1
|
Wang J, Liu Z. Research progress on molecular mechanisms of general anesthetic-induced neurotoxicity and cognitive impairment in the developing brain. Front Neurol 2022; 13:1065976. [PMID: 36504660 PMCID: PMC9729288 DOI: 10.3389/fneur.2022.1065976] [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: 10/10/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
General anesthetics-induced neurotoxicity and cognitive impairment in developing brains have become one of the current research hotspots in the medical science community. The underlying mechanisms are complex and involve various related molecular signaling pathways, cell mediators, autophagy, and other pathological processes. However, few drugs can be directly used to treat neurotoxicity and cognitive impairment caused by general anesthetics in clinical practice. This article reviews the molecular mechanism of general anesthesia-induced neurotoxicity and cognitive impairment in the neonatal brain after surgery in the hope of providing critical references for the treatments of clinical diseases.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Department of Anesthesiology, Baotou Central Hospital, Baotou, China,Baotou Clinical Medical College, Inner Mongolia Medical University, Baotou, China
| | - Zhihui Liu
- Department of Anesthesiology, Baotou Central Hospital, Baotou, China,*Correspondence: Zhihui Liu
| |
Collapse
|
2
|
Wong-Kee-You AMB, Loveridge-Easther C, Mueller C, Simon N, Good WV. The impact of early exposure to general anesthesia on visual and neurocognitive development. Surv Ophthalmol 2022; 68:539-555. [PMID: 35970232 DOI: 10.1016/j.survophthal.2022.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022]
Abstract
Every year millions of children are exposed to general anesthesia while undergoing surgical and diagnostic procedures. In the field of ophthalmology, 44,000 children are exposed to general anesthesia annually for strabismus surgery alone. While it is clear that general anesthesia is necessary for sedation and pain minimization during surgical procedures, the possibility of neurotoxic impairments from its exposure is of concern. In animals there is strong evidence linking early anesthesia exposure to abnormal neural development. but in humans the effects of anesthesia are debated. In humans many aspects of vision develop within the first year of life, making the visual system vulnerable to early adverse experiences and potentially vulnerable to early exposure to general anesthesia. We attempt to address whether the visual system is affected by early postnatal exposure to general anesthesia. We first summarize key mechanisms that could account for the neurotoxic effects of general anesthesia on the developing brain and review existing literature on the effects of early anesthesia exposure on the visual system in both animals and humans and on neurocognitive development in humans. Finally, we conclude by proposing future directions for research that could address unanswered questions regarding the impact of general anesthesia on visual development.
Collapse
Affiliation(s)
| | - Cam Loveridge-Easther
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA; University of Auckland, Auckland, New Zealand
| | - Claudia Mueller
- Sutter Health, San Francisco, CA, USA; Stanford Children's Health, Palo Alto, CA, USA
| | | | - William V Good
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA.
| |
Collapse
|
3
|
Uchida Y, Hashimoto T, Saito H, Takita K, Morimoto Y. Neonatal isoflurane exposure disturbs granule cell migration in the rat dentate gyrus. Biomed Res 2022; 43:1-9. [PMID: 35173111 DOI: 10.2220/biomedres.43.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
It has been reported that neonatal isoflurane exposure causes behavioral abnormalities following neurodegeneration in animals and gamma-aminobutyric acid type A (GABAA) receptor activation during the synaptogenesis is considered to be one possible trigger. Additionally, the inhibitory effect of excitatory GABAA receptor signaling on the granule cell (GC) migration in the neonatal rat dentate gyrus (DG) was reported in a febrile seizure model. Then, we hypothesized that neonatal isoflurane exposure, which activates GABAA receptor, causes GC migration disturbances in the neonatal rat. Rat pups were injected with 5-bromo-2'-deoxyuridine (BrdU) and divided into five treatment groups, and double immunofluorescent staining targeting BrdU and homeobox prospero-like protein 1 (Prox1) was performed to examine the localization of BrdU/Prox1 colabeled cells, and then the GC migration was assessed. As a result, we found that the ectopic migration of GC after 2% isoflurane exposure on postnatal day 7 significantly increased after P21. The number of hilar ectopic GCs was influenced by the concentration of isoflurane and the exposure day but not by carbon dioxide exposure. Our main finding is that neonatal isoflurane anesthesia disturbs the migration of GCs in the rat DG, which may be one possible mechanism underlying the neurotoxicity following neonatal isoflurane anesthesia.
Collapse
Affiliation(s)
- Yosuke Uchida
- Department of Anesthesiology, Hokkaido University Hospital
| | | | - Hitoshi Saito
- Department of Anesthesiology, Hokkaido University Hospital
| | - Koichi Takita
- Department of Anesthesiology, Hokkaido University Hospital
| | - Yuji Morimoto
- Department of Anesthesiology, Hokkaido University Hospital
| |
Collapse
|
4
|
Jiang T, Xu S, Shen Y, Xu Y, Li Y. Genistein Attenuates Isoflurane-Induced Neuroinflammation by Inhibiting TLR4-Mediated Microglial-Polarization in vivo and in vitro. J Inflamm Res 2021; 14:2587-2600. [PMID: 34168482 PMCID: PMC8216758 DOI: 10.2147/jir.s304336] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background Isoflurane, a widely used anesthetic in surgery, has been found to induce neurotoxicity. In parallel, genistein is thought to attenuate isoflurane-induced neurotoxicity, although underlying molecular mechanisms are still unclear. In this study, we studied the protective effects of genistein on isoflurane-induced neuroinflammation in rats and BV2 cells. Methods Sprague-Dawley rat pups were exposed to 0.75% isoflurane for 6 hours at postnatal day 7 (P7), and genistein (20, 40, or 80 mg/kg/day) or saline administered from P3 to P15. Hippocampal single-cell suspensions were prepared and apoptosis analyzed by flow cytometry. mRNA expression was determined by RT-qPCR, while protein expression was assessed using Western blot, immunochemistry and immunofluorescence. TLR4 was knocked-out in BV2 cells through CRISPR-Cas9. Results Genistein treatment reduced isoflurane-induced apoptosis and inflammation in rat hippocampus. Importantly, genistein promoted M2 and suppressed M1 microglia polarization in rat hippocampus after stimulation with isoflurane. In addition, genistein reduced isoflurane-induced protein expression levels of TLR4, MyD88, TRAF6, p-TAK1, p-p38, p-ERK, p-IκBα and p-NF-κB in rat hippocampus. In BV2 cells exposed to isoflurane, genistein treatment decreased IL-1β, TNF-α, IL-6 and IL-8 mRNA expressions, promoted M2 and suppressed M1 microglia polarization. Similarly, genistein also decreased TLR4 protein levels in isoflurane-induced BV2 cells. However, genistein did not affect CD16, iNOS, CD206 and Arg1 protein levels in TLR4-KO BV2 cells exposed to isoflurane. Conclusion Genistein attenuates isoflurane-induced neurotoxicity by inhibiting TLR4-mediated microglial inflammation in vivo and in vitro.
Collapse
Affiliation(s)
- Tao Jiang
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Shoucai Xu
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yangyang Shen
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yong Xu
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| | - Yuwen Li
- Shandong Cancer Research Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, People's Republic of China
| |
Collapse
|
5
|
Alipour M, Asl MK, Khordad E, Alipour F, Raoofi A, Ebrahimzadeh-Bideskan A, Ebrahimi V. Histopathological study on neuroapoptotic alterations induced by etomidate in rat hippocampus. Acta Histochem 2021; 123:151693. [PMID: 33601320 DOI: 10.1016/j.acthis.2021.151693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 11/19/2022]
Abstract
In human, there is substantial neurogenesis in the hippocampus that is implicated in memory formation and learning. These new-born neurons can be affected by neuropathological conditions. Anesthesia and surgical procedures are associated with postoperative cognitive changes particularly, impaired memory and learning. Therefore, the aim of this study was to evaluate the possible neurodegenerative effects of etomidate in rat hippocampus. Thirty male Wistar rats weighing 250 ± 30 g were randomly divided into 3 groups: 1) Etomidate group; four times 20 mg intraperitoneal injection with 1-h intervals, 2) Control group; the equal volume of normal saline, and 3) Normal group; without any intervention. 6 h after the last injection, the brains were removed and processed according to routine histological methods. TUNEL assay and toluidine blue staining were performed to evaluate neuro-histopathological changes in different regions of hippocampus. Our results showed that the number of TUNEL positive cells and dark neurons (DNs) in etomidate group were significantly higher in the CA1, CA2, CA3, and dentate gyrus (DG) of hippocampus compared with the control and normal groups (p < 0.05). While, there was no significant difference between the various regions of hippocampus in control and normal groups. Our findings showed that etomidate can increase apoptotic cells and dark neurons induction in different regions of hippocampus mainly in DG.
