sub-lethal dose of soman [80-90 µg/kg; subcutaneous]. pO2 changes can provide information regarding oxygen delivery and utilization and may be indicative of a disruption in cerebral blood flow and/or metabolism. Changes in oxygenation were measured with chronically implanted oxygen sensors in awake and freely moving rats. Measurements were taken before, during, and after soman-induced convulsive seizures. Soman exposure resulted in an immediate increase in pO2 in the cortex, followed by an even greater increase that precedes the onset of soman-induced convulsive seizures. The rise in hippocampus pO2 was delayed relative to the cortex, although the general pattern of brain oxygenation between these two regions was similar. After convulsive seizures began, pO2 levels declined but usually remained hyperoxygenated. Following the decline in pO2, low frequency cycles of large amplitude changes were observed in both the cortex and hippocampus. This pattern is consistent with recurring seizures. Measuring real-time changes in brain pO2 provides new information on the physiological status of the brain following soman exposure. These results highlight that the measurement of brain oxygenation could provide a sensitive marker of nerve agent exposure and serve as a biomarker for treatment studies.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Oxygen/metabolism , Soman/toxicity , Animals , Implants, Experimental , Male , Monitoring, Ambulatory , Rats , Seizures/chemically induced , Seizures/metabolism , Time Factors
4.
J Neurotrauma ; 35(1): 174-186, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28726571

ABSTRACT

Previous work in this laboratory used underwater explosive exposures to isolate the effects of shock-induced principle stress without shear on rat brain aggregate cultures. The current study has utilized simulated air blast to expose aggregates in suspension and enclosed within a spherical shell, enabling the examination of a much more complex biomechanical insult. Culture medium-filled spheres were exposed to single pulse overpressures of 15-30 psi (∼6-7 msec duration) and measurements within the sphere at defined sites showed complex and spatially dependent pressure changes. When brain aggregates were exposed to similar conditions, no cell death was observed and no changes in several commonly used biomarkers of traumatic brain injury (TBI) were noted. However, similarly to underwater blast, immediate and transient increases in the protein kinase B signaling pathway were observed at early time-points (3 days). In contrast, the oligodendrocyte marker 2',3'-cyclic nucleotide 3'-phosphodiesterase, as well as vascular endothelial growth factor, both displayed markedly delayed (14-28 days) and pressure-dependent responses. The imposition of a spherical shell between the single pulse shock wave and the target brain tissue introduces greatly increased complexity to the insult. This work shows that brain tissue can not only discriminate the nature of the pressure changes it experiences, but that a portion of its response is significantly delayed. These results have mechanistic implications for the study of primary blast-induced TBI and also highlight the importance of rigorously characterizing the actual pressure variations experienced by target tissue in primary blast studies.


Subject(s)
Blast Injuries/pathology , Brain Injuries, Traumatic/pathology , Brain/pathology , Disease Models, Animal , Animals , Brain Injuries, Traumatic/etiology , Cell Death , In Vitro Techniques , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
5.
J Neurotrauma ; 33(13): 1181-93, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26582146

ABSTRACT

The role of primary blast in blast-induced traumatic brain injury (bTBI) is controversial in part due to the technical difficulties of generating free-field blast conditions in the laboratory. The use of traditional shock tubes often results in artifacts, particularly of dynamic pressure, whereas the forces affecting the head are dependent on where the animal is placed relative to the tube, whether the exposure is whole-body or head-only, and on how the head is actually exposed to the insult (restrained or not). An advanced blast simulator (ABS) has been developed that enables high-fidelity simulation of free-field blastwaves, including sharply defined static and dynamic overpressure rise times, underpressures, and secondary shockwaves. Rats were exposed in head-only fashion to single-pulse blastwaves of 15 to 30 psi static overpressure. Head restraints were configured so as to eliminate concussive and minimize whiplash forces exerted on the head, as shown by kinematic analysis. No overt signs of trauma were present in the animals post-exposure. However, significant changes in brain 2',3'-cyclic nucleotide 3'-phosphohydrolase (CNPase) and neurofilament heavy chain levels were evident by 7 days. In contrast to most studies of primary blast-induced TBI (PbTBI), no elevation of glial fibrillary acidic protein (GFAP) levels was noted when head movement was minimized. The ABS described in this article enables the generation of shockwaves highly representative of free-field blast. The use of this technology, in concert with head-only exposure, minimized head movement, and the kinematic analysis of the forces exerted on the head provide convincing evidence that primary blast directly causes changes in brain function and that GFAP may not be an appropriate biomarker of PbTBI.


