Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 5673, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707598

ABSTRACT

The potential adverse impact of inhalational anesthetics on the developing brain was highlighted by the addition of a medication warning by the U.S. Food and Drug Administration for their use in the pediatric population. To investigate mechanisms by which early life anesthesia exposure could induce long-term neuronal dysfunction, we exposed rats to 1 minimum alveolar concentration sevoflurane at 7 days of life. The animals were raised normally until adulthood (P300) prior to sacrifice and analysis of cortical tissue structure (TEM), mitochondrial quality control and biogenesis pathways (Western blot, ELISA, ADP/ATP content), and markers of oxidative stress, proteotoxicity and inflammation (Western blot, ELISA). We found that early life anesthesia exposure led to adverse changes in mitochondrial quality maintenance pathways, autophagy and mitochondrial biogenesis. Although there was an escalation of oxidative stress markers and an increase in the nuclear localization of stress-related transcription factors, cellular redox compensatory responses were blunted, and oxidative phosphorylation was reduced. We found upregulation of mitochondrial stress and proteotoxicity markers, but a significant reduction of mitochondrial unfolded protein response end-effectors, contributing to an increase in inflammation. Contrary to acute exposure, we did not find an increase in apoptosis. Our findings suggest that a limited, early exposure to anesthesia may produce lasting cellular dysfunction through the induction of a sustained energy deficient state, resulting in persistent neuroinflammation and altered proteostasis/toxicity, mimicking aspects of chronic neurodegenerative diseases.


Subject(s)
Anesthesia/adverse effects , Brain/embryology , Neurodegenerative Diseases/pathology , Neurons/pathology , Animals , Animals, Newborn , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Chronic Disease , Homeostasis/drug effects , Inflammation Mediators/metabolism , Male , Mitochondria/ultrastructure , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Models, Biological , Neurons/drug effects , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Sevoflurane/toxicity , Signal Transduction/drug effects , Time Factors , Unfolded Protein Response/drug effects
2.
Int J Dev Neurosci ; 37: 87-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25003987

ABSTRACT

BACKGROUND: With growing evidence that anesthesia exposure in infancy affects cognitive development, it is important to understand how distinct anesthetic agents and combinations can alter long-term memory. Investigations of neuronal death suggest that combining anesthetic agents increases the extent of neuronal injury. However, it is unclear how the use of simultaneously combined anesthetics affects cognitive outcome relative to the use of a single agent. METHODS: Postnatal day 7 (P7) male rats were administered either sevoflurane as a single agent or the combined delivery of sevoflurane with nitrous oxide at 1 Minimum Alveolar Concentration for 4 h. Behavior was assessed in adulthood using the forced alternating T-maze, social recognition, and context-specific object recognition tasks. RESULTS: Animals exposed to either anesthetic were unimpaired in the forced alternating T-maze test and had intact social recognition. Subjects treated with the combined anesthetic displayed a deficit, however, in the object recognition task, while those treated with sevoflurane alone were unaffected. CONCLUSION: A combined sevoflurane and nitrous oxide anesthetic led to a distinct behavioral outcome compared with sevoflurane alone, suggesting that the simultaneous use of multiple agents may uniquely influence early neural and cognitive development and potentially impacts associative memory.


Subject(s)
Anesthetics, Inhalation/pharmacology , Cognition/drug effects , Methyl Ethers/pharmacology , Nitrous Oxide/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Body Weight/drug effects , Habituation, Psychophysiologic/drug effects , Interpersonal Relations , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Sevoflurane , Time Factors
3.
Neuropsychopharmacology ; 39(10): 2275-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24910347

