Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Br J Anaesth ; 111(2): 143-51, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23722106

ABSTRACT

Although previously considered entirely reversible, general anaesthesia is now being viewed as a potentially significant risk to cognitive performance at both extremes of age. A large body of preclinical as well as some retrospective clinical evidence suggest that exposure to general anaesthesia could be detrimental to cognitive development in young subjects, and might also contribute to accelerated cognitive decline in the elderly. A group of experts in anaesthetic neuropharmacology and neurotoxicity convened in Salzburg, Austria for the BJA Salzburg Seminar on Anaesthetic Neurotoxicity and Neuroplasticity. This focused workshop was sponsored by the British Journal of Anaesthesia to review and critically assess currently available evidence from animal and human studies, and to consider the direction of future research. It was concluded that mounting evidence from preclinical studies reveals general anaesthetics to be powerful modulators of neuronal development and function, which could contribute to detrimental behavioural outcomes. However, definitive clinical data remain elusive. Since general anaesthesia often cannot be avoided regardless of patient age, it is important to understand the complex mechanisms and effects involved in anaesthesia-induced neurotoxicity, and to develop strategies for avoiding or limiting potential brain injury through evidence-based approaches.


Subject(s)
Anesthesia, General/adverse effects , Anesthetics, General/adverse effects , Brain/drug effects , Neuronal Plasticity/drug effects , Neurotoxicity Syndromes/etiology , Periodicals as Topic , Aged , Aged, 80 and over , Animals , Austria , Cognition Disorders/chemically induced , Humans , Infant , United Kingdom
2.
Neurotoxicol Teratol ; 97: 107173, 2023.
Article in English | MEDLINE | ID: mdl-36893929

ABSTRACT

Attention-deficit hyperactivity disorder (ADHD) is a common human neurobehavioral disorder that usually begins in early childhood. Methylphenidate (MPH) has been used extensively as a first-line medicine for the treatment of ADHD. Since ADHD is often diagnosed in early childhood and can persist for the entire lifespan, individuals may take MPH for many years. Given that in the course of one's lifetime a person may stop taking MPH for periods of time, or may implement lifestyle changes that may reduce the need for MPH entirely, it is important to understand how cessation of MPH affects the adult brain following long-term use of MPH. The blockage of the dopamine transporter (DAT) and the norepinephrine transporter (NET) by MPH may help with ADHD symptoms by boosting monoamine levels in the synapse. In the present study, microPET/CT was used to investigate possible neurochemical alterations in the cerebral dopamine system after cessation of long-term MPH administration in nonhuman primates. MicroPET/CT images were collected from adult male rhesus monkeys 6Ā months after they stopped receiving vehicle or MPH following 12Ā years of chronic treatment. The neurochemical status of brain dopaminergic systems was evaluated using the vesicular monoamine transporter 2 (VMAT2) ligand [18F]-AV-133 and a tracer for imaging dopamine subtype 2 (D2) and serotonin subfamily 2 (5HT2) receptors, [18F]-FESP. Each tracer was injected intravenously and ten minutes later microPET/CT images were obtained over 120Ā min. The binding potential (BP) of each tracer in the striatum was obtained using the Logan reference tissue model with the cerebellar cortex time activity curve (TAC) as an input function. Brain metabolism was also evaluated using microPET/CT images of [18F]-FDG. [18F]-FDG was injected intravenously, and ten minutes later, microPET/CT images were obtained over 120Ā min. Radiolabeled tracer accumulation in regions of interest (ROIs) in the prefrontal cortex, temporal cortex, striatum, and cerebellum were converted into standard uptake values (SUVs). Compared to the vehicle control group, the BPs of [18F] AV-133 and [18F]-FESP in the striatum were not significantly altered in MPH treated groups. Additionally, no significant differences were detected in the SUVs of [18F]-FDG in the MPH treated group compared with control. This study demonstrates that 6Ā months after cessation of long-term, chronic MPH treatment, there are no significant neurochemical or neural metabolic changes in the central nervous system (CNS) of non-human primates (NHPs) and suggests that microPET imaging is helpful in assessing the status of biomarkers of neurochemical processes linked to chronic CNS drug exposure. (Supported by NCTR).