Collapse
Affiliation(s)
- Mohammad Alipour
- Department of Anesthesia, School of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Kamkar Asl
- Department of Anesthesia, School of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Khordad
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Raoofi
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran; Department of Anatomy, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Liu F, Qiu F, Chen H. miR-124-3p Ameliorates Isoflurane-Induced Learning and Memory Impairment via Targeting STAT3 and Inhibiting Neuroinflammation. Neuroimmunomodulation 2021; 28:248-254. [PMID: 34392240 DOI: 10.1159/000515661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/02/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Substantial evidence has indicated that isoflurane leads to learning and memory impairment. This study was designed to investigate the potential role of microRNA-124-3p (miR-124-3p) in isoflurane-induced learning and memory impairment in rats. METHODS Spatial learning and memory of rats were estimated by the Morris water maze (MWM) test after the construction of isoflurane-treated models. qRT-PCR was performed to assess the expression levels of miR-124-3p. The levels of interleukin-1β, interleukin-6, and tumor necrosis factor-α in the hippocampal tissues were determined by enzyme-linked immunosorbent assay. The luciferase activity was determined by using a dual-luciferase reporter assay system. RESULTS The higher escape latency and lower time spent in the original quadrant were shown in isoflurane-treated rats compared with the control rats. Moreover, treatment with isoflurane could induce neuroinflammation, and miR-124-3p was poorly expressed in the hippocampal tissue of isoflurane-treated rats. Furthermore, STAT3 is a functional target of miR-124-3p, and inflammatory cytokine level was downregulated by miR-124-3p. DISCUSSION/CONCLUSION Combining the results of the current study demonstrates that miR-124-3p may have pivotal roles in improving isoflurane-induced learning and memory impairment via targeting STAT3 and inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Fenghua Liu
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, China
| | - Fengyu Qiu
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, China
| | - Huayong Chen
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, China
| |
Collapse
|
7
|
Zhao S, Yuan C, Tuo X, Zhou C, Zhao Q, Shen T. MCLR induces dysregulation of calcium homeostasis and endoplasmic reticulum stress resulting in apoptosis in Sertoli cells. CHEMOSPHERE 2021; 263:127868. [PMID: 32828052 DOI: 10.1016/j.chemosphere.2020.127868] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
Microcystins-LR (MCLR) is a potent reproductive system toxin. We have previously shown that MCLR induced endoplasmic reticulum (ER) stress and apoptosis in testis. ER is the main calcium storage site in cells, and its calcium homeostasis plays an important role in the regulation of apoptosis. Hence, in the present study, we have investigated the role of calcium (Ca2+) in inducing apoptosis and how it affect the mitochondria and endoplasmic reticulum in TM4 cells. Our study found that MCLR induced an increase in Ca2+ concentration in TM4 cells. Compared to the controls, MCLR induced phosphorylation of calmodulin-dependent protein kinase II (CaMKII) which was involved in MAPKs activation, resulting in the induction of mitochondrial apoptosis pathways. Ca2+ chelator Bapta-AM partially reversed MCLR-induced apoptosis, confirming the possible involvement of calcium homeostasis disruption after MCLR exposure. Meanwhile, MCLR activated unfolded protein response and activated the ER apoptotic pathway by activating caspase-12. In addition, exposure to MCLR causes mitochondrial defects and increased apoptosis by up-regulating caspase 3 and cytosol cytochrome c expression. Collectively, these results demonstrated that MCLR disturbed calcium homeostasis, which caused ER-mitochondria dysfunction, ultimately promoted cell apoptosis in Sertoli cells.
Collapse
Affiliation(s)
- Sujuan Zhao
- School of Public Health, Anhui Medical University, Hefei, China
| | - Chunyang Yuan
- School of Public Health, Anhui Medical University, Hefei, China
| | - Xun Tuo
- College of Chemistry, Nanchang University, Nanchang, 330031, China
| | - Chengfan Zhou
- School of Public Health, Anhui Medical University, Hefei, China
| | - Qihong Zhao
- School of Public Health, Anhui Medical University, Hefei, China
| | - Tong Shen
- School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Huang L, Fang HB, Cheng HH, Mei SL, Cheng YP, Lv Y, Meng QT, Xia ZY. Epigenetic modulation of the MAPK pathway prevents isoflurane-induced neuronal apoptosis and cognitive decline in aged rats. Exp Ther Med 2020; 20:35. [PMID: 32952626 PMCID: PMC7480129 DOI: 10.3892/etm.2020.9162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Isoflurane is a broadly used inhalation anesthetic that causes cognitive impairment in rodent models as well as humans. Although previous studies suggested an association between isoflurane exposure and neuro-inflammation, apoptosis and mitochondrial dysfunction, the pathogenesis of isoflurane-induced cognitive decline remains elusive. In the present study, 22-month-old male Sprague-Dawley male rats (n=96) were divided into three groups: Control (Cont), isoflurane (ISO) and MS-275 pre-treated groups. The rats were sacrificed following exposure to isoflurane and a cognitive test. The hippocampus of each animal was harvested for quantitative PCR, TUNEL staining and western blot analysis. Histone deacetylases (HDAC)-1, -2 and -3 exhibited a significant increase at the gene and protein expression levels, whereas negligible mRNA expressions were observed for genes HDAC 4-11 (P>0.05; compared with Cont). Pre-treatment with the HDAC inhibitor MS-275 significantly inhibited the increase in TUNEL-positive cells induced by isoflurane exposure (70.72% decrease; P<0.001; compared with ISO). Furthermore, MS-275 significantly decreased caspase-3 and Bax expression levels while increasing Bcl-2 protein expression. The isoflurane-induced changes in the MAPK pathway signaling proteins ERK1/2, JNK and p38 were also reversed with MS-275 pre-treatment. Finally, in a Morris water maze test, the time to find a hidden platform was reduced in MS-275 pre-treated rats, compared with the ISO group. Therefore, the present study provided insight into the effect of isoflurane exposure on neuronal apoptosis pathways, as well as cognitive decline via epigenetic programming of MAPK signaling in aged rats.
Collapse
Affiliation(s)
- Lei Huang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hai-Bin Fang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hui-Hui Cheng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sheng-Lan Mei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yun-Ping Cheng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yao Lv
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
9
|
Xu W, Li XP, Li EZ, Liu YF, Zhao J, Wei LN, Ma L. Protective Effects of Allicin on ISO-Induced Rat Model of Myocardial Infarction via JNK Signaling Pathway. Pharmacology 2020; 105:505-513. [PMID: 32784309 DOI: 10.1159/000503755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/27/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This research was aimed to explore protective effects of allicin on rat model of myocardial infarction via JNK signaling pathway. METHODS Rat myocardial ischemia model was established with subcutaneous injection of isoproterenol (ISO). Seventy-five rats were randomly divided into 5 groups (n = 15): sham group, ISO group, low-dose group (1.2 mg/kg/days for 7 days), medium-dose group (1.8 mg/kg/days for 7 days), and high-dose group (3.6 mg/kg/days for 7 days). Routine HE staining and Masson staining were performed to observe myocardial histopathology. The expression of oxidative stress-related indicators, heart tissue apoptosis-related proteins, and JNK and p-JNK proteins were measured for different groups. RESULTS Compared with the sham group, the T wave value of the ISO group was significantly increased (p < 0.01). When allicin was administered, the T wave values at different time points in all groups were all decreased. Compared with the sham group, the ratio of eNOS, Bcl-2/Bax was significantly decreased, and p-eNOS, iNOS, caspase-3, caspase-9, and Cyt-c were significantly elevated in the ISO group (p < 0.05). After allicin was administered, significant changes in these proteins were observed in the medium- and high-dose groups. There was no significant change in the expression of JNK protein in the ISO group compared with the sham group; however, the expression of eNOS and p-JNK protein were significantly upregulated (p < 0.01) and the expression of p-eNOS and iNOS were significantly downregulated (p < 0.01). When allicin was administered, expression of p-JNK protein was significantly downregulated. CONCLUSION Allicin can reduce oxidative stress damage and cardiomyocyte apoptosis in rat model of myocardial infarction and can significantly regulate JNK signaling pathway.
Collapse
Affiliation(s)
- Wen Xu
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiang-Peng Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - En-Ze Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue-Fen Liu
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Zhao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li-Na Wei
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Ma
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China,
| |
Collapse
|
10
|
Aksenov DP, Miller MJ, Dixon CJ, Drobyshevsky A. Impact of anesthesia exposure in early development on learning and sensory functions. Dev Psychobiol 2020; 62:559-572. [PMID: 32115695 DOI: 10.1002/dev.21963] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/27/2020] [Accepted: 02/08/2020] [Indexed: 12/11/2022]
Abstract
Each year, millions of children undergo anesthesia, and both human and animal studies have indicated that exposure to anesthesia at an early age can lead to neuronal damage and learning deficiency. However, disorders of sensory functions were not reported in children or animals exposed to anesthesia during infancy, which is surprising, given the significant amount of damage to brain tissue reported in many animal studies. In this review, we discuss the relationship between the systems in the brain that mediate sensory input, spatial learning, and classical conditioning, and how these systems could be affected during anesthesia exposure. Based on previous reports, we conclude that anesthesia can induce structural, functional, and compensatory changes in both sensory and learning systems. Changes in myelination following anesthesia exposure were observed as well as the neurodegeneration in the gray matter across variety of brain regions. Disproportionate cell death between excitatory and inhibitory cells induced by anesthesia exposure can lead to a long-term shift in the excitatory/inhibitory balance, which affects both learning-specific networks and sensory systems. Anesthesia may directly affect synaptic plasticity which is especially critical to learning acquisition. However, sensory systems appear to have better ability to compensate for damage than learning-specific networks.
Collapse
Affiliation(s)
| | | | - Conor J Dixon
- NorthShore University HealthSystem, Evanston, IL, USA
| | | |
Collapse
|
11
|
Lei S, Lu P, Lu Y, Zheng J, Li W, Wang N, Zhang H, Li R, Wang K, Wen J, Wei H, Zhang Y, Qiu Z, Xu J, Lv H, Chen X, Liu Y, Zhang P. Dexmedetomidine Alleviates Neurogenesis Damage Following Neonatal Midazolam Exposure in Rats through JNK and P38 MAPK Pathways. ACS Chem Neurosci 2020; 11:579-591. [PMID: 31999428 DOI: 10.1021/acschemneuro.9b00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Midazolam, a widely used anesthetic, inhibits proliferation of neural stem cells (NSCs) and induces neuroapoptosis in neonates. Dexmedetomidine, an effective auxiliary medicine in clinical anesthesia, protects the developing brain against volatile anesthetic-induced neuroapoptosis. Whether dexmedetomidine protects against neurogenesis damage induced by midazolam remains unknown. This study aims to clarify the protective effect of dexmedetomidine on midazolam-induced neurogenesis damage and explore its potential mechanism. Postnatal 7-day-old Sprague-Dawley (SD) rats and cultured NSCs were treated with either normal saline, midazolam, or dexmedetomidine combined with midazolam. The rats were sacrificed at 1, 3, and 7 days after treatment. Cell proliferation was assessed by 5-bromodeoxyurdine (BrdU) incorporation. Cell viability was determined using MTT assay. Cell differentiation and apoptosis were detected by immunofluorescent staining and terminal dUTP nick-end labeling (TUNEL), respectively. The protein levels of p-JNK, p-P38, and cleaved caspase-3 were quantified using Western blotting. Midazolam decreased cell proliferation and increased cell apoptosis in the subventricular zone (SVZ), the subgranular zone (SGZ) of the hippocampus, and cultured NSCs. Moreover, midazolam decreased cell viability and increased the expression of p-JNK and p-P38 in cultured NSCs. Co-treatment with dexmedetomidine attenuated midazolam-induced changes in cell proliferation, viability, apoptosis, and protein expression of p-JNK and p-P38 in cultured NSCs. Midazolam and dexmedetomidine did not affect the differentiation of the cultured NSCs. These results indicate that dexmedetomidine alleviated midazolam-induced neurogenesis damage via JNK and P38 MAPK pathways.