ABSTRACT

Anesthesia in infancy impairs performance in recognition memory tasks in mammalian animals, but it is unknown if this occurs in humans. Successful recognition can be based on stimulus familiarity or recollection of event details. Several brain structures involved in recollection are affected by anesthesia-induced neurodegeneration in animals. Therefore, we hypothesized that anesthesia in infancy impairs recollection later in life in humans and rats. Twenty eight children ages 6-11 who had undergone a procedure requiring general anesthesia before age 1 were compared with 28 age- and gender-matched children who had not undergone anesthesia. Recollection and familiarity were assessed in an object recognition memory test using receiver operator characteristic analysis. In addition, IQ and Child Behavior Checklist scores were assessed. In parallel, thirty three 7-day-old rats were randomized to receive anesthesia or sham anesthesia. Over 10 months, recollection and familiarity were assessed using an odor recognition test. We found that anesthetized children had significantly lower recollection scores and were impaired at recollecting associative information compared with controls. Familiarity, IQ, and Child Behavior Checklist scores were not different between groups. In rats, anesthetized subjects had significantly lower recollection scores than controls while familiarity was unaffected. Rats that had undergone tissue injury during anesthesia had similar recollection indices as rats that had been anesthetized without tissue injury. These findings suggest that general anesthesia in infancy impairs recollection later in life in humans and rats. In rats, this effect is independent of underlying disease or tissue injury.


Subject(s)
Anesthesia, General/adverse effects , Memory, Long-Term/drug effects , Recognition, Psychology/drug effects , Animals , Association Learning/drug effects , Brain/drug effects , Brain/growth & development , Child , Female , Humans , Intelligence Tests , Male , Mental Recall/drug effects , Methyl Ethers/adverse effects , Neuropsychological Tests , Olfactory Perception/drug effects , ROC Curve , Random Allocation , Rats, Sprague-Dawley , Sevoflurane
4.
J Neurosurg Anesthesiol ; 25(3): 306-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23752046

ABSTRACT

BACKGROUND: Isoflurane exposure causes improvement in long-term neurocognitive function in young adult rats; this is associated with an increase in dentate gyrus (DG) progenitor proliferation 4 days after anesthesia. However, the number of new neurons that were born from cells that incorporated bromodeoxyuridine (BrdU) 4 days after anesthesia is not affected by anesthesia. We tested the hypothesis that progenitor proliferation continues to increase past 4 days, which would imply the possibility that the number of new neurons after anesthesia could be increased if BrdU labeling occurred at a later time point. METHODS: BrdU was injected at 0, 1, 2, 4, 9, 16 days after 4 hours of isoflurane exposure to 60-day old rats. Brains were harvested 2 hours later, immunohistochemically stained, and the number of BrdU+ cells in the DG was assessed microscopically. RESULTS: After 4 hours of exposure to isoflurane in 60-day old rats, the number of BrdU+ cells decreased on days 0 to 2, then increased on day 4 significantly, and regressed toward the control level on days 9 and 16. CONCLUSIONS: Anesthesia-induced progenitor proliferation in the DG was not sustained 9 days after anesthesia. We interpret these results to signify that an anesthetic effect on neurogenesis likely does not play a critical role in the previously observed isoflurane-induced long-term improvement in neurocognitive function in 60-day old rats and that the transient increase in progenitor proliferation serves to replenish the pool of neural stem cells. The mechanism of anesthesia-induced improvement in cognition of young adult rats remains elusive.


Subject(s)
Anesthetics, Inhalation/pharmacology , Dentate Gyrus/cytology , Isoflurane/pharmacology , Neural Stem Cells/drug effects , Anesthesia, General , Animals , Antimetabolites , Bromodeoxyuridine , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Neurogenesis/drug effects , Rats , Sample Size
5.
Anesthesiology ; 117(5): 1080-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001052