Subject(s)
Attention Deficit Disorder with Hyperactivity , Central Nervous System Stimulants , Methylphenidate , Child, Preschool , Adult , Animals , Male , Humans , Dopamine , Fluorodeoxyglucose F18/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy
3.
J Neural Transm (Vienna) ; 118(2): 203-11, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20963452

ABSTRACT

Recent reports indicate that 6-12 h of ketamine anesthesia can trigger neuronal apoptosis in postnatal day (PND) 7 rats. In vitro, ex vivo, and confocal fluorescent imaging studies suggest that dansyl compounds can accumulate within the cytoplasm of the apoptotic cell. High-resolution positron emission tomography (microPET) imaging has been proposed as a minimally invasive method for detecting apoptosis in the rat brain. Compared with [(18)F]-labeled annexin V, which binds to externalized phosphatidylserine (PS) on the outer membrane of apoptotic cells, intracellular uptake of the dansylhydrazone of p-fluorobenzaldehyde (DFNSH) may lead to improved target-to-background contrast ratios. In this study, the effect of ketamine on the uptake and retention of [(18)F]-DFNSH in the rat brain was investigated using microPET imaging. On PND 7, rat pups in the experimental group were exposed, at 2-h intervals, to six subcutaneous injections of ketamine (20 mg/kg) and control rat pups received six injections of saline. On PND 35, [(18)F]-DFNSH (37 MBq) was injected into the tail vein of rats and microPET images were obtained over 2 h following the injection. Radiolabeled tracer accumulation in the region of interest (ROI) in the frontal cortex was converted into standard uptake values (SUVs). The radiotracer was quickly distributed into the brains of both ketamine- and saline-treated rats. Compared with the control group, the uptake of [(18)F]-DFNSH was significantly increased in the ROI, frontal cortex area of ketamine-treated rats. In addition, the wash-out duration of the tracer was prolonged in the ketamine-treated animals. This study demonstrates that microPET imaging is capable of distinguishing differences in retention of [(18)F]-DFNSH in ROI and suggests that this compound may serve as a minimally invasive biomarker of neuronal apoptosis in rodents.


Subject(s)
Anesthetics, Dissociative/toxicity , Benzaldehydes/pharmacokinetics , Brain/diagnostic imaging , Ketamine/toxicity , Neurons/pathology , Positron-Emission Tomography/methods , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorine Radioisotopes/pharmacokinetics , Image Processing, Computer-Assisted , Male , Neurons/drug effects , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
4.
Neurotoxicol Teratol ; 87: 107017, 2021.
Article in English | MEDLINE | ID: mdl-34265415

ABSTRACT

Methylphenidate (MPH) is a psychostimulant approved by the FDA to treatment Attention-Deficit Hyperactivity Disorder (ADHD). MPH is believed to exert its pharmacological effects via preferential blockade of the dopamine transporter (DAT) and the norepinephrine transporter (NET), resulting in increased monoamine levels in the synapse. We used a quantitative non-invasive PET imaging technique to study the effects of long-term methylphenidate use on the central nervous system (CNS). We conducted microPET/CT scans on young adult male rhesus monkeys to monitor changes in the dopaminergic system. We used [18F] AV-133, a ligand for the vesicular monoamine transporter 2 (VMAT2), and [18F]FESP a ligand for the D2 and 5HT2 receptors. In this study we evaluated the effects if chronic MPH treatment in the nonhuman primates (NHP). Two-year-old, male rhesus monkeys were orally administered MPH diluted in the electrolyte replenisher, Prang, twice a day, five days per week (M-F) over an 8-year period. The dose of MPH was gradually escalated from 0.15Ā mg/kg initially to 2.5Ā mg/kg/dose for the low dose group, and 1.5Ā mg/kg to 12.5Ā mg/kg/dose for the high dose group (Rodriguez et al., 2010). Scans were performed on Mondays, about 60Ā h after their last treatment, to avoid the acute effects of MPH. Tracers were injected intravenously ten minutes before microPET/CT scanning. Sessions lasted about 120Ā min. The Logan reference tissue model was used to determine the Binding Potential (BP) of each tracer in the striatum with the cerebellar cortex time activity curve as an input function. Both MP treatment groups had a lower [18F] AV-133 BP, although this failed to reach statistical significance. MPH treatment did not have a significant effect on The BP of [18F] FESP in the striatum. Long-term administration of MPH did not significant change any of the marker of monoamine function used here. These data suggest that, despite lingering concerns, long-term use of methylphenidate does not negatively impact monoamine function. This study also demonstrates that microPET imaging can distinguish differences in binding potentials of a variety of radiotracers in the CNS of NHPs. This approach may provide minimally-invasive biomarkers of neurochemical processes associated with chronic exposure to CNS medications. (Supported by NCTR).