Collapse
Affiliation(s)
- Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jing Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Haixia Lv
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi’an Jiaotong University, Xi’an 710016, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
12
|
Hypoxia, hypercarbia, and mortality reporting in studies of anaesthesia-related neonatal neurodevelopmental delay in rodent models. Eur J Anaesthesiol 2020; 37:70-84. [DOI: 10.1097/eja.0000000000001105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
13
|
General anesthetic neurotoxicity in the young: Mechanism and prevention. Neurosci Biobehav Rev 2019; 107:883-896. [PMID: 31606415 DOI: 10.1016/j.neubiorev.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/27/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022]
Abstract
General anesthesia (GA) is usually considered to safely induce a reversible unconscious state allowing surgery to be performed without pain. A growing number of studies, in particular pre-clinical studies, however, demonstrate that general anesthetics can cause neuronal death and even long-term neurological deficits. Herein, we report our literature review and meta-analysis data of the neurological outcomes after anesthesia in the young. We also review available mechanistic and epigenetic data of GA exposure related to cognitive impairment per se and the potential preventive strategies including natural herbal compounds to attenuate those side effects. In summary, anesthetic-induced neurotoxicity may be treatable and natural herbal compounds and other medications may have great potential for such use but warrants further study before clinical applications can be initiated.
Collapse
|
14
|
Ko MJ, Mulia GE, van Rijn RM. Commonly Used Anesthesia/Euthanasia Methods for Brain Collection Differentially Impact MAPK Activity in Male and Female C57BL/6 Mice. Front Cell Neurosci 2019; 13:96. [PMID: 30983972 PMCID: PMC6447702 DOI: 10.3389/fncel.2019.00096] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/27/2019] [Indexed: 01/01/2023] Open
Abstract
The mitogen-activated protein kinases (MAPKs) are a family of protein kinases that regulate crucial neuronal functions such as neuronal differentiation, proliferation, and apoptosis through phosphorylation of subsequent protein kinases. The three classical MAPK subfamilies, extracellular signal-regulated kinase 1 and 2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 kinase have been linked to various neurological disorders often in conjunction with activation of a wide range of G protein-coupled receptors and receptor tyrosine kinases. Many studies investigating MAPK function in these disorders rely on histochemistry or immunoblotting that require brain isolation following euthanasia. Here, we evaluated to what degree different modes of anesthesia/euthanasia impact MAPK activity in adult male and female C57BL/6 mice. Mice were decapitated following ketamine/xylazine or isoflurane anesthesia, carbon dioxide asphyxiation, or without anesthesia. We selectively chose five brain regions (the prefrontal cortex, the dorsal hippocampus, the dorsal striatum, the nucleus accumbens, and the amygdala) that are heavily implicated in neuropsychiatric disorders. We found that relative to carbon dioxide asphyxiation, the other methods displayed significantly stronger ERK1/2 phosphorylation in select brain regions of male and female mice, with no pronounced sex difference. A similar, yet, less pronounced trend was observed for JNK activity, whereas the choice of euthanasia method did not differentially impact p38 phosphorylation. Our study results reveal how small differences in experimental design may impact whether one will be able to detect drug- or disease-related changes in MAPK activity. These findings are timely in a period where experimental rigor is emphasized to increase reproducibility of research.
Collapse
Affiliation(s)
- Mee Jung Ko
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States
| | - Grace E Mulia
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
15
|
Jin H, Wang M, Wang J, Cao H, Niu W, Du L. Paeonol attenuates isoflurane anesthesia-induced hippocampal neurotoxicity via modulation of JNK/ERK/P38MAPK pathway and regulates histone acetylation in neonatal rat. J Matern Fetal Neonatal Med 2018; 33:81-91. [PMID: 29886761 DOI: 10.1080/14767058.2018.1487396] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective: Volatile anesthetic such as isoflurane causes widespread neurodegeneration in the developing animal brains and also induces cognitive impairments. Paeonol is a plant-derived phenolic compound possessing numerous bioactive properties. The study investigates the neuroprotective effects of paeonol against isoflurane-induced neurodegeneration and cognitive disturbances in neonatal rats.Methods: Paeonol (50, 100, and 150 mg/kg body weight/day) was given orally to separate groups of neonatal rats from postnatal day 3 (P3) to P21 and were exposed to isoflurane (0.75%; 6 h) on P7.Results: Neuroapoptosis following isoflurane exposure was remarkably reduced by paeonol. Isoflurane-induced elevated cleaved caspase-3, Bad, and Bax expression, were down-regulated on paeonol administration. Paeonol significantly enhanced expression of antiapoptotic proteins (Bcl-2, Bcl-xL, xIAP, c-IAP-1, c-IAP-2, and survivin) and improved acetylation of HK39 and HK412. The expression of histone deacetylases (HDACs)-HDAC2 and HDAC-3 were down-regulated. Isoflurane-induced activation of JNK/p38MAPK signaling and suppressed ERK signaling and were effectively regulated by paeonol. General behavior and freezing responses of the rats were improved. Results of the Morris Water Maze tests revealed improved learning and memory retention on paeonol treatment.Conclusions: Paeonol effectively inhibited neuroapoptosis and improved isoflurane-induced cognitive dysfunctions via regulating histone acetylation and JNK/ERK1/2/p38MAPK signaling pathways.
Collapse
Affiliation(s)
- Haiyan Jin
- Department of Anesthesiology, The Children's Hospital, School of Medicine, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Minyan Wang
- Department of Anesthesiology, The Children's Hospital, School of Medicine, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jiangmei Wang
- Department of Anesthesiology, The Children's Hospital, School of Medicine, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hongmin Cao
- Department of Anesthesiology, The Children's Hospital, School of Medicine, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Wanting Niu
- Tissue Engineering Laboratories, VA Boston Healthcare System, Boston, MA, USA.,Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lizhong Du
- Department of Neonatology, The Children's Hospital, School of Medicine, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
16
|
Wang S, Zhou Y. Baicalein Inhibits Neuroapoptosis Via Pathways in Sevoflurane Induced Rats. Transl Neurosci 2018; 9:88-98. [PMID: 30042862 PMCID: PMC6057263 DOI: 10.1515/tnsci-2018-0015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/29/2018] [Indexed: 01/15/2023] Open
Abstract
Background Baicalein, a bioactive flavonoid was explored for its capability to attenuate sevoflurane induced neuronal apoptosis and to improve behavioural and cognitive impairments. Sevoflurane is a frequently used inhalation anesthetic in neonates and children. Neonatal sevoflurane exposure causes widespread neurodegeneration and cognitive impairments. Development of compounds that could effectively prevent/reduce the adverse effects is of tremendous medical value. Methods Isolated groups of neonatal rats were regulated with baicalein (25, 50 or 100 mg/kg b.wt) from postnatal day 3 (P3) to P21 and were exposed to sevoflurane (3%; 6 h) on P7. Results: Baicalein inhibited sevoflurane induced neuroapoptosis significantly as assessed by TUNEL assay. The raised levels of cleaved caspase-3, Bad and Bax were down-regulated by baicalein with enhanced Bcl-2, Bcl-xL, xIAP, c-IAP-1, c-IAP-2 and survivin expression. Baicalein regulated JNK/ERK signalling and also activated the PI3K/Akt pathway effectively as evident from the increased Akt, phospho-Akt, GSK-3β, phospho-GSK-3β levels. Baicalein, also improved the behaviour of animals in open filed and olfactory tests. The freezing responses and the performance in Morris Water Maze tests were enhanced. Conclusion Baicalein reduced neurodegeneration and improved learning and memory retention of rats and as well modulated PI3/Akt/GSK-3β and JNK/ERK signalling pathways.
Collapse
Affiliation(s)
- Si Wang
- Department of Pediatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China, 637000
| | - Yu Zhou
- Department of Pediatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China, 637000
| |
Collapse
|
17
|
All-Trans Retinoic Acid Ameliorates the Early Experimental Cerebral Ischemia-Reperfusion Injury in Rats by Inhibiting the Loss of the Blood-Brain Barrier via the JNK/P38MAPK Signaling Pathway. Neurochem Res 2018; 43:1283-1296. [PMID: 29802528 DOI: 10.1007/s11064-018-2545-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/09/2018] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
All-trans retinoic acid (ATRA) influences the outcomes of cerebral ischemic reperfusion (CIR) injury, but the mechanism remains unclear. The present study aimed to investigate the effects of ATRA on loss of the blood brain barrier (BBB) following CIR and to explore the possible mechanisms. Transient middle cerebral artery occlusion was performed on male SD rats to construct an in vivo CIR model. Neurological deficits, BBB permeability, brain edema, MRI and JNK/P38 MAPK proteins were detected at 24 h following CIR. We demonstrated that ATRA pretreatment could alleviate CIR-induced neurological deficits, increase of BBB permeability, infarct volume, degradation of tight junction proteins, inhibit MMP-9 protein expression and activity. ATRA treatment also reduced the p-P38 and p-JNK protein level. However the protective effect of ATRA on CIR could be reversed by administration of retinoic acid alpha receptor antagonist Ro41-5253. SP600125 and SB203580, which is the JNK/P38 pathway inhibitors has the same protective effect as ATRA. These results indicated that ATRA may inhibit the JNK/P38 MAPK pathway to alleviate BBB disruption and improve CIR outcomes.
