Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 37(2): 605-613, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26911894

ABSTRACT

The changes in the availability of striatal dopamine transporter and dopamine D2 receptor after mild focal ischemia in rats were measured using a small animal positron emission tomography system. Mild focal ischemia was induced by 20-minute middle cerebral artery occlusion. [11C]PE2I binding to dopamine transporter was transiently increased on the ipsilateral side of the striatum at 2 days after middle cerebral artery occlusion. On day 7 and 14 after middle cerebral artery occlusion, [11C]PE2I binding levels were decreased. In contrast, [11C]raclopride binding to dopamine D2 receptor in the ipsilateral striatum had not changed at 2 days after middle cerebral artery occlusion. [11C]Raclopride binding was significantly decreased on the ischemic side of the striatum at 7 and 14 days after middle cerebral artery occlusion. Moreover, on day 1 and 2 after middle cerebral artery occlusion, significant circling behavior to the contralateral direction was induced by amphetamine challenge. This behavior disappeared at 7 days after middle cerebral artery occlusion. At 14 days, circling behavior to the ipsilateral direction (middle cerebral artery occlusion side) was significantly increased, and that to the contralateral direction also appeared again. The present study suggested that amphetamine-induced circling behavior indicated striatal dopaminergic alterations and that dopamine transporter and dopamine D2 receptor binding could be key markers for predicting motor dysfunction after mild focal ischemia.


Subject(s)
Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Infarction, Middle Cerebral Artery/metabolism , Receptors, Dopamine D2/metabolism , Animals , Corpus Striatum/diagnostic imaging , Dopamine Plasma Membrane Transport Proteins/analysis , Infarction, Middle Cerebral Artery/diagnostic imaging , Male , Positron-Emission Tomography , Raclopride/metabolism , Rats, Wistar , Receptors, Dopamine D2/analysis
2.
EJNMMI Res ; 5(1): 115, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26160496

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) have been implicated in the pathophysiology of the brain after ischemic stroke. In this study, we investigate the generation of brain ROS after transient focal ischemia in mice using a radical trapping radiotracer, [(3)H]-labeled N-methyl-2,3-diamino-6-phenyl-dihydrophenanthridine ([(3)H]hydromethidine), which we recently reported as a ROS imaging probe. We also examined the effect of dimethylthiourea (DMTU), a hydroxyl radical scavenger, on brain ROS generation and infarct volume after transient focal ischemia in mice. METHODS: [(3)H]Hydromethidine was intravenously injected into mice at 1, 2, 5, and 7 h after transient middle cerebral artery occlusion (tMCAO), and then, the brain autoradiogram was acquired at 60 min after tracer injection. Brain infarct volumes at 24 h after tMCAO were assessed by 2,3,5-triphenyltetrazolium chloride staining. RESULTS: Accumulation of radioactivity was observed in the ipsilateral striatum and cortex at 1 h after tMCAO. The increase of radioactivity was attenuated at 2 h after tMCAO and then became maximized at 5 h. The high accumulation of radioactivity remained until 7 h after tMCAO. DMTU treatment significantly attenuated the accumulation of radioactivity in the ipsilateral hemisphere at 1, 5, and 7 h after tMCAO. Brain infarct volumes were also significantly reduced in DMTU-treated mice at 24 h after tMCAO. CONCLUSIONS: These results indicated that [(3)H]hydromethidine is a useful radiotracer for detecting in vivo brain ROS generation such as hydroxyl radical after ischemic injury.

3.
EJNMMI Res ; 5(1): 116, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26160497

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) have been implicated in cisplatin-induced nephrotoxicity. The aim of this study was to investigate the potential of using [(3)H]-labeled N-methyl-2,3-diamino-6-phenyl-dihydrophenanthridine ([(3)H]hydromethidine) for ex vivo imaging of regional ROS overproduction in mouse kidney induced by cisplatin. METHODS: Male C57BL/6 J mice were intraperitoneally administered with a single dose of cisplatin (30 mg/kg). Renal function was assessed by measuring serum creatinine and blood urea nitrogen (BUN) levels and morphology by histological examination. Renal malondialdehyde levels were measured as a lipid peroxidation marker. Autoradiographic studies were performed with kidney sections from mice at 60 min after [(3)H]hydromethidine injection. RESULTS: Radioactivity accumulation after [(3)H]hydromethidine injection was observed in the renal corticomedullary area of cisplatin-treated mice and was attenuated by pretreatment with dimethylthiourea (DMTU), a hydroxyl radical scavenger. Cisplatin administration significantly elevated serum creatinine and BUN levels, caused renal tissue damage, and promoted renal lipid peroxidation. These changes were significantly suppressed by DMTU pretreatment. CONCLUSIONS: The present study showed that [(3)H]hydromethidine was rapidly distributed to the kidney after its injection and trapped there in the presence of ROS such as hydroxyl radicals, suggesting that [(3)H]hydromethidine is useful for assessment of the renal ROS amount in cisplatin-induced nephrotoxicity.

