Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Women Birth ; 33(1): e48-e58, 2020 Feb.
Article in English | MEDLINE | ID: mdl-30545755

ABSTRACT

PROBLEM AND BACKGROUND: Psychotropic medication use is increasingly common among pregnant women. Many women solicit information from other mothers about the safety of these medications for use during pregnancy, yet little is known about the specific advice they receive. AIM: The purpose of the current study was to examine the type of feedback women receive on a popular internet message board about psychotropic medication use during pregnancy. METHODS: A modified Consensual Qualitative Research approach was used to analyze 1728 comments posted by Babycenter.com users about the safety of the use of six common psychotropic medications during pregnancy. Researchers analyzed the comments for overall themes and core ideas. FINDINGS: Results found that comments were comprised of six themes: (1) Personal Anecdotes, (2) Suggesting Alternative Solutions, (3) Directives, (4) Judgement, (5) Social Support, (6) Skepticism & Mistrust, and (7) Risks vs. Benefits. While many comments conveyed emotional support, or encouraged women to seek professional advice, others contained inaccurate and/or contradictory information, or harsh criticism. CONCLUSION: Given that the decision about the use of medication during pregnancy has implications for the health of the mother and fetus, it is important for care providers to be aware of what feedback women may receive from this source. Providers should address questions and concerns that women have about safety of these medications and recognize how the social context of the internet impacts the emotional health of pregnant women faced with these decisions.


Subject(s)
Internet , Pregnant Women/psychology , Social Media , Social Support , Female , Humans , Maternal Health , Pregnancy , Social Learning
2.
Brain Sci ; 9(9)2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31540060

ABSTRACT

This paper reviews the findings from preclinical animal and human clinical research investigating maternal/fetal, neonatal, and child neurodevelopmental outcomes following prenatal exposure to psychotropic drugs. Evidence for the risks associated with prenatal exposure was examined, including teratogenicity, neurodevelopmental effects, neonatal toxicity, and long-term neurobehavioral consequences (i.e., behavioral teratogenicity). We conducted a comprehensive review of the recent results and conclusions of original research and reviews, respectively, which have investigated the short- and long-term impact of drugs commonly prescribed to pregnant women for psychological disorders, including mood, anxiety, and sleep disorders. Because mental illness in the mother is not a benign event, and may itself pose significant risks to both mother and child, simply discontinuing or avoiding medication use during pregnancy may not be possible. Therefore, prenatal exposure to psychotropic drugs is a major public health concern. Decisions regarding drug choice, dose, and duration should be made carefully, by balancing severity, chronicity, and co-morbidity of the mental illness, disorder, or condition against the potential risk for adverse outcomes due to drug exposure. Globally, maternal mental health problems are considered as a major public health challenge, which requires a stronger focus on mental health services that will benefit both mother and child. More preclinical and clinical research is needed in order to make well-informed decisions, understanding the risks associated with the use of psychotropic medications during pregnancy.

3.
Sci Rep ; 9(1): 2779, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808927

ABSTRACT

Epidemiological studies suggest exposures to anesthetic agents and/or sedative drugs (AASDs) in children under three years old, or pregnant women during the third trimester, may adversely affect brain development. Evidence suggests lengthy or repeated AASD exposures are associated with increased risk of neurobehavioral deficits. Animal models have been valuable in determining the type of acute damage in the developing brain induced by AASD exposures, as well as in elucidating long-term functional consequences. Few studies examining very early exposure to AASDs suggest this may be a critical period for inducing long-term functional consequences, but the impact of repeated exposures at these ages has not yet been assessed. To address this, we exposed mouse pups to a prototypical general anesthetic, isoflurane (ISO, 1.5% for 3 hr), at three early postnatal ages (P3, P5 and P7). We quantified the acute neuroapoptotic response to a single versus repeated exposure, and found age- and brain region-specific effects. We also found that repeated early exposures to ISO induced subtle, sex-specific disruptions to activity levels, motor coordination, anxiety-related behavior and social preference. Our findings provide evidence that repeated ISO exposures may induce behavioral disturbances that are subtle in nature following early repeated exposures to a single AASD.