Collapse
|
18
|
Liu Y, Liu C, Zeng M, Han X, Zhang K, Fu Y, Li J, Li Y. Influence of sevoflurane exposure on mitogen-activated protein kinases and Akt/GSK-3β/CRMP-2 signaling pathways in the developing rat brain. Exp Ther Med 2018; 15:2066-2073. [PMID: 29434807 PMCID: PMC5776508 DOI: 10.3892/etm.2017.5651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/20/2017] [Indexed: 01/06/2023] Open
Abstract
Prolonged exposure to volatile anesthetics causes neurodegeneration in developing animal brains. However, their underlying mechanisms of action remain unclear. The current study investigated the expression of proteins associated with the mitogen-activated protein kinases (MAPK) and protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β)/collapsin response mediator protein 2 (CRMP-2) signaling pathways in the cortices of neonatal mice following exposure to sevoflurane. Seven-day-old (P7) neonatal C57BL/6 mice were randomly divided into 2 groups and either exposed to 2.6% sevoflurane or air for 6 h. Terminal deoxyribonucleotide transferase mediated dUTP nick end labeling (TUNEL) staining, as well as the expression of activated caspase-3 and α-fodrin, was used to detect neuronal apoptosis in the cortices of mice. MAPK signaling pathways were investigated by detecting the expression of phosphorylated (p-) extracellular signal-regulated kinase 1/2 (ERK1/2), p-cyclic adenosine monophosphate response element-binding protein (CREB), p-p38, p-nuclear factor (NF-κB) and p-c-Jun N-terminal kinase (p-JNK). Akt/GSK-3β/CRMP-2 signaling pathways were assessed by detecting the expression of p-Akt, p-GSK-3β and p-CRMP-2 in the cortices of P7 mice 2 h following exposure to sevoflurane. The results demonstrated that sevoflurane significantly increased the apoptosis of cells in the retrosplenial cortex (RS), frontal cortex (FC) and parietal association cortex (PtA), increased the expression of cleaved caspase-3 expression and promoted the formation of 145 kDa and 120 kDa fragments from α-fodrin. Sevoflurane inhibited the phosphorylation of ERK1/2 and CREB, stimulated the phosphorylation of p38 and NF-κB, but did not significantly affect the phosphorylation of JNK. Furthermore, sevoflurane inhibited the phosphorylation of Akt, decreased the phosphorylation of GSK-3β at ser9 and increased the phosphorylation of CRMP2 at Thr514. These results suggest that multiple signaling pathways, including ERK1/2, P38 and Akt/GSK-3β/CRMP-2 may be involved in sevoflurane-induced neuroapoptosis in the developing brain.
Collapse
Affiliation(s)
- Yafang Liu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chuiliang Liu
- Department of Anesthesiology, Chancheng Center Hospital, Guangdong Medical College, Foshan, Guangdong 528030, P.R. China
| | - Minting Zeng
- Department of Anesthesiology, Guangzhou Women and Children's Medical Centre of Guangzhou Medical University, Guangzhou, Guangdong 510523, P.R. China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jue Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
19
|
Prolonged duration of isoflurane anesthesia impairs spatial recognition memory through the activation of JNK1/2 in the hippocampus of mice. Neuroreport 2018; 28:386-390. [PMID: 28240723 DOI: 10.1097/wnr.0000000000000760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Postoperative cognitive dysfunction is a frequent complication with surgery and anesthesia, and the underlying mechanism is unclear. Our aim was to investigate the effect of different durations of isoflurane anesthesia on spatial recognition memory and activation of JNK1/2 in the hippocampus of mice. In the present study, adult male mice were anesthetized with isoflurane for different durations (1.5% isoflurane for 1, 2, and 4 h). Spatial recognition memory was determined using spontaneous alternation and two-trial recognition memory in Y-maze at 24 h after anesthesia. The activation of JNK1/2 in the hippocampus was tested using western blot. Mice treated with isoflurane for 4 h showed significantly decreased spontaneous alternations and decreased exploration parameters compared with the no anesthesia group, but this was not observed in mice treated with isoflurane for 1 or 2 h. The protein levels of p-JNK1/2 in the hippocampus were significantly increased at 10 min after isoflurane anesthesia for 1, 2, and 4 h compared with no anesthesia. However, only isoflurane anesthesia for 4 h still increased JNK1/2 and p-JNK1/2 levels at 24 h after anesthesia. We concluded that prolonged duration of isoflurane anesthesia maintained the activation of JNK1/2, which led to memory impairment at 24 h after anesthesia.
Collapse
|
20
|
Yang Z, Lv J, Lu X, Li X, An X, Wang J, Weng H, Li Y. Emulsified isoflurane induces release of cytochrome C in human neuroblastoma SHSY-5Y cells via JNK (c-Jun N-terminal kinases) signaling pathway. Neurotoxicol Teratol 2018; 65:19-25. [DOI: 10.1016/j.ntt.2017.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/27/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023]
|
21
|
Han X, Liu C, Zhang K, Guo M, Shen Z, Liu Y, Zuo Z, Cao M, Li Y. Calpain and JNK pathways participate in isoflurane - induced nucleus translocation of apoptosis-inducing factor in the brain of neonatal rats. Toxicol Lett 2017; 285:60-73. [PMID: 29289695 DOI: 10.1016/j.toxlet.2017.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/11/2017] [Accepted: 12/27/2017] [Indexed: 01/26/2023]
Abstract
Recent studies have demonstrated that volatile anesthetic causes caspase-dependent neuroapoptosis and persistent cognitive deficits in young animals. Apoptosis-inducing factor (AIF) can trigger apoptosis by caspase-independent pathway. Whether isoflurane induces neuroapoptosis by activation of AIF and its possible mechanism are underdetermined. Rats at postnatal day 7 were exposed to 1.1% isoflurane for 4 h and the expression of AIF, cytochrome c, caspase-3, μ-calpain, m-calpain, Bcl-2 and Bax in the mitochondrial, cytosolic, and nuclear fraction, as well as the number of both AIF and TUNEL positive neurons in the cortices of rats were measured. Moreover, the effects of calpain inhibitor MDL-28170 or JNK inhibitor SP600125 on isoflurane-induced AIF release, caspase activation and cognitive deficits were assessed. We found isoflurane activated CytC-caspase-3 dependent apoptosis pathway mainly in the early phase (0-6 h after exposure). Moreover, isoflurane activated mitochondrial μ-calpain, induced AIF truncation during early phase and activated m-calpain, induced AIF release from the mitochondria to cytosol and translocation into the nucleus in the late phase (6-24 h after exposure). MDL-28170 attenuated the isoflurane-induced mitochondrial AIF truncation, release and nuclear translocation, but did not change the expression of cleaved-caspase-3 and mitochondrial Bax and Bcl-2 proteins. SP600125 attenuated isoflurane-induced neuroapoptosis by inhibiting both AIF and caspase-3 pathways and reduced cognitive impairment in neonatal rats. This is the first study to provide the evidence that isoflurane induced AIF-dependent neuroapoptosis by activation of mitochondrial μ-calpain and m-calpain in neonatal rats. JNK inhibition reversed isoflurane-induced neuroapoptosis and subsequent long-term neurocognitive impairment, acting via inhibiting activation of both AIF and caspase-3 pathways.
Collapse
Affiliation(s)
- Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Chuiliang Liu
- Department of Anesthesiology, ChanCheng Center Hospital, Guangdong Medical College, Foshan, 528030, PR China
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Mingyan Guo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Zhiwen Shen
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Yafang Liu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Zhiyi Zuo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908-0710, USA
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China.
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
22
|
Liu L, Chang X, Zhang Y, Wu C, Li R, Tang L, Zhou Z. Fluorochloridone induces primary cultured Sertoli cells apoptosis: Involvement of ROS and intracellular calcium ions-mediated ERK1/2 activation. Toxicol In Vitro 2017; 47:228-237. [PMID: 29248592 DOI: 10.1016/j.tiv.2017.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/18/2017] [Accepted: 12/12/2017] [Indexed: 02/06/2023]
Abstract
Fluorochloridone (FLC) is a widely used pyrrolidone selective herbicide and reported to induce testis injuries in male rats, but the underlying mechanism is largely unknown. In the present study, primary-cultured Sertoli cells were exposed to FLC at the concentration of 0-10.00μM to study the mechanism of FLC-induced apoptosis. The roles of ROS, intracellular calcium, endoplasmic reticulum (ER), and ERK1/2 were looked at with ROS scavenger N-acetyl-cysteine (NAC), intracellular calcium chelator BAPTA-AM, ER calcium depleting agent thapsigargin (TG), and ERK1/2 inhibitor U0126, respectively. FLC induced dose-dependent apoptosis increase as well as the elevation in levels of ROS, intracellular calcium, and ERK1/2 activation. FLC treatment led to constantly increasing apoptotic rates and ERK1/2 activation over time, while inversed-V shaped change tendencies of ROS and intracellular calcium levels were observed. FLC-induced ROS generation disrupted the intracellular calcium homeostasis by attacking the ER, and the elevated intracellular calcium levels resulted in ERK1/2 over-phosphorylation and consequently promoted Sertoli cell apoptosis. Taken together, ROS and intracellular calcium-mediated ERK1/2 activation led to FLC-induced Sertoli cell apoptosis.