Subject(s)
Brain/drug effects , Dopamine Plasma Membrane Transport Proteins/drug effects , Methylphenidate/pharmacology , Time Factors , Animals , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Macaca mulatta , Methylphenidate/administration & dosage , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/pharmacology , Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Vesicular Monoamine Transport Proteins/drug effects , Vesicular Monoamine Transport Proteins/metabolism
5.
Neuroscience ; 151(4): 1053-65, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18201836

ABSTRACT

The anesthetic gas nitrous oxide (N2O) and the volatile anesthetic isoflurane (ISO) are commonly used in surgical procedures for human infants and in veterinary and laboratory animal practice to produce loss of consciousness and analgesia. Recent reports indicate that exposure of the developing brain to general anesthetics that block N-methyl-D-aspartate (NMDA) glutamate receptors or potentiate GABA(A) receptors can trigger widespread apoptotic neurodegeneration. In the present study, the question arises whether a relatively low dose of ISO alone or its combination with N2O entails significant risk of inducing enhanced apoptosis. In addition, the role of L-carnitine to attenuate these effects was also examined. Postnatal day 7 (PND-7) rat pups were exposed to N2O (75%) or a low dose of ISO (0.55%) alone, or N2O plus ISO for 2, 4, 6 or 8 h with or without L-carnitine. The neurotoxic effects were evaluated 6 h after completion of anesthetic administration. No significant neurotoxic effects were observed for the animals exposed to N2O or ISO alone. However, enhanced apoptotic cell death was apparent when N2O was combined with ISO at exposure durations of 6 h or more. Co-administration of L-carnitine (300 or 500 mg/kg, i.p.) effectively protected neurons from the anesthetic-induced damage. These data indicate that 6 h or more of inhaled anesthetic exposure consisting of a combination of N2O and ISO results in enhanced neuronal apoptosis, and L-carnitine effectively blocks the neuronal apoptosis caused by inhalation anesthetics in the developing rat brain.


Subject(s)
Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Carnitine/pharmacology , Frontal Lobe/cytology , Neurons/drug effects , Vitamin B Complex/pharmacology , Animals , Animals, Newborn , Caspase 3/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Fluoresceins , Isoflurane/toxicity , Neural Cell Adhesion Molecule L1/metabolism , Neurons/cytology , Nitrous Oxide/toxicity , Organic Chemicals , Rats , Rats, Sprague-Dawley , Sialic Acids/metabolism , Time Factors , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
6.
Neuroscience ; 144(1): 46-55, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17084538

ABSTRACT

1-Methyl-4-phenylpyridinium ion (MPP+), an inhibitor of mitochondrial complex I, has been widely used as a neurotoxin because it elicits a severe Parkinson's disease-like syndrome with an elevation of intracellular reactive oxygen species (ROS) and apoptosis. L-carnitine plays an integral role in attenuating the brain injury associated with mitochondrial neurodegenerative disorders. The present study investigates the effects of L-carnitine against the toxicity of MPP+ in rat forebrain primary cultures. Cells in culture were treated for 24 h with 100, 250, 500 and 1000 microM MPP+ alone or co-incubated with L-carnitine. MPP+ produced a dose-related increase in DNA fragmentation as measured by cell death ELISA (enzyme-linked immunosorbent assay), an increase in the number of TUNEL (terminal dUTP nick-end labeling)-positive cells and a reduction in the mitochondrial metabolism of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). No significant effect was observed with the release of lactate dehydrogenase (LDH), indicating that cell death presumably occurred via apoptotic mechanisms. Co-incubation of MPP+ with L-carnitine significantly reduced MPP+-induced apoptosis. Western blot analyses showed that neurotoxic concentrations of MPP+ decreased the ratio of BCL-X(L) to Bax and decreased the protein levels of polysialic acid neural cell adhesion molecules (PSA-NCAM), a neuron specific marker. L-carnitine blocked these effects of MPP+ suggesting its potential therapeutic utility in degenerative disorders such as Parkinson's disease, Alzheimer's disease, ornithine transcarbamylase deficiency and other mitochondrial diseases.