Subject(s)
Anesthetics, Inhalation/toxicity , Behavior, Animal/drug effects , Brain/pathology , Isoflurane/toxicity , Animals , Animals, Newborn , Apoptosis , Brain/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy
4.
Neurobiol Learn Mem ; 165: 106834, 2019 11.
Article in English | MEDLINE | ID: mdl-29550366

ABSTRACT

Fifteen years ago Olney and colleagues began using animal models to evaluate the effects of anesthetic and sedative agents (ASAs) on neurodevelopment. The results from ongoing studies indicate that, under certain conditions, exposure to these drugs during development induces an acute elevated apoptotic neurodegenerative response in the brain and long-term functional impairments. These animal models have played a significant role in bringing attention to the possible adverse effects of exposing the developing brain to ASAs when few concerns had been raised previously in the medical community. The apoptotic degenerative response resulting from neonatal exposure to ASAs has been replicated in many studies in both rodents and non-human primates, suggesting that a similar effect may occur in humans. In both rodents and non-human primates, significantly increased levels of apoptotic degeneration are often associated with functional impairments later in life. However, behavioral deficits following developmental ASA exposure have not been consistently reported even when significantly elevated levels of apoptotic degeneration have been documented in animal models. In the present work, we review this literature and propose a rodent model for assessing potential functional deficits following neonatal ASA exposure with special reference to experimental design and procedural issues. Our intent is to improve test sensitivity and replicability for detecting subtle behavioral effects, and thus enhance the translational significance of ASA models.


Subject(s)
Anesthesia/adverse effects , Neurodevelopmental Disorders/chemically induced , Anesthetics/adverse effects , Animals , Apoptosis/drug effects , Disease Models, Animal
5.
Brain Sci ; 6(3)2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27537919

ABSTRACT

The fetal and neonatal periods are critical and sensitive periods for neurodevelopment, and involve rapid brain growth in addition to natural programmed cell death (i.e., apoptosis) and synaptic pruning. Apoptosis is an important process for neurodevelopment, preventing redundant, faulty, or unused neurons from cluttering the developing brain. However, animal studies have shown massive neuronal cell death by apoptosis can also be caused by exposure to several classes of drugs, namely gamma-aminobutyric acid (GABA) agonists and N-methyl-d-aspartate (NMDA) antagonists that are commonly used in pediatric anesthesia. This form of neurotoxic insult could cause a major disruption in brain development with the potential to permanently shape behavior and cognitive ability. Evidence does suggest that psychoactive drugs alter neurodevelopment and synaptic plasticity in the animal brain, which, in the human brain, may translate to permanent neurodevelopmental changes associated with long-term intellectual disability. This paper reviews the seminal animal research on drug-induced developmental apoptosis and the subsequent clinical studies that have been conducted thus far. In humans, there is growing evidence that suggests anesthetics have the potential to harm the developing brain, but the long-term outcome is not definitive and causality has not been determined. The consensus is that there is more work to be done using both animal models and human clinical studies.

6.
Anesthesiology ; 120(3): 626-38, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24158051

ABSTRACT

BACKGROUND: The authors have previously shown that exposure of the neonatal nonhuman primate (NHP) brain to isoflurane for 5 h causes widespread acute apoptotic degeneration of neurons and oligodendrocyte. The current study explored the potential apoptogenic action of isoflurane in the fetal NHP brain. METHODS: Fetal rhesus macaques at gestational age of 120 days (G120) were exposed in utero for 5 h to isoflurane anesthesia (n = 5) or to no anesthesia (control condition; n = 4), and all regions of the brain were systematically evaluated 3 h later for evidence of apoptotic degeneration of neurons or glia. RESULTS: Exposure of the G120 fetal NHP brain to isoflurane caused a significant increase in apoptosis of neurons and of oligodendrocytes at a stage when oligodendrocytes were just beginning to myelinate axons. The neuroapoptosis response was most prominent in the cerebellum, caudate, putamen, amygdala, and several cerebrocortical regions. Oligodendrocyte apoptosis was diffusely distributed over many white matter regions. The total number of apoptotic profiles (neurons + oligodendrocytes) in the isoflurane-exposed brains was increased 4.1-fold, compared with the brains from drug-naive controls. The total number of oligodendrocytes deleted by isoflurane was higher than the number of neurons deleted. CONCLUSIONS: Isoflurane anesthesia for 5 h causes death of neurons and oligodendrocytes in the G120 fetal NHP brain. In the fetal brain, as the authors previously found in the neonatal NHP brain, oligodendrocytes become vulnerable when they are just achieving myelination competence. The neurotoxic potential of isoflurane increases between the third trimester (G120) and the neonatal period in the NHP brain.