Collapse
Affiliation(s)
- Luqing Liu
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Xiuli Chang
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Chunhua Wu
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Rui Li
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China; Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| | - Liming Tang
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| | - Zhijun Zhou
- School of Public Health/MOE Key Laboratory for Public Health Safety/Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
23
|
Jiang T, Wang XQ, Ding C, Du XL. Genistein attenuates isoflurane-induced neurotoxicity and improves impaired spatial learning and memory by regulating cAMP/CREB and BDNF-TrkB-PI3K/Akt signaling. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:579-589. [PMID: 29200900 PMCID: PMC5709474 DOI: 10.4196/kjpp.2017.21.6.579] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/02/2016] [Accepted: 08/18/2016] [Indexed: 01/16/2023]
Abstract
Anesthetics are used extensively in surgeries and related procedures to prevent pain. However, there is some concern regarding neuronal degeneration and cognitive deficits arising from regular anesthetic exposure. Recent studies have indicated that brain-derived neurotrophic factor (BDNF) and cyclic AMP response element-binding protein (CREB) are involved in learning and memory processes. Genistein, a plant-derived isoflavone, has been shown to exhibit neuroprotective effects. The present study was performed to examine the protective effect of genistein against isoflurane-induced neurotoxicity in rats. Neonatal rats were exposed to isoflurane (0.75%, 6 hours) on postnatal day 7 (P7). Separate groups of rat pups were orally administered genistein at doses of 20, 40, or 80 mg/kg body weight from P3 to P15 and then exposed to isoflurane anesthesia on P7. Neuronal apoptosis was detected by TUNEL assay and FluoroJade B staining following isoflurane exposure. Genistein significantly reduced apoptosis in the hippocampus, reduced the expression of proapoptotic factors (Bad, Bax, and cleaved caspase-3), and increased the expression of Bcl-2 and Bcl-xL. RT-PCR analysis revealed enhanced BDNF and TrkB mRNA levels. Genistein effectively upregulated cAMP levels and phosphorylation of CREB and TrkB, leading to activation of cAMP/CREB-BDNF-TrkB signaling. PI3K/Akt signaling was also significantly activated. Genistein administration improved general behavior and enhanced learning and memory in the rats. These observations suggest that genistein exerts neuroprotective effects by suppressing isoflurane-induced neuronal apoptosis and by activating cAMP/CREB-BDNF-TrkB-PI3/Akt signaling.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Anesthesiology, Shandong Cancer Hospital, Jinan 250117, Shandong Province, China
| | - Xiu-Qin Wang
- Department of Anesthesiology, Shandong Cancer Hospital, Jinan 250117, Shandong Province, China
| | - Chuan Ding
- Department of Anesthesiology, Shandong Cancer Hospital, Jinan 250117, Shandong Province, China
| | - Xue-Lian Du
- Department of Gynecology, Shandong Cancer Hospital, Jinan 250117, Shandong Province, China
| |
Collapse
|
24
|
Ding ML, Ma H, Man YG, Lv HY. Protective effects of a green tea polyphenol, epigallocatechin-3-gallate, against sevoflurane-induced neuronal apoptosis involve regulation of CREB/BDNF/TrkB and PI3K/Akt/mTOR signalling pathways in neonatal mice. Can J Physiol Pharmacol 2017; 95:1396-1405. [PMID: 28679060 DOI: 10.1139/cjpp-2016-0333] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), a polyphenol in green tea, is an effective antioxidant and possesses neuroprotective effects. Brain-derived neurotrophic factor (BDNF) and cyclic AMP response element-binding protein (CREB) are crucial for neurogenesis and synaptic plasticity. In this study, we aimed to assess the protective effects of EGCG against sevoflurane-induced neurotoxicity in neonatal mice. Distinct groups of C57BL/6 mice were given EGCG (25, 50, or 75 mg/kg body weight) from postnatal day 3 (P3) to P21 and were subjected to sevoflurane (3%; 6 h) exposure on P7. EGCG significantly inhibited sevoflurane-induced neuroapoptosis as determined by Fluoro-Jade B staining and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL). Increased levels of cleaved caspase-3, downregulated Bad and Bax, and significantly enhanced Bcl-2, Bcl-xL, xIAP, c-IAP-1, and survivin expression were observed. EGCG induced activation of the PI3K/Akt pathway as evidenced by increased Akt, phospho-Akt, GSK-3β, phospho-GSK-3β, and mTORc1 levels. Sevoflurane-mediated downregulation of cAMP/CREB and BDNF/TrkB signalling was inhibited by EGCG. Reverse transcription PCR analysis revealed enhanced BDNF and TrkB mRNA levels upon EGCG administration. Improved performance of mice in Morris water maze tests suggested enhanced learning and memory. The study indicates that EGCG was able to effectively inhibit sevoflurane-induced neurodegeneration and improve learning and memory retention of mice via activation of CREB/BDNF/TrkB-PI3K/Akt signalling.
Collapse
Affiliation(s)
- Mei-Li Ding
- a Department of Pediatrics, Shandong Jining No. 1 People's Hospital, Shandong 272011, China
| | - Hui Ma
- b Department of Neurosurgery, Shandong Jining No. 1 People's Hospital, Shandong 272011, China
| | - Yi-Gang Man
- a Department of Pediatrics, Shandong Jining No. 1 People's Hospital, Shandong 272011, China
| | - Hong-Yan Lv
- a Department of Pediatrics, Shandong Jining No. 1 People's Hospital, Shandong 272011, China
| |
Collapse
|
25
|
Yuan JH, Pan F, Chen J, Chen CE, Xie DP, Jiang XZ, Guo SJ, Zhou J. Neuroprotection by plumbagin involves BDNF-TrkB-PI3K/Akt and ERK1/2/JNK pathways in isoflurane-induced neonatal rats. ACTA ACUST UNITED AC 2017; 69:896-906. [PMID: 28464236 DOI: 10.1111/jphp.12681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/12/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES This study was designed to assess the effects of plumbagin on isoflurane-induced neurotoxicity. METHODS Neonatal Sprague Dawley rat pups were treated with plumbagin (50, 100 or 150 mg/kg body weight, orally) from postnatal day 2. The pups on postnatal day 7 were subjected to isoflurane (0.75%) exposure for 6 h. Neuronal apoptosis in the hippocampal tissues was detected by TUNEL assay and FluroJade B staining following isoflurane exposure. Protein expressions were analysed by immunoblotting. RT-PCR was performed to assess mRNA levels of brain-derived neurotrophic factor (BDNF) and TrkB. KEY FINDINGS We observed reduced apoptosis in hippocampal CA1, CA3 and dentate gyrus regions along with severely reduced pro-apoptotic factors (Bad, Bax and cleaved caspase-3) expression and raised levels of Bcl-2, Bcl-xL, survivin, xIAP and cIAPs (cell survival proteins) in plumbagin supplemented rats. Decrease in the levels of JNK, phospho-JNK, c-Jun and phospho-c-Jun with enhanced ERK1/2 levels was observed on plumbagin pretreatment. Down-regulated PI3K/Akt signalling following isoflurane was activated by plumbagin as evidenced by raised PI3K/Akt pathway proteins - mTORc1, Akt, phospho-Akt, GSK-3β, phospho-GSK-3β, PTEN and NF-κBp65 in the hippocampal tissues as detected by Western blotting. The mRNA levels were enhanced on plumbagin supplementation. CONCLUSIONS Plumbagin exerted its neuroprotective effects by effectively suppressing isoflurane-induced neuronal apoptosis via regulating BDNF-TrkB-PI3/Akt and ERK/JNK signalling.
Collapse
Affiliation(s)
- Jun-Hui Yuan
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Feng Pan
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Jie Chen
- Taizhou University Medical School, Taizhou, Zhejiang, China
| | - Cai-Er Chen
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Deng-Pan Xie
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Xing-Zhu Jiang
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Su-Juan Guo
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Jun Zhou
- Taizhou University Medical School, Taizhou, Zhejiang, China
| |
Collapse
|
26
|
Xu G, Huang YL, Li PL, Guo HM, Han XP. Neuroprotective effects of artemisinin against isoflurane-induced cognitive impairments and neuronal cell death involve JNK/ERK1/2 signalling and improved hippocampal histone acetylation in neonatal rats. ACTA ACUST UNITED AC 2017; 69:684-697. [PMID: 28294340 DOI: 10.1111/jphp.12704] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/12/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study was performed to assess the effect of artemisinin against isoflurane-induced neuronal apoptosis and cognitive impairment in neonatal rats. METHODS Artemisinin (50, 100 or 200 mg/kg b.wt/day; oral gavage) was administered to separate groups of neonatal rats starting from postnatal day 3 (P3) to postnatal day 21 (P21). On postnatal day 7 (P7), animals were exposed to inhalation anaesthetic isoflurane (0.75%) for 6 h. KEY FINDINGS Neuronal apoptosis following anaesthetic exposure was significantly reduced by artemisinin. Isoflurane-induced upregulated cleaved caspase-3, Bax and Bad expression were downregulated. Western blotting analysis revealed that treatment with artemisinin significantly enhanced the expression of anti-apoptotic proteins (Bcl-2, Bcl-xL, c-IAP-1, c-IAP-2, xIAP and survivin). Artemisinin increased the acetylation of H3K9 and H4K12 while reducing the expression of histone deacetlyases (HDACs) - HDAC-2 and HDAC-3. Isoflurane-induced activation of JNK signalling and downregulated ERK1/2 expression was effectively modulated by artemisinin. General behaviour of the animals in open-field and T-maze test were improved. Morris water maze test and object recognition test revealed better learning, working memory and also better memory retention on artemisinin treatment. CONCLUSIONS Artemisinin effectively inhibited neuronal apoptosis and improved cognition and memory via regulating histone acetylation and JNK/ERK1/2 signalling.