Subject(s)
1-Methyl-4-phenylpyridinium/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/toxicity , Apoptosis/drug effects , Carnitine/pharmacology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents , Prosencephalon/pathology , Animals , Blotting, Western , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Prosencephalon/drug effects , Rats , Rats, Sprague-Dawley , Sialic Acids/metabolism , Tetrazolium Salts , Thiazoles , bcl-2-Associated X Protein/biosynthesis , bcl-X Protein/biosynthesis
7.
Exp Biol Med (Maywood) ; 242(16): 1586-1592, 2017 10.
Article in English | MEDLINE | ID: mdl-28658972

ABSTRACT

Microphysiological systems (MPS) and computer simulation models that recapitulate the underlying biology and toxicology of critical developmental transitions are emerging tools for developmental effects assessment of drugs/chemicals. Opportunities and challenges exist for their application to alternative, more public health relevant and efficient chemical toxicity testing methods. This is especially pertinent to children's health research and the evaluation of complex embryological and reproductive impacts of drug/chemical exposure. Scaling these technologies to higher throughput is a key challenge and drives the need for in silico models for quantitative prediction of developmental toxicity to inform safety assessments. One example is cellular agent-based models, constructed from extant embryology, that produce data useful to simulate critical developmental transitions and thereby predict phenotypic consequences of disruption in silico. Biologically inspired MPS models built from human induced pluripotent stem (iPS)-derived cells and synthetic matrices that recapitulate organ-specific physiologies and native tissue architectures are providing exciting new research opportunities to advance the assessment of developmental toxicity and offer the possibility of deriving a full 'human on a chip' system, or a 'Homunculus.' Impact statement This 'commentary' summarizes research needs and opportunities for engineered MPS models for developmental and reproductive toxicity testing. Emerging concepts can be taken forward to a virtual tissue modeling framework for assessing chemical (and non-chemical) stressors on human development. These models will advance children's health research, both basic and translational and new ways to evaluate complex embryological and reproductive impacts of drug and chemical exposures to inform safety assessments.


Subject(s)
Child Health , Microchip Analytical Procedures/methods , Microtechnology/methods , Models, Biological , Computer Simulation , Humans , Induced Pluripotent Stem Cells/cytology , Toxicity Tests/methods
8.
Neurotoxicol Teratol ; 60: 95-101, 2017.
Article in English | MEDLINE | ID: mdl-27784630

ABSTRACT

The association of general anesthesia with developmental neurotoxicity, while nearly impossible to study in pediatric populations, is clearly demonstrable in a variety of animal models from rodents to nonhuman primates. Nearly all general anesthetics tested have been shown to cause abnormal brain cell death in animals when administered during periods of rapid brain growth. The ability to repeatedly assess in the same subjects adverse effects induced by general anesthetics provides significant power to address the time course of important events associated with exposures. Minimally-invasive procedures provide the opportunity to bridge the preclinical/clinical gap by providing the means to more easily translate findings from the animal laboratory to the human clinic. Positron Emission Tomography or PET is a tool with great promise for realizing this goal. PET for small animals (microPET) is providing valuable data on the life cycle of general anesthetic induced neurotoxicity. PET radioligands (annexin V and DFNSH) targeting apoptotic processes have demonstrated that a single bout of general anesthesia effected during a vulnerable period of CNS development can result in prolonged apoptotic signals lasting for several weeks in the rat. A marker of cellular proliferation (FLT) has demonstrated in rodents that general anesthesia-induced inhibition of neural progenitor cell proliferation is evident when assessed a full 2weeks after exposure. Activated glia express Translocator Protein (TSPO) which can be used as a marker of presumed neuroinflammatory processes and a PET ligand for the TSPO (FEPPA) has been used to track this process in both rat and nonhuman primate models. It has been shown that single bouts of general anesthesia can result in elevated TSPO expression lasting for over a week. These examples demonstrate the utility of specific PET tracers to inform, in a minimally-invasive fashion, processes associated with general anesthesia-induced developmental neurotoxicity. The fact that PET procedures are also used clinically suggests an opportunity to confirm in humans what has been repeatedly observed in animals.


Subject(s)
Anesthetics, General/adverse effects , Brain/drug effects , Brain/physiopathology , Ligands , Neurotoxicity Syndromes/physiopathology , Animals , Atrophy/physiopathology , Biomarkers/metabolism , Brain/metabolism , Cell Proliferation/physiology , Functional Neuroimaging , Inflammation/physiopathology , Neurotoxicity Syndromes/metabolism , Positron-Emission Tomography
9.
Neurotoxicol Teratol ; 56: 68-74, 2016.
Article in English | MEDLINE | ID: mdl-27307090

