1
|
Borzage MT, Peterson BS. A Scoping Review of the Mechanisms Underlying Developmental Anesthetic Neurotoxicity. Anesth Analg 2025; 140:409-426. [PMID: 38536739 PMCID: PMC11427602 DOI: 10.1213/ane.0000000000006897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 09/28/2024]
Abstract
Although anesthesia makes painful or uncomfortable diagnostic and interventional health care procedures tolerable, it may also disrupt key cellular processes in neurons and glia, harm the developing brain, and thereby impair cognition and behavior in children. Many years of studies using in vitro, animal behavioral, retrospective database studies in humans, and several prospective clinical trials in humans have been invaluable in discerning the potential toxicity of anesthetics. The objective of this scoping review was to synthetize the evidence from preclinical studies for various mechanisms of toxicity across diverse experimental designs and relate their findings to those of recent clinical trials in real-world settings.
Collapse
Affiliation(s)
- Matthew Thomas Borzage
- From the Fetal and Neonatal Institute, Division of Neonatology, Children’s Hospital Los Angeles, Los Angeles, California
| | - Bradley S. Peterson
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for the Developing Mind, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, California
| |
Collapse
|
2
|
Cheng H, Men Y, An Y, Yu J, Zhang G, Li J, Wang X, Sun G, Wu Y. Overexpression of endothelial S1pr2 promotes blood-brain barrier disruption via JNK/c-Jun/MMP-9 pathway after traumatic brain injury in both in vivo and in vitro models. Front Pharmacol 2024; 15:1448570. [PMID: 39679379 PMCID: PMC11637860 DOI: 10.3389/fphar.2024.1448570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/25/2024] [Indexed: 12/17/2024] Open
Abstract
Objectives The disruption of blood-brain barrier (BBB) is associated with poor outcomes of TBI patients. Sphingosine-1-phosphate receptor 2 (S1pr2), a member of the G protein-coupled receptor family, is involved in endothelial activation and the regulation of vascular integrity. We hypothesized that the inhibition of S1pr2 may alleviate BBB disruption and explored potential underlying molecular mechanisms. Methods Lesion volumes were assessed utilizing Nissl staining; neurological outcomes were evaluated through a battery of neurobehavioral assessments; phenotype-associated proteins were scrutinized via Western blot analysis; levels of reactive oxygen species (ROS), neuronal apoptosis, and S1pr2 expression were determined using immunofluorescence staining. The impact of S1pr2 inhibition after TBI and its underlying mechanism were elucidated using the selective S1pr2 inhibitor JTE-013, the JNK phosphorylation inhibitor SP600125, and cellular models. Chip-qPCR was employed to further elucidate the binding sites of the transcription factor c-Jun. Results The expression of S1pr2 significantly increased following TBI in mice. Pharmacological inhibition of S1pr2 alleviated secondary injury with reduced lesion volume, ROS generation, cerebral oedema, neurological deficits, and neuronal apoptosis; BBB disruption was also mitigated, accompanied by reduced degradation of tight junction proteins and decreased induction of matrix metalloproteinases-9 (MMP-9) post-TBI. Mechanistically, TBI induces an increase in S1pr2 specifically in endothelial cells, leading to the promotion of MMP-9 transactivation by enhancing JNK/c-Jun signaling. This results in the degradation of tight junction proteins and increased BBB permeability. Through in vitro and in vivo Chip-qPCR experiments, we verified that AP-1a and AP-1b of MMP-9 promoter function as binding sites for phosphorylated c-Jun. Conclusion Our findings identify a previously undisclosed role of S1pr2 in the pathophysiology of TBI. The S1pr2 inhibition presents a novel approach to alleviate BBB disruption after TBI through regulating the JNK/c-Jun/MMP-9 pathway.
Collapse
Affiliation(s)
- Hongbo Cheng
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yijiao Men
- Emergency Department, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaqing An
- Emergency Department, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiegang Yu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gengshen Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaming Li
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoliang Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
3
|
Hao X, Li Y, Gao H, Wang Z, Fang B. Inhalation Anesthetics Play a Janus-Faced Role in Self-Renewal and Differentiation of Stem Cells. Biomolecules 2024; 14:1167. [PMID: 39334933 PMCID: PMC11430341 DOI: 10.3390/biom14091167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Inhalation anesthesia stands as a pivotal modality within clinical anesthesia practices. Beyond its primary anesthetic effects, inhaled anesthetics have non-anesthetic effects, exerting bidirectional influences on the physiological state of the body and disease progression. These effects encompass impaired cognitive function, inhibition of embryonic development, influence on tumor progression, and so forth. For many years, inhaled anesthetics were viewed as inhibitors of stem cell fate regulation. However, there is now a growing appreciation that inhaled anesthetics promote stem cell biological functions and thus are now regarded as a double-edged sword affecting stem cell fate. In this review, the effects of inhaled anesthetics on self-renewal and differentiation of neural stem cells (NSCs), embryonic stem cells (ESCs), and cancer stem cells (CSCs) were summarized. The mechanisms of inhaled anesthetics involving cell cycle, metabolism, stemness, and niche of stem cells were also discussed. A comprehensive understanding of these effects will enhance our comprehension of how inhaled anesthetics impact the human body, thus promising breakthroughs in the development of novel strategies for innovative stem cell therapy approaches.