4.
J Cereb Blood Flow Metab ; 34(12): 1907-13, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25227606

ABSTRACT

To assess reactive oxygen species (ROS) production by detecting the fluorescent oxidation product, hydroethidine has been used extensively. The present study was undertaken to evaluate the potential of the hydroethidine derivative as a radiotracer to measure in vivo brain ROS production. [(3)H]-labeled N-methyl-2,3-diamino-6-phenyl-dihydrophenanthridine ([(3)H]Hydromethidine) was synthesized, and evaluated using in vitro radical-induced oxidization and in vivo brain ROS production model. In vitro studies have indicated that [(3)H]Hydromethidine is converted to oxidized products by a superoxide radical (O(2)(•)-) and a hydroxyl radical (OH(•)-) but not hydrogen peroxide (H(2)O(2)). In vivo whole-body distribution study showed that [(3)H]Hydromethidine rapidly penetrated the brain and then was washed out in normal mice. Microinjection of sodium nitroprusside (SNP) into the brain was performed to produce ROS such as OH(•)- via Fenton reaction. A significant accumulation of radioactivity immediately after [(3)H]Hydromethidine injection was seen in the side of the brain treated with SNP (5 and 20 nmol) compared with that in the contralateral side. These results indicated that [(3)H]Hydromethidine freely penetrated into the brain where it was rapidly converted to oxidized forms, which were trapped there in response to the production of ROS. Thus, [(3)H]Hydromethidine should be useful as a radical trapping radiotracer in the brain.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Hydroxyl Radical/chemical synthesis , Phenanthridines/chemical synthesis , Reactive Oxygen Species/metabolism , Animals , Autoradiography/methods , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Hydroxyl Radical/metabolism , Injections, Intravenous , Male , Methylation , Mice, Inbred C57BL , Microinjections , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Nitroso Compounds/pharmacology , Phenanthridines/chemistry , Phenanthridines/metabolism , Radionuclide Imaging , Tritium
5.
Neuroimage ; 79: 121-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23611861

ABSTRACT

The role of glial activation has been implicated in the development and persistence of neuropathic pain after nerve injury by recent studies. PK11195 binding to the translocator protein 18kDa (TSPO) has been shown to be enhanced in activated microglia. This study was designed to assess PK11195 imaging in spinal microglia during activation after nerve injury. The development of neuropathic pain was induced by partial sciatic nerve ligation (PSL). PSL rats on days 7 and 14 after nerve injury were subjected to imaging with a small-animal positron emission tomography/computed tomography (PET/CT) scanner using [(11)C]PK11195 to detect spinal microglial activation by means of noninvasive in vivo imaging. Spinal [(3)H]PK11195 autoradiography was performed to confirm the results of [(11)C]PK11195 PET in PSL rats. Quantitative RT-PCR of CD11b and GFAP mRNA, and the immunohistochemistry of Iba1 and GFAP were investigated to detect activated microglia and astrocytes. Mechanical allodynia was observed in the ipsilateral paw of PSL rats from day 3 after nerve injury and stably persisted from days 7 to 14. PET/CT fusion images clearly showed large amounts of accumulation of [(11)C]PK11195 in the lumbar spinal cord on days 7 and 14 after nerve injury. [(11)C]PK11195 enhanced images were restricted to the L3-L6 area of the spinal cord. The standardized uptake value (SUV) of [(11)C]PK11195 was significantly increased in the lumbar spinal cord compared to that of the thoracic region. Increased specific binding of [(11)C]PK11195 to TSPO in the spinal cord of PSL rats was confirmed by competition studies using unlabeled (R, S)-PK11195. Increased [(3)H]PK11195 binding was also observed in the ipsilateral dorsal horn of the L3-L6 spinal cord on days 7 and 14 after nerve injury. CD11b mRNA and Iba1 immunoreactive cells increased significantly on days 7 and 14 after nerve injury by PSL. However, changes in GFAP mRNA and immunoreactivity were slight in the ipsilateral side of PSL rats. In the present study, we showed that glial activation could be quantitatively imaged in the spinal cord of neuropathic pain rats using [(11)C]PK11195 PET, suggesting that high resolution PET using TSPO-specific radioligands might be useful for imaging to assess the role of glial activation, including neuroinflammatory processes, in neuropathic pain patients.