Subject(s)
Biomarkers , Blast Injuries , Brain Injuries, Traumatic , Disease Models, Animal , Equipment and Supplies , Animals , Male , Rats , Rats, Sprague-Dawley
6.
J Neurotrauma ; 32(1): 58-65, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25058115

ABSTRACT

Traumatic brain injury (TBI) is deemed the "signature injury" of recent military conflicts in Afghanistan and Iraq, largely because of increased blast exposure. Injuries to the brain can often be misdiagnosed, leading to further complications in the future. Therefore, the use of protein biomarkers for the screening and diagnosis of TBI is urgently needed. In the present study, we have investigated the plasma levels of soluble cellular prion protein (PrPC) as a novel biomarker for the diagnosis of primary blast-induced TBI (bTBI). We hypothesize that the primary blast wave can disrupt the brain and dislodge extracellular localized PrPC, leading to a rise in concentration within the systemic circulation. Adult male Sprague-Dawley rats were exposed to single pulse shockwave overpressures of varying intensities (15-30 psi or 103.4-206.8 kPa] using an advanced blast simulator. Blood plasma was collected 24 h after insult, and PrPC concentration was determined with a modified commercial enzyme-linked immunosorbent assay (ELISA) specific for PrPC. We provide the first report that mean PrPC concentration in primary blast exposed rats (3.97 ng/mL ± 0.13 SE) is significantly increased compared with controls (2.46 ng/mL ± 0.14 SE; two tailed test p < 0.0001). Furthermore, we report a mild positive rank correlation between PrPC concentration and increasing blast intensity (psi) reflecting a plateaued response at higher pressure magnitudes, which may have implications for all military service members exposed to blast events. In conclusion, it appears that plasma levels of PrPC may be a novel biomarker for the detection of primary bTBI.


Subject(s)
Blast Injuries/blood , Brain Injuries/diagnosis , PrPC Proteins/blood , Animals , Biomarkers/blood , Blast Injuries/complications , Brain Injuries/blood , Brain Injuries/etiology , Explosions , Male , Rats , Rats, Sprague-Dawley
7.
Toxicol Appl Pharmacol ; 247(3): 179-90, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20600214

ABSTRACT

The effect of ionic environment on sulphur mustard (bis 2-chloroethyl sulphide; HD) toxicity was examined in CHO-K1 cells. Cultures were treated with HD in different ionic environments at constant osmolar conditions (320 mOsM, pH 7.4). The cultures were refed with fresh culture medium 1h after HD exposure, and viability was assessed. Little toxicity was apparent when HD exposures were carried out in ion-free sucrose buffer compared to LC(50) values of approximately 100-150 microM when the cultures were treated with HD in culture medium. Addition of NaCl to the buffer increased HD toxicity in a salt concentration-dependent manner to values similar to those obtained in culture medium. HD toxicity was dependent on both cationic and anionic species with anionic environment playing a much larger role in determining toxicity. Substitution of NaI for NaCl in the treatment buffers increased HD toxicity by over 1000%. The activity of the sodium hydrogen exchanger (NHE) in recovering from cytosolic acidification in salt-free and in different chloride salts did not correlate with the HD-induced toxicity in these buffers. However, the inhibition by HD of intracellular pH regulation correlated with its toxicity in NaCl, NaI and sucrose buffers. Analytical chemical studies and the toxicity of the iodine mustard derivative ruled out the role of chemical reactions yielding differentially toxic species as being responsible for the differences in HD toxicity observed. This work demonstrates that the early events that HD sets into motion to cause toxicity are dependent on ionic environment, possibly due to intracellular pH deregulation.