Subject(s)
Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Brain/drug effects , Isoflurane/toxicity , Neurons/drug effects , Oligodendroglia/drug effects , Animals , Animals, Newborn , Brain/embryology , Brain/pathology , Disease Models, Animal , Female , Macaca mulatta , Neurons/pathology , Oligodendroglia/pathology
7.
Acta Neuropathol Commun ; 1: 23, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-24252271

ABSTRACT

BACKGROUND: In utero exposure of the fetal non-human primate (NHP) brain to alcohol on a single occasion during early or late third-trimester gestation triggers widespread acute apoptotic death of cells in both gray and white matter (WM) regions of the fetal brain. In a prior publication, we documented that the dying gray matter cells are neurons, and described the regional distribution and magnitude of this cell death response. Here, we present new findings regarding the magnitude, identity and maturational status of the dying WM cells in these alcohol-exposed fetal NHP brains. RESULTS: Our findings document that the dying WM cells belong to the oligodendrocyte (OL) lineage. OLs become vulnerable when they are just beginning to generate myelin basic protein in preparation for myelinating axons, and they remain vulnerable throughout later stages of myelination. We found no evidence linking astrocytes, microglia or OL progenitors to this WM cell death response. The mean density (profiles per mm3) of dying WM cells in alcohol-exposed brains was 12.7 times higher than the mean density of WM cells dying by natural apoptosis in drug-naive control brains. CONCLUSIONS: In utero exposure of the fetal NHP brain to alcohol on a single occasion triggers widespread acute apoptotic death of neurons (previous study) and of OLs (present study) throughout WM regions of the developing brain. The rate of OL apoptosis in alcohol-exposed brains was 12.7 times higher than the natural OL apoptosis rate. OLs become sensitive to the apoptogenic action of alcohol when they are just beginning to generate constituents of myelin in their cytoplasm, and they remain vulnerable throughout later stages of myelination. There is growing evidence for a similar apoptotic response of both neurons and OLs following exposure of the developing brain to anesthetic and anticonvulsant drugs. Collectively, this body of evidence raises important questions regarding the role that neuro and oligo apoptosis may play in the human condition known as fetal alcohol spectrum disorder (FASD), and also poses a question whether other apoptogenic drugs, although long considered safe for pediatric/obstetric use, may have the potential to cause iatrogenic FASD-like developmental disability syndromes.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Brain/embryology , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Oligodendroglia/drug effects , Animals , Apoptosis/physiology , Brain/pathology , Brain/physiopathology , Fetal Alcohol Spectrum Disorders , Macaca fascicularis , Myelin Basic Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/physiology , Oligodendroglia/pathology , Oligodendroglia/physiology , White Matter/drug effects , White Matter/embryology , White Matter/pathology , White Matter/physiopathology
8.
Brain Sci ; 3(3): 1153-81, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24587895

ABSTRACT

Maternal ingestion of alcohol during pregnancy can cause a disability syndrome termed Fetal Alcohol Spectrum Disorder (FASD), which may include craniofacial malformations, structural pathology in the brain, and a variety of long-term neuropsychiatric disturbances. There is compelling evidence that exposure to alcohol during early embryogenesis (4th week of gestation) can cause excessive death of cell populations that are essential for normal development of the face and brain. While this can explain craniofacial malformations and certain structural brain anomalies that sometimes accompany FASD, in many cases these features are absent, and the FASD syndrome manifests primarily as neurobehavioral disorders. It is not clear from the literature how alcohol causes these latter manifestations. In this review we will describe a growing body of evidence documenting that alcohol triggers widespread apoptotic death of neurons and oligodendroglia (OLs) in the developing brain when administered to animals, including non-human primates, during a period equivalent to the human third trimester of gestation. This cell death reaction is associated with brain changes, including overall or regional reductions in brain mass, and long-term neurobehavioral disturbances. We will also review evidence that many drugs used in pediatric and obstetric medicine, including general anesthetics (GAs) and anti-epileptics (AEDs), mimic alcohol in triggering widespread apoptotic death of neurons and OLs in the third trimester-equivalent animal brain, and that human children exposed to GAs during early infancy, or to AEDs during the third trimester of gestation, have a significantly increased incidence of FASD-like neurobehavioral disturbances. These findings provide evidence that exposure of the developing human brain to GAs in early infancy, or to alcohol or AEDs in late gestation, can cause FASD-like neurodevelopmental disability syndromes. We propose that the mechanism by which alcohol, GAs and AEDs produce neurobehavioral deficit syndromes is by triggering apoptotic death and deletion of neurons and OLs (or their precursors) from the developing brain. Therefore, there is a need for research aimed at deciphering mechanisms by which these agents trip the apoptosis trigger, the ultimate goal being to learn how to prevent these agents from causing neurodevelopmental disabilities.