ABSTRACT

BACKGROUND: Propofol in the early postnatal period has been shown to cause brain cell death. One proposed mechanism for cognitive dysfunction after anesthesia is alteration of neural stem cell function and neurogenesis. We examined the effect of propofol on neural precursor or stem cells (NPCs) grown in vitro. METHODS: Hippocampal-derived NPCs from postnatal day 2 rats were exposed to propofol or Diprivan. NPCs were then analyzed for bromodeoxyuridine incorporation to measure proliferation. Cell death was measured by lactate dehydrogenase release. Immunocytochemistry was used to evaluate the expression of neuronal and glial markers in differentiating NPCs exposed to propofol. RESULTS: Propofol dose dependently increases the release of lactate dehydrogenase from NPCs under both proliferating and differentiating conditions at supraclinical concentrations (more than 7.1 µM). Both Diprivan and propofol had the same effect on NPCs. Propofol-mediated release of lactate dehydrogenase is not inhibited by blocking the γ-aminobutyric acid type A receptor or extracellular calcium influx and is not mediated by caspase-3/7. Direct γ-aminobutyric acid type A receptor activation did not have the same effect. In differentiating NPCs, 6 h of propofol at 2.1 µM increased the number neurons but not glial cells 4 days later. Increased neuronal differentiation was not blocked by bicuculline. CONCLUSIONS: Only supraclinical concentrations of propofol or Diprivan kill NPCs in culture by a non-γ-aminobutyric acid type A, noncaspase-3 mechanism. Clinically relevant doses of propofol increase neuronal fate choice by a non-γ-aminobutyric acid type A mechanism.


Subject(s)
Cell Differentiation/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Neural Stem Cells/drug effects , Neurons/cytology , Propofol/pharmacology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/growth & development , Neural Stem Cells/physiology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Propofol/toxicity , Rats , Rats, Sprague-Dawley
6.
Anesthesiology ; 116(3): 586-602, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22354242

ABSTRACT

BACKGROUND: Anesthesia given to immature rodents causes cognitive decline, raising the possibility that the same might be true for millions of children undergoing surgical procedures under general anesthesia each year. We tested the hypothesis that anesthesia-induced cognitive decline in rats is treatable. We also tested if anesthesia-induced cognitive decline is aggravated by tissue injury. METHODS: Seven-day old rats underwent sevoflurane anesthesia (1 minimum alveolar concentration, 4 h) with or without tail clamping. At 4 weeks, rats were randomized to environmental enrichment or normal housing. At 8 weeks rats underwent neurocognitive testing, which consisted of fear conditioning, spatial reference memory, and water maze-based memory consolidation tests, and interrogated working memory, short-term memory, and early long-term memory. RESULTS: Sevoflurane-treated rats had a greater escape latency when the delay between memory acquisition and memory retrieval was increased from 1 min to 1 h, indicating that short-term memory was impaired. Delayed environmental enrichment reversed the effects of sevoflurane on short-term memory and generally improved many tested aspects of cognitive function, both in sevoflurane-treated and control animals. The performance of tail-clamped rats did not differ from those rats receiving anesthesia alone. CONCLUSION: Sevoflurane-induced cognitive decline in rats is treatable. Delayed environmental enrichment rescued the sevoflurane-induced impairment in short-term memory. Tissue injury did not worsen the anesthesia-induced memory impairment. These findings may have relevance to neonatal and pediatric anesthesia.


Subject(s)
Housing, Animal , Memory Disorders/chemically induced , Memory Disorders/therapy , Methyl Ethers/toxicity , Age Factors , Animals , Animals, Newborn , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Sevoflurane , Time Factors
8.
Anesth Analg ; 113(5): 1170-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21965351

ABSTRACT

Anesthesia kills neurons in the brain of infantile animals, including primates, and causes permanent and progressive neurocognitive decline. The anesthesia community and regulatory authorities alike are concerned that is also true in humans. In this review, I summarize what we currently know about the risks of pediatric anesthesia to long-term cognitive function. If anesthesia is discovered to cause cognitive decline in humans, we need to know how to prevent and treat it. Prevention requires knowledge of the mechanisms of anesthesia-induced cognitive decline. This review gives an overview of some of the mechanisms that have been proposed for anesthesia-induced cognitive decline and discusses possible treatment options. If anesthesia induces cognitive decline in humans, we need to know what type and duration of anesthetic is safe, and which, if any, is not safe. This review discusses early results of comparative animal studies of anesthetic neurotoxicity. Until we know if and how pediatric anesthesia affects cognition in humans, a change in anesthetic practice would be premature, not guided by evidence of better alternatives, and therefore potentially dangerous. The SmartTots initiative jointly supported by the International Anesthesia Research Society and the Food and Drug Administration aims to fund research designed to shed light on these issues that are of high priority to the anesthesia community and the public alike and therefore deserves the full support of these interest groups.