Subject(s)
Chemical Warfare Agents/toxicity , Mustard Gas/toxicity , Salts/pharmacology , Ammonium Chloride/chemistry , Ammonium Chloride/pharmacology , Animals , Buffers , CHO Cells , Caspase 3/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Cricetinae , Cricetulus , Culture Media/chemistry , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Intracellular Fluid/chemistry , Salts/chemistry , Sodium Chloride/chemistry , Sodium Chloride/pharmacology , Sodium Iodide/chemistry , Sodium Iodide/pharmacology , Sucrose
8.
J Chromatogr B Analyt Technol Biomed Life Sci ; 878(17-18): 1407-13, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20395179

ABSTRACT

A rapid and small volume assay to quantify HI-6 in plasma was developed to further the development and licensing of an intravenous formulation of HI-6. The objective of this method was to develop a sensitive and rapid assay that clearly resolved HI-6 and an internal standard in saline and plasma matrices. A fully validated method using ion-pair HPLC and 2-PAM as the internal standard fulfilled these requirements. Small plasma samples of 35 microL were extracted using acidification, filtration and neutralization. Linearity was shown for over 4 microg/mL to 1mg/mL with accuracy and precision within 6% relative error at the lower limit of detection. This method was utilized in the pharmacokinetic analysis HI-6 dichloride (2Cl) and HI-6 dimethane sulfonate (DMS) in anaesthetized guinea pigs and domestic swine following an intravenous bolus administration. From the resultant pharmacokinetic parameters a target plasma concentration of 100 microM was established and maintained in guinea pigs receiving an intravenous infusion. This validated method allows for the analysis of low volume samples, increased sample numbers and is applicable to the determination of pharmacokinetic profiles and parameters.


Subject(s)
Cholinesterase Reactivators/blood , Chromatography, High Pressure Liquid/methods , Oximes/blood , Pyridinium Compounds/blood , Swine/blood , Animals , Cholinesterase Reactivators/administration & dosage , Cholinesterase Reactivators/pharmacokinetics , Drug Stability , Guinea Pigs , Linear Models , Male , Oximes/administration & dosage , Oximes/pharmacokinetics , Pralidoxime Compounds/analysis , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
9.
Toxicon ; 54(2): 95-102, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19328212

ABSTRACT

Maitotoxin (MTX) is one of the most potent toxins known to date. It causes massive calcium (Ca(2+)) influx and necrotic cell death in various tissues. However, the exact mechanism(s) underlying its cellular toxicity is not fully understood. In the present study, the role of the sodium hydrogen exchanger (NHE) in MTX-induced increases in intracellular Ca(2+) and subsequent cell death were investigated in cultured rat cortical neurons. Intracellular Ca(2+) concentrations ([Ca(2+)](i)) were measured fluorimetrically using FURA-2 as the fluorescence indicator. Cell death was measured with the alamarBlue cell viability assay and the vital dye ethidium bromide (EB) uptake assay. Results showed that MTX increased, in a concentration dependent manner, both [Ca(2+)](i) and cell death in cortical neurons. Decreasing the pH of the treatment medium from 7.5 to 6.0 diminished MTX-induced cell death. The protection offered by lowering extracellular pH was not due to MTX degradation, because it was still effective even if the cells were treated with MTX in normal pH and then switched to a lower pH. Pretreatment of cells with the specific NHE inhibitor, 5-(N-ethyl-N-isopropyl)-amiloride (EIPA), prevented MTX-induced increases in [Ca(2+)](i), as well as cell death in a concentration dependent manner. Furthermore, knockdown of NHE1 by SiRNA transfection suppressed MTX-induced cell death in human embryonic kidney (HEK) cells. Together, these results suggest that NHE1 plays a major role in MTX-induced neurotoxicity.