9.
Brain Sci ; 3(3): 1128-52, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-24734165

ABSTRACT

Anesthetic and anti-epileptic drugs used in pediatric and obstetric medicine and several drugs, including alcohol, that are abused by pregnant women, trigger widespread neuroapoptosis in the developing brain of several animal species, including non-human primates. Caffeine (CAF) is often administered to premature infants to stimulate respiration, and these infants are also exposed simultaneously to anesthetic drugs for procedural sedation and/or surgical procedures. Pregnant women who abuse alcohol or other apoptogenic drugs also may heavily consume CAF. We administered CAF to infant mice alone or in combination with alcohol, phencyclidine, diazepam, midazolam, ketamine, or isoflurane, which are drugs of abuse and/or drugs frequently used in pediatric medicine, and found that CAF weakly triggers neuroapoptosis by itself and markedly potentiates the neuroapoptogenic action of each of these other drugs. Exposure of infant mice to CAF + phencyclidine resulted in long-term impairment in behavioral domains relevant to attention deficit/hyperactivity disorder, whereas exposure to CAF + diazepam resulted in long-term learning/memory impairment. At doses used in these experiments, these behavioral impairments either did not occur or were substantially less pronounced in mice exposed to CAF alone or to phencyclidine or diazepam alone. CAF currently enjoys the reputation of being highly beneficial and safe for use in neonatal medicine. Our data suggest the need to consider whether CAF may have harmful as well as beneficial effects on the developing brain, and the need for research aimed at understanding the full advantage of its beneficial effects while avoiding its potentially harmful effects.

10.
Ann Neurol ; 72(4): 525-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23109147

ABSTRACT

OBJECTIVE: Previously we reported that exposure of 6-day-old (P6) rhesus macaques to isoflurane for 5 hours triggers a robust neuroapoptosis response in developing brain. We have also observed (unpublished data) that isoflurane causes apoptosis of cellular profiles in the white matter that resemble glia. We analyzed the cellular identity of the apoptotic white matter profiles and determined the magnitude of this cell death response to isoflurane. METHODS: Neonatal (P6) rhesus macaques were exposed for 5 hours to isoflurane anesthesia according to current clinical standards in pediatric anesthesia. Brains were collected 3 hours later and examined immunohistochemically to analyze apoptotic neuronal and glial death. RESULTS: Brains exposed to isoflurane displayed significant apoptosis in both the white and gray matter throughout the central nervous system. Approximately 52% of the dying cells were glia, and 48% were neurons. Oligodendrocytes (OLs) engaged in myelinogenesis were selectively vulnerable, in contrast to OL progenitors, astrocytes, microglia, and interstitial neurons. When adjusted for control rates of OL apoptosis, the percentage of OLs that degenerated in the forebrain white matter of the isoflurane-treated group was 6.3% of the total population of myelinating OLs. INTERPRETATION: Exposure of the infant rhesus macaque brain to isoflurane for 5 hours is sufficient to cause widespread apoptosis of neurons and OLs throughout the developing brain. Deletion of OLs at a stage when they are just beginning to myelinate axons could potentially have adverse long-term neurobehavioral consequences that might be additive to the potential consequences of isoflurane-induced neuroapoptosis.


Subject(s)
Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Brain/pathology , Isoflurane/toxicity , Oligodendroglia/drug effects , Animals , Animals, Newborn , Axons/drug effects , Axons/physiology , Caspases/physiology , Cell Death/physiology , Immunohistochemistry , Macaca mulatta , Myelin Sheath/physiology , Nerve Regeneration/physiology , Tissue Fixation
11.
Anesthesiology ; 116(2): 372-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22222480