Subject(s)
Carrier Proteins/metabolism , Isoquinolines/pharmacokinetics , Microglia/metabolism , Peripheral Nerve Injuries/metabolism , Positron-Emission Tomography/methods , Receptors, GABA-A/metabolism , Sciatic Neuropathy/metabolism , Spinal Cord/metabolism , Animals , Male , Microglia/diagnostic imaging , Peripheral Nerve Injuries/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sciatic Neuropathy/diagnostic imaging , Sensitivity and Specificity , Spinal Cord/diagnostic imaging
6.
Brain Res ; 1340: 18-23, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20435022

ABSTRACT

PK11195 was previously reported to attenuate the quinolinic acid (QUIN)-induced enhancement of glucose metabolism in rat brain. In the present study, the effect of PK11195 or anesthesia on [(14)C]2-deoxyglucose ([(14)C]DG) uptake was investigated in order to determine whether the QUIN-induced enhancement of glucose metabolism occurred in glial cells or neurons. We confirmed that the microinjection of QUIN caused a significant enhancement of [(14)C]DG uptake at 2h after the infusion, while the co-injection of PK11195 and QUIN almost completely suppressed this enhancement of [(14)C]DG uptake. No effect of chloral hydrate anesthesia on the QUIN-induced enhancement of [(14)C]DG uptake was observed. In contrast to rats treated with QUIN, PK11195 did not affect the enhancement of [(14)C]DG uptake induced by fluorocitrate (FC); however, chloral hydrate anesthesia completely suppressed the FC-induced increase in [(14)C]DG uptake. These results indicated that the enhancement of glucose metabolism induced by QUIN mainly occurred in glial cells, and the neuroprotective effect of PK11195 in rats injected with QUIN might be related to the suppression of anaerobic glycolysis in glial cells.


Subject(s)
Brain/cytology , Brain/drug effects , Down-Regulation/drug effects , Glycolysis/drug effects , Isoquinolines/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Quinolinic Acid/antagonists & inhibitors , Anaerobiosis/drug effects , Anaerobiosis/physiology , Animals , Antineoplastic Agents/pharmacology , Brain/metabolism , Down-Regulation/physiology , Glucose/metabolism , Male , Neuroprotective Agents/pharmacology , Quinolinic Acid/physiology , Rats , Rats, Wistar , Up-Regulation/drug effects , Up-Regulation/physiology
7.
Neuroreport ; 20(17): 1538-42, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19779316

ABSTRACT

The uptake of [14C]lactate was measured in the brains of mice anesthetized with pentobarbital or chloral hydrate. The results showed significant increase of the [14C]lactate uptake in the brain under both anesthesia. Despite energy metabolism in the brain being suppressed by both pentobarbital and chloral hydrate, the [14C]lactate uptake was unexpectedly increased under anesthesia. [14C]Lactate uptake in rat brain injured by infusion of quinolic acid was significantly decreased, and the reduction of [14C]lactate uptake was parallel to neural cell death, suggesting that exogenous lactate might be selectively taken up by neuron. These results indicated that lactate rather than glucose might serve as an energy substrate for neuron in intact brain under anesthesia.


Subject(s)
Anesthetics, General/pharmacology , Brain Chemistry/drug effects , Brain/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Lactic Acid/metabolism , Animals , Autoradiography , Brain/metabolism , Brain Chemistry/physiology , Carbon Radioisotopes , Cell Death/drug effects , Cell Death/physiology , Chloral Hydrate/pharmacology , Down-Regulation/drug effects , Down-Regulation/physiology , Energy Metabolism/physiology , Male , Mice , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Pentobarbital/pharmacology , Quinolinic Acid/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...