ABSTRACT

Methylphenidate (MPH) is a psychostimulant commonly used for the treatment of Attention-Deficit Hyperactivity Disorder (ADHD). Since the long-term effects of this drug on the central nervous system (CNS) are not well understood, we conducted microPET/CT scans on young adult male rhesus monkeys (n=4/group) to gather information on brain metabolism using the uptake of [(18)F]Fluoro-2-deoxy-2-d-glucose (FDG) as a marker. Approximately two-year old, male rhesus monkeys were treated orally with MPH twice per day, five days per week (M-F) over a 6-year period. Subjects received MPH at either 2.5 or 12.5mg/kg/dose or vehicle (Prang). To minimize the acute effects of MPH on FDG uptake, microPET/CT scans were scheduled on Mondays before their first daily dosing of the week (approximately 68h since their last treatment). FDG (370Ā±8.88MBq) was injected intravenously and 30min later microPET/CT images were obtained over 60min. Radiolabeled tracer accumulation in regions of interest (ROIs) in the prefrontal cortex, temporal cortex, striatum and cerebellum were converted into Standard Uptake Values (SUVs). Compared to the control group, the uptake of FDG in the cerebellum was significantly decreased in both the low and high dose groups. These preliminary data demonstrate that microPET imaging is capable of distinguishing differences in retention of FDG in the brains of NHPs treated chronically with MPH and suggests that this approach may provide a minimally invasive biomarker for exploring the effects of chronic MPH treatment on aspects of brain function.


Subject(s)
Brain/drug effects , Brain/metabolism , Central Nervous System Stimulants/administration & dosage , Methylphenidate/administration & dosage , Positron-Emission Tomography , Tomography, X-Ray Computed , Animals , Fluorodeoxyglucose F18/metabolism , Macaca mulatta , Male
10.
Neuroscience ; 132(4): 967-77, 2005.
Article in English | MEDLINE | ID: mdl-15857702

ABSTRACT

Recent data suggest that anesthetic drugs may cause widespread and dose-dependent apoptotic neurodegeneration during development. The window of vulnerability to this neurotoxic effect, particularly with N-methyl-D-aspartate (NMDA) antagonists such as ketamine, is restricted to the period of synaptogenesis. The purposes of this study are to determine whether treatment of forebrain cultures with ketamine results in a dose-related increase in neurotoxicity and whether upregulation of NMDA receptor subunit NR1 promotes ketamine-induced apoptosis. Forebrain cultures were treated for 12 h with 0.1, 1, 10 and 20 microM ketamine or co-incubated with NR1 antisense oligonucleotide (2 microM). After washout of the ketamine, cultures were kept in serum-containing medium (in presence of glutamate) for 24 h. Application of ketamine (10 and 20 microM) resulted in a substantial increase in DNA fragmentation as measured by cell death enzyme-linked immunosorbent assay, increased number of terminal dUTP nick-end labeling positive cells, and a reduction in mitochondrial metabolism of the dye 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide. No significant effect was seen in the release of lactate dehydrogenase, indicating that cell death presumably occurred via an apoptotic mechanism. Co-incubation of ketamine with NR1 antisense significantly reduced ketamine-induced apoptosis. Western analysis showed that neurotoxic concentrations of ketamine increased Bax and NR1 protein levels. NR1 antisense prevented this increase caused by ketamine, suggesting that ketamine-induced cell death is associated with a compensatory upregulation of the NMDA receptor. These data suggest that NR1 antisense offers neuroprotection from apoptosis in vitro, and that upregulation of the NR1 following ketamine administration is, at least, partially responsible for the observed apoptosis.


Subject(s)
Anesthetics, Dissociative/toxicity , Apoptosis/drug effects , Ketamine/toxicity , Prosencephalon/drug effects , Prosencephalon/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , Oligonucleotides, Antisense/pharmacology , Prosencephalon/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Thionucleotides/pharmacology , bcl-2-Associated X Protein
12.
Neurotoxicology ; 26(4): 729-37, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16112330

ABSTRACT

MPP(+) (1-methyl-4-phenylpyridinium; the active metabolite of the neurotoxin MPTP (1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine)) depletes dopamine (DA) content and elicits cell death in PC12 cells. However, the mechanism of MPP(+)-induced neurotoxicity is still unclear. In this study, the dose response and time-course of MPP(+)-induced DA depletion and decreased cell viability were determined in nerve growth factor (NGF)-differentiated PC12 cells. The alteration of transcription factors (TFs) induced by MPP(+) from a selected dose level and time point was then evaluated using protein/DNA-binding arrays. K-means clustering analysis identified four patterns of protein/DNA-binding changes. Three of the 28 TFs identified in PC12 cells increased by 100% (p53, PRE, Smad SBE) and 2 decreased by 50% (HSE, RXR(DR1)) of control with MPP(+) treatment. In addition, three TFs decreased within the range of 33-50% (TFIID, E2F1, CREB) and two TFs increased within the range of 50-100% (PAX-5, Stat4). An electrophoretic mobility shift assay (EMSA) was used to confirm the changes of p53 and HSE. The observed changes in TFs correlated with the alterations of DA and cell viability. The data indicates that selective transcription factors are involved in MPP(+)-induced neurotoxicity and it provides mechanistic information that may be applicable to animal studies with MPTP and clinical studies of Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Neurotoxicity Syndromes/metabolism , Transcription Factors/metabolism , Animals , Cell Survival/drug effects , DNA/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Kinetics , Neurotransmitter Agents/metabolism , Oligonucleotide Array Sequence Analysis , PC12 Cells , Protein Binding , Rats
13.
J Hand Surg Eur Vol ; 40(5): 450-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25294736

