1
|
Yang Y, Hang W, Li J, Liu T, Hu Y, Fang F, Yan D, McQuillan PM, Wang M, Hu Z. Effect of General Anesthetic Agents on Microglia. Aging Dis 2024; 15:1308-1328. [PMID: 37962460 PMCID: PMC11081156 DOI: 10.14336/ad.2023.1108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
The effects of general anesthetic agents (GAAs) on microglia and their potential neurotoxicity have attracted the attention of neuroscientists. Microglia play important roles in the inflammatory process and in neuromodulation of the central nervous system. Microglia-mediated neuroinflammation is a key mechanism of neurocognitive dysfunction during the perioperative period. Microglial activation by GAAs induces anti-inflammatory and pro-inflammatory effects in microglia, suggesting that GAAs play a dual role in the mechanism of postoperative cognitive dysfunction. Understanding of the mechanisms by which GAAs regulate microglia may help to reduce the incidence of postoperative adverse effects. Here, we review the actions of GAAs on microglia and the consequent changes in microglial function. We summarize clinical and animal studies associating microglia with general anesthesia and describe how GAAs interact with neurons via microglia to further explore the mechanisms of action of GAAs in the nervous system.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China.
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA.
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Patrick M. McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Center, Penn State College of Medicine, Hershey, PA, USA.
| | - Mi Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Borzage MT, Peterson BS. A Scoping Review of the Mechanisms Underlying Developmental Anesthetic Neurotoxicity. Anesth Analg 2024:00000539-990000000-00807. [PMID: 38536739 PMCID: PMC11427602 DOI: 10.1213/ane.0000000000006897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Although anesthesia makes painful or uncomfortable diagnostic and interventional health care procedures tolerable, it may also disrupt key cellular processes in neurons and glia, harm the developing brain, and thereby impair cognition and behavior in children. Many years of studies using in vitro, animal behavioral, retrospective database studies in humans, and several prospective clinical trials in humans have been invaluable in discerning the potential toxicity of anesthetics. The objective of this scoping review was to synthetize the evidence from preclinical studies for various mechanisms of toxicity across diverse experimental designs and relate their findings to those of recent clinical trials in real-world settings.
Collapse
Affiliation(s)
- Matthew Thomas Borzage
- From the Fetal and Neonatal Institute, Division of Neonatology, Children's Hospital Los Angeles, Los Angeles, California
| | - Bradley S Peterson
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, California
- Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, California
| |
Collapse
|
3
|
Xiao W, Chen S, Chen J, Huang J. Dexmedetomidine alleviates propofol-induced neural injury in developing rats. Int J Dev Neurosci 2023; 83:631-640. [PMID: 37550504 DOI: 10.1002/jdn.10291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/01/2023] [Accepted: 07/14/2023] [Indexed: 08/09/2023] Open
Abstract
Propofol, a commonly used intravenous anesthetic, has been associated with neurodegeneration in the developing brain upon repeated exposure. Dexmedetomidine is an α2 adrenoceptor agonist that was previously reported to possess neuroprotective properties. Here, we confirmed the impacts of dexmedetomidine on propofol-induced neuroapoptosis and subsequent spatial learning and memory deficits in neonatal rats. We found that dexmedetomidine effectively mitigated propofol-induced spatial learning and memory impairments and improved aversive memory in developing rats. Dexmedetomidine reduced propofol-induced cell apoptosis in the hippocampus and modulated the mRNA expression of Bcl-2 and Bax. Additionally, dexmedetomidine attenuated the propofol-induced increase of inflammatory factors IL-6 and TNF-α. The reduced phosphorylation levels of Akt and CREB levels by propofol were re-activated by dexmedetomidine. In conclusion, our findings demonstrated that dexmedetomidine effectively mitigated propofol-induced cognitive and memory impairments in developing rats by modulating apoptosis and reducing inflammation via activating the Akt/CREB/BDNF signaling pathway. These findings suggest potential strategies to protect the developing brain from the adverse effects of anesthetics and improve patient care in pediatric anesthesia practice.
Collapse
Affiliation(s)
- Weiren Xiao
- Department of Anesthesiology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shouren Chen
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Jinzhuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jianzhong Huang
- Department of Anesthesiology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| |
Collapse
|
4
|
Liu J, Lin D, Yau A, Cottrell JE, Kass IS. Early-life propofol exposure does not affect later-life GABAergic inhibition, seizure induction, or social behavior. IBRO Neurosci Rep 2023; 14:483-493. [PMID: 37252630 PMCID: PMC10220478 DOI: 10.1016/j.ibneur.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/13/2023] [Indexed: 05/31/2023] Open
Abstract
The early developing brain is especially vulnerable to anesthesia, which can result in long lasting functional changes. We examined the effects of early-life propofol on adult excitatory-inhibitory balance and behavior. Postnatal day 7 male mice were exposed to propofol (250 mg/kg i.p.) and anesthesia was maintained for 2 h; control mice were given the same volume of isotonic saline and treated identically. The behavior and electrophysiology experiments were conducted when the mice were adults. We found that a 2-h neonatal propofol exposure did not significantly reduce paired pulse inhibition, alter the effect of muscimol (3 µM) to inhibit field excitatory postsynaptic potentials or alter the effect of bicuculline (100 µM) to increase the population spike in the CA1 region of hippocampal slices from adult mice. Neonatal propofol did not alter the evoked seizure response to pentylenetetrazol in adult mice. Neonatal propofol did not affect anxiety, as measured in the open field apparatus, depression-like behavior, as measured by the forced swim test, or social interactions with novel mice, in either the three-chamber or reciprocal social tests. These results were different from those with neonatal sevoflurane which demonstrated reduced adult GABAergic inhibition, increased seizure susceptibility and reduced social interaction. Even though sevoflurane and propofol both prominently enhance GABA inhibition, they have unique properties that alter the long-term effects of early-life exposure. These results indicate that clinical studies grouping several general anesthetic agents in a single group should be interpreted with great caution when examining long-term effects.