Subject(s)
Cerebral Cortex/cytology , Marine Toxins/toxicity , Neurons/drug effects , Oxocins/toxicity , Sodium-Hydrogen Exchangers/physiology , Acidosis/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Antimetabolites, Antineoplastic/toxicity , Calcium/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Female , Fluorometry , Methotrexate/toxicity , Necrosis , Neuroprotective Agents/pharmacology , Pregnancy , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchangers/antagonists & inhibitors
10.
Toxicol Appl Pharmacol ; 221(3): 363-71, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17482225

ABSTRACT

The dependence of sulphur mustard (HD) toxicity on intracellular (pH(i)) and extracellular pH was examined in CHO-K1 cells. HD produced an immediate and significant concentration-dependent decline in cytosolic pH, and also inhibited the mechanisms responsible for restoring pH(i) to physiological values. The concentration-response of HD-induced cytosolic acidification, closely paralleled the acidification of the extracellular buffer through HD hydrolysis. A viability study was carried out in order to assess the importance of HD-induced cytosolic acidification. Cultures were exposed to HD for 1 h in media that were adjusted through a pH range (pH 5.0-10), and the 24 h LC(50) values were assessed using the viability indicator dye alamarBlue. The toxicity of HD was found to be dependent on extracellular pH, with a greater than eight-fold increase in LD(50) obtained in cultures treated with HD at pH 9.5, compared to those treated at pH 5.0. Assays of apoptotic cell death, including morphology, soluble DNA, caspase-3 activity and TUNEL also showed that as pH was increased, much greater HD concentrations were required to cause cell death. The modest decline in HD half-life measured in buffers of increasing pH, did not account for the protective effects of basic pH. The early event(s) that HD initiates to eventually culminate in cell death are not known. However, based on the data obtained in this study, we propose that HD causes an extracellular acidification through chemical hydrolysis and that this, in both a concentration and temporally related fashion, results in cytosolic acidification. Furthermore, HD also acts to poison the antiporter systems responsible for maintaining physiological pH(i), so that the cells are unable to recover from this insult. It is this irreversible decline in pH(i) that initiates the cascade of events that results in HD-induced cell death.


Subject(s)
Cell Death/drug effects , Chemical Warfare Agents/toxicity , Hydrogen-Ion Concentration/drug effects , Mustard Gas/toxicity , Sodium-Hydrogen Exchangers/drug effects , Adaptation, Physiological/drug effects , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , CHO Cells , Caspase 3/drug effects , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Extracellular Fluid/chemistry , Extracellular Fluid/drug effects , Female , Intracellular Fluid/chemistry , Intracellular Fluid/drug effects
11.
Toxicology ; 208(3): 399-409, 2005 Mar 30.
Article in English | MEDLINE | ID: mdl-15695025

ABSTRACT

Anesthetized pigs were injected i.m. with 500 mg HI-6 dichloride (HI-6 2Cl) (1-[[[4-(aminocarbonyl)-pyridinio]methoxy]methyl]-2[(hydroxyimino)methyl]pyridinium dichloride; CAS 34433-31-3)) or the molar equivalent of HI-6 dimethanesulphonate (HI-6 DMS) 633 mg. Plasma HI-6 concentrations were measured by HPLC (1, 3, 5, 10, 15, 30, 60 min and every 30 min until 4h or 6h following the i.v. or i.m. dose respectively) while a variety of physiological responses were continuously examined. HI-6 (500 mg 2Cl or 633 mg DMS) resulted in an identical pharmacokinetic profile unaffected by atropine co-administration. Neither HI-6 salt resulted in clinically significant changes in cardiovascular or respiratory function. HI-6 DMS (1899 mg i.v.) resulted in plasma HI-6 concentrations about 10 times higher than measured following i.m. 500 mg 2Cl or 633 mg DMS and resulted in small transitory effect on mean arterial pressure. Atropine plus HI-6 DMS (1-9 mg/kg or 127-172 mg/kg i.m.) protected up to 100% of guinea pigs exposed to 5 x LD50 of GF (cyclohexyl methyl phosphonoflouridate) or soman (pinacolyl methylphosphonofluoridate) (GD) respectively. The results suggest that the two HI-6 salts have a similar pharmacokinetic profile while HI-6 DMS appears extremely safe and effective against nerve agents and may be as suitable for human use.