Collapse
Affiliation(s)
- Guang Xu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yun-Li Huang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ping-le Li
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai-Ming Guo
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue-Ping Han
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Li W, Li DY, Zhao SM, Zheng ZJ, Hu J, Li ZZ, Xiong SB. Rutin attenuates isoflurane-induced neuroapoptosis via modulating JNK and p38 MAPK pathways in the hippocampi of neonatal rats. Exp Ther Med 2017; 13:2056-2064. [PMID: 28565808 DOI: 10.3892/etm.2017.4173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/06/2016] [Indexed: 12/20/2022] Open
Abstract
An increasing number of infants and children undergo surgery and are exposed to anesthesia as a part of medical care each year. Isoflurane is a commonly used anesthetic in the pediatric population. However, previous studies have reported widespread isoflurane-induced neuroapoptosis and cognitive impairments in neonatal animal models, raising concerns over the administration of isoflurane in the pediatric population. The current study investigated the effects of rutin, a flavonoid, on isoflurane-induced neuroapoptosis in a neonatal rodent model. Groups of neonatal rat pups were administered rutin at doses of 10, 20 or 40 mg/kg body weight from postnatal day 1 (P1) to P15. On P7, pups were exposed to 0.75% isoflurane for 6 h. Rat pups in the control groups did not receive rutin, and did not receive anesthesia in one group. Neuroapoptosis following isoflurane exposure was determined by TUNEL assay. The expression levels of cleaved caspase-3, apoptotic pathway proteins [Bcl2-associated agonist of cell death (Bad), phospho-Bad, Bax, B-cell lymphoma 2 (Bcl-2) and Bcl-xL and mitogen-activated protein kinases (MAPK)] signalling pathway proteins [c-Jun N-terminal kinase (JNK), phospho-JNK, extracellular-signal-regulated kinase 1/2 (ERK1/2), phosphoERK1/2, p38, phospho-p38 and phospho-c-Jun], were determined by western blot analysis. The Morris water maze test was used to assess the learning and memory of pups on P30 and P31. The present study found that rutin at the tested doses of 10, 20 and 40 mg significantly reduced (P<0.05) the isoflurane-induced elevation in apoptotic cell count. The expression levels of caspase-3, Bad, Bax and MAPK proteins, which were increased following isoflurane treatment, were rescued by rutin treatment. Furthermore, rutin prevented the increase in Bcl-xL, Bcl-2 and phospho-Bad expression following isoflurane treatment, and enhanced the memory of the rats. Rutin provided neuroprotection against isoflurane-induced neuronal apoptosis and improved the learning and memory of rats by effectively regulating the expression levels of proteins in the MAPK pathway.
Collapse
Affiliation(s)
- Wei Li
- Hubei Cooperative Innovation Center for Industrial Fermentation, Hubei University of Technology, Wuhan, Hubei 430035, P.R. China
| | - De-Yuan Li
- Department of Nutrition and Food Research Institute, Wuhan Economic College, Wuhan, Hubei 430035, P.R. China
| | - Si-Ming Zhao
- Department of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430035, P.R. China
| | - Zhe-Jun Zheng
- Department of Nutrition and Food Research Institute, Wuhan Economic College, Wuhan, Hubei 430035, P.R. China
| | - Jie Hu
- Department of Nutrition and Food Research Institute, Wuhan Economic College, Wuhan, Hubei 430035, P.R. China
| | - Zong-Zhe Li
- Department of Nutrition and Food Research Institute, Wuhan Economic College, Wuhan, Hubei 430035, P.R. China
| | - Shan-Bai Xiong
- Department of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430035, P.R. China
| |
Collapse
|
28
|
Su ZY, Ye Q, Liu XB, Chen YZ, Zhan H, Xu SY. Dexmedetomidine mitigates isoflurane-induced neurodegeneration in fetal rats during the second trimester of pregnancy. Neural Regen Res 2017; 12:1329-1337. [PMID: 28966649 PMCID: PMC5607829 DOI: 10.4103/1673-5374.213554] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dexmedetomidine has significant neuroprotective effects. However, whether its protective effects can reduce neurotoxicity caused by isoflurane in fetal brain during the second trimester of pregnancy remains unclear. In this study, timed-pregnancy rats at gestational day 14 spontaneously inhaled 1.5% isoflurane for 4 hours, and were intraperitoneally injected with dexmedetomidine at dosages of 5, 10, 20, and 20 μg/kg 15 minutes before inhalation and after inhalation for 2 hours. Our results demonstrate that 4 hours after inhaling isoflurane, 20 μg/kg dexmedetomidine visibly mitigated isoflurane-induced neuronal apoptosis, reversed downregulation of brain-derived neurotrophic factor expression, and lessened decreased spatial learning and memory ability in adulthood in the fetal rats. Altogether, these findings indicate that dexmedetomidine can reduce neurodegeneration induced by isoflurane in fetal rats during the second trimester of pregnancy. Further, brain-derived neurotrophic factor participates in this process.
Collapse
Affiliation(s)
- Zhi-Yuan Su
- Department of Anesthesia, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Qing Ye
- Department of Anesthesia, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xian-Bao Liu
- Department of Anesthesia, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yu-Zhong Chen
- Department of Anesthesia, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Hong Zhan
- Department of Anesthesia, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Shi-Yuan Xu
- Department of Anesthesia, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
29
|
Si Y, Zhang Y, Han L, Chen L, Xu Y, Sun F, Ji M, Yang J, Bao H. Dexmedetomidine Acts via the JAK2/STAT3 Pathway to Attenuate Isoflurane-Induced Neurocognitive Deficits in Senile Mice. PLoS One 2016; 11:e0164763. [PMID: 27768775 PMCID: PMC5074497 DOI: 10.1371/journal.pone.0164763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022] Open
Abstract
Background Previous studies showed that isoflurane-induced cognitive deficits could be alleviated by dexmedetomidine in young animal subjects. In the current study, we examine whether dexmedetomidine could also alleviate isoflurane-induced cognitive deficits in senile animals. Methods Senile male C57BL/6 mice (20 months) received dexmedetomidine (50 μg/kg, i.p.) or vehicle 30 minutes prior to isoflurane exposure (1.3% for 4 h). Cognitive function was assessed 19 days later using a 5-day testing regimen with Morris water maze. Some subjects also received pretreatment with α2 adrenoreceptor antagonist atipamezole (250 μg/kg, i.p.), JAK2 inhibitor AG490 (15 mg/kg i.p.) or STAT3 inhibitor WP1066 (40 mg/kg i.p.) 30 minutes prior to dexmedetomidine. Results Isoflurane exposure increased and reduced the time spent in the quadrant containing the target platform in training sessions. The number of crossings over the original target quadrant was also decreased. Dexmedotomidine attenuated such effects. Effects of dexmedotomidine were reduced by pretreatment with atipamezole, AG490 and WP1066. Increased phosphorylation of JAK2 and STAT3 in the hippocampus induced by isoflurane was augmented by dexmedetomidine. Effects of dexmedetomidine on JAK2/STAT3 phosphorylation were attenuated by atipamezole, AG490 and WP1066. Isoflurane promoted neuronal apoptosis and increased the expression of cleaved caspase-3 and BAD, and reduced Bcl-2 expression. Attenuation of such effects by dexmedotomidine was partially blocked by atipamezole, AG490 and WP1066. Conclusion Dexmedetomidine could protect against isoflurane-induced spatial learning and memory impairment in senile mice by stimulating the JAK2/STAT3 signaling pathway. Such findings encourage the use of dexmedetomidine in geriatric patients receiving isoflurane anesthesia.
Collapse
Affiliation(s)
- Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liu Han
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lihai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yajie Xu
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Sun
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Muhuo Ji
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jianjun Yang
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, College of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- * E-mail:
| |
Collapse
|
30
|
Chen B, Deng X, Wang B, Liu H. Etanercept, an inhibitor of TNF-a, prevents propofol-induced neurotoxicity in the developing brain. Int J Dev Neurosci 2016; 55:91-100. [PMID: 27756568 DOI: 10.1016/j.ijdevneu.2016.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/18/2016] [Accepted: 10/10/2016] [Indexed: 11/19/2022] Open
Abstract
Propofol can induce acute neuronal apoptosis, neuronal loss or long-term cognitive impairment when exposed in neonatal rodents, but the mechanisms by which propofol induces developmental neurotoxicity are unclear. Recent studies have demonstrated that propofol can increase the TNF-α level in the developing brain, but there is a lack of direct evidence to show whether TNF-α is partially or fully involved in propofol-induced neurotoxicity. The present study shows that propofol exposure in neonatal rats induces an increase of TNF-α in the cerebral spinal fluid, hippocampus and prefrontal cortex (PFC). Etanercept, a TNF-α inhibitor, prevents propofol-induced short- or long-term neuronal apoptosis, neuronal loss, synaptic loss and long-term cognitive impairment. Furthermore, mTNF-α (precursor of TNF-α) expression in microglia cells is increased after propofol anaesthesia in either the hippocampus or PFC, but mTNF-α expression in neurons is only increased in the PFC. These findings suggest that TNF-α may mediate propofol-induced developmental neurotoxicity, and etanercept can provide neural protection. Microglia are the main cellular source of TNF-α after propofol exposure, while the synthesis of TNF-α in neurons is brain-region selective.
Collapse
Affiliation(s)
- Bo Chen
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China; Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Xiaoyuan Deng
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China
| | - Bin Wang
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China.
| |
Collapse
|
31
|
Ezaki J, Shimada R, Shibuya M, Kibayashi K. Hippocampal neuronal degeneration in the traumatic brain injury mouse: non-trivial effect of scalp incision. Neurol Res 2016; 38:994-1002. [PMID: 27615406 DOI: 10.1080/01616412.2016.1228746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES In experimental models of traumatic brain injury (TBI), posttraumatic hippocampal neuronal degeneration in the cornu ammonis 1 (CA1), and/or the cornu ammonis 3 (CA3) regions are regarded as the most notable phenotypic appearances relating to the pathophysiology of human post-concussion syndrome. However, these morphological changes are often also seen in subjects without TBI, namely 'sham' groups. The frequencies and reasons of appearance of hippocampal neuronal degeneration in mice with TBI and/or sham are not clear. METHODS We compared the frequencies of hippocampal neuronal degeneration among three groups: TBI (mice with external force impact performed by Marmarou's weight drop model after scalp incision), sham (mice with scalp incision alone), and control (mice with neither external force impact nor scalp incision), using hematoxylin and eosin stain in day 6 (n = 5 in each group.) Isoflurane was used for anesthesia in all mice. RESULTS The frequencies were 80, 100, and 20% in CA1, and 20, 40, and 60% in CA3, for TBI, sham, and control, respectively. In CA1, a significant difference of the frequency was observed between sham and control (p = 0.048), but not, between TBI and sham (p = 1.000) in Fisher's exact test. In CA3, no significant difference in the frequency was observed between the three groups. CONCLUSION Scalp incision, rather than external impact force, might affect the CA1 hippocampal neuronal degeneration in mice with TBI. In addition, factor(s) other than external impact force or scalp incision may also cause hippocampal neuronal degeneration in both CA1 and CA3. Careful interpretation is needed concerning hippocampal neuronal degeneration induced by a weight drop device observed in mice with TBI.