Subject(s)
Anesthetics/toxicity , Brain/growth & development , Neurotoxicity Syndromes/pathology , Animals , Brain/drug effects , Brain/pathology , Cognition Disorders/chemically induced , Cognition Disorders/psychology , Humans , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology
10.
Anesthesiology ; 112(2): 305-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098132

ABSTRACT

BACKGROUND: Roughly, 10% of elderly patients develop postoperative cognitive dysfunction. General anesthesia impairs spatial memory in aged rats, but the mechanism is not known. Hippocampal neurogenesis affects spatial learning and memory in rats, and isoflurane affects neurogenesis in neonatal and young adult rats. We tested the hypothesis that isoflurane impairs neurogenesis and hippocampal function in aged rats. METHODS: Isoflurane was administered to 16-month-old rats at one minimum alveolar concentration for 4 h. FluoroJade staining was performed to assess brain cell death 16 h after isoflurane administration. Dentate gyrus progenitor proliferation was assessed by bromodeoxyuridine injection 4 days after anesthesia and quantification of bromodeoxyuridine+ cells 12 h later. Neuronal differentiation was studied by determining colocalization of bromodeoxyuridine with the immature neuronal marker NeuroD 5 days after anesthesia. New neuronal survival was assessed by quantifying cells coexpressing bromodeoxyuridine and the mature neuronal marker NeuN 5 weeks after anesthesia. Four months after anesthesia, associative learning was assessed by fear conditioning. Spatial reference memory acquisition and retention was tested in the Morris Water Maze. RESULTS: Cell death was sporadic and not different between groups. We did not detect any differences in hippocampal progenitor proliferation, neuronal differentiation, new neuronal survival, or in any of the tests of long-term hippocampal function. CONCLUSION: In aged rats, isoflurane does not affect brain cell death, hippocampal neurogenesis, or long-term neurocognitive outcome.


Subject(s)
Anesthetics, Inhalation/pharmacology , Brain/pathology , Cell Death/drug effects , Cognition/drug effects , Hippocampus/growth & development , Isoflurane/pharmacology , Neurons/physiology , Aging/physiology , Aging/psychology , Algorithms , Anesthetics, Inhalation/toxicity , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cognition Disorders/chemically induced , Cognition Disorders/psychology , Conditioning, Psychological/drug effects , Fear/drug effects , Fear/psychology , Hippocampus/cytology , Hippocampus/drug effects , Immunohistochemistry , Isoflurane/toxicity , Male , Maze Learning/drug effects , Memory/drug effects , Neurons/drug effects , Rats , Treatment Outcome
11.
Anesth Analg ; 110(2): 431-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19917621

ABSTRACT

Anesthetic drugs cause brain cell death and long-term neurocognitive dysfunction in neonatal rats. Recently, human data also suggest that anesthesia early in life may cause cognitive impairment. The connection between cell death and neurocognitive decline is uncertain. It is conceivable that mechanisms other than brain cell death contribute to neurocognitive outcome of neonatal anesthesia. In a series of experiments, we demonstrate that isoflurane exposure causes significant hypercarbia in postnatal day 7 rats and that exposure to isoflurane or carbon dioxide for 4 h provoked brain cell death. However, 1 h of isoflurane exposure was not sufficient to cause brain cell death. Moreover, only 4 h of isoflurane exposure, but not 1 or 2 h of exposure or 4 h of carbon dioxide, led to impaired hippocampal function,questioning the association between anesthesia-induced brain cell death and neurocognitive dysfunction. Neurogenesis both in the developing and adult dentate gyrus is important for hippocampal function, specifically learning and memory. γ-Amino-butyric-acid regulates proliferation and neuronal differentiation both in the developing and the adult brain. Inhaled anesthetics are γ-amino-butyric-acid-ergic and may therefore affect neurogenesis, which could be an alternative mechanism mediating anesthesia-induced neurocognitive decline in immature rats. Understanding the mechanism will help guide clinical trials aiming to define the scope of the problem in humans and may lead to preventive and therapeutic strategies.