Subject(s)
Chemical Warfare Agents/poisoning , Neuroprotective Agents/pharmacokinetics , Organophosphate Poisoning , Pyridinium Compounds/pharmacokinetics , Soman/poisoning , Animals , Atropine/pharmacology , Dose-Response Relationship, Drug , Guinea Pigs , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organophosphorus Compounds , Oximes , Pyridinium Compounds/pharmacology , Pyridinium Compounds/therapeutic use , Swine , Time Factors
12.
Eur J Pharmacol ; 487(1-3): 17-28, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-15033372

ABSTRACT

The ATP-P2X(7) receptor subtype and a maitotoxin-activated ion channel were studied to determine factors which identify them as separate entities in the control of a cytotolytic pore. Activation of ATP-P2X(7) receptors with 2'-3'-O-(benzylbenzyl) ATP (BzATP) or maitotoxin ion channels resulted in influx of ethidium bromide and cell death. Maitotoxin (25-250 pM)-induced ethidium bromide uptake and cell death was sensitive to extracellular Ca(2+), the ionic composition of the buffer, reduced by the calmodulin inhibitor W7, (N-(s-aminohexyl)-5-chloro-1-naphthalenesulfonamide), (10-100 microM) but unaffected by the ATP-P2X(7) receptor antagonist oxidized ATP, (adenosine 5'-triphosphate periodate oxidized sodium salt) (oATP). BzATP (10-200 microM)-induced ethidium bromide uptake and cell death were inhibited by oATP, unaffected by W7, inhibited by high ionic concentrations but only slightly dependant on external Ca(2+). These results are consistent with the existence of a pharmacological mechanism for controlling cell death consisting of an ATP-P2X(7) receptor, a maitotoxin-activated ion channel and a cytolytic pore.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Ion Channels/drug effects , Marine Toxins/pharmacology , Oxocins/pharmacology , Receptors, Purinergic P2/biosynthesis , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/toxicity , Animals , Buffers , CHO Cells , Calmodulin/antagonists & inhibitors , Cations/metabolism , Cell Survival/drug effects , Cricetinae , Enzyme Inhibitors/pharmacology , Ethidium/metabolism , Marine Toxins/antagonists & inhibitors , Marine Toxins/toxicity , Oxocins/antagonists & inhibitors , Oxocins/toxicity , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X7 , Sulfonamides/pharmacology
13.
Br J Pharmacol ; 135(7): 1616-26, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11934801

ABSTRACT

1. ATP receptors of the P2X class have previously been identified on autonomic nerve endings and on a limited population of CNS neurons. 2. In the present study P2X receptors on mammalian cortical synaptosomes have been identified by a variety of functional and biochemical studies. In choline buffer ATP analogues caused concentration/time dependent Ca(2+) influx. Relative to the effects caused by ATP, benzoylbenzoyl ATP (BzATP) was about seven times more active than ATP while 2-me-S-ATP and ATPgammaS were much less active. alpha,beta-me- ATP and beta,gamma-me-ATP were virtually inactive. In sucrose buffer, relative to choline buffer, the activity of BzATP was more than doubled while activity in sodium buffer was reduced. Moreover, the P2X antagonists PPADS or Brilliant Blue G both significantly attenuated influx. These observations suggest the presence of P2X receptors on synaptosomes which subserve Ca(2+) influx. This activity profile of the ATP analogues and the response to blocking agents are characteristic of responses of P2X(7) receptors. 3. Influx was unaffected by the VSCC inhibitors omega-CTx-MVIIC and (-) 202 - 791, indicating that ATP induced Ca(2+) influx occurred primarily through P2X receptors. 4. P2X(7) receptor protein was identified by Western blotting and immunohistochemical staining. Purified preparations were devoid of significant concentrations of GFAP or the microglial marker OX-42 but contained greatly enriched amounts of syntaxin and SNAP 25. 5. The various pharmacological and biochemical studies were all consistent with the presence of functional P2X(7) receptors.


Subject(s)
Calcium/metabolism , Receptors, Purinergic P2/classification , Receptors, Purinergic P2/metabolism , Synaptosomes/metabolism , Analysis of Variance , Animals , Blotting, Western , Brain , Immunohistochemistry , Male , PC12 Cells , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/isolation & purification , Receptors, Purinergic P2X7
SELECTION OF CITATIONS
SEARCH DETAIL
...