201
|
Repeated exposure to ketamine-xylazine during early development impairs motor learning-dependent dendritic spine plasticity in adulthood. Anesthesiology 2015; 122:821-31. [PMID: 25575163 DOI: 10.1097/aln.0000000000000579] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent studies in rodents suggest that repeated and prolonged anesthetic exposure at early stages of development leads to cognitive and behavioral impairments later in life. However, the underlying mechanism remains unknown. In this study, we tested whether exposure to general anesthesia during early development will disrupt the maturation of synaptic circuits and compromise learning-related synaptic plasticity later in life. METHODS Mice received ketamine-xylazine (20/3 mg/kg) anesthesia for one or three times, starting at either early (postnatal day 14 [P14]) or late (P21) stages of development (n = 105). Control mice received saline injections (n = 34). At P30, mice were subjected to rotarod motor training and fear conditioning. Motor learning-induced synaptic remodeling was examined in vivo by repeatedly imaging fluorescently labeled postsynaptic dendritic spines in the primary motor cortex before and after training using two-photon microscopy. RESULTS Three exposures to ketamine-xylazine anesthesia between P14 and P18 impair the animals' motor learning and learning-dependent dendritic spine plasticity (new spine formation, 8.4 ± 1.3% [mean ± SD] vs. 13.4 ± 1.8%, P = 0.002) without affecting fear memory and cell apoptosis. One exposure at P14 or three exposures between P21 and P25 has no effects on the animals' motor learning or spine plasticity. Finally, enriched motor experience ameliorates anesthesia-induced motor learning impairment and synaptic deficits. CONCLUSIONS Our study demonstrates that repeated exposures to ketamine-xylazine during early development impair motor learning and learning-dependent dendritic spine plasticity later in life. The reduction in synaptic structural plasticity may underlie anesthesia-induced behavioral impairment.
Collapse
|
202
|
Dittmar MS, Petermichl W, Lindner R, Sinner B, Graf BM, Schlachetzki F, Gruber M. In Vitro Induction of Endothelial Apoptosis of the Post-Hypoxic Blood-Brain Barrier by Isoflurane but Not by Sevoflurane and Midazolam. PLoS One 2015; 10:e0130408. [PMID: 26091107 PMCID: PMC4475016 DOI: 10.1371/journal.pone.0130408] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/20/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The effects of anesthetics on the injured brain continue to be the subject of controversial discussion. Since isoflurane has recently been shown to induce apoptosis of cerebral endothelial cells, this study compared different anesthetic compounds regarding their potential to induce cerebro-vascular apoptosis. METHODS The in vitro model of the blood-brain barrier used in this study consisted of astrocyte-conditioned human umbilical vein endothelial cells (AC-HUVEC) has been used. After 24 h of deep hypoxia and reoxygenation or control treatment, AC-HUVEC were exposed to 0, 0.5, 1.0, or 2.0 times the minimum alveolar concentration of isoflurane or sevoflurane, or 0, 75, 150, or 300 nM of midazolam for 2 h. After 24 h, AC-HUVEC were harvested, and the degree of apoptosis was assessed by means of Western blots for the Bax and Bcl-2 ratio and, for controls and the highest concentration groups, terminal deoxynucleotidyl-mediated dUTP-biotin nick end labeling (TUNEL). RESULTS Without hypoxic pretreatment, 2.0 MAC of isoflurane slightly increased TUNEL intensity compared to control and sevoflurane, but without any significant changes in the Bax and Bcl-2 ratio. After hypoxic pretreatment, exposure to isoflurane led to a multifold increase in the Bax and Bcl-2 ratio in a dose dependent manner, which was also significantly higher than the ratio observed in the 2 MAC sevoflurane group. TUNEL intensity in the post-hypoxic 2 MAC isoflurane group was increased by a factor of 11 vs. control and by 40 vs. sevoflurane. Sevoflurane and midazolam did not significantly alter these markers of apoptosis, when compared to the control group. CONCLUSIONS Isoflurane administered after hypoxia elevates markers of apoptosis in endothelial cells transdifferentiated to the cerebro-vascular endothelium. Endothelial apoptosis may be a previously underestimated mechanism of anesthetic neurotoxicity. Administration of high concentrations of isoflurane in experimental settings may have negative effects on the blood-brain barrier.
Collapse
Affiliation(s)
- Michael S. Dittmar
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
- * E-mail:
| | - Walter Petermichl
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
| | - Regina Lindner
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
| | - Barbara Sinner
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
| | - Bernhard M. Graf
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
| | - Felix Schlachetzki
- Department of Neurology, Bezirksklinikum Regensburg, University of Regensburg, Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, Regensburg University Medical Center, Regensburg, Germany
| |
Collapse
|
203
|
Slikker W, Liu F, Rainosek SW, Patterson TA, Sadovova N, Hanig JP, Paule MG, Wang C. Ketamine-Induced Toxicity in Neurons Differentiated from Neural Stem Cells. Mol Neurobiol 2015; 52:959-69. [DOI: 10.1007/s12035-015-9248-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 11/28/2022]
|
204
|
Joksovic PM, Lunardi N, Jevtovic-Todorovic V, Todorovic SM. Early Exposure to General Anesthesia with Isoflurane Downregulates Inhibitory Synaptic Neurotransmission in the Rat Thalamus. Mol Neurobiol 2015; 52:952-8. [PMID: 26048671 DOI: 10.1007/s12035-015-9247-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 11/27/2022]
Abstract
Recent evidence supports the idea that common general anesthetics (GAs) such as isoflurane (Iso) and nitrous oxide (N2O; laughing gas) are neurotoxic and may harm the developing mammalian brain, including the thalamus; however, to date very little is known about how developmental exposure to GAs may affect synaptic transmission in the thalamus which, in turn, controls the function of thalamocortical circuitry. To address this issue we used in vitro patch-clamp recordings of evoked inhibitory postsynaptic currents (eIPSCs) from intact neurons of the nucleus reticularis thalami (nRT) in brain slices from rat pups (postnatal age P10-P18) exposed at age of P7 to clinically relevant GA combinations of Iso and N2O. We found that rats exposed to a combination of 0.75 % Iso and 75 % N2O display lasting reduction in the amplitude and faster decays of eIPSCs. Exposure to sub-anesthetic concentrations of 75 % N2O alone or 0.75 % Iso alone at P7 did not affect the amplitude of eIPSCs; however, Iso alone, but not N2O, significantly accelerated decay of eIPSCs. Anesthesia with 1.5 % Iso alone decreased amplitudes, caused faster decay and decreased the paired-pulse ratio of eIPSCs. We conclude that anesthesia at P7 with Iso alone or in combination with N2O causes plasticity of eIPSCs in nRT neurons by both presynaptic and postsynaptic mechanisms. We hypothesize that changes in inhibitory synaptic transmission in the thalamus induced by GAs may contribute to altered neuronal excitability and consequently abnormal thalamocortical oscillations later in life.
Collapse
Affiliation(s)
- Pavle M Joksovic
- Department of Anesthesiology, University of Virginia School of Medicine, PO 800710, Charlottesville, VA, 22908-0710, USA
| | | | | | | |
Collapse
|
205
|
Lunardi N, Oklopcic A, Prillaman M, Erisir A, Jevtovic-Todorovic V. Early Exposure to General Anesthesia Disrupts Spatial Organization of Presynaptic Vesicles in Nerve Terminals of the Developing Rat Subiculum. Mol Neurobiol 2015; 52:942-51. [PMID: 26048670 DOI: 10.1007/s12035-015-9246-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 01/08/2023]
Abstract
Exposure to general anesthesia (GA) during critical stages of brain development induces widespread neuronal apoptosis and causes long-lasting behavioral deficits in numerous animal species. Although several studies have focused on the morphological fate of neurons dying acutely by GA-induced developmental neuroapoptosis, the effects of an early exposure to GA on the surviving synapses remain unclear. The aim of this study is to study whether exposure to GA disrupts the fine regulation of the dynamic spatial organization and trafficking of synaptic vesicles in presynaptic terminals. We exposed postnatal day 7 (PND7) rat pups to a clinically relevant anesthetic combination of midazolam, nitrous oxide, and isoflurane and performed a detailed ultrastructural analysis of the synaptic vesicle architecture at presynaptic terminals in the subiculum of rats at PND 12. In addition to a significant decrease in the density of presynaptic vesicles, we observed a reduction of docked vesicles, as well as a reduction of vesicles located within 100 nm from the active zone, in animals 5 days after an initial exposure to GA. We also found that the synaptic vesicles of animals exposed to GA are located more distally with respect to the plasma membrane than those of sham control animals and that the distance between presynaptic vesicles is increased in GA-exposed animals compared to sham controls. We report that exposure of immature rats to GA during critical stages of brain development causes significant disruption of the strategic topography of presynaptic vesicles within the nerve terminals of the subiculum.
Collapse
Affiliation(s)
- N Lunardi
- Department of Anesthesiology, University of Virginia Health System, PO Box 800710, Charlottesville, VA, 22908, USA,
| | | | | | | | | |
Collapse
|
206
|
Mikhailenko VA, Butkevich IP, Vershinina EA, Ulanova NA. Long-term changes in adaptive behavior of rats after inflammatory pain stimulation during neonatal development. J EVOL BIOCHEM PHYS+ 2015. [DOI: 10.1134/s0022093015020052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
207
|
Lin EP, Lee JR, Loepke AW. Anesthetics and the Developing Brain: The Yin and Yang. CURRENT ANESTHESIOLOGY REPORTS 2015. [DOI: 10.1007/s40140-015-0107-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
208
|
Abstract
The results of several retrospective clinical studies suggest that exposure to anesthetic agents early in life is correlated with subsequent learning and behavioral disorders. Although ongoing prospective clinical trials may help to clarify this association, they remain confounded by numerous factors. Thus, some of the most compelling data supporting the hypothesis that a relatively short anesthetic exposure can lead to a long-lasting change in brain function are derived from animal models. The mechanism by which such changes could occur remains incompletely understood. Early studies identified anesthetic-induced neuronal apoptosis as a possible mechanism of injury, and more recent work suggests that anesthetics may interfere with several critical processes in brain development. The function of the mature brain requires the presence of circuits, established during development, which perform the computations underlying learning and cognition. In this review, we examine the mechanisms by which anesthetics could disrupt brain circuit formation, including effects on neuronal survival and neurogenesis, neurite growth and guidance, formation of synapses, and function of supporting cells. There is evidence that anesthetics can disrupt aspects of all of these processes, and further research is required to elucidate which are most relevant to pediatric anesthetic neurotoxicity.
Collapse
|
209
|
Yufune S, Satoh Y, Takamatsu I, Ohta H, Kobayashi Y, Takaenoki Y, Pagès G, Pouysségur J, Endo S, Kazama T. Transient Blockade of ERK Phosphorylation in the Critical Period Causes Autistic Phenotypes as an Adult in Mice. Sci Rep 2015; 5:10252. [PMID: 25993696 PMCID: PMC4438718 DOI: 10.1038/srep10252] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/08/2015] [Indexed: 12/02/2022] Open
Abstract
The critical period is a distinct time-window during the neonatal stage when animals display elevated sensitivity to certain environmental stimuli, and particular experiences can have profound and long-lasting effects on behaviors. Increasing evidence suggests that disruption of neuronal activity during the critical period contributes to autistic phenotype, although the pathogenic mechanism is largely unknown. Herein we show that extracellular signal-regulated protein kinases (ERKs) play important roles in proper formation of neural circuits during the critical period. Transient blockade of ERKs phosphorylation at postnatal day 6 (P6) by intraperitoneal injection of blood-brain barrier-penetrating MEK inhibitor, α-[amino[(4-aminophenyl)thio]methylene]-2-(trifluoromethyl)benzeneacetonitrile (SL327) caused significant increase of apoptosis in the forebrain. Furthermore, this induced long-term deleterious effects on brain functioning later in adulthood, resulting in social deficits, impaired memory and reduced long-term potentiation (LTP). Conversely, blockade of ERK phosphorylation at P14 no longer induced apoptosis, nor behavioral deficits, nor the reduced LTP. Thus, surprisingly, these effects of ERKs are strongly age-dependent, indicating that phosphorylation of ERKs during the critical period is absolutely required for proper development of brain functioning. This study provides novel insight into the mechanistic basis for neurodevelopment disorders: various neurodevelopment disorders might be generally linked to defects in ERKs signaling during the critical period.
Collapse
Affiliation(s)
- Shinya Yufune
- Department of Anesthesiology National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan,
| | - Yasushi Satoh
- Department of Anesthesiology National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan,
| | - Isao Takamatsu
- Department of Anesthesiology National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan,
| | - Hiroyuki Ohta
- Department of Physiology, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan
| | - Yasushi Kobayashi
- Department of Anatomy and Neurobiology, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan
| | - Yumiko Takaenoki
- Department of Anesthesiology National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan,
| | - Gilles Pagès
- Institute for Research on Cancer and Aging (IRCAN), University of Nice Sophia Antipolis, Centre Antoine Lacassagne, 33 Avenue de Valombrose, Nice 06189, France
| | - Jacques Pouysségur
- 1] Institute for Research on Cancer and Aging (IRCAN), University of Nice Sophia Antipolis, Centre Antoine Lacassagne, 33 Avenue de Valombrose, Nice 06189, France [2] Centre Scientifique de Monaco (CSM) Biochemical Department, 8 Quai Antoine Ier, MC 98000, Monaco
| | - Shogo Endo
- Aging Regulation Research Team, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Tomiei Kazama
- Department of Anesthesiology National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan,
| |
Collapse
|
210
|
Induction of TNF-α signaling cascade in neonatal rat brain during propofol anesthesia. Int J Dev Neurosci 2015; 44:22-32. [PMID: 25980792 DOI: 10.1016/j.ijdevneu.2015.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/09/2015] [Accepted: 05/11/2015] [Indexed: 11/20/2022] Open
Abstract
Propofol anesthesia can trigger pro- and anti-apoptotic signaling pathways in the rat brain. In our previous work, we demonstrated that propofol causes widespread apoptotic neurodegeneration in 7-postnatal-day-old (PND7) but not in PND14 rat neurons. The mechanism responsible for these opposing outcomes is unknown, apparently linked to the specific stage of brain development. The present study aims to elucidate the anti-apoptotic process that is activated in the cortex and thalamus of PND14 Wistar rats during the first 48 h after the onset of propofol anesthesia. We showed that the expression of tumor necrosis factor-α (TNF-α) and several components of its pathway, TNFR1 and caspase-8, was significantly increased in the cortex and thalamus. Nuclear factor kappa B (NF-κB) p65 was downregulated in the cortex and upregulated in the thalamus. The expression of c-Fos was upregulated only in the cortex, showing opposed profile compared to NF-κB p65. Double immunofluorescence staining revealed the colocalization of NF-κB p65 with neuronal marker (NeuN), but with predominantly cytoplasmic localization. Finally, X-linked inhibitor of apoptosis protein (XIAP) was upregulated in both examined structures. Immunohistochemical staining with Iba-1 revealed that the treatment did not induce changes in microglial morphology. Our results (i) reveal that the simultaneous activation of pro- and anti-apoptotic signaling occurs after propofol anesthesia, and (ii) pinpoint the potential neuroprotective role of XIAP in anesthesia-induced neurotoxicity.
Collapse
|
211
|
Gonzales ELT, Yang SM, Choi CS, Mabunga DFN, Kim HJ, Cheong JH, Ryu JH, Koo BN, Shin CY. Repeated neonatal propofol administration induces sex-dependent long-term impairments on spatial and recognition memory in rats. Biomol Ther (Seoul) 2015; 23:251-60. [PMID: 25995824 PMCID: PMC4428718 DOI: 10.4062/biomolther.2014.120] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/31/2014] [Accepted: 02/07/2015] [Indexed: 11/05/2022] Open
Abstract
Propofol is an anesthetic agent that gained wide use because of its fast induction of anesthesia and rapid recovery post-anesthesia. However, previous studies have reported immediate neurodegeneration and long-term impairment in spatial learning and memory from repeated neonatal propofol administration in animals. Yet, none of those studies has explored the sex-specific long-term physical changes and behavioral alterations such as social (sociability and social preference), emotional (anxiety), and other cognitive functions (spatial working, recognition, and avoidance memory) after neonatal propofol treatment. Seven-day-old Wistar-Kyoto (WKY) rats underwent repeated daily intraperitoneal injections of propofol or normal saline for 7 days. Starting fourth week of age and onwards, rats were subjected to behavior tests including open-field, elevated-plus-maze, Y-maze, 3-chamber social interaction, novel-object-recognition, passive-avoidance, and rotarod. Rats were sacrificed at 9 weeks and hippocampal protein expressions were analyzed by Western blot. Results revealed long-term body weight gain alterations in the growing rats and sex-specific impairments in spatial (female) and recognition (male) learning and memory paradigms. A markedly decreased expression of hippocampal NMDA receptor GluN1 subunit in female- and increased expression of AMPA GluR1 subunit protein expression in male rats were also found. Other aspects of behaviors such as locomotor activity and coordination, anxiety, sociability, social preference and avoidance learning and memory were not generally affected. These results suggest that neonatal repeated propofol administration disrupts normal growth and some aspects of neurodevelopment in rats in a sex-specific manner.
Collapse
Affiliation(s)
- Edson Luck T Gonzales
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 143-701
| | - Sung Min Yang
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 143-701
| | - Chang Soon Choi
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 143-701
| | - Darine Froy N Mabunga
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 143-701
| | - Hee Jin Kim
- Department of Pharmacy, Sahmyook University, Seoul 139-742
| | | | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul 130-701
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 120-752, Repulic of Korea
| | - Chan Young Shin
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 143-701 ; Department of Pharmacology, School of Medicine, Konkuk University, Seoul 143-701
| |
Collapse
|
212
|
Lee JH, Zhang J, Wei L, Yu SP. Neurodevelopmental implications of the general anesthesia in neonate and infants. Exp Neurol 2015; 272:50-60. [PMID: 25862287 DOI: 10.1016/j.expneurol.2015.03.028] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Each year, about six million children, including 1.5 million infants, in the United States undergo surgery with general anesthesia, often requiring repeated exposures. However, a crucial question remains of whether neonatal anesthetics are safe for the developing central nervous system (CNS). General anesthesia encompasses the administration of agents that induce analgesic, sedative, and muscle relaxant effects. Although the mechanisms of action of general anesthetics are still not completely understood, recent data have suggested that anesthetics primarily modulate two major neurotransmitter receptor groups, either by inhibiting N-methyl-D-aspartate (NMDA) receptors, or conversely by activating γ-aminobutyric acid (GABA) receptors. Both of these mechanisms result in the same effect of inhibiting excitatory activity of neurons. In developing brains, which are more sensitive to disruptions in activity-dependent plasticity, this transient inhibition may have longterm neurodevelopmental consequences. Accumulating reports from preclinical studies show that anesthetics in neonates cause cellular toxicity including apoptosis and neurodegeneration in the developing brain. Importantly, animal and clinical studies indicate that exposure to general anesthetics may affect CNS development, resulting in long-lasting cognitive and behavioral deficiencies, such as learning and memory deficits, as well as abnormalities in social memory and social activity. While the casual relationship between cellular toxicity and neurological impairments is still not clear, recent reports in animal experiments showed that anesthetics in neonates can affect neurogenesis, which could be a possible mechanism underlying the chronic effect of anesthetics. Understanding the cellular and molecular mechanisms of anesthetic effects will help to define the scope of the problem in humans and may lead to preventive and therapeutic strategies. Therefore, in this review, we summarize the current evidence on neonatal anesthetic effects in the developmental CNS and discuss how factors influencing these processes can be translated into new therapeutic strategies.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James Zhang
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Visual and Neurocognitive Rehabilitation, VA Medical Center, Atlanta, GA 30033, USA.
| |
Collapse
|
213
|
Hyperexcitability of rat thalamocortical networks after exposure to general anesthesia during brain development. J Neurosci 2015; 35:1481-92. [PMID: 25632125 DOI: 10.1523/jneurosci.4883-13.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prevailing literature supports the idea that common general anesthetics (GAs) cause long-term cognitive changes and neurodegeneration in the developing mammalian brain, especially in the thalamus. However, the possible role of GAs in modifying ion channels that control neuronal excitability has not been taken into consideration. Here we show that rats exposed to GAs at postnatal day 7 display a lasting reduction in inhibitory synaptic transmission, an increase in excitatory synaptic transmission, and concomitant increase in the amplitude of T-type calcium currents (T-currents) in neurons of the nucleus reticularis thalami (nRT). Collectively, this plasticity of ionic currents leads to increased action potential firing in vitro and increased strength of pharmacologically induced spike and wave discharges in vivo. Selective blockade of T-currents reversed neuronal hyperexcitability in vitro and in vivo. We conclude that drugs that regulate thalamic excitability may improve the safety of GAs used during early brain development.
Collapse
|
214
|
Han T, Hu Z, Tang Y, Shrestha A, Ouyang W, Liao Q. Inhibiting Rho kinase 2 reduces memory dysfunction in adult rats exposed to sevoflurane at postnatal days 7-9. Biomed Rep 2015; 3:361-364. [PMID: 26137236 DOI: 10.3892/br.2015.429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/06/2015] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to investigate the roles of Rho protein A (RhoA) and Rho kinases 2 (ROCK2) in the memory dysfunction of adult rats exposed to sevoflurane at postnatal days 7-9 (P7-9). One-week-old Sprague-Dawley rats were divided into four groups known as C, S1, S3 and F. Rats in the S1 (2 h at P7) and S3 groups (2 h/day at P7-9) were exposed to sevoflurane. The rats in the F group were treated with the ROCK2 inhibitor and subsequent sevoflurane exposure (2 h/day at P7-9). The rats in the C group received no sevoflurane. The protein levels of RhoA, ROCK2 and cleaved caspase-3 (Cl-Csp3) in the adult hippocampus were assessed by western blot analysis. Learning and memory of rats at postnatal 45-50 days (P45-50) were detected by the Morris water maze (MWM) test. During the training of MWM, the latency and distance of rats in the S3 group were significantly longer than that of the C group (P<0.05, respectively). In the probe test, the percentages of time and distance in the target quadrant for the S3 group were evidently less than that of the C group (P<0.05). There was no significant difference in the behaviors between the C and S1 groups (P>0.05, respectively). Corresponding to the behavioral changes, the levels of RhoA, ROCK2 and Cl-Csp3 in the hippocampus of the S3 group significantly increased, compared to that of the C and S1 groups (P<0.05). Additionally, the ROCK2 inhibitor clearly decreased ROCK2 and Cl-Csp3 expression and shortened the latency during the training (P<0.05, P46-49 respectively) and probe test (P<0.05) in the F group, compared to that of the S3 group. Compared to the C group, the expression of RhoA, ROCK2 and Cl-Csp3 in the hippocampus of the S1 group had no significant difference (P>0.05). Multiple inhalation of sevoflurane can induce neurotoxicity and memory dysfunction. RhoA and ROCK2 played important roles in the impairment of learning and memory of adults rats exposed to sevoflurane at the postnatal early stage.
Collapse
Affiliation(s)
- Tao Han
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhonghua Hu
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yongzhong Tang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Alisha Shrestha
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Wen Ouyang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Qin Liao
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
215
|
Altered metabolomic profiles may be associated with sevoflurane-induced neurotoxicity in neonatal rats. Neurochem Res 2015; 40:788-99. [PMID: 25663300 DOI: 10.1007/s11064-015-1529-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/23/2015] [Accepted: 01/28/2015] [Indexed: 12/25/2022]
Abstract
Experimental studies demonstrate that inhaled anesthetics can cause neurodegeneration and neurobehavioral dysfunctions. Evidence suggests changes in cerebral metabolism following inhaled anesthetics treatment can perturb cerebral homeostasis, which may be associated with their induced neurotoxicity. Seven-day-old rat pups were divided into two groups: control group (Group C) and sevoflurane group (Group S, 3 % sevoflurane exposure for 6 h). Gas chromatography-mass spectrometry (GC-MS) was used for analyzed differential metabolites of cerebral cortex in both groups, Also western blot, flow cytometry, enzymatic methods and electron microscopy were performed in various biochemical and anatomical assays. Sevoflurane exposure significantly elevated caspase-3 activation and ROS levels, decreased mitochondrial cardiolipin contents, and changed cellular ultrastructure in the cerebral cortex. Correspondingly, these results corroborated the GC-MS findings which showed altered metabolic pathways of glucose, amino acids, and lipids, as well as intracellular antioxidants and osmolyte systems in neonatal brain following prolonged exposure to high sevoflurane concentration. Our data indicate that sevoflurane anesthesia causes significant oxidative stress, neuroapoptosis, and cellular ultrastructure damage which is associated with altered brain metabotype in the neonatal rat. Our study also confirmed that GC-MS is a strategic and complementary platform for the metabolomic characterization of sevoflurane-induced neurotoxicity in the developing brain.
Collapse
|
216
|
Abstract
BACKGROUND Transplants cause ischemia-reperfusion (IR) injury that can affect distant organs. Liver is particularly sensitive to IR injury. The present randomized experimental study was designed to investigate a possible protective effect of sevoflurane against liver inflammatory response to lung IR in a lung upper lobe left autotransplant model. METHODS Two groups (sevoflurane and control) of eight swines each were submitted to upper lobe left lung autotransplant. Hypnotic maintenance was performed with sevoflurane 3% or propofol 8 to 10 mg/kg per hr until pneumonectomy was done; then propofol was used for all animals. Blood and liver samples were taken in four different moments: prepneumonectomy, prereperfusion, 10 min postreperfusion and 30 min postreperfusion to measure levels of interleukin (IL)-1β, IL-10, tumor necrosis factor (TNF)-α, monocyte chemotactic protein (MCP)-1, nuclear factor (NF)-κB, C-reactive protein, ferritin and caspase 3. Non-parametric test was used to find statistical meaning. RESULTS Lung IR markedly increased the expression of TNF-α, IL-1β, MCP-1, NF-κB and caspase activity in control livers compared with basal levels, whereas liver IL-10 expression decreased 10 and 30 min post-reperfusion. Sevoflurane significantly decreased TNF-α, IL-1β, MCP-1, NF-κB liver expression and caspase 3 activity. Sevoflurane also reverted the lung IR-induced decrease in IL-10 expression. CONCLUSIONS The present results indicate that lung IR caused hepatic injury. Sevoflurane attenuated liver injury in a model of upper lobe left lung autotransplant in pigs.
Collapse
|
217
|
YU X, LIU Y, BO S, QINGHUA L. Effects of sevoflurane on learning, memory, and expression of pERK1/2 in hippocampus in neonatal rats. Acta Anaesthesiol Scand 2015; 59:78-84. [PMID: 25349022 DOI: 10.1111/aas.12433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/22/2014] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sevoflurane may be associated with neural toxicity in the developing brain, but the mechanism is still unclear. Phosphorylated extracellular signal-regulated kinases 1/2 (pERK1/2) are important for developing neurons. The aim of our study was to investigate the effects of sevoflurane on spatial learning and memory and on expression of pERK1/2 in hippocampus of neonatal rats. METHODS Sixty-three neonatal rats were randomly divided into three groups: control group, sevoflurane (sevo) group, and sham group. Rats in the control group were placed in a plastic chamber flushed continuously for 4 h with air alone, rats in the sevo group were exposed in 5% sevoflurane and air for 4 h, and rats in the sham group were exposed in 5% carbon dioxide and air for 4 h, with identical flow rates for all groups. All three groups were subjected to Morris water maze test 1 day after sevoflurane exposure. Moreover, expression of pERK1/2 was determined by immunochemistry and Western blot at 1, 3, and 6 weeks after exposure. RESULTS Compared with the control group, the escape latency was longer in sevo group and the expression of pERK1/2 was significantly inhibited in the sevo group (P < 0.01); no differences between control and sham groups were observed. CONCLUSION Our study demonstrated that neonatal rats exposed to sevoflurane had impaired spatial learning and memory, and this may be attributed to decreased pERK1/2 in the hippocampus.
Collapse
Affiliation(s)
- X. YU
- Department of Anesthesiology; The First College of Clinical Medical Science; China Three Gorges University; Yichang China
- Department of Anesthesiology; Yichang Central People's Hospital; Yichang China
| | - Y. LIU
- Department of Anesthesiology; The First College of Clinical Medical Science; China Three Gorges University; Yichang China
- Department of Anesthesiology; Yichang Central People's Hospital; Yichang China
| | - S. BO
- Department of Anesthesiology; The First College of Clinical Medical Science; China Three Gorges University; Yichang China
- Department of Anesthesiology; Yichang Central People's Hospital; Yichang China
| | - L. QINGHUA
- Department of Anesthesiology; The First College of Clinical Medical Science; China Three Gorges University; Yichang China
- Department of Anesthesiology; Yichang Central People's Hospital; Yichang China
| |
Collapse
|
218
|
Long-term Effects of Single or Multiple Neonatal Sevoflurane Exposures on Rat Hippocampal Ultrastructure. Anesthesiology 2015; 122:87-95. [DOI: 10.1097/aln.0000000000000477] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Background:
Neonatal exposure to general anesthetics may pose significant neurocognitive risk. Human epidemiological studies demonstrate higher rates of learning disability among children with multiple, but not single, exposures to anesthesia. The authors employ a rat model to provide a histological correlate for these population-based observations. The authors examined long-term differences in hippocampal synaptic density, mitochondrial density, and dendritic spine morphology.
Methods:
Twenty male rat pups (n = 5/condition) were exposed to 2.5% sevoflurane under one of four conditions: single 2-h exposure on postnatal day 7 (P7); single 6-h exposure on P7; repeated 2-h exposures on P7, P10, and P13 for a cumulative 6 h of general anesthetics; or control exposure to 30% oxygen on P7, P10, and P13.
Results:
Repeated exposure to general anesthetics resulted in greater synaptic loss relative to a single 2-h exposure (P < 0.001). The magnitude of synaptic loss induced by three 2-h exposures (1.977 ± 0.040 μm3 [mean ± SEM]) was more profound than that of a single 6-h exposure (2.280 ± 0.045 μm3, P = 0.022). Repeated exposures did not alter the distribution of postsynaptic density length, indicating a uniform pattern of loss across spine types. In contrast, mitochondrial toxicity was best predicted by the cumulative duration of exposure. Relative to control (0.595 ± 0.017), both repeated 2-h exposures (0.479 ± 0.015) and a single 6-h exposure (0.488 ± 0.013) were associated with equivalent reductions in the fraction of presynaptic terminals containing mitochondria (P < 0.001).
Conclusion:
This suggests a “threshold effect” for general anesthetic–induced neurotoxicity, whereby even brief exposures induce long-lasting alterations in neuronal circuitry and sensitize surviving synapses to subsequent loss.
Collapse
|
219
|
Shan H, Yan R, Diao J, Lin L, Wang S, Zhang M, Zhang R, Wei J. Involvement of caspases and their upstream regulators in myocardial apoptosis in a rat model of selenium deficiency-induced dilated cardiomyopathy. J Trace Elem Med Biol 2015; 31:85-91. [PMID: 26004897 DOI: 10.1016/j.jtemb.2015.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/24/2015] [Accepted: 03/24/2015] [Indexed: 12/19/2022]
Abstract
Keshan disease is an endemic dilated cardiomyopathy (DCM) which is closely related with selenium-deficient diet in China. In the previous study, we reported that the low selenium status plays a pivotal role in the myocardial apoptosis in the DCM rats, however, the underlying mechanism remains unclear. The present study aimed to determine whether the intrinsic, extrinsic pathways and the upstream regulators were involved in the myocardial apoptosis of selenium deficiency-induced DCM rats. Therefore, the rat model of endemic DCM was induced by a selenium-deficient diet for 12 weeks. Accompanied with significant dilation and impaired systolic function of left ventricle, an enhanced myocardial apoptosis was detected by TUNEL assay. Western blot analysis showed remarkably increased protein levels of cleaved caspase-3, caspase-8, caspase-9, and cytosolic cytochrome c released from the mitochondria. In addition, the immunoreactivities of p53 and Bax were significantly up-regulated, while the anti-apoptotic Bcl-2 family members Bcl-2 and Bcl-X(L) were down-regulated. Furthermore, appropriate selenium supplement for another 4 weeks could partially reverse all the above changes. In conclusion, the intrinsic, extrinsic pathways and the upstream regulators such as p53, Bax, Bcl-2, and Bcl-X(L )were all involved in selenium deficiency-induced myocardial apoptosis.
Collapse
Affiliation(s)
- Hu Shan
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China; Department of Endemic Disease, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China
| | - Rui Yan
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China; Department of Endemic Disease, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China
| | - Jiayu Diao
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China
| | - Lin Lin
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China
| | - Suqin Wang
- Department of Cardiology, Henan Provincial People's Hospital, 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Ming Zhang
- Key Laboratory of Trace Elements and Endemic Disease of Ministry of Health, Xi'an Jiaotong University School of Medicine, 76 Yantaxi Road, Xi'an, Shaanxi 710061, China
| | - Rong Zhang
- Key Laboratory of Trace Elements and Endemic Disease of Ministry of Health, Xi'an Jiaotong University School of Medicine, 76 Yantaxi Road, Xi'an, Shaanxi 710061, China
| | - Jin Wei
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China; Department of Endemic Disease, The Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, 157 Xiwu Road, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
220
|
Stratmann G, Sall JW, May LDV, Loepke AW, Lee MT. Beyond anesthetic properties: the effects of isoflurane on brain cell death, neurogenesis, and long-term neurocognitive function. Anesth Analg 2014; 110:431-7. [PMID: 19917621 DOI: 10.1213/ane.0b013e3181af8015] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Greg Stratmann
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA.
| | | | | | | | | |
Collapse
|
221
|
Location- and Subunit-Specific NMDA Receptors Determine the Developmental Sevoflurane Neurotoxicity Through ERK1/2 Signaling. Mol Neurobiol 2014; 53:216-230. [DOI: 10.1007/s12035-014-9005-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|
222
|
Peng J, Drobish JK, Liang G, Wu Z, Liu C, Joseph DJ, Abdou H, Eckenhoff MF, Wei H. Anesthetic preconditioning inhibits isoflurane-mediated apoptosis in the developing rat brain. Anesth Analg 2014; 119:939-946. [PMID: 25099925 DOI: 10.1213/ane.0000000000000380] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND We hypothesized that preconditioning (PC) with a short exposure to isoflurane (ISO) would reduce neurodegeneration induced by prolonged exposure to ISO in neonatal rats, as previously shown in neuronal cell culture. METHODS We randomly divided 7-day-old Sprague-Dawley rats into 3 groups: control, 1.5% ISO, and PC + 1.5% ISO. The control group was exposed to carrier gas (30% oxygen balanced in nitrogen) for 30 minutes and then to carrier gas again for 6 hours the following day. The 1.5% ISO group was exposed to carrier gas for 30 minutes and then to 1.5% ISO for 6 hours the following day. The PC + 1.5% ISO group was preconditioned with a 30-minute 1.5% ISO exposure and then exposed to 1.5% ISO for 6 hours the following day. Blood and brain samples were collected 2 hours after the exposures for determination of neurodegenerative biomarkers, including caspase-3, S100β, caspase-12, and an autophagy biomarker Beclin-1. RESULTS Prolonged exposure to ISO significantly increased cleaved caspase-3 expression in the cerebral cortex of 7-day-old rats compared with the group preconditioned with ISO and the controls using Western blot assays. However, significant differences were not detected for other markers of neuronal injury. CONCLUSIONS The ISO-mediated increase in cleaved caspase-3 in the postnatal day 7 rat brain is ameliorated by PC with a brief anesthetic exposure, and differences were not detected in other markers of neuronal injury.
Collapse
Affiliation(s)
- Jun Peng
- From the Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Han XD, Li M, Zhang XG, Xue ZG, Cang J. Single sevoflurane exposure increases methyl-CpG island binding protein 2 phosphorylation in the hippocampus of developing mice. Mol Med Rep 2014; 11:226-30. [PMID: 25338822 DOI: 10.3892/mmr.2014.2751] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 06/09/2014] [Indexed: 11/06/2022] Open
Abstract
Sevoflurane is an inhaled anesthetic that is widely used in clinical practice, particularly for pediatric anesthesia. Previous studies have suggested that sevoflurane may induce neurotoxicity in the brains of neonatal mice. In the present study, the possible mechanism of neurodegeneration induced by sevoflurane in the developing brain, and the possibility that memantine treatment is able to reverse this phenomenon, were investigated. On postnatal day 7 (P7) C57BL/6 mice were continuously exposed to 1.5% sevoflurane for 2 h following pre-injection of saline or memantine. Methyl-CpG island binding protein 2 (MeCP2), cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) expression in the hippocampus was measured by western blotting. Exposure to 1.5% sevoflurane resulted in increased MeCP2 phosphorylation in the hippocampus, which was reversed by memantine injection. However, neither CREB phosphorylation nor BDNF expression were significantly altered by sevoflurane treatment. The current study indicated that sevoflurane causes neurotoxicity in the developing brain, and that this may be attributed to increased MeCP2 phosphorylation in the hippocampus. It was also demonstrated that this neurotoxicity can be prevented by the N-methyl-D-aspartate glutamate receptor inhibitor memantine.
Collapse
Affiliation(s)
- Xiao-Dan Han
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Min Li
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiao-Guang Zhang
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Zhang-Gang Xue
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jing Cang
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
224
|
Wang W, Wang Y, Wu H, Lei L, Xu S, Shen X, Guo X, Shen R, Xia X, Liu Y, Wang F. Postoperative cognitive dysfunction: current developments in mechanism and prevention. Med Sci Monit 2014; 20:1908-12. [PMID: 25306127 PMCID: PMC4206478 DOI: 10.12659/msm.892485] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a subtle disorder of thought processes, which may influence isolated domains of cognition and has a significant impact on patient health. The reported incidence of POCD varies enormously due to lack of formal criteria for the assessment and diagnosis of POCD. The significant risk factors of developing POCD mainly include larger and more invasive operations, duration of anesthesia, advanced age, history of alcohol abuse, use of anticholinergic medications, and other factors. The release of cytokines due to the systemic stress response caused by anesthesia and surgical procedures might induce the changes of brain function and be involved in the development of postoperative cognitive dysfunction. The strategies for management of POCD should be a multimodal approach involving close cooperation between the anesthesiologist, surgeon, geriatricians, and family members to promote early rehabilitation and avoid loss of independence in these patients.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yan Wang
- Department of Anesthesiology, Affiliated Chaohu Hospital, Anhui Medical University, Chaohu, Anhui, China (mainland)
| | - Haibo Wu
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Liming Lei
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Shiqin Xu
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xiaofeng Shen
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xirong Guo
- Pediatric Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Rong Shen
- Pediatric Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xiaoqiong Xia
- Department of Anesthesiology, Affiliated Chaohu Hospital, Anhui Medical University, Chaohu, Anhui, China (mainland)
| | - Yusheng Liu
- Department of Anesthesiology, Nanjing Medical University, Nanjing, China (mainland)
| | - Fuzhou Wang
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
225
|
Cheng Y, Levy RJ. Subclinical carbon monoxide limits apoptosis in the developing brain after isoflurane exposure. Anesth Analg 2014; 118:1284-92. [PMID: 24413549 DOI: 10.1213/ane.0000000000000030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Volatile anesthetics cause widespread apoptosis in the developing brain. Carbon monoxide (CO) has antiapoptotic properties, and exhaled endogenous CO is commonly rebreathed during low-flow anesthesia in infants and children, resulting in subclinical CO exposure. Thus, we aimed to determine whether CO could limit isoflurane-induced apoptosis in the developing brain. METHODS Seven-day-old male CD-1 mouse pups underwent 1-hour exposure to 0 (air), 5, or 100 ppm CO in air with or without isoflurane (2%). We assessed carboxyhemoglobin levels, cytochrome c peroxidase activity, and cytochrome c release from forebrain mitochondria after exposure and quantified the number of activated caspase-3 positive cells and TUNEL positive nuclei in neocortex, hippocampus, and hypothalamus/thalamus. RESULTS Carboxyhemoglobin levels approximated those expected in humans after a similar time-weighted CO exposure. Isoflurane significantly increased cytochrome c peroxidase activity, cytochrome c release, the number of activated caspase-3 cells, and TUNEL positive nuclei in the forebrain of air-exposed mice. CO, however, abrogated isoflurane-induced cytochrome c peroxidase activation and cytochrome c release from forebrain mitochondria and decreased the number of activated caspase-3 positive cells and TUNEL positive nuclei after simultaneous exposure with isoflurane. CONCLUSIONS Taken together, the data indicate that CO can limit apoptosis after isoflurane exposure via inhibition of cytochrome c peroxidase depending on concentration. Although it is unknown whether CO directly inhibited isoflurane-induced apoptosis, it is possible that low-flow anesthesia designed to target rebreathing of specific concentrations of CO may be a desired strategy to develop in the future in an effort to prevent anesthesia-induced neurotoxicity in infants and children.
Collapse
Affiliation(s)
- Ying Cheng
- From the Division of Anesthesiology and Pain Medicine, Children's National Medical Center, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | | |
Collapse
|
226
|
Giles EK, Lawrence AJ, Duncan JR. Exploring the Modulation of Hypoxia-Inducible Factor (HIF)-1α by Volatile Anesthetics as a Possible Mechanism Underlying Volatile Anesthetic-Induced CNS Injury. Neurochem Res 2014; 39:1640-7. [DOI: 10.1007/s11064-014-1379-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 06/26/2014] [Accepted: 06/30/2014] [Indexed: 12/11/2022]
|
227
|
Neira A, Aguirre LF, Gómez RM, Barbosa FR, Pérez JA, Muñoz AM. Effects of multiple exposures to sevoflurane at sub-MAC doses on neuroapoptosis and cognitive function during the neonatal period. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2014. [DOI: 10.1016/j.rcae.2014.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
228
|
Pellegrini L, Bennis Y, Velly L, Grandvuillemin I, Pisano P, Bruder N, Guillet B. Erythropoietin protects newborn rat against sevoflurane-induced neurotoxicity. Paediatr Anaesth 2014; 24:749-59. [PMID: 24725211 DOI: 10.1111/pan.12372] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2014] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Recent data on newborn animals exposed to anesthetics have raised safety concerns regarding anesthesia practices in young children. Indeed, studies on rodents have demonstrated a widespread increase in brain apoptosis shortly after exposure to sevoflurane, followed by long-term neurologic impairment. In this context, we aimed to evaluate the protective effect of rh-EPO, a potent neuroprotective agent, in rat pups exposed to sevoflurane. MATERIAL AND METHODS At postnatal day 7, 75 rat pups were allocated into three groups: SEVO + EPO (n = 27) exposed to sevoflurane 2 vol% (0.5 MAC) for 6 h in an air/O2 mixture (60/40) + 5000 UI.kg(-1) rh-EPO IP; SEVO (n = 27) exposed to sevoflurane + vehicle IP; and CONTROL (n = 21) exposed to the mixture without sevoflurane + vehicle IP. Three days after anesthesia (D10), apoptosis was quantified on brain extract with TUNEL method and caspase 3. NGF and BDNF expression was determined by Western blotting. Rats reaching adulthood were evaluated in terms of exploration capacities (object exploration duration) together with spatial and object learning (water maze and novel object test). RESULTS Sevoflurane exposure impaired normal behavior in adult rats by reducing the exploratory capacities during the novel object test and impaired both spatial and object learning capacities in adult rats (water maze, ratio time to find platform 3rd trial/1st trial: 1.1 ± 0.2 vs 0.4 ± 0.1; n = 9, SEVO vs CONTROL; P = 0.01). Rh-EPO reduced sevoflurane-induced behavior and learning abnormalities in adult rats (water maze, ratio time to find platform 3rd trial/1st trial: 0.3 ± 0.1 vs 1.1 ± 0.2; n = 9, SEVO + EPO vs SEVO; P = 0.01). Three days after anesthesia, rh-EPO prevented sevoflurane-induced brain apoptosis (5 ± 3 vs 35 ± 6 apoptotic cells·mm(-2) ; n = 6, SEVO + EPO vs SEVO; P = 0.01) and elevation of caspase three level and significantly increased the brain expression of BDNF and NGF (n = 6, SEVO + EPO vs SEVO; P = 0.01). CONCLUSION Six hours of sevoflurane anesthesia in newborn rats induces significant long-term cognitive impairment. A single administration of rh-EPO immediately after postnatal exposure to sevoflurane reduces both early activation of apoptotic phenomenon and late onset of neurologic disorders.
Collapse
Affiliation(s)
- Lionel Pellegrini
- Department of Anesthesia, APHM, CHU Timone, Marseille, France; INSERM UMR_S 1076, Aix-Marseille University, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
229
|
Neira A, Aguirre LF, Gómez RM, Ríos Barbosa F, Pérez JA, Muñoz AM. Efectos de múltiples exposiciones a sevoflurano a dosis sub-CAM en la neuroapoptosis y la función cognitiva en el periodo neonatal. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2014. [DOI: 10.1016/j.rca.2014.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
230
|
Jevtovic-Todorovic V. Good gas, bad gas: isoflurane, carbon monoxide, and which is which? Anesth Analg 2014; 118:1160-2. [PMID: 24842171 DOI: 10.1213/ane.0000000000000235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Vesna Jevtovic-Todorovic
- From the Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
231
|
Sinner B, Becke K, Engelhard K. General anaesthetics and the developing brain: an overview. Anaesthesia 2014; 69:1009-22. [DOI: 10.1111/anae.12637] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2014] [Indexed: 12/17/2022]
Affiliation(s)
- B. Sinner
- Department of Anaesthesiology; University of Regensburg; Regensburg Germany
| | - K. Becke
- Department of Anesthesiology and Intensive Care; Cnopf Childrens’ Hospital/Hospital Hallerwiese; Nuremberg Germany
| | - K. Engelhard
- Department of Anaesthesiology; University Medical Center of the Johannes Gutenberg University; Mainz Germany
| |
Collapse
|
232
|
Milanović D, Pešić V, Popić J, Tanić N, Kanazir S, Jevtović-Todorović V, Ruždijić S. Propofol anesthesia induces proapoptotic tumor necrosis factor-α and pro-nerve growth factor signaling and prosurvival Akt and XIAP expression in neonatal rat brain. J Neurosci Res 2014; 92:1362-73. [PMID: 24827783 DOI: 10.1002/jnr.23409] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/27/2014] [Accepted: 04/08/2014] [Indexed: 11/12/2022]
Abstract
Previously we observed that prolonged exposure to propofol anesthesia causes caspase-3- and calpain-mediated neuronal death in the developing brain. The present study examines the effects of propofol anesthesia on the expression of tumor necrosis factor-α (TNFα), pro-nerve growth factor (NGF), and their receptors in the cortex and the thalamus. We also investigated how propofol influences the expression of Akt and X-linked inhibitor of apoptosis (XIAP) expression, proteins that promote prosurvival pathways. Seven-day-old rats (P7) were exposed to propofol anesthesia lasting 2, 4, or 6 hr and killed 0, 4, 16, or 24 hr after anesthesia termination. The relative levels of mRNA and protein expression were estimated by RT-PCR and Western blot analysis, respectively. The treatments caused marked activation of TNFα and its receptor TNFR-1 and pro-NGF and p75(NTR) receptor expression. In parallel with the induction of these prodeath signals, we established that propofol anesthesia promotes increased expression of the prosurvival molecules pAkt and XIAP during the 24-hr postanesthesia period. These results show that different brain structures respond to propofol anesthesia with a time- and duration of exposure-dependent increase in proapoptotic signaling and with concomitant increases in activities of prosurvival proteins. We hypothesized that the fine balance between these opposing processes sustains homeostasis in the immature rat brain and prevents unnecessary damage after exposure to an injurious stimulus. The existence of this highly regulated process provides a time frame for potential therapeutic intervention directed toward suppressing the deleterious component of propofol anesthesia.
Collapse
Affiliation(s)
- Desanka Milanović
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | | | | | | | | | | | | |
Collapse
|
233
|
Dingley J, Tooley J, Liu X, Scull-Brown E, Elstad M, Chakkarapani E, Sabir H, Thoresen M. Xenon ventilation during therapeutic hypothermia in neonatal encephalopathy: a feasibility study. Pediatrics 2014; 133:809-18. [PMID: 24777219 DOI: 10.1542/peds.2013-0787] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Therapeutic hypothermia has become standard of care in newborns with moderate and severe neonatal encephalopathy; however, additional interventions are needed. In experimental models, breathing xenon gas during cooling offers long-term additive neuroprotection. This is the first xenon feasibility study in cooled infants. Xenon is expensive, requiring a closed-circuit delivery system. METHODS Cooled newborns with neonatal encephalopathy were eligible for this single-arm, dose-escalation study if clinically stable, under 18 hours of age and requiring less than 35% oxygen. Xenon duration increased stepwise from 3 to 18 hours in 14 subjects; 1 received 25% xenon and 13 received 50%. Respiratory, cardiovascular, neurologic (ie, amplitude-integrated EEG, seizures), and inflammatory (C-reactive protein) effects were examined. The effects of starting or stopping xenon rapidly or slowly were studied. Three matched control subjects per xenon treated subject were selected from our cooling database. Follow-up was at 18 months using mental developmental and physical developmental indexes of the Bayley Scales of Infant Development II. RESULTS No adverse respiratory or cardiovascular effects, including post-extubation stridor, were seen. Xenon increased sedation and suppressed seizures and background electroencephalographic activity. Seizures sometimes occurred during rapid weaning of xenon but not during slow weaning. C-reactive protein levels were similar between groups. Hourly xenon consumption was 0.52 L. Three died, and 7 of 11 survivors had mental and physical developmental index scores ≥70 at follow-up. CONCLUSIONS Breathing 50% xenon for up to 18 hours with 72 hours of cooling was feasible, with no adverse effects seen with 18 months' follow-up.
Collapse
Affiliation(s)
- John Dingley
- College of Medicine, Swansea University, Swansea, United Kingdom
| | - James Tooley
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and
| | - Xun Liu
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and
| | - Emma Scull-Brown
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and
| | - Maja Elstad
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ela Chakkarapani
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and
| | - Hemmen Sabir
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and
| | - Marianne Thoresen
- Neonatal Neuroscience, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; and Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
234
|
Li Y, Zeng M, Chen W, Liu C, Wang F, Han X, Zuo Z, Peng S. Dexmedetomidine reduces isoflurane-induced neuroapoptosis partly by preserving PI3K/Akt pathway in the hippocampus of neonatal rats. PLoS One 2014; 9:e93639. [PMID: 24743508 PMCID: PMC3990549 DOI: 10.1371/journal.pone.0093639] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/04/2014] [Indexed: 12/03/2022] Open
Abstract
Prolonged exposure to volatile anesthetics, such as isoflurane and sevoflurane, causes neurodegeneration in the developing animal brains. Recent studies showed that dexmedetomidine, a selective α2-adrenergic agonist, reduced isoflurane-induced cognitive impairment and neuroapoptosis. However, the mechanisms for the effect are not completely clear. Thus, we investigated whether exposure to isoflurane or sevoflurane at an equivalent dose for anesthesia during brain development causes different degrees of neuroapoptosis and whether this neuroapoptosis is reduced by dexmedetomidine via effects on PI3K/Akt pathway that can regulate cell survival. Seven-day-old (P7) neonatal Sprague-Dawley rats were randomly exposed to 0.75% isoflurane, 1.2% sevoflurane or air for 6 h. Activated caspase-3 was detected by immunohistochemistry and Western blotting. Phospho-Akt, phospho-Bad, Akt, Bad and Bcl-xL proteins were detected by Western blotting in the hippocampus at the end of exposure. Also, P7 rats were pretreated with various concentrations of dexmedetomidine alone or together with PI3K inhibitor LY294002, and then exposed to 0.75% isoflurane. Terminal deoxyribonucleotide transferase-mediated dUTP nick end labeling (TUNEL) and activated caspase-3 were used to detect neuronal apoptosis in their hippocampus. Isoflurane, not sevoflurane at the equivalent dose, induced significant neuroapoptosis, decreased the levels of phospho-Akt and phospho-Bad proteins, increased the expression of Bad protein and reduced the ratio of Bcl-xL/Bad in the hippocampus. Dexmedetomidine pretreatment dose-dependently inhibited isoflurane-induced neuroapoptosis and restored protein expression of phospho-Akt and Bad as well as the Bcl-xL/Bad ratio induced by isoflurane. Pretreatment with single dose of 75 µg/kg dexmedetomidine provided a protective effect similar to that with three doses of 25 µg/kg dexmedetomidine. Moreover, LY294002, partly inhibited neuroprotection of dexmedetomidine. Our results suggest that dexmedetomidine pretreatment provides neuroprotection against isoflurane-induced neuroapoptosis in the hippocampus of neonatal rats by preserving PI3K/Akt pathway activity.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (YL); (SP)
| | - Minting Zeng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiqiang Chen
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Anesthesiology, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Chuiliang Liu
- Department of Anesthesiology, ChanCheng Center Hospital, Foshan, Guangdong, China
| | - Fei Wang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhiyi Zuo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Shuling Peng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (YL); (SP)
| |
Collapse
|
235
|
Chiao S, Zuo Z. A double-edged sword: volatile anesthetic effects on the neonatal brain. Brain Sci 2014; 4:273-94. [PMID: 24961761 PMCID: PMC4101477 DOI: 10.3390/brainsci4020273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 02/01/2023] Open
Abstract
The use of volatile anesthetics, a group of general anesthetics, is an exceedingly common practice. These anesthetics may have neuroprotective effects. Over the last decade, anesthetic induced neurotoxicity in pediatric populations has gained a certain notoriety based on pre-clinical cell and animal studies demonstrating that general anesthetics may induce neurotoxicity, including neuroapoptosis, neurodegeneration, and long-term neurocognitive and behavioral deficits. With hundreds of millions of people having surgery under general anesthesia worldwide, and roughly six million children annually in the U.S. alone, the importance of clearly defining toxic or protective effects of general anesthetics cannot be overstated. Yet, with our expanding body of knowledge, we have come to learn that perhaps not all volatile anesthetics have the same pharmacological profiles; certain ones may have a more favorable neurotoxic profile and may actually exhibit neuroprotection in specific populations and situations. Thus far, very few clinical studies exist, and have not yet been convincing enough to alter our practice. This review will provide an update on current data regarding volatile anesthetic induced neurotoxicity and neuroprotection in neonatal and infant populations. In addition, this paper will discuss ongoing studies and the trajectory of further research over the coming years.
Collapse
Affiliation(s)
- Sunny Chiao
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA.
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
236
|
Abstract
Preclinical and clinical studies have demonstrated the adverse consequences of untreated pain and stress on brain development in the preterm infant. Sucrose has widely been implemented as standard therapy for minor procedural pain. Anesthetics are commonly utilized in preterm infants during major surgery. Pharmacologic agents (benzodiazepines and opioids) have been examined in clinical trials of preterm infants requiring invasive mechanical ventilation. Controversy exists regarding the safety and long-term impact of these interventions. Ongoing multidisciplinary research will help define the impact of these agents and identify potential alternative therapies.
Collapse
|
237
|
Wang M, Zhang JH, Applegate RL. Adverse effect of inhalational anesthetics on the developing brain. Med Gas Res 2014; 4:2. [PMID: 24528992 PMCID: PMC3926857 DOI: 10.1186/2045-9912-4-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 02/11/2014] [Indexed: 11/10/2022] Open
Abstract
We did a PubMed search and summarized studies on the potential adverse effect of anesthetics especially neurotoxicity in the developing brain, so named anesthesia-induced developmental neurotoxicity. Even though many experimental studies using animal models indicated some adverse effect of anesthetics, more evidence is needed before a recommendation can be made to change the way those anesthetics are used in the pediatric population. Two large clinical trials are underway and may provide insight to the potential human neurotoxic effect of anesthetics.
Collapse
Affiliation(s)
| | | | - Richard L Applegate
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
238
|
Li G, Yu B. Elevation of protective autophagy as a potential way for preventing developmental neurotoxicity of general anesthetics. Med Hypotheses 2014; 82:177-80. [DOI: 10.1016/j.mehy.2013.11.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/25/2013] [Indexed: 12/19/2022]
|
239
|
Savage S, Ma D. The neurotoxicity of nitrous oxide: the facts and "putative" mechanisms. Brain Sci 2014; 4:73-90. [PMID: 24961701 PMCID: PMC4066238 DOI: 10.3390/brainsci4010073] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/08/2014] [Accepted: 01/16/2014] [Indexed: 02/03/2023] Open
Abstract
Nitrous oxide is a widely used analgesic agent, used also in combination with anaesthetics during surgery. Recent research has raised concerns about possible neurotoxicity of nitrous oxide, particularly in the developing brain. Nitrous oxide is an N-methyl-d-aspartate (NMDA)-antagonist drug, similar in nature to ketamine, another anaesthetic agent. It has been linked to post-operative cardiovascular problems in clinical studies. It is also widely known that exposure to nitrous oxide during surgery results in elevated homocysteine levels in many patients, but very little work has investigated the long term effect of these increased homocysteine levels. Now research in rodent models has found that homocysteine can be linked to neuronal death and possibly even cognitive deficits. This review aims to examine the current knowledge of mechanisms of action of nitrous oxide, and to describe some pathways by which it may have neurotoxic effects.
Collapse
Affiliation(s)
- Sinead Savage
- Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, UK.
| | - Daqing Ma
- Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, UK.
| |
Collapse
|
240
|
Deng M, Hofacer RD, Jiang C, Joseph B, Hughes EA, Jia B, Danzer SC, Loepke AW. Brain regional vulnerability to anaesthesia-induced neuroapoptosis shifts with age at exposure and extends into adulthood for some regions. Br J Anaesth 2014; 113:443-51. [PMID: 24431386 DOI: 10.1093/bja/aet469] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND General anaesthesia facilitates surgical operations and painful interventions in millions of patients every year. Recent observations of anaesthetic-induced neuronal cell death in newborn animals have raised substantial concerns for young children undergoing anaesthesia. However, it remains unclear why some brain regions are more affected than others, why certain neurones are eliminated while neighbouring cells are seemingly unaffected, and what renders the developing brain exquisitely vulnerable, while the adult brain apparently remains resistant to the phenomenon. METHODS Neonatal (P7), juvenile (P21), and young adult mice (P49) were anaesthetized with 1.5% isoflurane. At the conclusion of anaesthesia, activated cleaved caspase 3 (AC3), a marker of apoptotic cell death, was quantified in the neocortex (RSA), caudoputamen (CPu), hippocampal CA1 and dentate gyrus (DG), cerebellum (Cb), and olfactory bulb (GrO) and compared with that found in unanaesthetized littermates. RESULTS After anaesthetic exposure, increased AC3 was detected in neonatal mice in RSA (11-fold, compared with controls), CPu (10-fold), CA1 (three-fold), Cb (four-fold), and GrO (four-fold). Surprisingly, AC3 continued to be elevated in the DG and GrO of juvenile (15- and 12-fold, respectively) and young adult mice (two- and four-fold, respectively). CONCLUSIONS The present study confirms the findings of previous studies showing peak vulnerability to anaesthesia-induced neuronal cell death in the newborn forebrain. It also shows sustained susceptibility into adulthood in areas of continued neurogenesis, substantially expanding the previously observed age of vulnerability. The differential windows of vulnerability among brain regions, which closely follow regional peaks in neurogenesis, may explain the heightened vulnerability of the developing brain because of its increased number of immature neurones.
Collapse
Affiliation(s)
- M Deng
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai 201102, China Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - R D Hofacer
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Program in Neuroscience, University of Cincinnati, Cincinnati, OH 45221, USA
| | - C Jiang
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - B Joseph
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - E A Hughes
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - B Jia
- Heart Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - S C Danzer
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Program in Neuroscience, University of Cincinnati, Cincinnati, OH 45221, USA Department of Anesthesiology, University of Cincinnati, Cincinnati, OH 45221, USA Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45221, USA
| | - A W Loepke
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Program in Neuroscience, University of Cincinnati, Cincinnati, OH 45221, USA Department of Anesthesiology, University of Cincinnati, Cincinnati, OH 45221, USA Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
241
|
McCann ME, Schouten ANJ. Beyond survival; influences of blood pressure, cerebral perfusion and anesthesia on neurodevelopment. Paediatr Anaesth 2014; 24:68-73. [PMID: 24267703 DOI: 10.1111/pan.12310] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2013] [Indexed: 02/04/2023]
Abstract
Neonates have a higher perioperative mortality risk largely due to the degree of prior illness of the infants, the complexity of their surgeries, and infant physiology. It is important to consider contributing anesthetic factors during the perioperative period that may affect cerebral perfusion and neurocognitive outcome, such as alterations in hemodynamics and ventilation. Limitations of blood pressure as a marker for cerebral perfusion are discussed, as well as the effect of hypocapnia on the brain.
Collapse
Affiliation(s)
- Mary Ellen McCann
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
242
|
Effects of multiple exposures to sevoflurane at sub-MAC doses on neuroapoptosis and cognitive function during the neonatal period☆. COLOMBIAN JOURNAL OF ANESTHESIOLOGY 2014. [DOI: 10.1097/01819236-201442030-00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
243
|
Abstract
All routinely utilized sedatives and anesthetics have been found neurotoxic in a wide variety of animal species, including non-human primates. Neurotoxic effects observed in animals include histologic evidence for apoptotic neuronal cell death and subsequent learning and memory impairment. Several cohort studies in neonates with significant comorbidities requiring surgical procedures early in life have also demonstrated abnormal neurodevelopmental outcomes. This article provides an overview of the currently available data from both animal experiments and human clinical studies regarding the effects of sedatives and anesthetics on the developing brain.
Collapse
Affiliation(s)
- Erica P Lin
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 2001, Cincinnati, OH 45229, USA.
| | - Sulpicio G Soriano
- Department of Anesthesiology, Perioperative, and Pain Medicine, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Andreas W Loepke
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 2001, Cincinnati, OH 45229, USA
| |
Collapse
|
244
|
Yu Y, Zhang Y. Desflurane accelerates neuronal cytotoxicity of Aβ by downregulating miR-214. Neurosci Lett 2013; 554:28-33. [PMID: 24021802 DOI: 10.1016/j.neulet.2013.08.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/19/2013] [Accepted: 08/27/2013] [Indexed: 10/26/2022]
Abstract
Since more and more evidence suggests anesthetics serve as a risk factor of Alzheimer's disease (AD), we examine the neurotoxicity of a commonly used inhalational anesthetics, desflurane, and its potential toxicity mechanism in primary rat hippocampal neurons. Here we show that desflurane increase the cytotoxicity of intracellular and extracellular amyloid β (Aβ) in the presence or absence of serum. It is also demonstrated that the cytotoxicity of desflurane is caused by the reduction of miR-214 which binds to Bax 3'UTR and results in the increased expression of Bax. Therefore, we conclude that desflurane accelerates neuronal cytotoxicity of Aβ by downregulating miR-214. Our study sheds a light on the therapy of cytotoxicity induced by inhaled anesthetics, especially in patients of AD.
Collapse
Affiliation(s)
- You Yu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, China
| | | |
Collapse
|
245
|
Perna RB, Loughan AR, Le JA, Hertza J. Prenatal and Perinatal Anesthesia and the Long-Term Cognitive Sequelae: A Review. APPLIED NEUROPSYCHOLOGY-CHILD 2013; 4:65-71. [DOI: 10.1080/21622965.2013.779275] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
246
|
Li J, Wu H, Xue G, Wang P, Hou Y. 17β-Oestradiol Protects Primary-Cultured Rat Cortical Neurons from Ketamine-Induced Apoptosis by Activating PI3K/Akt/Bcl-2 Signalling. Basic Clin Pharmacol Toxicol 2013; 113:411-8. [PMID: 23981522 DOI: 10.1111/bcpt.12124] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/12/2013] [Indexed: 12/30/2022]
Affiliation(s)
- Jianli Li
- Hebei Medical University; Shijiazhuang Hebei Province China
- Hebei General Hospital; Shijiazhuang Hebei Province China
| | - Honghai Wu
- Department of Pharmacy; Bethune International Peace Hospital of Chinese PLA; Shijiazhuang Hebei Province China
| | - Gai Xue
- Department of Pharmacy; Bethune International Peace Hospital of Chinese PLA; Shijiazhuang Hebei Province China
| | - Pei Wang
- Hebei General Hospital; Shijiazhuang Hebei Province China
| | - Yanning Hou
- Hebei Medical University; Shijiazhuang Hebei Province China
- Department of Pharmacy; Bethune International Peace Hospital of Chinese PLA; Shijiazhuang Hebei Province China
| |
Collapse
|
247
|
Wang WY, Luo Y, Jia LJ, Hu SF, Lou XK, Shen SL, Lu H, Zhang HH, Yang R, Wang H, Ma ZW, Xue QS, Yu BW. Inhibition of aberrant cyclin-dependent kinase 5 activity attenuates isoflurane neurotoxicity in the developing brain. Neuropharmacology 2013; 77:90-9. [PMID: 24055498 DOI: 10.1016/j.neuropharm.2013.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/29/2022]
Abstract
Aberrant CDK5 activity is implicated in a number of neurodegenerative disorders. Isoflurane exposure leads to neuronal apoptosis, and subsequent learning and memory defects in the developing brain. The present study was designed to examine whether and how CDK5 activity plays a role in developmental isoflurane neurotoxicity. Rat pups and hippocampal neuronal cultures were exposed to 1.5% isoflurane for 4 h. The protein and mRNA levels of CDK5, p35 and p25 were detected by western blot and QReal-Time PCR. CDK5 activity was evaluated in vitro using Histone H1 as a substrate. Roscovitine (an inhibitor of CDK5) was applied before isoflurane treatment, cleaved Caspase-3, Bcl-2, Bax, MEF2 and phospho-MEF2A-Ser-408 expressions were determined. Dominant-Negative CDK5 was transfected before isoflurane treatment. Neuronal apoptosis was evaluated by Flow cytometry (FCM) and TUNEL-staining. Cognitive functions were assessed by Morris water maze. We found that isoflurane treatment led to an aberrant CDK5 activation due to its activator p25 that was cleaved from p35 by calpain. Inhibition of CDK5 activity with Roscovitine enhanced Bcl-2, and decreased cleaved Caspase-3 and Bax expressions. In addition, isoflurane exposure resulted in a decrease of MEF2 and increase of phospho-MEF2A-Ser-408, which were rescued by Roscovitine or Dominant-Negative CDK5 transfection. Dominant-Negative CDK5 transfection also decreased the percentage of TUNEL-positive cells in isoflurane neurotoxicity. Moreover, Roscovitine remarkably alleviated the learning and memory deficits induced by postnatal isoflurane exposure. These results indicated that aberrant CDK5 activity-dependent MEF2 phosphorylation mediates developmental isoflurane neurotoxicity. Inhibition of CDK5 overactivation contributes to the relief of isoflurane neurotoxicity in the developing brain.
Collapse
Affiliation(s)
- Wen-Yuan Wang
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, Shangtang Road 158, Hangzhou 310014, China.
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Ruijin Er Road 197, Shanghai 200025, China
| | - Li-Jie Jia
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Ruijin Er Road 197, Shanghai 200025, China
| | - Shuang-Fei Hu
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, Shangtang Road 158, Hangzhou 310014, China
| | - Xiao-Kan Lou
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, Shangtang Road 158, Hangzhou 310014, China
| | - She-Liang Shen
- Department of Anesthesiology, Zhejiang Provincial People's Hospital, Shangtang Road 158, Hangzhou 310014, China
| | - Han Lu
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Ruijin Er Road 197, Shanghai 200025, China
| | - Hong-Hai Zhang
- Department of Anesthesiology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Rui Yang
- Department of Pharmacy, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Hua Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zheng-Wen Ma
- Department of Neurobiology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qing-Sheng Xue
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Ruijin Er Road 197, Shanghai 200025, China
| | - Bu-Wei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Ruijin Er Road 197, Shanghai 200025, China.
| |
Collapse
|
248
|
Patkai J, Zana-Taieb E, Didier C, Jarreau PH, Lopez E. Aspects fondamentaux de la toxicite éventuelle des drogues anesthésiques. Arch Pediatr 2013; 20:1059-66. [DOI: 10.1016/j.arcped.2013.06.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 06/21/2013] [Accepted: 06/24/2013] [Indexed: 01/08/2023]
|
249
|
Mishra SK, Kang JH, Lee CW, Oh SH, Ryu JS, Bae YS, Kim HM. Midazolam induces cellular apoptosis in human cancer cells and inhibits tumor growth in xenograft mice. Mol Cells 2013; 36:219-26. [PMID: 24008365 PMCID: PMC3887981 DOI: 10.1007/s10059-013-0050-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 07/03/2013] [Accepted: 07/03/2013] [Indexed: 12/18/2022] Open
Abstract
Midazolam is a widely used anesthetic of the benzodiazepine class that has shown cytotoxicity and apoptosisinducing activity in neuronal cells and lymphocytes. This study aims to evaluate the effect of midazolam on growth of K562 human leukemia cells and HT29 colon cancer cells. The in vivo effect of midazolam was investigated in BALB/c-nu mice bearing K562 and HT29 cells human tumor xenografts. The results show that midazolam decreased the viability of K562 and HT29 cells by inducing apoptosis and S phase cell-cycle arrest in a concentration-dependent manner. Midazolam activated caspase-9, capspase-3 and PARP indicating induction of the mitochondrial intrinsic pathway of apoptosis. Midazolam lowered mitochondrial membrane potential and increased apoptotic DNA fragmentation. Midazolam showed reactive oxygen species (ROS) scavenging activity through inhibition of NADPH oxidase 2 (Nox2) enzyme activity in K562 cells. Midazolam caused inhibition of pERK1/2 signaling which led to inhibition of the anti-apoptotic proteins Bcl-XL and XIAP and phosphorylation activation of the pro-apoptotic protein Bid. Midazolam inhibited growth of HT29 tumors in xenograft mice. Collectively our results demonstrate that midazolam caused growth inhibition of cancer cells via activation of the mitochondrial intrinsic pathway of apoptosis and inhibited HT29 tumor growth in xenograft mice. The mechanism underlying these effects of midazolam might be suppression of ROS production leading to modulation of apoptosis and growth regulatory proteins. These findings present possible clinical implications of midazolam as an anesthetic to relieve pain during in vivo anticancer drug delivery and to enhance anticancer efficacy through its ROS-scavenging and pro-apoptotic properties.
Collapse
Affiliation(s)
| | - Ju-Hee Kang
- Division of Cancer Biology, National Cancer Center, Goyang 410-769, Korea
- Department of Food and Nutrition, Division of Natural Sciences, Chung-Ang University, Ansung 456-756, Korea
| | - Chang Woo Lee
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 363-883, Korea
| | - Seung Hyun Oh
- Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 406-840, Korea
| | - Jun Sun Ryu
- Head and Neck Oncology Clinic, Center for Specific Organs Cancer, Center for Thyroid Cancer, National Cancer Center, Goyang 410-769, Korea
| | - Yun Soo Bae
- Department of Life Science, College of Natural Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Hwan Mook Kim
- Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 406-840, Korea
| |
Collapse
|
250
|
Abstract
Laboratory studies have shown that general anesthetics may cause accelerated apoptosis and other adverse morphologic changes in neurons of the developing brain. The mechanism may be related to the neuronal quiescence or inactivity associated with anesthetic exposure. Few data exist on how brief anesthetic exposure may affect neurodevelopment in the newborn. Good evidence however shows that untreated pain and stress have an adverse effect on neurodevelopment, and therefore, at this stage, providing effective analgesia, sedation, and anesthesia would seem to be more important than concern over neurotoxicity.
Collapse
|