ABSTRACT

BACKGROUND: Exposure of rhesus macaque fetuses for 24 h or neonates for 9 h to ketamine anesthesia causes neuroapoptosis in the developing brain. The current study clarifies the minimum exposure required for and the extent and spatial distribution of ketamine-induced neuroapoptosis in rhesus fetuses and neonates. METHOD: Ketamine was administered by IV infusion for 5 h to postnatal day 6 rhesus neonates or to pregnant rhesus females at 120 days' gestation (full term = 165 days). Three hours later, fetuses were delivered by cesarean section, and the fetal and neonatal brains were studied for evidence of apoptotic neurodegeneration, as determined by activated caspase-3 staining. RESULTS: Both the fetal (n = 3) and neonatal (n = 4) ketamine-exposed brains had a significant increase in apoptotic profiles compared with drug-naive controls (fetal n = 4; neonatal n = 5). Loss of neurons attributable to ketamine exposure was 2.2 times greater in fetuses than in neonates. The pattern of neurodegeneration in fetuses was different from that in neonates, and all subjects exposed at either age had a pattern characteristic for that age. CONCLUSION: The developing rhesus macaque brain is sensitive to the apoptogenic action of ketamine at both a fetal and neonatal age, and exposure duration of 5 h is sufficient to induce a significant neuroapoptosis response at either age. The pattern of neurodegeneration induced by ketamine in fetuses was different from that in neonates, and loss of neurons attributable to ketamine exposure was 2.2 times greater in the fetal than neonatal brains.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Fetus/drug effects , Ketamine/toxicity , Nerve Degeneration/chemically induced , Animals , Animals, Newborn , Apoptosis/physiology , Brain/pathology , Female , Fetus/pathology , Infusions, Intravenous , Ketamine/administration & dosage , Macaca mulatta , Nerve Degeneration/pathology , Pregnancy , Random Allocation
12.
Neurotoxicol Teratol ; 33(4): 473-80, 2011.
Article in English | MEDLINE | ID: mdl-21640820

ABSTRACT

While the toxic effects of lead have been recognized for millennia, it has remained a significant public health concern due to its continued use and toxicological potential. Of particular interest is the increased susceptibility of young children to the toxic effects of lead. Although the exact mechanism(s) for lead toxicity is currently not well understood, research has established that it can be a potent NMDA antagonist. Previous research has established that exposure to NMDA antagonists during the brain growth spurt period (first 2 weeks of life in mice) can produce apoptotic neurodegeneration throughout the brain. Based on this information, the ability of lead exposure (two injections of 350 mg/kg lead 4h apart) to produce apoptosis in the neonatal mouse brain was assessed histologically 8-24h after treatment using activated caspase-3 immunohistochemistry, De Olmos silver technique, Nissl staining, and electron microscopy. Lead exposure produced significant neurodegeneration in the caudate/putamen, hippocampus, subiculum, and superficial and deep cortical layers of the frontal cortical regions. Further ultrastructural examination revealed cellular profiles consistent with apoptotic cell death. Statistical results showed that lead exposure significantly increased apoptotic neurodegeneration above that seen in normal controls in animals treated at postnatal day 7, but not on day 14. The results of this study may provide a basis for further elucidation of mechanisms through which the immature nervous system may be particularly susceptible to lead exposure.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Environmental Pollutants/toxicity , Lead/toxicity , Neurons/drug effects , Animals , Animals, Newborn , Brain/growth & development , Brain/ultrastructure , Caspase 3/metabolism , Cell Count , Dose-Response Relationship, Drug , Environmental Pollutants/blood , Immunohistochemistry , Lead/blood , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neurons/ultrastructure
13.
PLoS One ; 5(6): e11374, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20613880

ABSTRACT

BACKGROUND: Exposure to NMDA glutamate antagonists during the brain growth spurt period causes widespread neuroapoptosis in the rodent brain. This period in rodents occurs during the first two weeks after birth, and corresponds to the third trimester of pregnancy and several years after birth in humans. The developing human brain may be exposed to NMDA antagonists through drug-abusing mothers or through anesthesia. METHODOLOGY/PRINCIPAL FINDINGS: We evaluated the long-term neurobehavioral effects of mice exposed to a single dose of the NMDA antagonist, phencyclidine (PCP), or saline, on postnatal day 2 (P2) or P7, or on both P2 and P7. PCP treatment on P2 + P7 caused more severe cognitive impairments than either single treatment. Histological examination of acute neuroapoptosis resulting from exposure to PCP indicated that the regional pattern of degeneration induced by PCP in P2 pups was different from that in P7 pups. The extent of damage when evaluated quantitatively on P7 was greater for pups previously treated on P2 compared to pups treated only on P7. CONCLUSIONS: These findings signify that PCP induces different patterns of neuroapoptosis depending on the developmental age at the time of exposure, and that exposure at two separate developmental ages causes more severe neuropathological and neurobehavioral consequences than a single treatment.