Collapse
Affiliation(s)
- Jinyang Liu
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Daisy Lin
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Alice Yau
- State University of New York Downstate Health Sciences University College of Medicine, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - James E. Cottrell
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Ira S. Kass
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| |
Collapse
|
5
|
Chen J, Xiao F, Chen L, Zhou Z, Wei Y, Zhong Y, Li L, Xie Y. Role of ferroptosis in hypoxic preconditioning to reduce propofol neurotoxicity. Front Pharmacol 2023; 14:1121280. [PMID: 36817119 PMCID: PMC9932196 DOI: 10.3389/fphar.2023.1121280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Background: An increasing number of studies have reported that neurotoxicity of propofol may cause long-term learning and cognitive dysfunction. Hypoxic preconditioning has been shown to have neuroprotective effects, reducing the neurotoxicity of propofol. Ferroptosis is a new form of death that is different from apoptosis, necrosis, autophagy and pyroptosis. However, it is unclear whether hypoxic preconditioning reduces propofol neurotoxicity associated with ferroptosis. Thus, we aimed to evaluate the effect of propofol on primary hippocampal neurons in vitro to investigate the neuroprotective mechanism of hypoxic preconditioning and the role of ferroptosis in the reduction of propofol neurotoxicity by hypoxic preconditioning. Methods: Primary hippocampal neurons were cultured for 8 days in vitro and pretreated with or without propofol, hypoxic preconditioning, agonists or inhibitors of ferroptosis. Cell counting kit-8, Calcein AM, Reactive oxygen species (ROS), Superoxide dismutase (SOD), Ferrous iron (Fe2+), Malondialdehyde (MDA) and Mitochondrial membrane potential assay kit with JC-1 (JC-1) assays were used to measure cell viability, Reactive oxygen species level, Superoxide dismutase content, Fe2+ level, MDA content, and mitochondrial membrane potential. Cell apoptosis was evaluated using flow cytometry analyses, and ferroptosis-related proteins were determined by Western blot analysis. Results: Propofol had neurotoxic effects that led to decreased hippocampal neuronal viability, reduced mitochondrial membrane potential, decreased SOD content, increased ROS level, increased Fe2+ level, increased MDA content, increased neuronal apoptosis, altered expression of ferroptosis-related proteins and activation of ferroptosis. However, hypoxic preconditioning reversed these effects, inhibited ferroptosis caused by propofol and reduced the neurotoxicity of propofol. Conclusion: The neurotoxicity of propofol in developing rats may be related to ferroptosis. Propofol may induce neurotoxicity by activating ferroptosis, while hypoxic preconditioning may reduce the neurotoxicity of propofol by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fei Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lifei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhan Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi Wei
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Li
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,*Correspondence: Yubo Xie, ; Li Li,
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China,*Correspondence: Yubo Xie, ; Li Li,
| |
Collapse
|
6
|
Zhang M, Yin Y. Dual roles of anesthetics in postoperative cognitive dysfunction: Regulation of microglial activation through inflammatory signaling pathways. Front Immunol 2023; 14:1102312. [PMID: 36776829 PMCID: PMC9911670 DOI: 10.3389/fimmu.2023.1102312] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent clinical entity following surgery and is characterized by declined neurocognitive function. Neuroinflammation mediated by microglia is the essential mechanism of POCD. Anesthetics are thought to be a major contributor to the development of POCD, as they promote microglial activation and induce neuroinflammation. However, this claim remains controversial. Anesthetics can exert both anti- and pro-inflammatory effects by modulating microglial activation, suggesting that anesthetics may play dual roles in the pathogenesis of POCD. Here, we review the mechanisms by which the commonly used anesthetics regulate microglial activation via inflammatory signaling pathways, showing both anti- and pro-inflammatory properties of anesthetics, and indicating how perioperative administration of anesthetics might either relieve or worsen POCD development. The potential for anesthetics to enhance cognitive performance based on their anti-inflammatory properties is further discussed, emphasizing that the beneficial effects of anesthetics vary depending on dose, exposure time, and patients' characteristics. To minimize the incidence of POCD, we recommend considering these factors to select appropriate anesthetics.
Collapse
Affiliation(s)
- Mengxue Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
7
|
Chen B, Qin G, Xiao J, Deng X, Lin A, Liu H. Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor. J Neuroinflammation 2022; 19:181. [PMID: 35831873 PMCID: PMC9281167 DOI: 10.1186/s12974-022-02528-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/12/2022] [Indexed: 11/27/2022] Open
Abstract
Background Perioperative neurocognitive disorders (PNDs) are considered the most common postoperative complication in geriatric patients. However, its pathogenesis is not fully understood. Surgery-triggered neuroinflammation is a major contributor to the development of PNDs. Neuroinflammation can influence N-methyl-D-aspartate receptor (NMDAR) expression or function which is closely associated with cognition. We, therefore, hypothesized that the persistent changes in NMDAR expression or function induced by transient neuroinflammation after surgery were involved in the development of PNDs. Methods Eighteen-month-old male Sprague–Dawley rats were subjected to abdominal surgery with sevoflurane anesthesia to establish the PNDs animal model. Then, we determined the transient neuroinflammation by detecting the protein levels of proinflammatory cytokines and microglia activation using ELISA, western blot, immunohistochemistry, and microglial morphological analysis from postoperative days 1–20. Persistent changes in NMDAR expression were determined by detecting the protein levels of NMDAR subunits from postoperative days 1–59. Subsequently, the dysfunction of synaptic NMDAR was evaluated by detecting the structural plasticity of dendritic spine using Golgi staining. Pull-down assay and western blot were used to detect the protein levels of Rac1-GTP, phosphor-cofilin, and Arp3, which contribute to the regulation of the structural plasticity of dendritic spine. Finally, glycyrrhizin, an anti-inflammatory agent, was administered to further explore the role of synaptic NMDAR dysfunction induced by transient neuroinflammation in the neuropathogenesis of PNDs. Results We showed that transient neuroinflammation induced by surgery caused sustained downregulation of synaptic NR2A and NR2B subunits in the dorsal hippocampus and led to a selective long-term spatial memory deficit. Meanwhile, the detrimental effect of neuroinflammation on the function of synaptic NMDARs was shown by the impaired structural plasticity of dendritic spines and decreased activity of the Rac1 signaling pathways during learning. Furthermore, anti-inflammatory treatment reversed the downregulation and hypofunction of synaptic NR2A and NR2B and subsequently rescued the long-term spatial memory deficit. Conclusions Our results identify sustained synaptic NR2A and NR2B downregulation and hypofunction induced by transient neuroinflammation following surgery as important contributors to the development of PNDs in elderly rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02528-5.