ABSTRACT

We conducted a systematic review of studies reporting clinical outcomes after proximal row carpectomy or to four-corner arthrodesis for scaphoid non-union advanced collapse or scapholunate advanced collapse arthritis. Seven studies (Levels I-III; 240 patients, 242 wrists) were evaluated. Significantly different post-operative values were as follows for four-corner arthrodesis versus proximal row carpectomy groups: wrist extension, 39 (SD 11Āŗ) versus 43 (SD 11Āŗ); wrist flexion, 32 (SD 10Āŗ) versus 36 (SD 11Āŗ); flexion-extension arc, 62 (SD 14Āŗ) versus 75 (SD 10Āŗ); radial deviation, 14 (SD 5Āŗ) versus 10 (SD 5Āŗ); hand grip strength as a percentage of contralateral side, 74% (SD 13) versus 67% (SD 16); overall complication rate, 29% versus 14%. The most common post-operative complications were non-union (grouped incidence, 7%) after four-corner arthrodesis and synovitis and clinically significant oedema (3.1%) after proximal row carpectomy. Radial deviation and post-operative hand grip strength (as a percentage of the contralateral side) were significantly better after four-corner arthrodesis. Four-corner arthrodesis gave significantly greater post-operative radial deviation and grip strength as a percentage of the opposite side. Wrist flexion, extension, and the flexion-extension arc were better after proximal row carpectomy, which also had a lower overall complication rate.


Subject(s)
Arthritis/physiopathology , Arthritis/surgery , Arthrodesis/methods , Carpal Bones/surgery , Orthopedic Procedures/methods , Wrist Joint , Hand Strength , Humans , Range of Motion, Articular , Treatment Outcome , Wrist Joint/physiopathology , Wrist Joint/surgery
14.
J Clin Endocrinol Metab ; 50(5): 811-8, 1980 May.
Article in English | MEDLINE | ID: mdl-6768757

ABSTRACT

Experiments were performed to describe and compare the transplacental pharmacokinetics of the teratogen and transplacental carcinogen, diethylstilbestrol [4,4'-dihydroxy-alpha,alpha'-diethyl-trans, cis-stilbene (DES)], and the endogenous estrogen, 17 beta-estradiol (E2). Timed mated pregnant rhesus monkeys (119-137 days gestation) were anesthetized, and catheters were implanted in the maternal femoral artery and the interplacental fetal artery and vein using an extraamniotic technique. Single doses of either radiolabeled DES or E2 were administered via the maternal radial vein. Maternal plasma levels of labeled compound decreased rapidly after dose administration. Fetal plasma levels of radioactivity derived from either DES or E2 increased rapidly and then plateaued higher than maternal levels 1--2 h after dose administration. High pressure liquid chromatography of maternal and fetal plasma samples revealed both parent and conjugated metabolites of DES and E2. The principal metabolite of DES (DES monglucuronide) was radiolabeled and given to either the mother or the fetus iv. There was no significant cross-over of this metabolite in either direction. It is concluded that DES crosses the primate placenta in an unconjugated form and that, based on total radioactivity, placental transfer is similar to that of E2. The extensive fetoplacental metabolism of DES appears to be responsible for the greater half-life of this agent and its metabolites in the fetal circulation compared with the maternal circulation.


Subject(s)
Diethylstilbestrol/metabolism , Estradiol/metabolism , Placenta/metabolism , Animals , Carbon Radioisotopes , Female , Haplorhini , Kinetics , Macaca mulatta , Mass Spectrometry , Pregnancy , Tritium
15.
Mol Neurobiol ; 11(1-3): 121-43, 1995.
Article in English | MEDLINE | ID: mdl-8561957