Subject(s)
Apoptosis/drug effects , Behavior, Animal/drug effects , Brain/drug effects , N-Methylaspartate/antagonists & inhibitors , Animals , Animals, Newborn , Body Weight/drug effects , Brain/cytology , Brain/growth & development , Conditioning, Classical , Fear , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Phencyclidine/pharmacology , Reflex, Startle
14.
Neurobiol Dis ; 40(1): 200-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20580929

ABSTRACT

The ability of brief exposure to alcohol to cause widespread neuroapoptosis in the developing rodent brain and subsequent long-term neurocognitive deficits has been proposed as a mechanism underlying the neurobehavioral deficits seen in fetal alcohol spectrum disorder (FASD). It is unknown whether brief exposure to alcohol causes apoptosis in the fetal primate brain. Pregnant fascicularis macaques at various stages of gestation (G105 to G155) were exposed to alcohol for 8h, then the fetuses were delivered by caesarean section and their brains perfused with fixative and evaluated for apoptosis. Compared to saline control brains, the ethanol-exposed brains displayed a pattern of neuroapoptosis that was widespread and similar to that caused by alcohol in infant rodent brain. The observed increase in apoptosis was on the order of 60-fold. We propose that the apoptogenic action of alcohol could explain many of the neuropathological changes and long-term neuropsychiatric disturbances associated with human FASD.


Subject(s)
Alcohol-Induced Disorders, Nervous System/pathology , Apoptosis/drug effects , Brain/drug effects , Brain/pathology , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/pathology , Animals , Apoptosis/physiology , Brain/physiopathology , Cell Count , Central Nervous System Depressants/toxicity , Disease Models, Animal , Drug Administration Schedule , Female , Macaca fascicularis , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Time
15.
Anesthesiology ; 112(4): 834-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20234312

ABSTRACT

BACKGROUND: Brief isoflurane anesthesia induces neuroapoptosis in the developing rodent brain, but susceptibility of non-human primates to the apoptogenic action of isoflurane has not been studied. Therefore, we exposed postnatal day 6 (P6) rhesus macaques to a surgical plane of isoflurane anesthesia for 5 h, and studied the brains 3 h later for histopathologic changes. METHOD: With the same intensity of physiologic monitoring typical for human neonatal anesthesia, five P6 rhesus macaques were exposed for 5 h to isoflurane maintained between 0.7 and 1.5 end-tidal Vol% (endotracheally intubated and mechanically ventilated) and five controls were exposed for 5 h to room air without further intervention. Three hours later, the brains were harvested and serially sectioned across the entire forebrain and midbrain, and stained immunohistochemically with antibodies to activated caspase-3 for detection and quantification of apoptotic neurons. RESULTS: Quantitative evaluation of brain sections revealed a median of 32.5 (range, 18.0-48.2) apoptotic cells/mm of brain tissue in the isoflurane group and only 2.5 (range, 1.1-5.2) in the control group (difference significant at P = 0.008). Apoptotic neuronal profiles were largely confined to the cerebral cortex. In the control brains, they were sparse and randomly distributed, whereas in the isoflurane brains they were abundant and preferentially concentrated in specific cortical layers and regions. CONCLUSION: The developing non-human primate brain is sensitive to the apoptogenic action of isoflurane and displays a 13-fold increase in neuroapoptosis after 5 h exposure to a surgical plane of isoflurane anesthesia.


Subject(s)
Anesthetics, Inhalation/toxicity , Animals, Newborn/physiology , Apoptosis/drug effects , Brain/cytology , Isoflurane/toxicity , Neurons/drug effects , Animals , Brain/drug effects , Caspase 3/metabolism , Hemodynamics/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , Intubation, Intratracheal , Macaca mulatta , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure
16.
Anesth Analg ; 110(2): 442-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19955510

ABSTRACT

Millions of human fetuses, infants, and children are exposed to anesthetic drugs every year in the United States and throughout the world. Anesthesia administered during critical stages of neurodevelopment has been considered safe and without adverse long-term consequences. However, recent reports provide mounting evidence that exposure of the immature animal brain to anesthetics during the period of rapid synaptogenesis, also known as the brain growth spurt period, triggers widespread apoptotic neurodegeneration, inhibits neurogenesis, and causes significant long-term neurocognitive impairment. Herein, we summarize currently available evidence for anesthesia-induced pathological changes in the brain and associated long-term neurocognitive deficits and discuss promising strategies for protecting the developing brain from the potentially injurious effects of anesthetic drugs while allowing the beneficial actions of these drugs to be realized.