Collapse
Affiliation(s)
- Jiro Ezaki
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Ryo Shimada
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Misato Shibuya
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Kazuhiko Kibayashi
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| |
Collapse
|
32
|
Hua FZ, Ying J, Zhang J, Wang XF, Hu YH, Liang YP, Liu Q, Xu GH. Naringenin pre-treatment inhibits neuroapoptosis and ameliorates cognitive impairment in rats exposed to isoflurane anesthesia by regulating the PI3/Akt/PTEN signalling pathway and suppressing NF-κB-mediated inflammation. Int J Mol Med 2016; 38:1271-80. [DOI: 10.3892/ijmm.2016.2715] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 07/21/2016] [Indexed: 11/06/2022] Open
|
33
|
Wang LY, Tang ZJ, Han YZ. Neuroprotective effects of caffeic acid phenethyl ester against sevoflurane‑induced neuronal degeneration in the hippocampus of neonatal rats involve MAPK and PI3K/Akt signaling pathways. Mol Med Rep 2016; 14:3403-12. [PMID: 27498600 DOI: 10.3892/mmr.2016.5586] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/24/2016] [Indexed: 11/06/2022] Open
Abstract
Millions of infants and children are exposed to anesthesia every year during medical care. Sevoflurane is a volatile anesthetic that is frequently used for pediatric anesthesia. However, previous reports have suggested that the administration of sevoflurane promotes neurodegeneration, raising concerns regarding the safety of its usage. The present study aimed to investigate caffeic acid phenethyl ester (CAPE) and its protective effect against sevoflurane‑induced neurotoxicity in neonatal rats. Rat pups were administered with CAPE at 10, 20 or 40 mg/kg body weight from postnatal day 1 (P1) to P15. The P7 rats were exposed to sevoflurane (2.9%) for 6 h. Control group rats received no sevoflurane or CAPE. Neuronal apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick‑end labeling assay. The expression levels of caspases (caspase‑3, ‑8 and ‑9), apoptotic pathway proteins [Bcl‑2‑associated X protein (Bax), B cell CCL/lymphoma 2 (Bcl‑2), Bcl‑2‑like 1 (Bcl‑xL), Bcl‑2‑associated agonist of cell death (Bad) and phosphorylated (p)‑Bad], mitogen‑activated protein kinases (MAPK) signaling pathway proteins [c‑Jun N‑terminal kinase (JNK), p‑JNK, extracellular signal‑regulated kinase (ERK)1/2, p‑ERK1/2, p38, p‑p38 and p‑c‑Jun] and the phosphoinositide 3‑kinase (PI3K)/Akt cascade were evaluated by western blotting following sevoflurane and CAPE treatment. In addition, the expression of cleaved caspase‑3 was analyzed by immunohistochemistry. CAPE significantly reduced sevoflurane‑induced apoptosis, downregulated the expression levels of caspases and pro‑apoptotic proteins (Bax and Bad) and elevated the expression levels of Bcl‑2 and Bcl‑xL when compared with sevoflurane treatment. Furthermore, CAPE appeared to modify the expression levels of MAPKs and activate the PI3K/Akt signaling pathway. Thus, the present study demonstrated that CAPE effectively inhibited sevoflurane‑induced neuroapoptosis by modulating the expression and phosphorylation of apoptotic pathway proteins and MAPKs, and by regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Li-Yan Wang
- Department of Pediatric Surgery, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Zhi-Jun Tang
- Department of Orthopedics in Repair and Reconstruction, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Yu-Zeng Han
- Department of Pediatric Internal Medicine, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
34
|
Yarza R, Vela S, Solas M, Ramirez MJ. c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for Alzheimer's Disease. Front Pharmacol 2016; 6:321. [PMID: 26793112 PMCID: PMC4709475 DOI: 10.3389/fphar.2015.00321] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/28/2015] [Indexed: 01/08/2023] Open
Abstract
c-Jun N-terminal kinases (JNKs) are a family of protein kinases that play a central role in stress signaling pathways implicated in gene expression, neuronal plasticity, regeneration, cell death, and regulation of cellular senescence. It has been shown that there is a JNK pathway activation after exposure to different stressing factors, including cytokines, growth factors, oxidative stress, unfolded protein response signals or Aβ peptides. Altogether, JNKs have become a focus of screening strategies searching for new therapeutic approaches to diabetes, cancer or liver diseases. In addition, activation of JNK has been identified as a key element responsible for the regulation of apoptosis signals and therefore, it is critical for pathological cell death associated with neurodegenerative diseases and, among them, with Alzheimer’s disease (AD). In addition, in vitro and in vivo studies have reported alterations of JNK pathways potentially associated with pathogenesis and neuronal death in AD. JNK’s, particularly JNK3, not only enhance Aβ production, moreover it plays a key role in the maturation and development of neurofibrillary tangles. This review aims to explain the rationale behind testing therapies based on inhibition of JNK signaling for AD in terms of current knowledge about the pathophysiology of the disease. Keeping in mind that JNK3 is specifically expressed in the brain and activated by stress-stimuli, it is possible to hypothesize that inhibition of JNK3 might be considered as a potential target for treating neurodegenerative mechanisms associated with AD.
Collapse
Affiliation(s)
- Ramon Yarza
- Department of Pharmacology and Toxicology, University of Navarra Pamplona, Spain
| | - Silvia Vela
- Department of Pharmacology and Toxicology, University of Navarra Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of NavarraPamplona, Spain; Navarra Institute for Health ResearchPamplona, Spain
| | - Maria J Ramirez
- Department of Pharmacology and Toxicology, University of NavarraPamplona, Spain; Navarra Institute for Health ResearchPamplona, Spain
| |
Collapse
|
35
|
Su SH, Wu YF, Lin Q, Yu F, Hai J. Cannabinoid receptor agonist WIN55,212-2 and fatty acid amide hydrolase inhibitor URB597 suppress chronic cerebral hypoperfusion-induced neuronal apoptosis by inhibiting c-Jun N-terminal kinase signaling. Neuroscience 2015; 301:563-75. [DOI: 10.1016/j.neuroscience.2015.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/13/2015] [Accepted: 03/11/2015] [Indexed: 11/15/2022]
|
36
|
Toxic and protective effects of inhaled anaesthetics on the developing animal brain: systematic review and update of recent experimental work. Eur J Anaesthesiol 2015; 31:669-77. [PMID: 24922049 DOI: 10.1097/eja.0000000000000073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Accumulating preclinical data indicate that neonatal exposure to general anaesthetics is detrimental to the central nervous system. Some studies, however, display potential protective effects of exactly the same anaesthetic agents on the immature brain. The effects of inhaled anaesthetics on the developing brain have received close attention from researchers, clinicians and the public in recent decades. OBJECTIVES To summarise the preclinical evidence reported in the last 5 years on both the deleterious effects and the neuroprotective potential in special indications, of inhaled anaesthetics on the developing brain. DESIGN A systematic review. DATA SOURCES PubMed search performed in June 2013. ELIGIBILITY CRITERIA Search terms included brain, development, inhaled anaesthetic, toxicity and protection within the scope of the last 5 years with animals. The reference lists of relevant articles and recent reviews were also hand-searched for additional studies. The type, dose and exposure duration of anaesthetics, species and age of animals, histopathologic indicators, outcomes and affected brain areas, neuro developmental test modules and outcomes, as well as other outcomes and comments were summarised. RESULTS Two hundred and nineteen relevant titles were initially revealed. In total, 81 articles were identified, with 68 articles assessing the detrimental effects induced by inhaled anaesthetics in the immature brain along with possible treatments. The remaining 13 articles focused on the protective profile of inhaled anaesthetics on perinatal hypoxic-ischaemic brain injury. Administration of inhaled anaesthetic agents to the immature brain was shown to be deleterious in several preclinical studies. In perinatal hypoxic-ischaemic brain injury models, pre- and postconditioning of inhalational anaesthetics exerted neuroprotective effects. CONCLUSION The majority of studies have linked inhaled anaesthetics to toxic effects in the neonatal brain of rodents, piglets and primates. Only a few studies, however, could demonstrate long-lasting cognitive impairment. The results of inhalational anaesthetic-induced neuroprotection in perinatal hypoxic-ischaemic brain injury are a promising basis for more research in this field. In general, prospective clinical trials are needed to further differentiate the effects of inhaled anaesthetics on the immature brain.