ABSTRACT

Two experiments were performed to investigate the interactive effects of prenatal coadministration of cocaine hydrochloride (C) and nicotine tartrate (N). Experiment I was designed to determine doses of C and N that could be coadministered without altering maternal gestational parameters and/or fetal viability. Exposure of Sprague-Dawley rats to combined high-dose C (20 mg/kg) and high-dose N (5.0 mg/kg) on gestation days 8-21 was not more toxic to dam or fetus that that of exposure to C alone. Experiment II investigated pregnancy outcome, postnatal development, and behavior of the offspring following drug exposure to either high-dose cocaine (20 mg/kg: CS), high-dose nicotine (5.0 mg/kg: NS), or both (NC) on gestation days 8-21. N was administered by osmotic minipump and C by sc injection. Saline-injected dams, fitted with saline-fitted pumps (SS), and untreated dams, pair-fed (PF) to NC females, served as controls. Alterations in maternal variables were limited to a 10-15% decrease in food consumption in NC and CS groups. Pregnancy outcome and birth statistics were unaffected by prenatal treatment, as was offspring body weight during the first four postnatal weeks. However, the development of surface righting was delayed inC CS pups, and only CS offspring were underresponsive to the stimulatory effects of dopamine agonists on activity and stereotypy. Behavioral responses to N challenge were similar in all groups. In addition, only CS offspring showed altered behavioral responses in a spontaneous alternation task. Treatment effects on dopamine D1 and D2 binding in the caudate nucleus were not observed. The combination of N and C did not exacerbate any of the behavioral changes seen in CS offspring. These results support the hypothesis that C is a behavioral teratogen in rodents, and suggest that in the present model, nicotine can mitigate some of the consequences of in utero exposure to cocaine.


Subject(s)
Cocaine/toxicity , Maze Learning/drug effects , Motor Activity/drug effects , Nicotine/toxicity , Pregnancy, Animal/drug effects , Prenatal Exposure Delayed Effects , Reflex/drug effects , Stereotyped Behavior/drug effects , Aging , Animals , Avoidance Learning/drug effects , Body Weight/drug effects , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Cocaine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Female , Fetal Blood/metabolism , Nicotine/pharmacokinetics , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Reference Values , Reflex, Startle/drug effects , Sex Characteristics , Weight Gain/drug effects
16.
Free Radic Biol Med ; 31(3): 405-11, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11461779

ABSTRACT

Oxidative stress is one of the major causes of cellular injury. Various reactive oxygen (ROS) and nitrogen (RNS) species such as superoxide, hydroxyl radical, peroxynitrite, and nitric oxide are involved in the manifestations of different types of organ toxicity and the resultant syndromes, symptoms, or diseases. Hypothermic conditions have been reported to reduce the oxidative stress in various in vitro and in vivo studies. In the present study, we sought to determine the effect of lowered temperatures on oxidative stress-induced cell death in Chinese hamster ovary (CHO) cells. We also investigated the oxidative stress-induced alterations in the expression of anti-apoptotic protein, bcl-2, in CHO cells at lowered temperatures. CHO cells were incubated at four different temperatures of 30, 32, 35, and 37 degrees C (control temperature) from 1 to 4 d. In another set, the cells were incubated with 100 microM hydrogen peroxide (H(2)O(2)) for 30 min before harvesting at different time points. The cells were harvested at 1, 2, 3, and 4 d. Cell survival was significantly higher at 30 degrees C as compared to 37 degrees C over 4 d of incubation. In cells incubated with H(2)O(2), significantly higher cell viability was observed at lower temperatures as compared to the cells incubated at 37 degrees C. The activity of glutathione peroxidase (GSH-Px) also increased significantly at lower temperatures. Lowered temperature also provided a significant increase in the expression of anti-apoptotic protein, bcl-2 after 4 d of incubation. These data suggest that hypothermic conditions lowers the risk of oxidative stress-induced cellular damage and programmed cell death by increasing the activity of GSH-Px and by the induction in the expression of the anti-apoptotic protein, bcl-2.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , CHO Cells , Cold Temperature , Cricetinae , Genes, bcl-2 , Glutathione Peroxidase/metabolism , Hypothermia , Kinetics , L-Lactate Dehydrogenase/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors
17.
Mech Ageing Dev ; 48(2): 157-66, 1989 May.
Article in English | MEDLINE | ID: mdl-2500570

ABSTRACT

The effects of long-term caloric restriction on the hepatic phase II drug metabolizing enzymes were investigated in the male Fischer 344 rat. Rats that had been restricted to 60% of their pair-fed control consumption from 14 weeks post-partum exhibited altered conjugating enzyme activities at 22 months. Caloric restriction significantly reduced the age-related decrease in glutathione-S-transferase activity towards 1,2-dichloro-4-nitrobenzene, but did not significantly alter the age-related changes in UDP-glucuronyltransferase or sulfotransferase activities towards hydroxysteroids. Caloric restriction appeared to increase hepatic microsomal UDP-glucuronyltransferase activity toward bilirubin and gamma-glutamyltranspeptidase activities. These observations suggest that caloric restriction has multiple effects on the hepatic phase II drug metabolizing enzymes in the rat. Such effects may alter hepatic metabolism and activation or detoxification of drugs and carcinogens.