Subject(s)
Anesthetics, Inhalation/pharmacology , Brain/pathology , Cell Death/drug effects , Cognition/drug effects , Isoflurane/pharmacology , Nervous System Diseases/chemically induced , Nervous System Diseases/pathology , Neurogenesis/drug effects , Anesthetics, Inhalation/toxicity , Animals , Humans , Isoflurane/toxicity , Rats
12.
Anesth Analg ; 110(2): 431-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-25508825

ABSTRACT

Anesthetic drugs cause brain cell death and long-term neurocognitive dysfunction in neonatal rats. Recently, human data also suggest that anesthesia early in life may cause cognitive impairment. The connection between cell death and neurocognitive decline is uncertain. It is conceivable that mechanisms other than brain cell death contribute to neurocognitive outcome of neonatal anesthesia. In a series of experiments, we demonstrate that isoflurane exposure causes significant hypercarbia in postnatal day 7 rats and that exposure to isoflurane or carbon dioxide for 4 h provoked brain cell death. However, 1 h of isoflurane exposure was not sufficient to cause brain cell death. Moreover, only 4 h of isoflurane exposure, but not 1 or 2 h of exposure or 4 h of carbon dioxide, led to impaired hippocampal function,questioning the association between anesthesia-induced brain cell death and neurocognitive dysfunction. Neurogenesis both in the developing and adult dentate gyrus is important for hippocampal function, specifically learning and memory. γ-Amino-butyric-acid regulates proliferation and neuronal differentiation both in the developing and the adult brain. Inhaled anesthetics are γ-amino-butyric-acid-ergic and may therefore affect neurogenesis, which could be an alternative mechanism mediating anesthesia-induced neurocognitive decline in immature rats. Understanding the mechanism will help guide clinical trials aiming to define the scope of the problem in humans and may lead to preventive and therapeutic strategies.


Subject(s)
Anesthetics, Inhalation/pharmacology , Brain/cytology , Cognition/drug effects , Isoflurane/pharmacology , Nervous System Diseases/chemically induced , Nervous System Diseases/physiopathology , Neurogenesis/drug effects , Animals , Brain/drug effects , Cell Death/drug effects , Humans , Rats
13.
Anesth Analg ; 109(3): 801-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19690249

ABSTRACT

BACKGROUND: While studying neurotoxicity in rats, we observed that the anesthetic minimum alveolar anesthetic concentration (MAC) of isoflurane decreases with increasing duration of anesthesia in 7-day-old but not in 60-day-old rats. After 15 min of anesthesia in 7-day-old rats, MAC was 3.5% compared with 1.3% at 4 h. We investigated whether kinetic or dynamic factors mediated this decrease. METHODS: In 7-day-old rats, we measured inspired and cerebral partial pressures of isoflurane at MAC as a function of duration of anesthesia. In 60-day-old rats, we measured inspired partial pressures of isoflurane at MAC as a function of duration of anesthesia. Finally, we determined the effect of administering 1 mg/kg naloxone and of delaying the initiation of the MAC determination (pinching the tail) on MAC in 7-day-old rats. RESULTS: In 7-day-old rats, both inspired and cerebral measures of MAC decreased from 1 to 4 h. The inspired MAC decreased 56%, whereas the cerebral MAC decreased 33%. At 4 h, the inspired MAC approximated the cerebral MAC (i.e., the partial pressures did not differ appreciably). Neither administration of 1 mg/kg naloxone nor delaying tail clamping until 3 h reversed the decrease in MAC. In 60-day-old rats, inspired MAC of isoflurane was stable from 1 to 4 h of anesthesia. CONCLUSIONS: MAC of isoflurane decreases over 1-4 h of anesthesia in 7-day-old but not in 60-day-old rats. Both pharmacodynamic and a pharmacokinetic components contribute to the decrease in MAC in 7-day-old rats. Neither endorphins nor sensory desensitization mediate the pharmacodynamic component.