Collapse
|
37
|
McAdams RM, McPherson RJ, Beyer RP, Bammler TK, Farin FM, Juul SE. Dose-dependent effects of morphine exposure on mRNA and microRNA (miR) expression in hippocampus of stressed neonatal mice. PLoS One 2015; 10:e0123047. [PMID: 25844808 PMCID: PMC4386824 DOI: 10.1371/journal.pone.0123047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/18/2015] [Indexed: 12/02/2022] Open
Abstract
Morphine is used to sedate critically ill infants to treat painful or stressful conditions associated with intensive care. Whether neonatal morphine exposure affects microRNA (miR) expression and thereby alters mRNA regulation is unknown. We tested the hypothesis that repeated morphine treatment in stress-exposed neonatal mice alters hippocampal mRNA and miR expression. C57BL/6 male mice were treated from postnatal day (P) 5 to P9 with morphine sulfate at 2 or 5 mg/kg ip twice daily and then exposed to stress consisting of hypoxia (100% N2 1 min and 100% O2 5 min) followed by 2h maternal separation. Control mice were untreated and dam-reared. mRNA and miR expression profiling was performed on hippocampal tissues at P9. Overall, 2 and 5 mg/kg morphine treatment altered expression of a total of 150 transcripts (>1.5 fold change, P<0.05) from which 100 unique mRNAs were recognized (21 genes were up- and 79 genes were down-regulated), and 5 mg/kg morphine affected 63 mRNAs exclusively. The most upregulated mRNAs were fidgetin, arginine vasopressin, and resistin-like alpha, and the most down-regulated were defensin beta 11, aquaporin 1, calmodulin-like 4, chloride intracellular channel 6, and claudin 2. Gene Set Enrichment Analysis revealed that morphine treatment affected pathways related to cell cycle, membrane function, signaling, metabolism, cell death, transcriptional regulation, and immune response. Morphine decreased expression of miR-204-5p, miR-455-3p, miR-448-5p, and miR-574-3p. Nine morphine-responsive mRNAs that are involved in neurodevelopment, neurotransmission, and inflammation are predicted targets of the aforementioned differentially expressed miRs. These data establish that morphine produces dose-dependent changes in both hippocampal mRNA and miR expression in stressed neonatal mice. If permanent, morphine–mediated neuroepigenetic effects may affect long-term hippocampal function, and this provides a mechanism for the neonatal morphine-related impairment of adult learning.
Collapse
Affiliation(s)
- Ryan M. McAdams
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Ronald J. McPherson
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| | - Richard P. Beyer
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K. Bammler
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Frederico M. Farin
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Sandra E. Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
38
|
Li L, Yu Q, Liang W. Molecular pathways of mitochondrial dysfunctions: Possible cause of cell death in anesthesia-induced developmental neurotoxicity. Brain Res Bull 2015; 110:14-9. [DOI: 10.1016/j.brainresbull.2014.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 02/05/2023]
|
39
|
22nd annual meeting of Chinese Society of Anesthesiology. Br J Anaesth 2014. [DOI: 10.1093/bja/aeu337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
40
|
Liao Z, Cao D, Han X, Liu C, Peng J, Zuo Z, Wang F, Li Y. Both JNK and P38 MAPK pathways participate in the protection by dexmedetomidine against isoflurane-induced neuroapoptosis in the hippocampus of neonatal rats. Brain Res Bull 2014; 107:69-78. [PMID: 25026397 DOI: 10.1016/j.brainresbull.2014.07.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 12/12/2022]
Abstract
Dexmedetomidine, a highly selective α2-adrenergic agonist, has been reported to attenuate isoflurane-induced cognitive impairment and neuroapoptosis. However, the underlying molecular mechanisms remain poorly understood. The aim of this study was to investigate whether mitogen-activated protein kinase (MAPK) pathway was involved in dexmedetomidine-induced neuroprotection against isoflurane effects. Seven-day-old (P7) neonatal Sprague-Dawley rats were pretreated with various concentrations of dexmedetomidine, and then exposed to 0.75% isoflurane or air for 6h. Terminal deoxyribonucleotide transferase-mediated dUTP nick end labeling (TUNEL) was used to detect neuronal apoptosis in their hippocampus. Activated caspase-3, extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun NH2-terminal kinases (JNK), p38, phospho-ERK1/2, phospho-JNK and phospho-p38 proteins were detected by Western blotting in the hippocampus at the end of exposure. Also, P7 rats were pretreated with 75 μg/kg dexmedetomidine alone, or given the ERK inhibitor U0126 before dexmedetomidine pretreatment, or pretreated with the p38 MAPK inhibitor SB203580 or JNK inhibitor SP600125 alone, and then exposed to 0.75% isoflurane for 6h. Isoflurane induced significant neuroapoptosis, increased the protein expression of phospho-JNK, phospho-c-Jun, phospho-p38 and phospho-nuclear factor-κB (NF-κB), decreased the level of phospho-ERK1/2 protein and reduced the ratio of Bcl-2/Bax in the hippocampus. Dexmedetomidine pretreatment inhibited isoflurane-induced neuroapoptosis and restored proteins expression of MAPK pathways and the Bcl-2/Bax ratio after isoflurane exposure. Moreover, SB203580 and SP600125 also partly attenuated the isoflurane-induced protein changes. However, U0126 did not reverse dexmedetomidine-induced neuroprotection. Our results indicate that the JNK and p38 pathways, not the ERK pathway are involved in dexmedetomidine-induced neuroprotection against isoflurane effects.
Collapse
Affiliation(s)
- Zhaoxia Liao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Dexiong Cao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Chuiliang Liu
- Department of Anesthesiology, ChanCheng Center Hospital, Foshan 528030, China.
| | - Jun Peng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Zhiyi Zuo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Anesthesiology, University of Virginia Health System, PO Box 800710, Charlottesville, VA 22908-0710, USA.
| | - Fei Wang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
41
|
Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya YK, Chauhan LKS, Dwivedi HN, Pant AB, Khanna VK. Unraveling the mechanism of neuroprotection of curcumin in arsenic induced cholinergic dysfunctions in rats. Toxicol Appl Pharmacol 2014; 279:428-440. [PMID: 24952339 DOI: 10.1016/j.taap.2014.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/20/2022]
Abstract
Earlier, we found that arsenic induced cholinergic deficits in rat brain could be protected by curcumin. In continuation to this, the present study is focused to unravel the molecular mechanisms associated with the protective efficacy of curcumin in arsenic induced cholinergic deficits. Exposure to arsenic (20mg/kg body weight, p.o) for 28 days in rats resulted to decrease the expression of CHRM2 receptor gene associated with mitochondrial dysfunctions as evident by decrease in the mitochondrial membrane potential, activity of mitochondrial complexes and enhanced apoptosis both in the frontal cortex and hippocampus in comparison to controls. The ultrastructural images of arsenic exposed rats, assessed by transmission electron microscope, exhibited loss of myelin sheath and distorted cristae in the mitochondria both in the frontal cortex and hippocampus as compared to controls. Simultaneous treatment with arsenic (20mg/kg body weight, p.o) and curcumin (100mg/kg body weight, p.o) for 28 days in rats was found to protect arsenic induced changes in the mitochondrial membrane potential and activity of mitochondrial complexes both in frontal cortex and hippocampus. Alterations in the expression of pro- and anti-apoptotic proteins and ultrastructural damage in the frontal cortex and hippocampus following arsenic exposure were also protected in rats simultaneously treated with arsenic and curcumin. The data of the present study reveal that curcumin could protect arsenic induced cholinergic deficits by modulating the expression of pro- and anti-apoptotic proteins in the brain. More interestingly, arsenic induced functional and ultrastructural changes in the brain mitochondria were also protected by curcumin.
Collapse
Affiliation(s)
- Pranay Srivastava
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajesh S Yadav
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India; Department of Crimnology and Forensic Science, Harisingh Gour University, Sagar 470 003, India
| | - Lalit P Chandravanshi
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajendra K Shukla
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Yogesh K Dhuriya
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Lalit K S Chauhan
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Hari N Dwivedi
- Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow 227 015, India
| | - Aditiya B Pant
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Vinay K Khanna
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India.
| |
Collapse
|
42
|
Li Y, Zeng M, Chen W, Liu C, Wang F, Han X, Zuo Z, Peng S. Dexmedetomidine reduces isoflurane-induced neuroapoptosis partly by preserving PI3K/Akt pathway in the hippocampus of neonatal rats. PLoS One 2014; 9:e93639. [PMID: 24743508 PMCID: PMC3990549 DOI: 10.1371/journal.pone.0093639] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/04/2014] [Indexed: 12/03/2022] Open
Abstract
Prolonged exposure to volatile anesthetics, such as isoflurane and sevoflurane, causes neurodegeneration in the developing animal brains. Recent studies showed that dexmedetomidine, a selective α2-adrenergic agonist, reduced isoflurane-induced cognitive impairment and neuroapoptosis. However, the mechanisms for the effect are not completely clear. Thus, we investigated whether exposure to isoflurane or sevoflurane at an equivalent dose for anesthesia during brain development causes different degrees of neuroapoptosis and whether this neuroapoptosis is reduced by dexmedetomidine via effects on PI3K/Akt pathway that can regulate cell survival. Seven-day-old (P7) neonatal Sprague-Dawley rats were randomly exposed to 0.75% isoflurane, 1.2% sevoflurane or air for 6 h. Activated caspase-3 was detected by immunohistochemistry and Western blotting. Phospho-Akt, phospho-Bad, Akt, Bad and Bcl-xL proteins were detected by Western blotting in the hippocampus at the end of exposure. Also, P7 rats were pretreated with various concentrations of dexmedetomidine alone or together with PI3K inhibitor LY294002, and then exposed to 0.75% isoflurane. Terminal deoxyribonucleotide transferase-mediated dUTP nick end labeling (TUNEL) and activated caspase-3 were used to detect neuronal apoptosis in their hippocampus. Isoflurane, not sevoflurane at the equivalent dose, induced significant neuroapoptosis, decreased the levels of phospho-Akt and phospho-Bad proteins, increased the expression of Bad protein and reduced the ratio of Bcl-xL/Bad in the hippocampus. Dexmedetomidine pretreatment dose-dependently inhibited isoflurane-induced neuroapoptosis and restored protein expression of phospho-Akt and Bad as well as the Bcl-xL/Bad ratio induced by isoflurane. Pretreatment with single dose of 75 µg/kg dexmedetomidine provided a protective effect similar to that with three doses of 25 µg/kg dexmedetomidine. Moreover, LY294002, partly inhibited neuroprotection of dexmedetomidine. Our results suggest that dexmedetomidine pretreatment provides neuroprotection against isoflurane-induced neuroapoptosis in the hippocampus of neonatal rats by preserving PI3K/Akt pathway activity.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (YL); (SP)
| | - Minting Zeng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiqiang Chen
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Anesthesiology, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Chuiliang Liu
- Department of Anesthesiology, ChanCheng Center Hospital, Foshan, Guangdong, China
| | - Fei Wang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhiyi Zuo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Shuling Peng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (YL); (SP)
| |
Collapse
|