Subject(s)
Aging/metabolism , Energy Intake , Glutathione Transferase/metabolism , Liver/enzymology , Animals , Epoxide Hydrolases/metabolism , Glucuronosyltransferase/metabolism , Inactivation, Metabolic , Male , Pharmaceutical Preparations/metabolism , Rats , Rats, Inbred F344 , Sulfotransferases/metabolism
18.
J Endocrinol ; 130(3): 339-45, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1940712

ABSTRACT

A synthetic version of the human relaxin encoded by the human gene 2 (hR1x-2) was administered to pregnant rhesus monkeys (Macaca mulatta) on gestational days 141-158. Monkeys (three per group) received doses of 100 micrograms/kg or 2000 micrograms/kg as a continuous i.v. infusion over 2 h into a radial vein. One monkey in the low-dose group received, along with the unlabelled hR1x-2, 25.5 microCi/kg of the test material internally labelled with [35S]cysteine. Immunoreactive hR1x-2, as measured by enzyme-linked immunosorbent assay, appeared in all fetuses within 30 min (the first sampling time) of beginning the infusions. Peak fetal plasma levels of hR1x-2 were only 0.8-1.5% of the maternal values. Only 8-15% of the fetal serum radioactivity was hR1x-2. Radioactivity from maternal urine pooled over the 4-h experiment did not elute at the volume corresponding to hR1x-2, but near the column volume.


Subject(s)
Maternal-Fetal Exchange/physiology , Placenta/metabolism , Pregnancy, Animal/metabolism , Relaxin/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Female , Fetal Blood/chemistry , Humans , Macaca mulatta , Pregnancy , Relaxin/administration & dosage , Relaxin/blood , Sulfur Radioisotopes , Time Factors
19.
Brain Res Mol Brain Res ; 91(1-2): 174-8, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11457507

ABSTRACT

Methamphetamine (METH)-induced alterations in the expression of p53 and bcl-2 protein were studied in the striatum of wild type, neuronal nitric oxide synthase knockout (nNOS -/-) and copper zinc superoxide dismutase overexpressed (SOD-Tg) mice. METH treatment up-regulated p53 and down-regulated bcl-2 expression in the striatum of wild type mice. No significant alterations were observed in the expression of these proteins in the nNOS -/- or SOD-Tg mice. These data suggest that METH might cause its neurotoxic effects via the production of free radicals and secondary perturbations in the expression of genes known to be involved in apoptosis and cell death machinery.


Subject(s)
Central Nervous System Stimulants/pharmacology , Corpus Striatum/physiology , Methamphetamine/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics , Animals , Corpus Striatum/drug effects , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Superoxide Dismutase/genetics
20.
Psychopharmacology (Berl) ; 127(3): 187-94, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8912396

ABSTRACT

It has been postulated that differences in pharmacokinetics do not contribute to the well-known individual variability in response to amphetamine (AMPH), but this is yet to be investigated thoroughly. Therefore, rotational behavior of outbred rats (Sprague-Dawley, 4 months old) was recorded during microdialysis sessions and striatal microdialysate was analyzed concomitantly for AMPH and dopamine concentrations after a single injection of 2.5 mg/kg AMPH SC. Three hours later these rats received three doses of 5 mg/kg AMPH SC (spaced 2 h apart) and their brain temperature was recorded every 20 min. The most important findings were: 1) the increase in extracellular dopamine was highly correlated with the corresponding peak AMPH levels in the microdialysate; 2) the peak dopamine level in response to 2.5 mg/kg AMPH was predictive of the hyperthermic response observed during 3 x 5 mg/kg AMPH and 3) high versus low rotators differed neither in their AMPH nor in their dopamine extracellular striatal concentrations.


Subject(s)
Amphetamine/pharmacology , Caudate Nucleus/drug effects , Central Nervous System Stimulants/pharmacology , Dopamine Agents/pharmacology , Dopamine/metabolism , Motor Activity/drug effects , Amphetamine/metabolism , Amphetamine/pharmacokinetics , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/physiology , Caudate Nucleus/metabolism , Central Nervous System Stimulants/metabolism , Dopamine Agents/metabolism , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL