51
|
Neudecker V, Perez-Zoghbi JF, Brambrink AM. Recent advances in understanding cognitive and behavioural alterations after early-in-life anaesthesia exposure and new mitigation/alternative strategies in preclinical studies. Curr Opin Anaesthesiol 2021; 34:402-408. [DOI: 10.1097/aco.0000000000001016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
52
|
Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor and cognitive deficits. Cell Biol Toxicol 2021; 38:347-369. [PMID: 33928466 DOI: 10.1007/s10565-021-09601-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022]
Abstract
Clinical surgical practices have found that children who undergo multiple anesthesia may have an increased risk of deficiencies in cognition and fine motor control. Here, we report that YT521-B homology domain family 1 (YTHDF1), a critical reader protein for N6-methyladenosine-modified mRNA, was significantly downregulated in the prefrontal cortex of young mice after multiple sevoflurane anesthesia exposures. Importantly, sevoflurane led to a decrease in protein synthesis in mouse cortical neurons that was fully rescued by YTHDF1, suggesting that anesthesia may affect early brain development by affecting m6A-dependent mRNA translation. Transcriptome-wide experiments showed that numerous mRNA targets related to synaptic functions in the prefrontal mouse cortex were associated with m6A methylation and YTHDF1. In particular, we found that synaptophysin, a critical presynaptic protein, was specifically modified by m6A methylation and associated with YTHDF1, and m6A methylation of synaptophysin decreased with multiple sevoflurane exposures. Importantly, we showed that fine motor control skills and cognitive functions were impaired in mice with multiple anesthesia exposures, and these effects were fully reversed by reintroducing YTHDF1 through a blood-brain barrier (BBB)-crossing viral delivery system. Finally, we found that the fine motor skills in children who underwent prolonged anesthesia were compromised 6 months after surgery. Our findings indicated that impairment in the translational regulation of mRNA via N6-methyladenosine methylation is a potential mechanism underlying the effects of anesthesia on neural development in the young brain. 1. N6-methyladenosine (m6A) modifications were involved in anesthesia-induced neurotoxicity. 2. Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor deficits in young mice. 3. YTHDF1, a m6A reader protein, rescued sevoflurane-induced protein synthesis inhibition and fine motor deficits in young mice.
Collapse
|
53
|
Pei W, Fu L, Li SQ, Yu Y. Brain transcriptomics of nonhuman primates: A review. Neurosci Lett 2021; 753:135872. [PMID: 33812931 DOI: 10.1016/j.neulet.2021.135872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 11/12/2022]
Abstract
The brain is one of the most important and intricate organs in our bodies. Interpreting brain function and illustrating the changes and molecular mechanisms during physiological or pathological processes are essential but sometimes difficult to achieve. In addition to histology, ethology and pharmacology, the development of transcriptomics alleviates this condition by enabling high-throughput observation of the brain at various levels of anatomical specificity. Moreover, because human brain samples are scarce, the brains of nonhuman primates are important alternative models. Here in this review, we summarize the applications of transcriptomics in nonhuman primate brain studies, including investigations of brain development, aging, toxic effects and diseases. Overall, as a powerful tool with developmental potential, transcriptomics has been widely utilized in neuroscience.
Collapse
Affiliation(s)
- Wendi Pei
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China
| | - Lin Fu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China
| | - Shui-Qing Li
- Department of Pain, Peking University Third Hospital, Beijing, 100191, China.
| | - Yang Yu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
54
|
Young JT, Vlasova RM, Howell BR, Knickmeyer RC, Morin E, Kuitchoua KI, Lubach GR, Noel J, Hu X, Shi Y, Caudill G, Alexander AL, Niethammer M, Paule MG, Coe CL, Sanchez M, Styner M. General anaesthesia during infancy reduces white matter micro-organisation in developing rhesus monkeys. Br J Anaesth 2021; 126:845-853. [PMID: 33549320 DOI: 10.1016/j.bja.2020.12.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 12/04/2020] [Accepted: 12/24/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Non-human primates are commonly used in neuroimaging research for which general anaesthesia or sedation is typically required for data acquisition. In this analysis, the cumulative effects of exposure to ketamine, Telazol® (tiletamine and zolazepam), and the inhaled anaesthetic isoflurane on early brain development were evaluated in two independent cohorts of typically developing rhesus macaques. METHODS Diffusion MRI scans were analysed from 43 rhesus macaques (20 females and 23 males) at either 12 or 18 months of age from two separate primate colonies. RESULTS Significant, widespread reductions in fractional anisotropy with corresponding increased axial, mean, and radial diffusivity were observed across the brain as a result of repeated anaesthesia exposures. These effects were dose dependent and remained after accounting for age and sex at time of exposure in a generalised linear model. Decreases of up to 40% in fractional anisotropy were detected in some brain regions. CONCLUSIONS Multiple exposures to commonly used anaesthetics were associated with marked changes in white matter microstructure. This study is amongst the first to examine clinically relevant anaesthesia exposures on the developing primate brain. It will be important to examine if, or to what degree, the maturing brain can recover from these white matter changes.
Collapse
Affiliation(s)
- Jeffrey T Young
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Roza M Vlasova
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Brittany R Howell
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Rebecca C Knickmeyer
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elyse Morin
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kaela I Kuitchoua
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Gabriele R Lubach
- Harlow Center for Biological Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jean Noel
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaoping Hu
- Department of Bioengineering, University of California, Riverside, CA, USA
| | - Yundi Shi
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gibson Caudill
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew L Alexander
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA
| | - Marc Niethammer
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mar Sanchez
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Martin Styner
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
55
|
Raper J, Chahroudi A. Clinical and Preclinical Evidence for Adverse Neurodevelopment after Postnatal Zika Virus Infection. Trop Med Infect Dis 2021; 6:tropicalmed6010010. [PMID: 33445671 PMCID: PMC7838975 DOI: 10.3390/tropicalmed6010010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Although the Zika virus (ZIKV) typically causes mild or no symptoms in adults, during the 2015−2016 outbreak, ZIKV infection in pregnancy resulted in a spectrum of diseases in infants, including birth defects and neurodevelopmental disorders identified in childhood. While intense clinical and basic science research has focused on the neurodevelopmental outcomes of prenatal ZIKV infection, less is known about the consequences of infection during early life. Considering the neurotropism of ZIKV and the rapidly-developing postnatal brain, it is important to understand how infection during infancy may disrupt neurodevelopment. This paper reviews the current knowledge regarding early postnatal ZIKV infection. Emerging clinical evidence supports the hypothesis that ZIKV infection during infancy can result in negative neurologic consequences. However, clinical data regarding postnatal ZIKV infection in children are limited; as such, animal models play an important role in understanding the potential complications of ZIKV infection related to the vulnerable developing brain. Preclinical data provide insight into the potential behavioral, cognitive, and motor domains that clinical studies should examine in pediatric populations exposed to ZIKV during infancy.
Collapse
Affiliation(s)
- Jessica Raper
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA;
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ann Chahroudi
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA;
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
56
|
Piao M, Wang Y, Liu N, Wang X, Chen R, Qin J, Ge P, Feng C. Sevoflurane Exposure Induces Neuronal Cell Parthanatos Initiated by DNA Damage in the Developing Brain via an Increase of Intracellular Reactive Oxygen Species. Front Cell Neurosci 2020; 14:583782. [PMID: 33424554 PMCID: PMC7793874 DOI: 10.3389/fncel.2020.583782] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
The safety of volatile anesthetics in infants and young children has been drawing increasing concern due to its potential neurotoxicity in the developing brain. Neuronal death is considered a major factor associated with developmental neurotoxicity after exposure to volatile anesthetics sevoflurane, but its mechanism remains elusive. Parthanatos, a new type of programmed cell death, resulting from poly (ADP-ribose) polymerase 1 (PARP-1) hyperactivation in response to DNA damage, was found to account for the pathogenesis of multiple neurological disorders. However, the role of Parthanatos in sevoflurane-induced neonatal neuronal cell death has not been investigated. To test it, neuronal cells treated with 2, 4, and 8% sevoflurane for 6, 12, and 24 h and postnatal day 7 rats exposed to 2.5% sevoflurane for 6 h were used in the present study. Our results found sevoflurane exposure induced neuronal cell death, which was accompanied by PARP-1 hyperactivation, cytoplasmic polymerized ADP-ribose (PAR) accumulation, mitochondrial depolarization, and apoptosis-inducing factor (AIF) nuclear translocation in the neuronal cells and hippocampi of rats. Pharmacological or genetic inhibition of PAPR-1 significantly alleviated sevoflurane-induced neuronal cell death and accumulation of PAR polymer and AIF nuclear translocation, which were consistent with the features of Parthanatos. We observed in vitro and in vivo that sevoflurane exposure resulted in DNA damage, given that 8-hydroxydeoxyguanosine (8-OHdG) and phosphorylation of histone variant H2AX (γH2AX) were improved. Moreover, we detected that sevoflurane exposure was associated with an overproduction of intracellular reactive oxygen species (ROS). Inhibition of ROS with antioxidant NAC markedly alleviated DNA damage caused by sevoflurane, indicating that ROS participated in the regulation of sevoflurane-induced DNA damage. Additionally, sevoflurane exposure resulted in upregulation of Parthanatos-related proteins and neuronal cell death, which were significantly attenuated by pretreatment with NAC. Therefore, these results suggest that sevoflurane exposure induces neuronal cell Parthanatos initiated by DNA damage in the developing brain via the increase of intracellular ROS.
Collapse
Affiliation(s)
- Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Yingying Wang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Xuedong Wang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Rui Chen
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Jing Qin
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Pengfei Ge
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
57
|
Neudecker V, Perez-Zoghbi JF, Coleman K, Neuringer M, Robertson N, Bemis A, Glickman B, Schenning KJ, Fair DA, Martin LD, Dissen GA, Brambrink AM. Infant isoflurane exposure affects social behaviours, but does not impair specific cognitive domains in juvenile non-human primates. Br J Anaesth 2020; 126:486-499. [PMID: 33198945 DOI: 10.1016/j.bja.2020.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clinical studies show that children exposed to anaesthetics for short times at young age perform normally on intelligence tests, but display altered social behaviours. In non-human primates (NHPs), infant anaesthesia exposure for several hours causes neurobehavioural impairments, including delayed motor reflex development and increased anxiety-related behaviours assessed by provoked response testing. However, the effects of anaesthesia on spontaneous social behaviours in juvenile NHPs have not been investigated. We hypothesised that multiple, but not single, 5 h isoflurane exposures in infant NHPs are associated with impairments in specific cognitive domains and altered social behaviours at juvenile age. METHODS Eight Rhesus macaques per group were anaesthetised for 5 h using isoflurane one (1×) or three (3×) times between postnatal days 6 and 12 or were exposed to room air (control). Cognitive testing, behavioural assessments in the home environment, and provoked response testing were performed during the first 2 yr of life. RESULTS The cognitive functions tested did not differ amongst groups. However, compared to controls, NHPs in the 3× group showed less close social behaviour (P=0.016), and NHPs in the 1× group displayed increased anxiety-related behaviours (P=0.038) and were more inhibited towards novel objects (P<0.001). CONCLUSIONS 5 h exposures of NHPs to isoflurane during infancy are associated with decreased close social behaviour after multiple exposures and more anxiety-related behaviours and increased behavioural inhibition after single exposure, but they do not affect the cognitive domains tested. Our findings are consistent with behavioural alterations in social settings reported in clinical studies, which may guide future research.
Collapse
Affiliation(s)
- Viola Neudecker
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Jose F Perez-Zoghbi
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Kristine Coleman
- Division of Neuroscience, USA; Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | | | - Nicola Robertson
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | | | | | | | - Damien A Fair
- Department of Behavioural Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Lauren D Martin
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | | | - Ansgar M Brambrink
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
58
|
Isoflurane Exposure in Juvenile Caenorhabditis elegans Causes Persistent Changes in Neuron Dynamics. Anesthesiology 2020; 133:569-582. [PMID: 32452864 DOI: 10.1097/aln.0000000000003335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Animal studies demonstrate that anesthetic exposure during neurodevelopment can lead to persistent behavioral impairment. The changes in neuronal function underlying these effects are incompletely understood. Caenorhabditis elegans is well suited for functional imaging of postanesthetic effects on neuronal activity. This study aimed to examine such effects within the neurocircuitry underlying C. elegans locomotion. METHODS C. elegans were exposed to 8% isoflurane for 3 h during the neurodevelopmentally critical L1 larval stage. Locomotion was assessed during early and late adulthood. Spontaneous activity was measured within the locomotion command interneuron circuitry using confocal and light-sheet microscopy of the calcium-sensitive fluorophore GCaMP6s. RESULTS C. elegans exposed to isoflurane demonstrated attenuation in spontaneous reversal behavior, persisting throughout the animal's lifespan (reversals/min: untreated early adulthood, 1.14 ± 0.42, vs. isoflurane-exposed early adulthood, 0.83 ± 0.55; untreated late adulthood, 1.75 ± 0.64, vs. isoflurane-exposed late adulthood, 1.14 ± 0.68; P = 0.001 and 0.006, respectively; n > 50 animal tracks/condition). Likewise, isoflurane exposure altered activity dynamics in the command interneuron AVA, which mediates crawling reversals. The rate at which AVA transitions between activity states was found to be increased. These anesthetic-induced effects were more pronounced with age (off-to-on activity state transition time (s): untreated early adulthood, 2.5 ± 1.2, vs. isoflurane-exposed early adulthood, 1.9 ± 1.3; untreated late adulthood, 4.6 ± 3.0, vs. isoflurane-exposed late adulthood, 3.0 ± 2.4; P = 0.028 and 0.008, respectively; n > 35 traces acquired from more than 15 animals/condition). Comparable effects were observed throughout the command interneuron circuitry, indicating that isoflurane exposure alters transition rates between behavioral crawling states of the system overall. These effects were modulated by loss-of-function mutations within the FoxO transcription factor daf-16 and by rapamycin-mediated mechanistic Target of Rapamycin (mTOR) inhibition. CONCLUSIONS Altered locomotive behavior and activity dynamics indicate a persistent effect on interneuron dynamics and circuit function in C. elegansafter developmental exposure to isoflurane. These effects are modulated by a loss of daf-16 or mTOR activity, consistent with a pathologic activation of stress-response pathways.
Collapse
|
59
|
Yan J, Li J, Cheng Y, Zhang Y, Zhou Z, Zhang L, Jiang H. Dusp4 Contributes to Anesthesia Neurotoxicity via Mediated Neural Differentiation in Primates. Front Cell Dev Biol 2020; 8:786. [PMID: 32974341 PMCID: PMC7466444 DOI: 10.3389/fcell.2020.00786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background Children who are exposed to anesthesia multiple times may undergo cognitive impairment during development. The underlying mechanism has been revealed as anesthesia-induced cognitive deficiency in young rodents and monkeys. However, the molecular mechanism of sevoflurane-induced neural development toxicity is unclear. Methods By combining RNA sequencing analysis of macaques’ prefrontal cortex and human neural differentiation, this study investigates the mechanism of sevoflurane-induced neurotoxicity in primates. Results The level of dual specificity protein phosphatase 4 (Dusp4) was significantly downregulated in non-human primates after sevoflurane treatment. We further uncovered the dynamical expression of Dusp4 during the human neural differentiation of human embryonic stem cells and found that knockdown of Dusp4 could significantly inhibit human neural differentiation. Conclusion This study indicated that Dusp4 is critically involved in the sevoflurane-induced inhibition of neural differentiation in non-human primate and the regulation of human neural differentiation. It also suggested that Dusp4 is a potential therapeutic target for preventing the sevoflurane-induced neurotoxicity in primates.
Collapse
Affiliation(s)
- Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingjie Li
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyong Cheng
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenning Zhou
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
60
|
Soriano SG, McCann ME. Is Anesthesia Bad for the Brain? Current Knowledge on the Impact of Anesthetics on the Developing Brain. Anesthesiol Clin 2020; 38:477-492. [PMID: 32792178 DOI: 10.1016/j.anclin.2020.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There are compelling preclinical data that common general anesthetics cause increased neuroapoptosis in juvenile animals. Retrospective studies demonstrate that young children exposed to anesthesia have school difficulties, which could be caused by anesthetic neurotoxicity, perioperative hemodynamic and homeostatic instability, underlying morbidity, or the neuroinflammatory effects of surgical trauma. Unnecessary procedures should be avoided. Baseline measures of blood pressure are important in determining perioperative blood pressure goals. Inadvertent hypocapnia or moderate hypercapnia and hyperoxia or hypoxia should be avoided. Pediatric patients should be maintained in a normothermic, euglycemic state with neutral positioning. Improving outcomes of infants and children requires the collaboration of anesthesiologists, surgeons, pediatricians and neonatologists.
Collapse
Affiliation(s)
- Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Mary Ellen McCann
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
61
|
Dexmedetomidine Attenuates Neurotoxicity in Developing Rats Induced by Sevoflurane through Upregulating BDNF-TrkB-CREB and Downregulating ProBDNF-P75NRT-RhoA Signaling Pathway. Mediators Inflamm 2020; 2020:5458061. [PMID: 32655312 PMCID: PMC7322616 DOI: 10.1155/2020/5458061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/23/2020] [Indexed: 12/22/2022] Open
Abstract
To investigate the mechanism dexmedetomidine in relieving the neurotoxicity of a developing brain induced by sevoflurane. Sprague-Dawley rats, 6 days old, were randomly divided into three groups. Rats in the control group were inhaled with air after injection of normal saline; rats in the sevoflurane group were injected with normal saline and inhaled with 3% sevoflurane for 2 h in three consecutive day; rats in the dexmedetomidine group were inhaled with 3% sevoflurane after intraperitoneal injection of dexmedetomidine 25 μg/kg. WB results showed that mBDNF, pTrkB/TrkB, and CREB were significantly decreased in the hippocampus of the sevoflurane group, which are significantly upregulated in the dexmedetomidine group. In the sevoflurane group, proBDNF, P75NRT, and RhoA were significantly increased, which were significantly lower than those in the dexmedetomidine group than those in the sevoflurane group. The expression BDNF was downregulated in the sevoflurane group, while the proBDNF was upregulated in the sevoflurane group. In the Morris water maze test, the escape latency of the sevoflurane group was significantly prolonged. In sevoflurane groups, the number of crossing platform was significantly reduced, the synaptic protein decreased significantly, and this effect was reversed in rats of the dexmedetomidine group. Dexmedetomidine could reduce synaptic plasticity decline in developing rats induced by sevoflurane, through downregulating the proBDNF-p75NTR-RhoA pathway and upregulating BDNF-TrkB-CREB.
Collapse
|
62
|
Li T, Huang Z, Wang X, Zou J, Tan S. Role of the GABAA receptors in the long-term cognitive impairments caused by neonatal sevoflurane exposure. Rev Neurosci 2020; 30:869-879. [PMID: 31145696 DOI: 10.1515/revneuro-2019-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023]
Abstract
Sevoflurane is a widely used inhalational anesthetic in pediatric surgeries, which is considered reasonably safe and reversible upon withdrawal. However, recent preclinical studies suggested that peri-neonatal sevoflurane exposure may cause developmental abnormalities in the brain. The present review aimed to present and discuss the accumulating experimental data regarding the undesirable effects of sevoflurane on brain development as revealed by the laboratory studies. First, we summarized the long-lasting side effects of neonatal sevoflurane exposure on cognitive functions. Subsequently, we presented the structural changes, namely, neuroapoptosis, neurogenesis and synaptogenesis, following sevoflurane exposure in the immature brain. Finally, we also discussed the potential mechanisms underlying subsequent cognitive impairments later in life, which are induced by neonatal sevoflurane exposure and pointed out potential strategies for mitigating sevoflurane-induced long-term cognitive impairments. The type A gamma-amino butyric acid (GABAA) receptor, the main targets of sevoflurane, is excitatory rather than inhibitory in the immature neurons. The excitatory effects of the GABAA receptors have been linked to increased neuroapoptosis, elevated serum corticosterone levels and epigenetic modifications following neonatal sevoflurane exposure in rodents, which might contribute to sevoflurane-induced long-term cognitive abnormalities. We proposed that the excitatory GABAA receptor-mediated HPA axis activity might be a novel mechanism underlying sevoflurane-induced long-term cognitive impairments. More studies are needed to investigate the effectiveness and mechanisms by targeting the excitatory GABAA receptor as a prevention strategy to alleviate cognitive deficits induced by neonatal sevoflurane exposure in future.
Collapse
Affiliation(s)
- Tao Li
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Zeyi Huang
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Xianwen Wang
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Ju Zou
- Department of Parasitology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Sijie Tan
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| |
Collapse
|
63
|
Kang W, Lu D, Yang X, Ma W, Chen X, Chen K, Xu X, Zhou X, Zhou L, Feng X. Sevoflurane Induces Hippocampal Neuronal Apoptosis by Altering the Level of Neuropeptide Y in Neonatal Rats. Neurochem Res 2020; 45:1986-1996. [PMID: 32378074 DOI: 10.1007/s11064-020-03028-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022]
Abstract
Numerous studies have shown that the inhaled general anesthetic sevoflurane imposes toxicity on the central nervous system during the developmental period but the underlying mechanisms remain unclear. Neuropeptide Y (NPY) was reported to have important neuroprotective effects, which can attenuate neuronal loss under pathological conditions. However, the effects of NPY on sevoflurane-induced hippocampal neuronal apoptosis have not been investigated. In this study, postnatal day 7 (PND7) Sprague-Dawley rats and primary cultured cells separated from hippocampi were exposed to sevoflurane (2.4% for 4 h) and the NPY expression levels after treatment were analyzed. Furthermore, neuronal apoptosis assay was conducted via immunofluorescence staining of cleaved caspase-3 and flow cytometry after exogenous NPY administration to PND7 rats as well as cultured hippocampal neurons to elucidate the role of NPY in sevoflurane-induced neurotoxicity. Our results showed the level of NPY gradually decreased within 24 h after sevoflurane exposure in both the hippocampus of PND7 rats and cultured hippocampal neurons, but not in cultured astrocytes. In the exogenous NPY pretreatment study, the proportion of cleaved caspase-3 positive cells in the CA1 region of the hippocampus was increased significantly at 24 h after sevoflurane treatment, while NPY pretreatment could reduce it. Similarly, NPY could also reverse the apoptogenic effect of sevoflurane on cultured neurons. Herein, our results showed that sevoflurane caused a significant decrease in NPY expression, whereas exogenous NPY supplementation could reduce sevoflurane-induced hippocampal neuronal apoptosis both in vivo and in vitro.
Collapse
Affiliation(s)
- Wenbin Kang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Dihan Lu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xiaoyu Yang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Wudi Ma
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xi Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Keyu Chen
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xuanxian Xu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xue Zhou
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| | - Lihua Zhou
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| | - Xia Feng
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
64
|
Chinn GA, Pearn ML, Vutskits L, Mintz CD, Loepke AW, Lee JJ, Chen J, Bosnjak ZJ, Brambrink AM, Jevtovic-Todorovic V, Sun LS, Sall JW. Standards for preclinical research and publications in developmental anaesthetic neurotoxicity: expert opinion statement from the SmartTots preclinical working group. Br J Anaesth 2020; 124:585-593. [PMID: 32145876 PMCID: PMC7424895 DOI: 10.1016/j.bja.2020.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/06/2020] [Accepted: 01/24/2020] [Indexed: 12/16/2022] Open
Abstract
In March 2019, SmartTots, a public-private partnership between the US Food and Drug Administration and the International Anesthesia Research Society, hosted a meeting attended by research experts, anaesthesia journal editors, and government agency representatives to discuss the continued need for rigorous preclinical research and the importance of establishing reporting standards for the field of anaesthetic perinatal neurotoxicity. This group affirmed the importance of preclinical research in the field, and welcomed novel and mechanistic approaches to answer some of the field's largest questions. The attendees concluded that summarising the benefits and disadvantages of specific model systems, and providing guidance for reporting results, would be helpful for designing new experiments and interpreting results across laboratories. This expert opinion report is a summary of these discussions, and includes a focused review of current animal models and reporting standards for the field of perinatal anaesthetic neurotoxicity. This will serve as a practical guide and road map for novel and rigorous experimental work.
Collapse
Affiliation(s)
- Gregory A Chinn
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Matthew L Pearn
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Laszlo Vutskits
- Department of Anesthesiology, Clinical Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Cyrus D Mintz
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas W Loepke
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer J Lee
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Jerri Chen
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Zeljko J Bosnjak
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | - Lena S Sun
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Jeffrey W Sall
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA.
| |
Collapse
|
65
|
Yu W, Wu Z, Zhao P. Neurotoxicity effects of anesthetic exposure on the developing brain of non-human primates. Med Hypotheses 2020; 140:109647. [PMID: 32135447 DOI: 10.1016/j.mehy.2020.109647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 11/17/2022]
Abstract
Mounting evidence has shown that general anesthetic agents commonly used clinically can cause anesthetic-related neurotoxicity in the developing brains of mammals, potentially causing long-term neurological impairment. This results in growing interest and concern among the public. Here, we present an overview of the relevant findings from non-human primates, arguably the very best model for studies of developmental neurotoxicity. These studies have shown that varying degrees of neurodegeneration occur as a result of anesthesia type, duration/dose of exposure, the timing of exposure, and brain region of interest, combined with subsequent alterations in cognitive assessments. Specifically, the rapid advancement of minimally or non-invasive neuroimaging methodologies and availability provided more sophisticated techniques for investigating brain structure and function. Neuroimaging methodologies have shown some of their most significant promise in studies of anesthetic-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110001, Liaoning Province, China.
| |
Collapse
|
66
|
Multiple sevoflurane exposures don't disturb the T-cell receptor repertoire in infant rhesus monkeys' thymus. Life Sci 2020; 248:117457. [PMID: 32092334 DOI: 10.1016/j.lfs.2020.117457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
AIMS Multiple surgical procedures and anesthesia increase the risk of the development in children. However, the influence of such exposures on the developing childhood immunity organs is rarely reported. MATERIALS AND METHODS High-throughput sequencing of T-cell receptor (TCR) repertoires (TCRseq) from rhesus monkeys' thymus was performed to investigate whether anesthetics could induce de novo antigen recognition via TCR or TCR development impairments. KEY FINDINGS No significant difference between sevoflurane and control groups regarding VJ gene combinations and diversity of V and J gene was seen, nor was there an obvious change in similar average number of Complementarity Determining Region 3 (CDR3) aa clonotypes. Our analysis of Rank abundance, Gini coefficient, Simpson index, Normalized Shannon Diversity Entropy (NSDE), Morisita-Horn Similarity Index (MHSI) and Bhattacharyya Distance (BD) indicated there is no difference in TCR diversity and similarity. SIGNIFICANCE These results suggest early events in thymic T cell development and repertoire generation are not abnormality after multiple sevoflurane exposure during childhood. The stabilization of the immune repertoires suggested the safety of sevoflurane in host immune response in children.
Collapse
|
67
|
Liang L, Xie R, Lu R, Ma R, Wang X, Wang F, Liu B, Wu S, Wang Y, Zhang H. Involvement of homodomain interacting protein kinase 2-c-Jun N-terminal kinase/c-Jun cascade in the long-term synaptic toxicity and cognition impairment induced by neonatal Sevoflurane exposure. J Neurochem 2020; 154:372-388. [PMID: 31705656 PMCID: PMC7496229 DOI: 10.1111/jnc.14910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/23/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
Sevoflurane is one of the most widely used anesthetics with recent concerns rising about its pediatric application. The synaptic toxicity and mechanisms underlying its long‐term cognition impairment remain unclear. In this study, we investigated the expression and roles of homeodomain interacting protein kinase 2 (HIPK2), a stress activating kinase involved in neuronal survival and synaptic plasticity, and its downstream c‐Jun N‐terminal kinase (JNK)/c‐Jun signaling in the long‐term toxicity of neonatal Sevoflurane exposure. Our data showed that neonatal Sevoflurane exposure results in impairment of memory, enhancement of anxiety, less number of excitatory synapses and lower levels of synaptic proteins in the hippocampus of adult rats without significant changes of hippocampal neuron numbers. Up‐regulation of HIPK2 and JNK/c‐Jun was observed in hippocampal granular neurons shortly after Sevoflurane exposure and persisted to adult. 5‐((6‐Oxo‐5‐(6‐(piperazin‐1‐yl)pyridin‐3‐yl)‐1,6‐dihydropyridin‐3‐yl)methylene)thiazolidine‐2,4‐dione trifluoroacetate, antagonist of HIPK2, could significantly rescue the cognition impairment, decrease in long‐term potentiation, reduction in spine density and activation of JNK/c‐Jun induced by Sevoflurane. JNK antagonist SP600125 partially restored synapse development and cognitive function without affecting the expression of HIPK2. These data, in together, revealed a novel role of HIPK2‐JNK/c‐Jun signaling in the long‐term synaptic toxicity and cognition impairment of neonatal Sevoflurane exposure, indicating HIPK2‐JNK/c‐Jun cascade as a potential target for reducing the synaptic toxicity of Sevoflurane. ![]()
Cover Image for this issue: doi: 10.1111/jnc.14757.
Collapse
Affiliation(s)
- Lirong Liang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Rougang Xie
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Rui Lu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Ruixue Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Xiaoxia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Fengjuan Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Bing Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Hui Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
68
|
Conflicting Actions of Inhalational Anesthetics, Neurotoxicity and Neuroprotection, Mediated by the Unfolded Protein Response. Int J Mol Sci 2020; 21:ijms21020450. [PMID: 31936788 PMCID: PMC7013687 DOI: 10.3390/ijms21020450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/02/2020] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Preclinical studies have shown that exposure of the developing brain to inhalational anesthetics can cause neurotoxicity. However, other studies have claimed that anesthetics can exert neuroprotective effects. We investigated the mechanisms associated with the neurotoxic and neuroprotective effects exerted by inhalational anesthetics. Neuroblastoma cells were exposed to sevoflurane and then cultured in 1% oxygen. We evaluated the expression of proteins related to the unfolded protein response (UPR). Next, we exposed adult mice in which binding immunoglobulin protein (BiP) had been mutated, and wild-type mice, to sevoflurane, and evaluated their cognitive function. We compared our results to those from our previous study in which mice were exposed to sevoflurane at the fetal stage. Pre-exposure to sevoflurane reduced the expression of CHOP in neuroblastoma cells exposed to hypoxia. Anesthetic pre-exposure also significantly improved the cognitive function of adult wild-type mice, but not the mutant mice. In contrast, mice exposed to anesthetics during the fetal stage showed cognitive impairment. Our data indicate that exposure to inhalational anesthetics causes endoplasmic reticulum (ER) stress, and subsequently leads to an adaptive response, the UPR. This response may enhance the capacity of cells to adapt to injuries and improve neuronal function in adult mice, but not in developing mice.
Collapse
|
69
|
Luo A, Tang X, Zhao Y, Zhou Z, Yan J, Li S. General Anesthetic-Induced Neurotoxicity in the Immature Brain: Reevaluating the Confounding Factors in the Preclinical Studies. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7380172. [PMID: 31998797 PMCID: PMC6970503 DOI: 10.1155/2020/7380172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/17/2019] [Indexed: 01/30/2023]
Abstract
General anesthetic (GA) is used clinically to millions of young children each year to facilitate surgical procedures, relieve perioperative stress, and provide analgesia and amnesia. During recent years, there is a growing concern regarding a causal association between early life GA exposure and subsequently long-term neurocognitive abnormalities. To address the increasing concern, mounting preclinical studies and clinical trials have been undergoing. Until now, nearly all of the preclinical findings show that neonatal exposure to GA causally leads to acute neural cell injury and delayed cognitive impairment. Unexpectedly, several influential clinical findings suggest that early life GA exposure, especially brief and single exposure, does not cause adverse neurodevelopmental outcome, which is not fully in line with the experimental findings and data from several previous cohort trials. As the clinical data have been critically discussed in previous reviews, in the present review, we try to analyze the potential factors of the experimental studies that may overestimate the adverse effect of GA on the developing brain. Meanwhile, we briefly summarized the advance in experimental research. Generally, our purpose is to provide some useful suggestions for forthcoming preclinical studies and strengthen the powerfulness of preclinical data.
Collapse
Affiliation(s)
- Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Xiaole Tang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Zhiqiang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Jing Yan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| |
Collapse
|
70
|
Abstract
General anesthesia has been unequivocally linked to abnormal development of the central nervous system, leading to neurocognitive impairments in laboratory models. In vitro and in vivo studies have consistently shown that exposure to GABA agonists (eg, volatile anesthetics, midazolam, and propofol) or NMDA antagonists (eg, ketamine, isoflurane, and nitrous oxide) produces dose dependent and developmental age dependent effects on various neuronal transmission systems. Exposure to these drugs increases neuronal cell death in juvenile animals including rats, mice, and non-human primates. The possibility of anesthetic induced neurotoxicity occurring in children has led to concerns about the safety of pediatric anesthesia. A spectrum of behavioral changes has been documented after general anesthetic exposure in young children, including emergence delirium, which may be evidence of toxicity. Most clinical studies are retrospective; specifics about medications or monitoring are unavailable and many of the outcomes may not be sensitive to detect small neurocognitive deficits. Some of these retrospective studies have shown an association between anesthesia exposure at a young age and neurocognitive deficits, but others have not. Practitioners and families should be reassured that although general anesthetics have the potential to induce neurotoxicity, very little clinical evidence exists to support this.
Collapse
Affiliation(s)
- Mary Ellen McCann
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sulpicio G Soriano
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
71
|
Early Postnatal Exposure to Isoflurane Disrupts Oligodendrocyte Development and Myelin Formation in the Mouse Hippocampus. Anesthesiology 2019; 131:1077-1091. [PMID: 31436548 PMCID: PMC6800770 DOI: 10.1097/aln.0000000000002904] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Early postnatal exposure to general anesthetics may interfere with brain development. We tested the hypothesis that isoflurane causes a lasting disruption in myelin development via actions on the mammalian target of rapamycin pathway. METHODS Mice were exposed to 1.5% isoflurane for 4 h at postnatal day 7. The mammalian target of rapamycin inhibitor, rapamycin, or the promyelination drug, clemastine, were administered on days 21 to 35. Mice underwent Y-maze and novel object position recognition tests (n = 12 per group) on days 56 to 62 or were euthanized for either immunohistochemistry (n = 8 per group) or Western blotting (n = 8 per group) at day 35 or were euthanized for electron microscopy at day 63. RESULTS Isoflurane exposure increased the percentage of phospho-S6-positive oligodendrocytes in fimbria of hippocampus from 22 ± 7% to 51 ± 6% (P < 0.0001). In Y-maze testing, isoflurane-exposed mice did not discriminate normally between old and novel arms, spending equal time in both (50 ± 5% old:50 ± 5% novel; P = 0.999), indicating impaired spatial learning. Treatment with clemastine restored discrimination, as evidenced by increased time spent in the novel arm (43 ± 6% old:57 ± 6% novel; P < 0.001), and rapamycin had a similar effect (44 ± 8% old:56 ± 8% novel; P < 0.001). Electron microscopy shows a reduction in myelin thickness as measured by an increase in g-ratio from 0.76 ± 0.06 for controls to 0.79 ± 0.06 for the isoflurane group (P < 0.001). Isoflurane exposure followed by rapamycin treatment resulted in a g-ratio (0.75 ± 0.05) that did not differ significantly from the control value (P = 0.426). Immunohistochemistry and Western blotting show that isoflurane acts on oligodendrocyte precursor cells to inhibit both proliferation and differentiation. DNA methylation and expression of a DNA methyl transferase 1 are reduced in oligodendrocyte precursor cells after isoflurane treatment. Effects of isoflurane on oligodendrocyte precursor cells were abolished by treatment with rapamycin. CONCLUSIONS Early postnatal exposure to isoflurane in mice causes lasting disruptions of oligodendrocyte development in the hippocampus via actions on the mammalian target of rapamycin pathway.
Collapse
|
72
|
Li G, Du J, Wang L, Shi X. Developmental neurotoxicity in the context of multiple sevoflurane exposures: Potential role of histone deacetylase 6. Neurotoxicol Teratol 2019; 74:106813. [PMID: 31251981 DOI: 10.1016/j.ntt.2019.106813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 05/26/2019] [Accepted: 06/24/2019] [Indexed: 11/28/2022]
Abstract
Animal studies have demonstrated that multiple exposures to sevoflurane during the postnatal period lead to impaired synaptogenesis and cognitive deficits in adulthood. However, the underlying mechanisms remain unclear. Histone deacetylase 6 (HDAC6), a unique isoform of class II histone deacetylases (HDACs), mediates diverse cellular processes such as cell survival, inflammation, intracellular trafficking and protein degradation. Varieties of literature suggest the importance of HDAC6 in memory formation and abnormal neurodegenerative diseases. The aim of this study was to investigate potential roles of HDAC6 in sevoflurane-induced developmental neurotoxicity. Postnatal day 7 (P7) rat pups were randomly assigned to control group and sevoflurane group (n = 6 for each group). They were exposed to 60% oxygen and 40% nitrogen with or without 3% sevoflurane for 2 h daily for three consecutive days (P7, P8 and P9). Immediately after the last exposure, both hippocampi were harvested for detection of HDAC6 expression and activity. Next, P7 rat pups were divided into control group, sevoflurane group, sevoflurane + Tubastatin A, and Tubastatin A groups (n = 6 for each group in molecular experiments; n = 16 for each group in behavioral testing). A dose of 25 mg/kg body weight of Tubastatin A (a selective HDAC6 inhibitor) were administrated intraperitoneally 30 min prior to each sevoflurane exposure. After treatments, expression levels of synaptophysin and postsynaptic density 95 protein (PSD95) were quantified using Western blot, and synaptic ultrastructure was evaluated by transmission electron microscopy. Additional pups were raised until P49 to measure cognitive performance using the Morris water maze test. Our results demonstrated that multiple sevoflurane exposures enhanced HDAC6 expression and activity in hippocampi of the developing brain. Tubastatin A ameliorated sevoflurane-induced decreases in synaptophysin and PSD95 expression during development, as well as synaptic ultrastructural damage and cognitive deficits in adulthood. In conclusion, HDAC6 is involved in the developmental neurotoxicity caused by multiple sevoflurane exposures and its inhibition may prevent related damage.
Collapse
Affiliation(s)
- Guohui Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Jian'er Du
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Lai Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Xueyin Shi
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| |
Collapse
|
73
|
Iqbal F, Thompson AJ, Riaz S, Pehar M, Rice T, Syed NI. Anesthetics: from modes of action to unconsciousness and neurotoxicity. J Neurophysiol 2019; 122:760-787. [PMID: 31242059 DOI: 10.1152/jn.00210.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern anesthetic compounds and advanced monitoring tools have revolutionized the field of medicine, allowing for complex surgical procedures to occur safely and effectively. Faster induction times and quicker recovery periods of current anesthetic agents have also helped reduce health care costs significantly. Moreover, extensive research has allowed for a better understanding of anesthetic modes of action, thus facilitating the development of more effective and safer compounds. Notwithstanding the realization that anesthetics are a prerequisite to all surgical procedures, evidence is emerging to support the notion that exposure of the developing brain to certain anesthetics may impact future brain development and function. Whereas the data in support of this postulate from human studies is equivocal, the vast majority of animal research strongly suggests that anesthetics are indeed cytotoxic at multiple brain structure and function levels. In this review, we first highlight various modes of anesthetic action and then debate the evidence of harm from both basic science and clinical studies perspectives. We present evidence from animal and human studies vis-à-vis the possible detrimental effects of anesthetic agents on both the young developing and the elderly aging brain while discussing potential ways to mitigate these effects. We hope that this review will, on the one hand, invoke debate vis-à-vis the evidence of anesthetic harm in young children and the elderly, and on the other hand, incentivize the search for better and less toxic anesthetic compounds.
Collapse
Affiliation(s)
- Fahad Iqbal
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Thompson
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Saba Riaz
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Pehar
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Rice
- Department of Anesthesiology, Perioperative and Pain Medicine, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
74
|
Early Developmental Exposure to Repetitive Long Duration of Midazolam Sedation Causes Behavioral and Synaptic Alterations in a Rodent Model of Neurodevelopment. J Neurosurg Anesthesiol 2019; 31:151-162. [PMID: 30767941 DOI: 10.1097/ana.0000000000000541] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
There is a large body of preclinical literature suggesting that exposure to general anesthetic agents during early life may have harmful effects on brain development. Patients in intensive care settings are often treated for prolonged periods with sedative medications, many of which have mechanisms of action that are similar to general anesthetics. Using in vivo studies of the mouse hippocampus and an in vitro rat cortical neuron model we asked whether there is evidence that repeated, long duration exposure to midazolam, a commonly used sedative in pediatric intensive care practice, has the potential to cause lasting harm to the developing brain. We found that mice that underwent midazolam sedation in early postnatal life exhibited deficits in the performance on Y-maze and fear-conditioning testing at young adult ages. Labeling with a nucleoside analog revealed a reduction in the rate of adult neurogenesis in the hippocampal dentate gyrus, a brain region that has been shown to be vulnerable to developmental anesthetic neurotoxicity. In addition, using immunohistochemistry for synaptic markers we found that the number of presynaptic terminals in the dentate gyrus was reduced, while the number of excitatory postsynaptic terminals was increased. These findings were replicated in a midazolam sedation exposure model in neurons in culture. We conclude that repeated, long duration exposure to midazolam during early development has the potential to result in persistent alterations in the structure and function of the brain.
Collapse
|
75
|
Zhang L, Xue Z, Liu Q, Liu Y, Xi S, Cheng Y, Li J, Yan J, Shen Y, Xiao C, Xie Z, Qiu Z, Jiang H. Disrupted folate metabolism with anesthesia leads to myelination deficits mediated by epigenetic regulation of ERMN. EBioMedicine 2019; 43:473-486. [PMID: 31060905 PMCID: PMC6562069 DOI: 10.1016/j.ebiom.2019.04.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 11/22/2022] Open
Abstract
Background Exposure to anesthetics during early life may impair cognitive functions. However, the underlying mechanisms remain largely unknown. We set out to determine effects of sevoflurane anesthesia on folate metabolism and myelination in young non-human primates, mice and children. Methods Young rhesus macaque and mice received 2.5 to 3% sevoflurane daily for three days. DNA and RNA sequencing and immunohistochemistry among others were used in the studies. We performed unbiased transcriptome profiling in prefrontal cortex of rhesus macaques and mice after the sevoflurane anesthesia. We constructed a brain blood barrier-crossing AAV-PHP.EB vector to harbor ERMN expression in rescue studies. We measured blood folate levels in children after anesthesia and surgery. Findings We found that thymidylate synthase (TYMS) gene was downregulated after the sevoflurane anesthesia in both rhesus macaque and mice. There was a reduction in blood folate levels in children after the anesthesia and surgery. Combined with transcriptome and genome-wide DNA methylation analysis, we identified that ERMN was the primary target of the disrupted folate metabolism. Myelination was compromised by the anesthesia in the young mice, which was rescued by systematic administration of folic acid or expression of ERMN in the brain through brain-specific delivery of the adeno-associated virus. Moreover, folic acid and expression of ERMN alleviated the cognitive impairment caused by the sevoflurane anesthesia in the mice. Interpretation General anesthesia leads to disrupted folate metabolism and subsequently defects in myelination in the developmental brain, and ERMN is the important target affected by the anesthesia via epigenetic mechanisms.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Zhenyu Xue
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Qidong Liu
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Yunbo Liu
- The Institute of Laboratory Animal Science, CAMS & PUMC. Beijing, PR China
| | - Siwei Xi
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Yanyong Cheng
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Jingjie Li
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China
| | - Yuan Shen
- Department of Psychiatry, Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, PR China
| | - Chong Xiao
- The Institute of Laboratory Animal Science, CAMS & PUMC. Beijing, PR China
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | - Zilong Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, PR China.
| |
Collapse
|
76
|
Johnson SC, Pan A, Sun GX, Freed A, Stokes JC, Bornstein R, Witkowski M, Li L, Ford JM, Howard CRA, Sedensky MM, Morgan PG. Relevance of experimental paradigms of anesthesia induced neurotoxicity in the mouse. PLoS One 2019; 14:e0213543. [PMID: 30897103 PMCID: PMC6428290 DOI: 10.1371/journal.pone.0213543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/24/2019] [Indexed: 11/30/2022] Open
Abstract
Routine general anesthesia is considered to be safe in healthy individuals. However, pre-clinical studies in mice, rats, and monkeys have repeatedly demonstrated that exposure to anesthetic agents during early post-natal periods can lead to acute neurotoxicity. More concerning, later-life defects in cognition, assessed by behavioral assays for learning and memory, have been reported. Although the potential for anesthetics to damage the neonatal brain is well-documented, the clinical significance of the pre-clinical models in which damage is induced remains quite unclear. Here, we systematically evaluate critical physiological parameters in post-natal day 7 neonatal mice exposed to 1.5% isoflurane for 2–4 hours, the most common anesthesia induced neurotoxicity paradigm in this animal model. We find that 2 or more hours of anesthesia exposure results in dramatic respiratory and metabolic changes that may limit interpretation of this paradigm to the clinical situation. Our data indicate that neonatal mouse models of AIN are not necessarily appropriate representations of human exposures.
Collapse
Affiliation(s)
- Simon C. Johnson
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Neurology, University of Washington, Seattle, WA, United States of America
- * E-mail:
| | - Amanda Pan
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Grace X. Sun
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Arielle Freed
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- University of Washington School of Dentistry, Seattle, WA, United States of America
| | - Julia C. Stokes
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Rebecca Bornstein
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Pathology, University of Washington, Seattle, WA, United States of America
| | - Michael Witkowski
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Li Li
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Jeremy M. Ford
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Seattle Children's Imagination Lab, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Christopher R. A. Howard
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Seattle Children's Imagination Lab, Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Margaret M. Sedensky
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States of America
| | - Philip G. Morgan
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
77
|
Wu L, Zhao H, Weng H, Ma D. Lasting effects of general anesthetics on the brain in the young and elderly: "mixed picture" of neurotoxicity, neuroprotection and cognitive impairment. J Anesth 2019; 33:321-335. [PMID: 30859366 PMCID: PMC6443620 DOI: 10.1007/s00540-019-02623-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
Abstract
General anesthetics are commonly used in major surgery. To achieve the depth of anesthesia for surgery, patients are being subjected to a variety of general anesthetics, alone or in combination. It has been long held an illusory concept that the general anesthesia is entirely reversible and that the central nervous system is returned to its pristine state once the anesthetic agent is eliminated from the active site. However, studies indicate that perturbation of the normal functioning of these targets may result in long-lasting desirable or undesirable effects. This review focuses on the impact of general anesthetic exposure to the brain and summarizes the molecular and cellular mechanisms by which general anesthetics may induce long-lasting undesirable effects when exposed at the developing stage of the brain. The vulnerability of aging brain to general anesthetics, specifically in the context of cognitive disorders and Alzheimer’s disease pathogeneses are also discussed. Moreover, we will review emerging evidence regarding the neuroprotective property of xenon and anesthetic adjuvant dexmedetomidine in the immature and mature brains. In conclusion, “mixed picture” effects of general anesthetics should be well acknowledged and should be implemented into daily clinical practice for better patient outcome.
Collapse
Affiliation(s)
- Lingzhi Wu
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Hao Weng
- Department of Anesthesiology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Fengxian District, Shanghai, China
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| |
Collapse
|
78
|
Ryan AM, Freeman SM, Murai T, Lau AR, Palumbo MC, Hogrefe CE, Bales KL, Bauman MD. Non-invasive Eye Tracking Methods for New World and Old World Monkeys. Front Behav Neurosci 2019; 13:39. [PMID: 30890923 PMCID: PMC6412371 DOI: 10.3389/fnbeh.2019.00039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
Eye-tracking methods measure what humans and other animals visually attend to in the environment. In nonhuman primates, eye tracking can be used to test hypotheses about how primates process social information. This information can further our understanding of primate behavior as well as offer unique translational potential to explore causes of or treatments for altered social processing as seen in people with neurodevelopmental disorders such as autism spectrum disorder and schizophrenia. However, previous methods for collecting eye-tracking data in nonhuman primates required some form of head restraint, which limits the opportunities for research with respect to the number of or kinds of primates that can undergo an eye-tracking study. We developed a novel, noninvasive method for collecting eye tracking data that can be used both in animals that are difficult to restrain without sedation as well as animals that are of different ages and sizes as the box size can be adjusted. Using a transport box modified with a viewing window, we collected eye-tracking data in both New (Callicebus cupreus) and Old World monkeys (Macaca mulatta) across multiple developmental time points. These monkeys had the option to move around the box and avert their eyes from the screen, yet, they demonstrated a natural interest in viewing species-specific imagery with no previous habituation to the eye-tracking paradigm. Provided with opportunistic data from voluntary viewing of stimuli, we found that juveniles viewed stimuli more than other age groups, videos were viewed more than static photo imagery, and that monkeys increased their viewing time when presented with multiple eye tracking sessions. This noninvasive approach opens new opportunities to integrate eye-tracking studies into nonhuman primate research.
Collapse
Affiliation(s)
- Amy M. Ryan
- The UC Davis MIND Institute, University of California, Davis, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Sara M. Freeman
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Takeshi Murai
- Platform Technology Research Unit, Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Allison R. Lau
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Michelle C. Palumbo
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Casey E. Hogrefe
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Karen L. Bales
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Melissa D. Bauman
- The UC Davis MIND Institute, University of California, Davis, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| |
Collapse
|
79
|
Schaefer ML, Wang M, Perez PJ, Coca Peralta W, Xu J, Johns RA. Nitric Oxide Donor Prevents Neonatal Isoflurane-induced Impairments in Synaptic Plasticity and Memory. Anesthesiology 2019; 130:247-262. [PMID: 30601214 PMCID: PMC6538043 DOI: 10.1097/aln.0000000000002529] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC Some general anesthetics have been shown to have adverse effects on neuronal development that affect neural function and cognitive behavior.Clinically relevant concentrations of inhalational anesthetics inhibit the postsynaptic density (PSD)-95, discs large homolog, and zona occludens-1 (PDZ) domain-mediated protein-protein interaction between PSD-95 or PSD-93 and N-methyl-D-aspartate receptors or neuronal NO synthase. WHAT THIS ARTICLE TELLS US THAT IS NEW Neonatal PSD-95 PDZ2WT peptide treatment mimics the effects of isoflurane (~1 minimum alveolar concentration) by altering dendritic spine morphology, neural plasticity, and memory without inducing detectable increases in apoptosis or changes in synaptic density.These results indicate that a single dose of isoflurane (~1 minimum alveolar concentration) or PSD-95 PDZ2WT peptide alters dendritic spine architecture and functions important for cognition in the developing brain. This impairment can be prevented by administration of the NO donor molsidomine. BACKGROUND In humans, multiple early exposures to procedures requiring anesthesia constitute a significant risk factor for development of learning disabilities and disorders of attention. In animal studies, newborns exposed to anesthetics develop long-term deficits in cognition. Previously, our laboratory showed that postsynaptic density (PSD)-95, discs large homolog, and zona occludens-1 (PDZ) domains may serve as a molecular target for inhaled anesthetics. This study investigated a role for PDZ interactions in spine development, plasticity, and memory as a potential mechanism for early anesthetic exposure-produced cognitive impairment. METHODS Postnatal day 7 mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active PSD-95 PDZ2WT peptide. Apoptosis, hippocampal dendritic spine changes, synapse density, long-term potentiation, and cognition functions were evaluated (n = 4 to 18). RESULTS Exposure of postnatal day 7 mice to isoflurane or PSD-95 PDZ2WT peptide causes a reduction in long thin spines (median, interquartile range [IQR]: wild type control [0.54, 0.52 to 0.86] vs. wild type isoflurane [0.31, 0.16 to 0.38], P = 0.034 and PDZ2MUT [0.86, 0.67 to 1.0] vs. PDZ2WT [0.55, 0.53 to 0.59], P = 0.028), impairment in long-term potentiation (median, IQR: wild type control [123, 119 to 147] and wild type isoflurane [101, 96 to 118], P = 0.049 and PDZ2MUT [125, 119 to 131] and PDZ2WT [104, 97 to 107], P = 0.029), and deficits in acute object recognition (median, IQR: wild type control [79, 72 to 88] vs. wild type isoflurane [63, 55 to 72], P = 0.044 and PDZ2MUT [81, 69 to 84] vs. PDZ2WT [67, 57 to 77], P = 0.039) at postnatal day 21 without inducing detectable differences in apoptosis or changes in synaptic density. Impairments in recognition memory and long-term potentiation were preventable by introduction of a NO donor. CONCLUSIONS Early disruption of PDZ domain-mediated protein-protein interactions alters spine morphology, synaptic function, and memory. These results support a role for PDZ interactions in early anesthetic exposure-produced cognitive impairment. Prevention of recognition memory and long-term potentiation deficits with a NO donor supports a role for the N-methyl-D-aspartate receptor/PSD-95/neuronal NO synthase pathway in mediating these aspects of isoflurane-induced cognitive impairment.
Collapse
Affiliation(s)
- Michele L Schaefer
- From the Department Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
80
|
Neuropsychological and Behavioral Outcomes after Exposure of Young Children to Procedures Requiring General Anesthesia: The Mayo Anesthesia Safety in Kids (MASK) Study. Anesthesiology 2019; 129:89-105. [PMID: 29672337 DOI: 10.1097/aln.0000000000002232] [Citation(s) in RCA: 354] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Few studies of how exposure of children to anesthesia may affect neurodevelopment employ comprehensive neuropsychological assessments. This study tested the hypothesis that exposure to multiple, but not single, procedures requiring anesthesia before age 3 yr is associated with adverse neurodevelopmental outcomes. METHODS Unexposed, singly exposed, and multiply exposed children born in Olmsted County, Minnesota, from 1994 to 2007 were sampled using a propensity-guided approach and underwent neuropsychological testing at ages 8 to 12 or 15 to 20 yr. The primary outcome was the Full-Scale intelligence quotient standard score of the Wechsler Abbreviated Scale of Intelligence. Secondary outcomes included individual domains from a comprehensive neuropsychological assessment and parent reports. RESULTS In total, 997 children completed testing (411, 380, and 206 unexposed, singly exposed, and multiply exposed, respectively). The primary outcome of intelligence quotient did not differ significantly according to exposure status; multiply exposed and singly exposed children scoring 1.3 points (95% CI, -3.8 to 1.2; P = 0.32) and 0.5 points (95% CI, -2.8 to 1.9; P = 0.70) lower than unexposed children, respectively. For secondary outcomes, processing speed and fine motor abilities were decreased in multiply but not singly exposed children; other domains did not differ. The parents of multiply exposed children reported increased problems related to executive function, behavior, and reading. CONCLUSIONS Anesthesia exposure before age 3 yr was not associated with deficits in the primary outcome of general intelligence. Although secondary outcomes must be interpreted cautiously, they suggest the hypothesis that multiple, but not single, exposures are associated with a pattern of changes in specific neuropsychological domains that is associated with behavioral and learning difficulties.
Collapse
|
81
|
Soriano SG, Vutskits L, Jevtovic-Todorovic V, Hemmings HC. Thinking, fast and slow: highlights from the 2016 BJA seminar on anaesthetic neurotoxicity and neuroplasticity. Br J Anaesth 2019; 119:443-447. [PMID: 28969326 DOI: 10.1093/bja/aex238] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- S G Soriano
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - L Vutskits
- Département d'Anesthésiologie, Pharmacologie et Soins Intensifs, Hopitaux Universitaires de Geneve, Rue Willy-Donzé 6, CH-1205 Genève, Switzerland
| | - V Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Denver School of Medicine, 12631 E. 17th Ave. Suite 2001, Aurora, CO 80045, USA
| | - H C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
82
|
|
83
|
Walters JL, Zhang X, Talpos JC, Fogle CM, Li M, Chelonis JJ, Paule MG. Sevoflurane exposure has minimal effect on cognitive function and does not alter microglial activation in adult monkeys. Neurotoxicology 2018; 71:159-167. [PMID: 30605762 DOI: 10.1016/j.neuro.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
Postoperative Cognitive Dysfunction (POCD) is a complication that has been observed in a subset of adult and elderly individuals after general anesthesia and surgery. Although the pathogenesis of POCD is largely unknown, a growing body of preclinical research suggests that POCD may be caused by general anesthesia. A significant amount of research has examined the effects of general anesthesia on neurocognitive function in rodents, yet no studies have assessed the adverse effects of general anesthesia on brain function in adult nonhuman primates. Thus, this study sought to determine the effects of an extended exposure to sevoflurane anesthesia on cognitive function and neural inflammation in adult rhesus macaques. Five adult rhesus macaques (16-17 years of age) were exposed to sevoflurane anesthesia for 8 h and, and micro-positron emission tomography (PET)/computed tomography (CT) imaging and a battery of operant tasks were used to assess the effects of anesthesia exposure on 18F-labeled fluoroethoxybenzyl-N-(4-phenoxypyridin-3-yl) acetamide ([18F]-FEPPA) uptake, a biomarker of microglia activation, and aspects of complex cognitive function. Exposure to sevoflurane anesthesia for 8 h did not increase [18F]-FEPPA uptake in the adult monkey brain. Sevoflurane anesthesia significantly decreased accuracy (mean difference = 22.79) on a learning acquisition task 6 days after exposure [t(3) = 6.92, p = 0.006], but this effect did not persist when measured 1 week and 2 weeks after additional exposures. Further, sevoflurane anesthesia had no impact on performance in 4 additional cognitive tasks. These data suggest that exposure to anesthesia alone may not be sufficient to cause persistent POCD in adult populations.
Collapse
Affiliation(s)
- Jennifer L Walters
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| | - Xuan Zhang
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| | - John C Talpos
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States.
| | - Charles M Fogle
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| | - Mi Li
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| | - John J Chelonis
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| | - Merle G Paule
- National Center for Toxicological Research (NCTR)/FDA, Division of Neurotoxicology, 3900 NCTR Road, Jefferson, AR, 72079, United States
| |
Collapse
|
84
|
Jevtovic-Todorovic V. Anaesthesia-induced developmental neurotoxicity: reality or fiction? Br J Anaesth 2018; 119:455-457. [PMID: 28969321 DOI: 10.1093/bja/aex161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- V Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
85
|
Johnson SC, Pan A, Li L, Sedensky M, Morgan P. Neurotoxicity of anesthetics: Mechanisms and meaning from mouse intervention studies. Neurotoxicol Teratol 2018; 71:22-31. [PMID: 30472095 DOI: 10.1016/j.ntt.2018.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/02/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022]
Abstract
Volatile anesthetics are widely used in human medicine and generally considered to be safe in healthy individuals. In recent years, the safety of volatile anesthesia in pediatric patients has been questioned following reports of anesthetic induced neurotoxicity in pre-clinical studies. These studies in mice, rats, and primates have demonstrated that exposure to anesthetic agents during early post-natal periods can cause acute neurotoxicity, as well as later-life cognitive defects including deficits in learning and memory. In recent years, the focus of many pre-clinical studies has been on identifying candidate pathways or potential therapeutic targets through intervention trials. These reports have shed light on the mechanisms underlying anesthesia induced neurotoxicity as well as highlighting the challenges of pre-clinical modeling of anesthesia induced neurotoxicity in mice. Here, we summarize the data derived from intervention studies in neonatal mouse models of anesthetic exposure and provide an overview of mechanisms proposed to mediate anesthesia induced neurotoxicity in mice based on these reports. The majority of these studies implicate one of three mechanisms: reactive oxygen species (ROS) mediated stress and signaling, growth/nutrient signaling, or direct neuronal modulation.
Collapse
Affiliation(s)
- Simon C Johnson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America.
| | - Amanda Pan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America
| | - Li Li
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America; Department of Anesthesiology, University of Washington, Seattle, WA, United States of America
| | - Margaret Sedensky
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America; Department of Anesthesiology, University of Washington, Seattle, WA, United States of America
| | - Philip Morgan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America; Department of Anesthesiology, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
86
|
Ben-Ari Y. Oxytocin and Vasopressin, and the GABA Developmental Shift During Labor and Birth: Friends or Foes? Front Cell Neurosci 2018; 12:254. [PMID: 30186114 PMCID: PMC6110879 DOI: 10.3389/fncel.2018.00254] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/24/2018] [Indexed: 12/15/2022] Open
Abstract
Oxytocin (OT) and vasopressin (AVP) are usually associated with sociability and reduced stress for the former and antidiuretic agent associated with severe stress and pathological conditions for the latter. Both OT and AVP play major roles during labor and birth. Recent contradictory studies suggest that they might exert different roles on the GABA excitatory/inhibitory developmental shift. We reported (Tyzio et al., 2006) that at birth, OT exerts a neuro-protective action mediated by an abrupt reduction of intracellular chloride levels ([Cl-]i) that are high in utero, reinforcing GABAergic inhibition and modulating the generation of the first synchronized patterns of cortical networks. This reduction of [Cl-]i levels is abolished in rodent models of Fragile X Syndrome and Autism Spectrum Disorders, and its restoration attenuates the severity of the pathological sequels, stressing the importance of the shift at birth (Tyzio et al., 2014). In contrast, Kaila and co-workers (Spoljaric et al., 2017) reported excitatory GABA actions before and after birth that are modulated by AVP but not by OT, challenging both the developmental shift and the roles of OT. Here, I analyze the differences between these studies and suggest that the ratio AVP/OT like that of excitatory/inhibitory GABA depend on stress and pathological conditions.
Collapse
Affiliation(s)
- Yehezkel Ben-Ari
- Neurochlore and Ben-Ari Institute of Neuroarcheology (IBEN), Marseille, France
| |
Collapse
|
87
|
Early Developmental Exposure to General Anesthetic Agents in Primary Neuron Culture Disrupts Synapse Formation via Actions on the mTOR Pathway. Int J Mol Sci 2018; 19:ijms19082183. [PMID: 30049952 PMCID: PMC6121894 DOI: 10.3390/ijms19082183] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 12/05/2022] Open
Abstract
Human epidemiologic studies and laboratory investigations in animal models suggest that exposure to general anesthetic agents (GAs) have harmful effects on brain development. The mechanism underlying this putative iatrogenic condition is not clear and there are currently no accepted strategies for prophylaxis or treatment. Recent evidence suggests that anesthetics might cause persistent deficits in synaptogenesis by disrupting key events in neurodevelopment. Using an in vitro model consisting of dissociated primary cultured mouse neurons, we demonstrate abnormal pre- and post-synaptic marker expression after a clinically-relevant isoflurane anesthesia exposure is conducted during neuron development. We find that pharmacologic inhibition of the mechanistic target of rapamycin (mTOR) pathway can reverse the observed changes. Isoflurane exposure increases expression of phospho-S6, a marker of mTOR pathway activity, in a concentration-dependent fashion and this effect occurs throughout neuronal development. The mTOR 1 complex (mTORC1) and the mTOR 2 complex (mTORC2) branches of the pathway are both activated by isoflurane exposure and this is reversible with branch-specific inhibitors. Upregulation of mTOR is also seen with sevoflurane and propofol exposure, suggesting that this mechanism of developmental anesthetic neurotoxicity may occur with all the commonly used GAs in pediatric practice. We conclude that GAs disrupt the development of neurons during development by activating a well-defined neurodevelopmental disease pathway and that this phenotype can be reversed by pharmacologic inhibition.
Collapse
|
88
|
Lee JR, Loepke AW. Does pediatric anesthesia cause brain damage? - Addressing parental and provider concerns in light of compelling animal studies and seemingly ambivalent human data. Korean J Anesthesiol 2018; 71:255-273. [PMID: 29969889 PMCID: PMC6078876 DOI: 10.4097/kja.d.18.00165] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/04/2018] [Indexed: 02/07/2023] Open
Abstract
Anesthesia facilitates surgery in millions of young children every year. Structural brain abnormalities and functional impairment observed in animals have created substantial concerns among clinicians, parents, and government regulators. Clinical studies seemed ambivalent; it remains unclear whether differential species effects exist towards anesthetic exposure. The current literature search and analysis attempts to unify the available clinical and animal studies, which currently comprise of > 530 in vivo animal studies and > 30 clinical studies. The prevalence of abnormalities was lowest for exposures < 1 hour, in both animals and humans, while studies with injurious findings increased in frequency with exposure time. Importantly, no exposure time, anesthetic technique, or age during exposure was clearly identifiable to be entirely devoid of any adverse outcomes. Moreover, the age dependence of maximum injury clearly identified in animal studies, combined with the heterogeneity in age in most human studies, may impede the discovery of a specific human neurological phenotype. In summary, animal and human research studies identify a growing prevalence of injurious findings with increasing exposure times. However, the existing lack of definitive data regarding safe exposure durations, unaffected ages, and non-injurious anesthetic techniques precludes any evidence-based recommendations for drastically changing current clinical anesthesia management. Animal studies focusing on brain maturational states more applicable to clinical practice, as well as clinical studies focusing on prolonged exposures during distinct developmental windows of vulnerability, are urgently needed to improve the safety of perioperative care for thousands of young children requiring life-saving and quality of life-improving procedures daily.
Collapse
Affiliation(s)
- Jeong-Rim Lee
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Andreas W Loepke
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
89
|
Xu J, Kang E, Mintz CD. Anesthetics disrupt brain development via actions on the mTOR pathway. Commun Integr Biol 2018; 11:1-4. [PMID: 30083288 PMCID: PMC6067894 DOI: 10.1080/19420889.2018.1451719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/08/2018] [Indexed: 11/05/2022] Open
Abstract
Experiments conducted in non-human primates have recently provided new evidence supporting a longstanding concern that exposure to general anesthesia during late intrauterine life or early childhood can cause lasting cognitive deficits through harmful effects on brain development. The mammalian target of rapamycin (mTOR) signaling system plays a key role in both normal brain development and in a wide range of developmental disorders that are characterized by cognitive deficits. Intriguingly, our recently published work shows that anesthetics can chronically alter mTOR signaling in the hippocampal dentate gyrus and that normalization of mTOR signaling can prevent anesthesia-induced perturbation of structure and function. In this addendum, we briefly discuss the putative role of mTOR in developmental anesthetic neurotoxicity.
Collapse
Affiliation(s)
- Jing Xu
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China.,Department of Anesthesiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eunchai Kang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C David Mintz
- Department of Anesthesiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
90
|
Mavigner M, Raper J, Kovacs-Balint Z, Gumber S, O'Neal JT, Bhaumik SK, Zhang X, Habib J, Mattingly C, McDonald CE, Avanzato V, Burke MW, Magnani DM, Bailey VK, Watkins DI, Vanderford TH, Fair D, Earl E, Feczko E, Styner M, Jean SM, Cohen JK, Silvestri G, Johnson RP, O'Connor DH, Wrammert J, Suthar MS, Sanchez MM, Alvarado MC, Chahroudi A. Postnatal Zika virus infection is associated with persistent abnormalities in brain structure, function, and behavior in infant macaques. Sci Transl Med 2018; 10:eaao6975. [PMID: 29618564 PMCID: PMC6186170 DOI: 10.1126/scitranslmed.aao6975] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/07/2018] [Indexed: 12/22/2022]
Abstract
The Zika virus (ZIKV) epidemic is associated with fetal brain lesions and other serious birth defects classified as congenital ZIKV syndrome. Postnatal ZIKV infection in infants and children has been reported; however, data on brain anatomy, function, and behavioral outcomes following infection are absent. We show that postnatal ZIKV infection of infant rhesus macaques (RMs) results in persistent structural and functional alterations of the central nervous system compared to age-matched controls. We demonstrate ZIKV lymphoid tropism and neurotropism in infant RMs and histopathologic abnormalities in the peripheral and central nervous systems including inflammatory infiltrates, astrogliosis, and Wallerian degeneration. Structural and resting-state functional magnetic resonance imaging (MRI/rs-fMRI) show persistent enlargement of lateral ventricles, maturational changes in specific brain regions, and altered functional connectivity (FC) between brain areas involved in emotional behavior and arousal functions, including weakened amygdala-hippocampal connectivity in two of two ZIKV-infected infant RMs several months after clearance of ZIKV RNA from peripheral blood. ZIKV infection also results in distinct alterations in the species-typical emotional reactivity to acute stress, which were predicted by the weak amygdala-hippocampal FC. We demonstrate that postnatal ZIKV infection of infants in this model affects neurodevelopment, suggesting that long-term clinical monitoring of pediatric cases is warranted.
Collapse
Affiliation(s)
- Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jessica Raper
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Zsofia Kovacs-Balint
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sanjeev Gumber
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Siddhartha K Bhaumik
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaodong Zhang
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jakob Habib
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cameron Mattingly
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Victoria Avanzato
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC 20060, USA
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, FL 33146, USA
| | - Thomas H Vanderford
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Damien Fair
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Eric Earl
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Eric Feczko
- Oregon Health and Science University, Portland, OR 97239, USA
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sherrie M Jean
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce K Cohen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - R Paul Johnson
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jens Wrammert
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Vaccine Center, Atlanta, GA 30329, USA
| | - Mar M Sanchez
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria C Alvarado
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA.
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
91
|
Jevtovic-Todorovic V. Monkey business: the importance of mounting behavioural evidence for anaesthesia-induced developmental neurotoxicity. Br J Anaesth 2018; 120:617-619. [DOI: 10.1016/j.bja.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/15/2022] Open
|
92
|
Raper J, De Biasio JC, Murphy KL, Alvarado MC, Baxter MG. Persistent alteration in behavioural reactivity to a mild social stressor in rhesus monkeys repeatedly exposed to sevoflurane in infancy. Br J Anaesth 2018; 120:761-767. [PMID: 29576116 PMCID: PMC6200105 DOI: 10.1016/j.bja.2018.01.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 01/02/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Socio-emotional development is the expression and management of emotions, which in non-human primates can be examined using responses toward increasing levels of threat. Damage to the limbic system alters socio-emotional development in primates. Thus, neuronal and glial cell loss caused by exposure to general anaesthesia early in infancy might also impact socio-emotional development. We recently reported that repeated sevoflurane exposure in the first month of life alters emotional behaviours at 6 months of age and impairs visual recognition memory after the first year of life in rhesus monkeys. The present study evaluated socio-emotional behaviour at 1 and 2 yr of age in those same monkeys to determine the persistence of altered emotional behaviour. METHODS Rhesus monkeys of both sexes were exposed to sevoflurane anaesthesia three times for 4 h each time in the first 6 weeks of life. At 1 and 2 yr of age, they were tested on the human intruder task, a well-established mild acute social stressor. RESULTS Monkeys exposed to sevoflurane as infants exhibited normal fear and hostile responses, but exaggerated self-directed (displacement) behaviours, a general indicator of stress and anxiety in non-human primates. CONCLUSIONS Early repeated sevoflurane exposure in infant non-human primates results in an anxious phenotype that was first detected at 6 months, and persists for at least 2 yr of age. This is the first demonstration of such a prolonged impact of early anaesthesia exposure on emotional reactivity.
Collapse
Affiliation(s)
- J Raper
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA.
| | - J C De Biasio
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K L Murphy
- Comparative Biology Centre, Newcastle University, Newcastle, UK
| | - M C Alvarado
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - M G Baxter
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|