Subject(s)
Anesthetics/toxicity , Apoptosis/drug effects , Brain/drug effects , Neurons/drug effects , Anesthetics/adverse effects , Animals , Anticonvulsants/pharmacology , Body Temperature , Brain/growth & development , Brain/physiology , Ethanol/pharmacology , Fetus/drug effects , Humans , Hypothermia, Induced , Mice , Neurodegenerative Diseases/chemically induced
17.
Anesthesiology ; 110(4): 862-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19293695

ABSTRACT

BACKGROUND: Ethanol and anesthetic drugs trigger neuroapoptosis in the developing mouse brain. Recently, it was found that ethanol-induced neuroapoptosis is preceded by suppressed phosphorylation of extracellular signal-regulated protein kinase (ERK), and lithium counteracts both the phosphorylated ERK suppressant action and ethanol-induced neuroapoptosis. The current study was undertaken to address the following questions. (1) Do ketamine and propofol mimic ethanol in suppressing ERK phosphorylation? (2) If they do, does lithium prevent this suppressant action and also prevent these anesthetic drugs from triggering neuroapoptosis? METHOD: Postnatal day 5 mice were treated with propofol, ketamine, lithium, a combination of propofol or ketamine with lithium or saline, and their brains were prepared for Western blot analysis or histology. For Western blot, cytosolic lysates of caudate putamen were analyzed for expression of phosphorylated ERK and phosphorylated serine/threonine-specific protein kinase. For histology, brains were stained immunohistochemically with antibodies to activated caspase-3, and the density of activated caspase-3 positive cells was determined. RESULTS: Ketamine and propofol suppressed phosphorylated ERK, and lithium counteracted both the phosphorylated ERK suppressant action and neuroapoptotic action of these anesthetic drugs. CONCLUSION: If further testing finds lithium to be safe for use in pediatric/obstetric medicine, administration of a single dose of lithium before anesthesia induction may be a suitable means of mitigating the risk of anesthesia-induced developmental neuroapoptosis.


Subject(s)
Antipsychotic Agents/therapeutic use , Apoptosis/drug effects , Brain/drug effects , Lithium Carbonate/therapeutic use , Neurons/drug effects , Anesthetics, Intravenous/toxicity , Animals , Blotting, Western , Brain/embryology , Brain/pathology , Caspase 3/drug effects , Ketamine/toxicity , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Propofol/toxicity , Proto-Oncogene Proteins c-akt/drug effects , Random Allocation , Treatment Outcome
18.
Neonatology ; 96(1): 23-32, 2009.
Article in English | MEDLINE | ID: mdl-19204407

ABSTRACT

BACKGROUND: Magnesium sulfate (MgSO4) is often used as a treatment for pre-eclampsia/eclampsia and preterm labor, resulting in the exposure of a significant number of neonates to this drug despite a lack of evidence suggesting that it is safe, or effective as a tocolytic. While there is evidence that MgSO4 may be neuroprotective in perinatal brain injury, recent reviews have suggested that the effects are dependent upon dose, and that higher doses may actually increase neonatal morbidity and mortality. There is a lack of evidence investigating the neurotoxic effects of neonatal magnesium (Mg) exposure on the developing brain, specifically in terms of neurodevelopmental apoptosis, a cell-killing phenomenon known to be potentiated by other drugs with mechanisms of action at Mg-binding sites (i.e. NMDA receptor antagonists such as MK-801, ketamine, and PCP). OBJECTIVE: To investigate the effects of Mg exposure on the neonatal mouse brain at different postnatal ages to determine whether MgSO4 treatment causes significant cell death in the developing mouse brain. METHODS: C57Bl/6 mice were treated with four doses of MgSO4 (250 mg/kg) on postnatal days 3 (P3), 7 (P7) or 14 (P14). Caspase-3 immunohistochemistry, cupric silver staining, and electron microscopy techniques were used to examine Mg-treated brains for neurotoxic effects. RESULTS: Qualitative evaluation using cupric silver staining revealed widespread damage throughout the brain in P7 animals. Results of electron microscopy confirmed that the cell death process was apoptotic in nature. Quantitative evaluation of damage to the cortex, caudate-putamen, hippocampus, thalamus, and cerebellum showed that Mg treatment caused significant brain damage in animals treated on P3 and P7, but not P14. CONCLUSIONS: Administration of high doses of Mg may be detrimental to the fetal brain, particularly if exposure occurs during critical periods of neurodevelopment.