Subject(s)
Anesthesia/methods , Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Pulmonary Alveoli/drug effects , Algorithms , Animals , Brain/drug effects , Desflurane , Endorphins/metabolism , Gases , Isoflurane/analogs & derivatives , Kinetics , Methyl Ethers/pharmacology , Naloxone/pharmacology , Rats , Sevoflurane , Time Factors
14.
Anesthesiology ; 110(4): 849-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19293696

ABSTRACT

BACKGROUND: Millions of neonates undergo anesthesia each year. Certain anesthetic agents cause brain cell death and long-term neurocognitive dysfunction in postnatal day (P)7 rats. Despite its intuitive appeal, a causal link between cell death and neurocognitive decline after anesthesia has not been established. If one existed, the degree of cell death would be expected to correlate with the degree of neurocognitive dysfunction caused by anesthesia. The authors therefore tested if cell death caused by various durations of isoflurane at 1 minimum alveolar concentration causes duration-dependent long-term neurocognitive dysfunction. METHODS: Isoflurane was administered to P7 rats at 1 minimum alveolar concentration for 0, 1, 2, or 4 h. To control for the respiratory depressant effects of anesthesia, a group of rats was treated with 4 h of carbon dioxide. Cell death was assessed by FluoroJade staining 12 h after the end of each intervention, and neurocognitive outcome was assessed 8 weeks later by using fear conditioning, spatial reference memory, and spatial working memory tasks. RESULTS: Widespread brain cell death was caused by 2 h and 4 h of isoflurane and by 4 h of carbon dioxide. The degree and distribution of thalamic cell death was similar in 4 h isoflurane-treated and 4-h carbon dioxide-treated rats. Only 4 h of isoflurane caused a long-term neurocognitive deficit affecting both spatial reference memory and spatial working memory. Working memory was improved in carbon dioxide-treated rats. CONCLUSION: Isoflurane-induced brain cell death may be partly caused by hypercarbia. The inconsistencies between cell death and neurocognitive outcome suggest that additional or alternative mechanisms may mediate anesthesia-induced long-term neurocognitive dysfunction.


Subject(s)
Anesthetics, Inhalation/toxicity , Isoflurane/toxicity , Memory Disorders/chemically induced , Neurons/drug effects , Animals , Blood Gas Analysis , Carbon Dioxide/toxicity , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Fear , Female , Male , Neurons/cytology , Rats , Rats, Sprague-Dawley , Survival Rate , Time Factors , Treatment Outcome
15.
Anesthesiology ; 110(4): 826-33, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19293697

ABSTRACT

BACKGROUND: Isoflurane causes long-term hippocampal-dependent learning deficits in rats despite limited isoflurane-induced hippocampal cell death, raising questions about the causality between isoflurane-induced cell death and isoflurane-induced cognitive function. Neurogenesis in the dentate gyrus is required for hippocampal-dependent learning and thus constitutes a potential alternative mechanism by which cognition can be altered after neonatal anesthesia. The authors tested the hypothesis that isoflurane alters proliferation and differentiation of hippocampal neural progenitor cells. METHODS: Multipotent neural progenitor cells were isolated from pooled rat hippocampi (postnatal day 2) and grown in culture. These cells were exposed to isoflurane and evaluated for cell death using lactate dehydrogenase release, caspase activity, and immunocytochemistry for nuclear localization of cleaved caspase 3. Growth was assessed by cell counting and BrdU incorporation. Expression of markers of stemness (Sox2) and cell division (Ki67) were determined by quantitative polymerase chain reaction. Cell fate selection was assessed using immunocytochemistry to stain for neuronal and glial markers. RESULTS: Isoflurane did not change lactate dehydrogenase release, activity of caspase 3/7, or the amount of nuclear cleaved caspase 3. Isoflurane decreased caspase 9 activity, inhibited proliferation, and decreased the proportion of cells in s-phase. messenger ribonucleic acid expression of Sox2 (stem cells) and Ki67 (proliferation) were decreased. Differentiating neural progenitor cells more often select a neuronal fate after isoflurane exposure. CONCLUSIONS: The authors conclude that isoflurane does not cause cell death, but it does act directly on neural progenitor cells independently of effects on the surrounding brain to decrease proliferation and increase neuronal fate selection. These changes could adversely affect cognition after isoflurane anesthesia.