Collapse
Affiliation(s)
- Bo Chen
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Guangcheng Qin
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jingyu Xiao
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Xiaoyuan Deng
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Aolei Lin
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
8
|
Platholi J, Hemmings HC. Effects of general anesthetics on synaptic transmission and plasticity. Curr Neuropharmacol 2021; 20:27-54. [PMID: 34344292 PMCID: PMC9199550 DOI: 10.2174/1570159x19666210803105232] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
General anesthetics depress excitatory and/or enhance inhibitory synaptic transmission principally by modulating the function of glutamatergic or GABAergic synapses, respectively, with relative anesthetic agent-specific mechanisms. Synaptic signaling proteins, including ligand- and voltage-gated ion channels, are targeted by general anesthetics to modulate various synaptic mechanisms, including presynaptic neurotransmitter release, postsynaptic receptor signaling, and dendritic spine dynamics to produce their characteristic acute neurophysiological effects. As synaptic structure and plasticity mediate higher-order functions such as learning and memory, long-term synaptic dysfunction following anesthesia may lead to undesirable neurocognitive consequences depending on the specific anesthetic agent and the vulnerability of the population. Here we review the cellular and molecular mechanisms of transient and persistent general anesthetic alterations of synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Jimcy Platholi
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| | - Hugh C Hemmings
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| |
Collapse
|
9
|
Postconditioning with Sevoflurane or Propofol Alleviates Lipopolysaccharide-Induced Neuroinflammation but Exerts Dissimilar Effects on the NR2B Subunit and Cognition. Mol Neurobiol 2021; 58:4251-4267. [PMID: 33970453 DOI: 10.1007/s12035-021-02402-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023]
Abstract
Neuroinflammation can cause cognitive deficits, and preexisting neuroinflammation is observed frequently in the clinic after trauma, surgery, and infection. Patients with preexisting neuroinflammation often need further medical treatment under general anesthesia. However, the effects of postconditioning with general anesthetics on preexisting neuroinflammation have not been determined. In this study, adult rats were posttreated with sevoflurane or propofol after intracerebroventricular administration of lipopolysaccharide. The effects of sevoflurane or propofol postconditioning on neuroinflammation-induced recognition memory deficits were detected. Our results found that postconditioning with sevoflurane but not propofol reversed the selective spatial recognition memory impairment induced by neuroinflammation, and these differential effects did not appear to be associated with the similar anti-neuroinflammatory responses of general anesthetics. However, postconditioning with propofol induced a selective long-lasting upregulation of extrasynaptic NR2B-containing N-methyl-D-aspartate receptors in the dorsal hippocampus, which downregulated the cAMP response element-binding signaling pathway and impaired spatial recognition memory. Additionally, the NR2B antagonists memantine and Ro25-6981 reversed this neurotoxicity induced by propofol postconditioning. Taken together, these results indicate that under preexisting neuroinflammation, postconditioning with sevoflurane can provide reliable neuroprotection by attenuating lipopolysaccharide-induced neuroinflammation, apoptosis, and neuronal loss and eventually improving spatial recognition deficits. However, although posttreatment with propofol also has the same anti-neuroinflammatory effects, the neurotoxicity caused by propofol postconditioning following neuroinflammation warrants further consideration.
Collapse
|
10
|
Wan J, Shen CM, Wang Y, Wu QZ, Wang YL, Liu Q, Sun YM, Cao JP, Wu YQ. Repeated exposure to propofol in the neonatal period impairs hippocampal synaptic plasticity and the recognition function of rats in adulthood. Brain Res Bull 2021; 169:63-72. [PMID: 33450329 DOI: 10.1016/j.brainresbull.2021.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/09/2022]
Abstract
Anesthesia of neonates with propofol induces persistent behavioral abnormalities in adulthood. Although propofol-triggered apoptosis of neurons in the developing brain may contribute to the development of cognitive deficits, the mechanism of neurotoxicity induced by neonatal exposure to propofol remains unclear. In this study, the effects of neonatal propofol anesthesia on synaptic plasticity and neurocognitive function were investigated. Postnatal day 7 (PND-7) Sprague-Dawley rats were intraperitoneally injected with fat emulsion or 20, 40 or 60 mg/kg propofol for three consecutive days. The expression of brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB) and postsynaptic density protein 95 (PSD-95) in the rat hippocampus at PND-10 and PND-12 was measured by Western blotting. The number of dendritic branches, total dendritic length and dendritic spine density were observed by Golgi-Cox staining 24 h and 72 h after the last propofol administration. Long-term potentiation (LTP) was measured electrophysiologically in hippocampus of PND-60 rats to evaluate the synaptic function. The learning and memory abilities of rats were evaluated by Morris water maze (MWM) experiments, Novel object recognition test (NORT) and Object location test (OLT) at PND-60. Our results showed that neonatal exposure to propofol significantly inhibited the expression of BDNF, TrkB and PSD-95 in the rat hippocampus. The number of dendritic branches, total dendritic length and dendritic spine density of neurons in the rat hippocampus were markedly reduced after neonatal propofol anesthesia. LTP was significantly diminished in hippocampus of PND-60 rats after repeated exposure to propofol in the neonatal period. Morris water maze experiments showed that repeated neonatal exposure to propofol significantly prolonged the escape latency and decreased the time spent in the target quadrant and the number of platform crossings. NORT and OLT showed that repeated neonatal exposure to propofol markedly reduced the Investigation Time for novel object or location. All of the results above indicate that repeated exposure to propofol in the neonatal period can impair hippocampal synaptic plasticity and the recognition function of rats in adulthood.
Collapse
Affiliation(s)
- Jie Wan
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Chu-Meng Shen
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Yu Wang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Qing-Zi Wu
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Yi-Lei Wang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Yi-Man Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Jun-Ping Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, PR China.
| |
Collapse
|
11
|
Hypermethylation of EFEMP1 in the Hippocampus May Be Related to the Deficit in Spatial Memory of Rat Neonates Triggered by Repeated Administration of Propofol. BIOMED RESEARCH INTERNATIONAL 2021; 2020:8851480. [PMID: 33381589 PMCID: PMC7765714 DOI: 10.1155/2020/8851480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/22/2020] [Accepted: 11/30/2020] [Indexed: 11/17/2022]
Abstract
It has been confirmed that repeated application of propofol, as an intravenous and short-fast-acting anesthetic, in neonatal animals or humans may produce long-term deficits in cognitive functions. With the aim of explaining the neurotoxic effects of repeated administration of propofol on neonatal rat pups from P7 to P9 especially from an epigenetic perspective, the present study used the Morris water maze to detect cognitive deficits in spatial learning and memory, Sequenom methylation on the CpG island located in the promoter region of epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1) to assess the methylation level of this region, and Western blot to measure the expression of EFEMP1, TIMP-3, and MMP-9. As the results have shown, repeated propofol administration on neonatal rats caused significant systemic growth retardation, impairment of spatial learning and memory, and hypermethylation of the CpG sites in the promoter region of EFEMP1 accompanied by lower expression of EFEMP1 and TIMP-3 and enhanced expression of MMP-9. These data suggest that repeated propofol administration in neonatal rats may generate hypermethylation in the promoter region of EFEMP1 which results in downregulation of the expression of EFEMP1 and tissue inhibitor of metalloproteinase-3 (TIMP-3) but upregulation of the expression of matrix metalloproteinase-9 (MMP-9), which together may affect the stability of ECM to hamper the development of the central nervous system and therefore lead to deficits in cognitive functions.
Collapse
|
12
|
Feng L, Sun ZG, Liu QW, Ma T, Xu ZP, Feng ZG, Yuan WX, Zhang H, Xu LH. Propofol inhibits the expression of Abelson nonreceptor tyrosine kinase without affecting learning or memory function in neonatal rats. Brain Behav 2020; 10:e01810. [PMID: 32869521 PMCID: PMC7667295 DOI: 10.1002/brb3.1810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/17/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Propofol is one of the most commonly used intravenous drugs to induce and maintain general anesthesia. In vivo and in vitro studies have shown that propofol can affect neuronal growth, leading to apoptosis and impairing cognitive function. The Abelson nonreceptor tyrosine kinase (c-Abl) is associated with both neuritic plaques and neurofibrillary tangles in the brains of patients with Alzheimer's disease and other neurodegenerative diseases. This study aimed to explore the effect of propofol on apoptosis and neurocognition through its regulation of c-Abl expression in vivo and in vitro. MATERIALS AND METHODS In this study, primary hippocampal neurons were cultured and exposed to propofol at different concentrations. Protein expression was measured by Western blotting and coimmunoprecipitation. The c-Abl transcription level was verified by fluorescence quantitative PCR. Reactive oxygen species (ROS) levels were detected by flow cytometry. In addition, an animal experiment was conducted to assess neuronal apoptosis by immunofluorescence staining for caspase-3 and to evaluate behavioral changes by the Morris water maze (MWM) test. RESULTS The in vitro experiment showed that propofol significantly decreased c-Abl expression and ROS levels. In addition, propofol has no cytotoxic effect and does not affect cell activity. Moreover, in the animal experiment, intraperitoneal injection of 50 mg/kg propofol for 5 days obviously decreased the expression of c-Abl in the neonatal rat brain (p < .05) but did not significantly increase the number of caspase-3-positive cells. Propofol treatment did not significantly reduce the number of platform crossings (p > .05) or prolong the escape latency of neonatal rats (p > .05) in the MWM test. CONCLUSIONS The present data suggest that reduced expression of this nonreceptor tyrosine kinase through consecutive daily administration of propofol did not impair learning or memory function in neonatal rats.
Collapse
Affiliation(s)
- Long Feng
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China.,PLA general hospital of Hainan Hospital, Hainan, China
| | - Zhi-Gao Sun
- PLA general hospital of Hainan Hospital, Hainan, China
| | - Qiang-Wei Liu
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Tao Ma
- Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhi-Peng Xu
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Ze-Guo Feng
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Wei-Xiu Yuan
- PLA general hospital of Hainan Hospital, Hainan, China
| | - Hong Zhang
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Long-He Xu
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China.,PLA general hospital of Hainan Hospital, Hainan, China
| |
Collapse
|
13
|
Zhu X, Li H, Tian M, Zhou S, He Y, Zhou M. miR-455-3p alleviates propofol-induced neurotoxicity by reducing EphA4 expression in developing neurons. Biomarkers 2020; 25:685-692. [PMID: 33032457 DOI: 10.1080/1354750x.2020.1832147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Propofol, an aesthetic agent in paediatric patients, results in neurotoxicity in the developing neurons. To reduce side effects of propofol, the protective role of miR-455-3p (microRNA-455-3p) in developing rat brain was investigated. MATERIALS AND METHODS Primary hippocampal neurons were isolated from postnatal day 1 or 2 SD (Sprague-Dawley) rats. The neurons were exposed to various concentrations of propofol (0, 10, 30, or 50 μM) for 6 h. Propofol-induced cell viability was assessed by MTT assay, expression levels of miR-455-3p and EphA4 (erythropoietin-producing hepatocellular A4) in propofol-induced neurons were determined using qRT-PCR and western blot, respectively. Binding ability between miR-455-3p and EphA4 was predicted, and then validated by luciferase reporter assay. Neurons expressing miR-455-3p mimics, were treated with 50 μM propofol for 6 h, and apoptosis status was evaluated by flow cytometry. RESULTS Exposure to propofol significantly decreased cell viability of developing neurons isolated from neonatal rats. Propofol decreased miR-455-3p expression, while increased EphA4 level in the neurons. miR-455-3p mimics increased propofol-induced reduce in cell viability, and attenuated propofol-induced cell apoptosis of neurons. MiR-455-3p could target EphA4, and decreased expression of EphA4 in neurons exposure to propofol. EphA4 knockdown counteracted with the promotive effects of propofol on cell viability and apoptosis of neurons. CONCLUSION Propofol treatment induces neurotoxicity and suppresses miR-455-3p levels in the developing hippocampal neurons. However, miR-455-3p could alleviate such neurotoxicity by reducing EphA4 expression, provided new insights into miR-455-3p as novel therapeutic target to prevent propofol-induced damages from bench to clinic.
Collapse
Affiliation(s)
- Xiaojuan Zhu
- Department of Anesthesiology, The First People's Hospital of Kashi, Kashgar City, China
| | - Huifang Li
- Department of Anesthesiology, The First People's Hospital of Kashi, Kashgar City, China
| | - Ming Tian
- Department of Anesthesiology, The First People's Hospital of Kashi, Kashgar City, China
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Yuqin He
- Department of Anesthesiology, The First People's Hospital of Kashi, Kashgar City, China
| | - Ming Zhou
- Department of Anesthesiology, The First People's Hospital of Kashi, Kashgar City, China
| |
Collapse
|
14
|
Clausen NG, Hansen TG, Disma N. Anesthesia Neurotoxicity in the Developing Brain: Basic Studies Relevant for Neonatal or Perinatal Medicine. Clin Perinatol 2019; 46:647-656. [PMID: 31653300 DOI: 10.1016/j.clp.2019.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Diagnostic and invasive procedures in premature infants may require general anesthesia. General anesthetics interfere with the development of the immature animal brain. Accelerated apoptosis, disturbed synaptogenesis, and cytoarchitecture are among the mechanisms suspected to underlie this phenomenon. The implications for humans are unknown. This article presents current suspected mechanisms of anesthesia-induced neurotoxicity and elaborates on the difficulties in translating results from animal research to human. Ethical considerations limit the conduct of such experiments in human neonates, but the use of animal models is still considered feasible. Vulnerable periods in brain development need further identification as do neurotoxic and neuroprotective interventions.
Collapse
Affiliation(s)
- Nicola Groes Clausen
- Department of Anesthesiology and Intensive Care, Odense University Hospital, J.B. Winsløwsvej 4, Odense C 5000, Denmark
| | - Tom G Hansen
- Department of Anesthesiology and Intensive Care - Pediatrics, Odense University Hospital, J.B. Winsløwsvej 4, Odense C 5000, Denmark; Department of Clinical Research - Anesthesiology, University of Southern Denmark, Odense C 5000, Denmark
| | - Nicola Disma
- Department of Anesthesia, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, Genoa 16100, Italy.
| |
Collapse
|
15
|
Neonatal exposure to the experimental environment or ketamine can induce long-term learning dysfunction or overmyelination in female but not male rats. Neuroreport 2019; 30:491-497. [PMID: 30920435 PMCID: PMC6493779 DOI: 10.1097/wnr.0000000000001228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ketamine can induce neurotoxicity after exposures to the developing brain. To investigate whether ketamine at subanesthetic dosage or its environmental condition can cause long-term cognitive dysfunction after multiple exposures in male or female neonatal rats, postnatal day 5 (P5)-day-old Sprague-Dawley rats were randomized into three groups: ketamine group, vehicle group, and control group (no disturbance). Learning and memory abilities from P60 to P65 and immunofluorescence tests for myelin basic protein (MBP) in gray matter on P65 were conducted. The results showed that in female rats, the path length on day 1 in ketamine group and on days 1 and 2 in vehicle group was longer than that in control (P<0.05), but there was no difference between ketamine and vehicle groups (P>0.05). The mean density of MBP in the medial prefrontal cortex (mPFC) was significantly increased in vehicle and ketamine groups compared with that in control (P<0.05), and there was a significant difference between vehicle and ketamine groups (P<0.05), but MBP density was not changed in CA1 or CA3 region (P>0.05). In male rats, there were no significant differences in path length among the groups, and the density of MBP in the mPFC and hippocampus in vehicle or ketamine group was not different from that in control (P>0.05). Pearson’s correlation analysis showed that there was a positive correlation between MBP density in the mPFC and path length in adult female rats (r=0.753, P<0.01). Overall, the results suggested that neonatal female rats exposed to multiple episodes of the experimental environment can develop learning dysfunction in adulthood, which may result from overmyelination in the mPFC, but male rats were not affected. Ketamine could increase myelination in the mPFC in female rats, but it did not induce learning dysfunction in adulthood; therefore, ketamine may be a safe drug for pediatric anesthesia.
Collapse
|
16
|
Guo X, Cheng M, Ke W, Wang Y, Ji X. MicroRNA‑214 suppresses propofol‑induced neuroapoptosis through activation of phosphoinositide 3‑kinase/protein kinase B signaling by targeting phosphatase and tensin homolog expression. Int J Mol Med 2018; 42:2527-2537. [PMID: 30106086 PMCID: PMC6193586 DOI: 10.3892/ijmm.2018.3814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/31/2018] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to investigate the effects of microRNA (miR)‑214 on neuroapoptosis induced by propofol and the possible mechanism of its anti‑apoptotic effects. Initially, it was observed that miR‑214 expression was upregulated in propofol‑induced neuroapoptosis rats. Next, propofol‑treated nerve cells were transfected with miR‑214 mimics. The results revealed that miR‑214 overexpression induced apoptosis, inhibited cell proliferation, inhibited cyclin D1 protein expression, promoted caspase‑3 activity and B‑cell lymphoma 2‑associated X protein expression, and enhanced the levels of inflammation factors in nerve cells treated with propofol. In addition, miR‑214 overexpression suppressed phosphoinositide 3‑kinase/protein kinase B (PI3K/Akt) signaling by targeting the activation of phosphatase and tensin homolog (PTEN) and nuclear factor‑κB expression in nerve cells treated with propofol. Treatment with a PTEN inhibitor successfully suppressed the PTEN protein expression and decreased the apoptosis of propofol‑treated nerve cells subsequent to miR‑214 overexpression through PI3K/Akt signaling. In conclusion, the present study data revealed that miR‑214 suppressed propofol‑induced neuroapoptosis through the activation of PI3K/Akt signaling by targeting PTEN expression.
Collapse
Affiliation(s)
- Xukeng Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Minghua Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Weiqi Ke
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yuting Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xuan Ji
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
17
|
Signaling network between the dysregulated expression of microRNAs and mRNAs in propofol-induced developmental neurotoxicity in mice. Sci Rep 2018; 8:14172. [PMID: 30242182 PMCID: PMC6155049 DOI: 10.1038/s41598-018-32474-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
Mounting evidence has demonstrated that general anesthetics could induce acute neuroapoptosis in developing animals followed by long-term cognitive dysfunction, with the mechanisms remaining largely unknown. The aim of this study was to investigate the effect of the intravenous anesthetic propofol on the profiles of microRNAs (miRNAs) and messenger RNAs (mRNAs), and their interactive signaling networks in the developing mouse hippocampus. Postnatal day 7 (P7) mice were exposed to propofol for 3 hours. Hippocampi were harvested from both P7 (3 hours after exposure) and P60 mice for the analysis of the expression of 726 miRNAs and 24,881 mRNAs, and apoptosis. Long-term memory ability of P60 mice was analyzed using the Morris Water Maze. Propofol induced acute apoptosis in the hippocampus, and impaired memory function of mice. There were 100 altered mRNAs and 18 dysregulated miRNAs in the propofol-treated hippocampi compared with the intralipid-treated control tissues on P7. Bioinformatics analysis of these abnormally expressed genes on P7 indicated that 34 dysregulated miRNA-mRNA target pairs were related to pathological neurological and developmental disorder processes such as cell viability, cell morphology and migration, neural stem cell proliferation and neurogenesis, oligodendrocyte myelination, reactive oxygen species, and calcium signaling. Neonatal propofol exposure also resulted in the abnormal expression of 49 mRNAs and 4 miRNAs in P60 mouse hippocampi. Specifically, bioinformatics analysis indicates that among these dysregulated mRNAs and miRNAs, there were 2 dysregulated miRNA-mRNA targets pairs (Fam46a/miR-363-3p and Rgs3/miR-363-3p) that might be related to the effect of propofol on long-term cognitive function. Collectively, our novel investigation indicates that acute and long-term dysregulated miRNA-mRNA signaling networks potentially participate in propofol-induced developmental neurotoxicity.
Collapse
|
18
|
Li Y, Liu Y, Fan J, Zhou Q, Song X, Peng Z, Qin Z, Tao T. Validation and bioinformatic analysis of propofol-induced differentially expressed microRNAs in primary cultured neural stem cells. Gene 2018; 664:90-100. [PMID: 29679758 DOI: 10.1016/j.gene.2018.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/16/2018] [Indexed: 11/27/2022]
Abstract
Propofol, a widely used intravenous anesthetic, was previously considered as a neuroprotective agent. Recently, however, accumulating evidence suggests that it may cause neurotoxicity, especially in the development of neural stem cells (NSCs). The potential mechanisms contributing to propofol-induced neurotoxicity during neurogenesis, such as those involving microRNAs (miRNAs), are still unknown. In this study, a total of 27 differentially expressed miRNAs were identified in our initial screen and 6 miRNAs were validated by qRT-PCR. Three miRNAs were up-regulated (miR-377-5p, miR-194-3p and miR-143-5p), and three were down-regulated (miR-3583-3p, miR-466b-5p and miR-410-5p). Following gene ontology and KEGG pathway enrichment analysis, Gabbr1, Canca1b and Gabbr2, which are enriched in the GABAergic synapse pathway, were selected as genes potentially playing a role in propofol-induced neurotoxicity. Gabbr1 and Cacna1b, which are targeted by miRNAs that are up-regulated following propofol exposure, showed decreased expression at the mRNA and protein levels. Gabbr2, targeted by miRNAs that were down-regulated following treatment with propofol, was up-regulated at both the levels of mRNA and protein expression. The two clusters of miRNAs that show differential expression following propofol exposure may act in a synergistic manner to regulate several genes simultaneously during the development of NSCs. Our results may contribute to clarify the molecular mechanism and provide potential therapeutic targets for propofol induced neurotoxicity.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jun Fan
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangzhou, China
| | - Quan Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuling Song
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyong Peng
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Tao Tao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
19
|
Zhou J, Wang F, Zhang J, Li J, Ma L, Dong T, Zhuang Z. The interplay of BDNF-TrkB with NMDA receptor in propofol-induced cognition dysfunction : Mechanism for the effects of propofol on cognitive function. BMC Anesthesiol 2018; 18:35. [PMID: 29621970 PMCID: PMC5887174 DOI: 10.1186/s12871-018-0491-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 02/13/2018] [Indexed: 01/15/2023] Open
Abstract
Background The aim of the present study was to verify whether propofol impaired learning and memory through the interplay of N-methyl-D-aspartate (NMDA) receptor with brain-derived neurotrophic factor (BDNF)-tyrosine kinase B (TrkB) signaling pathway. Methods 120 Sprague-Dawley (SD) rats were randomly assigned into eight groups. Experimental drugs including saline, intralipid, propofol, N-methyl-D-aspartate (NMDA), 7,8-dihydroxyflavone (7,8-DHF), K252a and MK-801. Spatial learning and memory of rats were tested by the Morris water maze (MWM) test. The mRNA and protein expression were determined by immunohistochemistry, RT-PCR and western blot. Finally, hippocampus cells proliferation and apoptosis were examined by PCNA immunohistochemistry and TUNEL respectively. Results The memory and learning was diminished in the propofol exposure group, however, the impaired memory and learning of rats were improved with the addition of NMDA and 7,8-DHF, while the improvement of memory and learning of rats were reversed with the addition of K252a and MK-801. In addition, the mRNA and protein expression levels and hippocampus cells proliferation were the same trend with the results of the MWM test, while apoptosis in hippocampus was reversed. Conclusion The propofol can impair memory and learning of rats and induce cognition dysfunction through the interplay of NMDA receptor and BDNF-TrkB-CREB signaling pathway.
Collapse
Affiliation(s)
- Junfei Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Fang Wang
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China
| | - Jun Zhang
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China
| | - Jianfeng Li
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China
| | - Li Ma
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China
| | - Tieli Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China.
| | - Zhigang Zhuang
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, College of Medicine, No. 2 Jingba Road, Zhengzhou, 450003, China.
| |
Collapse
|
20
|
Wu L, Wang S, Feng Y, Zhao W, Zuo W, Zhong L, Lin J, Zhao W, Luo F. KIF17 mediates the learning and memory impairment in offspring induced by maternal exposure to propofol during middle pregnancy. Mol Med Rep 2018; 17:5428-5434. [PMID: 29393422 DOI: 10.3892/mmr.2018.8479] [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: 10/06/2017] [Accepted: 12/15/2017] [Indexed: 11/05/2022] Open
Abstract
Preclinical studies suggest that propofol may cause neuronal injury to the developing brain. A previous study demonstrated that, in a rat model, maternal exposure to propofol during early or late pregnancy caused learning and memory impairment in the offspring. However, whether propofol exposure during middle pregnancy can cause long‑term behavioral deficits in the offspring remains to be elucidated. N‑methyl‑D‑aspartate receptor 2B subunit (NR2B) serves a critical role in memory modulation. To exert its function, NR2B must be transported to the neuronal membrane by kinesin family member 17 (KIF17). The aim of the present study was to investigate the role of KIF17 in learning and memory impairment in rat offspring caused by propofol exposure during middle pregnancy. Pregnant rats were exposed to propofol on gestational day 14 (G14) for 4 and 8 h, with control pregnant rats receiving an equal volume of normal saline. The learning and memory of the offspring was assessed using Morris water maze tests from postnatal day 30 (P30) to P36. The levels of KIF17 protein, total NR2B (T‑NR2B) and membrane NR2B (M‑NR2B) in the hippocampus were detected using western blotting. The results demonstrated that propofol exposure caused learning and memory deficits and decreased KIF17 and M‑NR2B protein levels in the hippocampus; however, no but changes in the expression of T‑NR2B were observed. These results indicate that maternal propofol exposure during middle pregnancy impairs learning and memory in offspring rats by suppressing the expression of KIF17 and inhibiting the translocation of NR2B to the neuronal membrane.
Collapse
Affiliation(s)
- Liuqing Wu
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shengqiang Wang
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yunlin Feng
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Weihong Zhao
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wei Zuo
- Department of Pain Management, The People's Hospital of Jiujiang, Jiujiang, Jiangxi 332000, P.R. China
| | - Liang Zhong
- Department of Anesthesiology, The People's Hospital of Pingxiang, Pingxiang, Jiangxi 337000, P.R. China
| | - Jiamei Lin
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Weilu Zhao
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Foquan Luo
- Department of Anesthesiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
21
|
Xiao Y, Zhou L, Tu Y, Li Y, Liang Y, Zhang X, Lv J, Zhong Y, Xie Y. Dexmedetomidine attenuates the propofol-induced long-term neurotoxicity in the developing brain of rats by enhancing the PI3K/Akt signaling pathway. Neuropsychiatr Dis Treat 2018; 14:2191-2206. [PMID: 30214209 PMCID: PMC6118247 DOI: 10.2147/ndt.s169099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Propofol induces short- and long-term neurotoxicity. Our previous study showed that dexmedetomidine (Dex) can attenuate the propofol-induced acute neurotoxicity in rodents by enhancing the PI3K/Akt signaling. However, whether treatment of young rats with Dex could protect them from long-term neurotoxicity induced by propofol is unclear. MATERIALS AND METHODS Seven-day-old male Sprague Dawley rats were randomized and injected intraperitoneally with saline (100 μL, NS), propofol (100 mg/kg), Dex (75 μg/kg), propofol (100 mg/kg) plus Dex (25, 50 or 75 μg/kg), 10% dimethyl sulfoxide (DMSO, 100 μL) or TDZD-8 (a GSK3β inhibitor, 1 mg/kg), or intracerebroventricularly with DMSO (5 μL) or LY294002 (a PI3K inhibitor, 25 μg/5 μL DMSO). Other rats in the experimental group were injected with the same doses of propofol, Dex and LY294002 or TDZD-8. All the rats were monitored until they were 9 weeks old. Their spatial learning and memory were tested by Morris water maze. The neuronal apoptosis, expression of PSD95, expression and phosphorylation of Akt and GSK3β and synaptic ultrastructures were determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, immunohistochemistry, Western blot and transmission electron microscopy assays, respectively. RESULTS Compared with the NS control group, young rats injected with intralipid, Dex, TDZD-8, LY294002 or DMSO alone did not show any significant change as they aged. Propofol significantly increased the escape latency time, hippocampal neuroapoptosis and synaptic ultrastructural changes but decreased the relative levels of PSD95 expression, and Akt and GSK3β phosphorylation in the developing hippocampus of the rats. The neuronal toxic effects of propofol were significantly mitigated by the pretreatment with a higher dose of Dex. The neuroprotective effect of Dex was enhanced by the treatment with TDZD-8, but was completely abrogated by the treatment with LY294002. CONCLUSION Our results indicated that the pretreatment of young rats with Dex attenuated the propofol-induced long-term neurotoxicity in their developing hippocampus by enhancing the PI3K/Akt signaling.
Collapse
Affiliation(s)
- Yong Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| | - Lifang Zhou
- Department of Anesthesiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Youbing Tu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| | - Yuantao Li
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, People's Republic of China
| | - Yubing Liang
- Department of Anesthesiology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Xu Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| | - Jing Lv
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| | - Yu Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China,
| |
Collapse
|
22
|
Malhotra A, Yosh E, Xiong M. Propofol's Effects on the Fetal Brain for Non-Obstetric Surgery. Brain Sci 2017; 7:brainsci7080107. [PMID: 28820429 PMCID: PMC5575627 DOI: 10.3390/brainsci7080107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 11/16/2022] Open
Abstract
While the use of Propofol has been increasing in usage for general surgical procedures since its release to market, there has been little work done on its potential link to neurotoxicity in humans. Only recently, following the release of a warning label from the United States Food and Drug Administration (USFDA) regarding a potential link to "neurotoxicity" in the neonate, did the surgical and anesthesiology communities become more aware of its potential for harm. Given the widespread use of this drug in clinical practice, the warning label naturally raised controversy regarding intrapartum Propofol usage. While intended to generate further studies, the lack of a viable anesthetic alternative raises issues regarding its current usage for surgical procedures in pregnant women. To answer the question whether current evidence is supportive of Propofol usage at its current levels in pregnant women, this review summarizes available evidence of fetal Propofol exposure in animal studies.
Collapse
Affiliation(s)
- Ajay Malhotra
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Emily Yosh
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Ming Xiong
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| |
Collapse
|
23
|
Monni L, Ghezzi F, Corsini S, Nistri A. Neurotoxicity of propofol on rat hypoglossal motoneurons in vitro. Neurosci Lett 2017; 655:95-100. [PMID: 28676256 DOI: 10.1016/j.neulet.2017.06.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/15/2017] [Accepted: 06/22/2017] [Indexed: 01/26/2023]
Abstract
Although propofol is a widely used intravenous general anaesthetic, many studies report its toxic potential, particularly on the developing central nervous system. We investigated its action on hypoglossal motoneurons (HMs) that control two critical functions in neonates, namely tongue muscle activity and airway patency. Thus, clinically relevant concentrations of propofol (1 and 5μM) were applied (4h) to neonatal rat brainstem slices to evaluate the expression of apoptosis-inducing factor (AIF) as biomarker of toxicity. This anaesthetic strongly increased AIF in the cytoplasm and the nucleus, without early loss of HMs. Electrophysiological recordings from HMs showed that propofol (5μM) enhanced GABA- and glycine-evoked current amplitude and lengthened GABAergic current decay time. Propofol also depressed NMDA receptor-mediated responses without affecting AMPA receptors. Since GABA and glycine depolarize neonatal HMs, we propose that the damaging action by propofol on these motoneurons might arise from the facilitated action of these transmitters with subsequent cytoplasmic Ca2+ overload. This phenomenon, in turn, may trigger cell death mechanisms manifested as increased expression of AIF and its translocation into the nucleus. Since propofol is also employed for induction and maintenance of paediatric surgery, caution is needed because its potential neurotoxicity might negatively impact neurodevelopment.
Collapse
Affiliation(s)
- Laura Monni
- Department of Neuroscience, International School for Advanced Studies (SISSA), via Bonomea, 265, 34136 Trieste, Italy.
| | - Filippo Ghezzi
- Department of Neuroscience, International School for Advanced Studies (SISSA), via Bonomea, 265, 34136 Trieste, Italy.
| | - Silvia Corsini
- Department of Neuroscience, International School for Advanced Studies (SISSA), via Bonomea, 265, 34136 Trieste, Italy.
| | - Andrea Nistri
- Department of Neuroscience, International School for Advanced Studies (SISSA), via Bonomea, 265, 34136 Trieste, Italy.
| |
Collapse
|
24
|
TNF-α Mediates the Intrinsic and Extrinsic Pathway in Propofol-Induced Neuronal Apoptosis Via PI3K/Akt Signaling Pathway in Rat Prefrontal Cortical Neurons. Neurotox Res 2017; 32:409-419. [DOI: 10.1007/s12640-017-9751-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 12/26/2022]
|
25
|
Milanovic D, Pesic V, Loncarevic-Vasiljkovic N, Avramovic V, Tesic V, Jevtovic-Todorovic V, Kanazir S, Ruzdijic S. Neonatal Propofol Anesthesia Changes Expression of Synaptic Plasticity Proteins and Increases Stereotypic and Anxyolitic Behavior in Adult Rats. Neurotox Res 2017; 32:247-263. [DOI: 10.1007/s12640-017-9730-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/28/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
|