Subject(s)
Animals, Newborn , Apoptosis/drug effects , Brain/drug effects , Brain/growth & development , Magnesium Sulfate/toxicity , Aging , Animals , Brain/cytology , Caspase 3/analysis , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Copper , Hippocampus/cytology , Hippocampus/drug effects , Immunohistochemistry , Magnesium Sulfate/administration & dosage , Mice , Mice, Inbred C57BL , Microscopy, Electron , Neurons/drug effects , Neurons/enzymology , Neurons/ultrastructure , Silver , Staining and Labeling , Thalamus/cytology , Thalamus/drug effects
19.
Neurobiol Aging ; 29(2): 153-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17112636

ABSTRACT

The NMDA antagonist, memantine (Namenda), and the cholinesterase inhibitor, donepezil (Aricept), are currently being used widely, either individually or in combination, for treatment of Alzheimer's disease (AD). NMDA antagonists have both neuroprotective and neurotoxic properties; the latter is augmented by drugs, such as pilocarpine, that increase cholinergic activity. Whether donepezil, by increasing cholinergic activity, might augment memantine's neurotoxic potential has not been investigated. In the present study, we determined that a dose of memantine (20mg/kg, i.p.), considered to be in the therapeutic (neuroprotective) range for rats, causes a mild neurotoxic reaction in the adult rat brain. Co-administration of memantine (20 or 30 mg/kg) with donepezil (2.5-10mg/kg) markedly potentiated this neurotoxic reaction, causing neuronal injury at lower doses of memantine, and causing the toxic reaction to become disseminated and lethal to neurons throughout many brain regions. These findings raise questions about using this drug combination in AD, especially in the absence of evidence that the combination is beneficial, or that either drug arrests or reverses the disease process.


Subject(s)
Brain/pathology , Cholinesterase Inhibitors/adverse effects , Excitatory Amino Acid Antagonists/toxicity , Indans/adverse effects , Memantine/toxicity , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Piperidines/adverse effects , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/ultrastructure , Cell Death/drug effects , Donepezil , Dose-Response Relationship, Drug , Drug Synergism , Female , Rats , Rats, Sprague-Dawley
20.
Acad Emerg Med ; 11(8): 809-19, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15289185

ABSTRACT

OBJECTIVES: Results from recent studies on animal models of concussion suggest that multiple, rather than single, episodes of mild traumatic brain injury result in impaired cognitive performance in mice. The objective of the present study was to administer multiple impacts to the heads of mice while directly measuring the force of the impacts to determine how these parameters are related to transient loss of consciousness, cognitive deficits, and potential neuropathologic effects. METHODS: even-week-old male C57BL/6 mice were randomly assigned to experimental conditions involving three impacts (weight-drop method) to the head to induce mild traumatic brain injury or to sham control procedures. Some impacted (n = 10) and sham control (n = 10) mice were evaluated behaviorally and tested for spatial learning using the Morris water maze (MWM), whereas other impacted (n = 10) and sham control (n = 5) mice were used for histopathologic analysis. RESULTS: The mean ( +/- SD) force of impact was 19 ( +/- 3.5) N. Impacted mice took longer to regain consciousness compared with sham control mice (p < 0.0005). Behavioral test results showed that the groups did not differ on activity or sensorimotor tests or during cued trials in the MWM. Impacted mice exhibited impaired spatial learning performance during place trials in the MWM (p < 0.05). Silver staining revealed a contra-coup type of injury involving ventral brain structures in contact with or in close proximity to the skull. CONCLUSIONS: This multiple-impact model, delivered within a specifiable force range, results in transient, reversible loss of consciousness, a contra-coup brain injury, and cognitive impairment.


Subject(s)
Brain Concussion/complications , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Multiple Trauma/complications , Animals , Brain/pathology , Brain Concussion/pathology , Brain Concussion/physiopathology , Disease Models, Animal , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Multiple Trauma/pathology , Multiple Trauma/physiopathology , Recurrence , Reference Values , Reflex/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...