Subject(s)
Anesthetics, Inhalation/adverse effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Isoflurane/adverse effects , Neurons/drug effects , Animals , Antimetabolites/metabolism , Bromodeoxyuridine/metabolism , Caspases/metabolism , Cell Death , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Immunohistochemistry , Ki-67 Antigen/metabolism , L-Lactate Dehydrogenase/metabolism , Neurons/cytology , Pluripotent Stem Cells/drug effects , Rats , Rats, Sprague-Dawley , SOXB1 Transcription Factors/metabolism , Treatment Outcome
16.
Anesthesiology ; 110(4): 834-48, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19293705

ABSTRACT

BACKGROUND: Anesthetic agents cause cell death in the developing rodent brain and long-term, mostly hippocampal-dependent, neurocognitive dysfunction. However, a causal link between these findings has not been shown. Postnatal hippocampal neurogenesis affects hippocampal function into adulthood; therefore, the authors tested the hypothesis that isoflurane affects long-term neurocognitive function via an effect on dentate gyrus neurogenesis. METHODS: The S-phase marker 5-bromodeoxyuridine was administered at various times before, during, and after 4 h of isoflurane given to postnatal day (P)60 and P7 rats to assess dentate gyrus progenitor proliferation, early neuronal lineage selection, and long-term survival of new granule cell neurons. Fear conditioning and spatial reference memory was tested at various intervals from 2 weeks until 8 months after anesthesia. RESULTS: In P60 rats, isoflurane increased early neuronal differentiation as assessed by BrdU/NeuroD costaining, decreased progenitor proliferation for 1 day, and subsequently increased progenitor proliferation 5-10 days after anesthesia. In P7 rats, isoflurane did not induce neuronal lineage selection but decreased progenitor proliferation until at least 5 days after anesthesia. Isoflurane improved spatial reference memory of P60 rats long-term, but it caused a delayed-onset, progressive, persistent hippocampal deficit in P7 rats in fear conditioning and spatial reference memory tasks. CONCLUSION: The authors conclude that isoflurane differentially affects both neurogenesis and long-term neurocognitive function in P60 and P7 rats. Neurogenesis might mediate the long-term neurocognitive outcome after isoflurane at different ages.


Subject(s)
Anesthetics, Inhalation/adverse effects , Cognition/drug effects , Dentate Gyrus/drug effects , Isoflurane/adverse effects , Neurogenesis/drug effects , Age Factors , Animals , Bromodeoxyuridine , Cell Death , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Conditioning, Psychological/drug effects , Dentate Gyrus/cytology , Male , Memory Disorders/chemically induced , Neurons/cytology , Neurons/drug effects , Rats , Treatment Outcome
19.
Anesth Analg ; 101(5): 1497-1498, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16244018

ABSTRACT

Headaches complicating lumboperitoneal (LP) shunt placement have been attributed to shunt failure with resultant high intracranial pressure or to overdrainage with resultant low intracranial pressure. In this case, a 17-yr-old girl had symptoms of a low-pressure headache after LP shunt placement alleviated by an epidural blood patch. The success of this therapy suggests postdural puncture as a possible cause for low-pressure headache after LP shunt placement. Epidural blood patch may be an alternative initial therapy for some low-pressure headaches after LP shunt placement.


Subject(s)
Blood Patch, Epidural , Cerebrospinal Fluid Shunts/adverse effects , Post-Dural Puncture Headache/therapy , Adolescent , Female , Humans , Post-Dural Puncture Headache/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...