Collapse
Affiliation(s)
- Xiaotong Hao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Hairong Gao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
4
|
Lu P, Liang F, Dong Y, Xie Z, Zhang Y. Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration. Int J Mol Sci 2023; 24:ijms24076746. [PMID: 37047719 PMCID: PMC10095407 DOI: 10.3390/ijms24076746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
Collapse
Affiliation(s)
- Pan Lu
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
5
|
Jin Y, Hu X, Meng F, Luo Q, Liu H, Yang Z. Sevoflurane Exposure of Clinical Doses in Pregnant Rats Induces Vcan Changes without Significant Neural Apoptosis in the Offspring. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020190. [PMID: 36837392 PMCID: PMC9965787 DOI: 10.3390/medicina59020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/17/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023]
Abstract
Background and Objectives: Sevoflurane is a commonly used inhalational anaesthetic in clinics. Prolonged exposure to sevoflurane can induce significant changes in lipid metabolism and neuronal damage in the developing brain. However, the effect of exposure of pregnant rats to clinical doses of sevoflurane remains unclear. Materials and Methods: Twenty-eight pregnant rats were randomly and equally divided into sevoflurane exposure (S) group, control (C) and a blank group at gestational day (G) 18; Rats in S group received 2% sevoflurane with 98% oxygen for 6 h in an anesthetizing chamber, while C group received 100% oxygen at an identical flow rate for 6 h in an identical chamber. Partial least squares discriminant analysis (PLS-DA), ultra performance liquid chromatography/time-of-flight mass spectrometry(UPLC/TOF-MS) and MetaboAnalyst were used to analysis acquire metabolomics profiles, and immunohistochemical changes of neuronalapoptosis in hippocampus and cortex of neonatal rats were also analyzed. Results: This study aimed to explore lipidomics and transcriptomics changes related to 2% sevoflurane exposure for 6 h in the developing brains of newborn offspring rats. Ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC/TOF-MS) and RNA sequencing (RNA-seq) analyses were used to acquire metabolomics and transcriptomics profiles. We used RNA-seq to analyse the expression of the coding and non-coding transcripts in neural cells of the cerebral cortex. No significant differences in arterial oxygen tension (PaO2), arterial carbon dioxide tension (PaCO2), or arterial blood gas were found between the groups. The relative standard deviation (RSD) of retention times was <1.53%, and the RSDs of peak areas ranged from 2.13% to 8.51%. Base peak chromatogram (BPC) profiles showed no differences between the groups. We evaluated the partial least square-discriminant analysis (PLS-DA) model. In negative ion mode, R2X was over 70%, R2Y was over 93%, and Q2 (cum) was over 80%. Cell apoptosis was not remarkably enhanced by TUNEL and haematoxylin and eosin (HE) staining in the sevoflurane-exposed group compared to the control group (p > 0.05). Glycerophospholipid (GP) and sphingolipid metabolism disturbances might adversely influence neurodevelopment in offspring. The expression of mRNAs (Vcan gene, related to neuronal development, function and repair) of the sevoflurane group was significantly increased in the differential genes by qRT-PCR verification. Conclusions: GP and sphingolipid metabolism homeostasis may be potential therapeutic approaches against inhalational anaesthetic-induced neurodegenerative disorders. Meanwhile, sevoflurane-induced Vcan changes indicated some lipidomic and transcriptomic changes, even if neural cell apoptosis was not significantly changed in the usual clinical dose of sevoflurane exposure.
Collapse
Affiliation(s)
- Yi Jin
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200025, China
- Shanghai Municipal Key Clinical Specialty, Shanghai 200025, China
- Department of Anesthesiology, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Xiaoxue Hu
- Department of Anesthesiology, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Fanhua Meng
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qing Luo
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Henry Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, The University of Pennsylvania, 3401 Spruce Street, Philadelphia, PA 19104, USA
- Correspondence: (H.L.); (Z.Y.)
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200025, China
- Shanghai Municipal Key Clinical Specialty, Shanghai 200025, China
- Correspondence: (H.L.); (Z.Y.)
| |
Collapse
|
6
|
Lee JA, Bae DH, Choi WH, Cho CH, Bang YS, Yoo J. Effects of Sevoflurane Exposure on Fetal Brain Development Using Cerebral Organoids. J Mol Neurosci 2022; 72:2440-2450. [PMID: 36478139 DOI: 10.1007/s12031-022-02080-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
Sevoflurane is a safe and well-known inhaled anesthetic. Given that sevoflurane can be delivered to developing fetuses through the mother, it is critical to determine whether this agent affects fetal neurodevelopment. Recent research has sought to determine whether sevoflurane affects fetal brain development when the mother is exposed during the second to third trimester of pregnancy, considered to be the crucial period for the development of nervous system. However, even though the first trimester is a critical period for fetal organogenesis and the most susceptible time to teratogen exposure, research regarding the effects of sevoflurane on organogenesis, especially on brain development, is insufficient. In the present study, human embryonic stem cells (hESC)-derived cerebral organoids were exposed to sevoflurane during the time corresponding to the first trimester to investigate the effect of early sevoflurane exposure on fetal brain development, specifically the processes of neuronal differentiation and maturation. Organoid size exposed to the intermediate concentration of sevoflurane did not differ from control, immunofluorescence demonstrated that sevoflurane temporarily decreased the size of SOX2 + /N-cad + ventricular zone structures only during the mid-time point, and upregulated expression of TUJ1 and MAP2 only during the early time point. However, all markers returned to normal levels, and organoids formed normal cortical structures at the late time point. Our results suggest that maternal sevoflurane exposure during the first trimester of pregnancy can cause abnormal neuronal differentiation in the fetal brain. However, considering the recovery observed in later periods, sevoflurane exposure might not have lasting impacts on fetal brain development.
Collapse
Affiliation(s)
- Jae A Lee
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Dong Hyuck Bae
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Woo Hee Choi
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea
| | - Chang-Hoon Cho
- R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea
| | - Yun-Sic Bang
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, CHA Bundang Medical Center, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| | - Jongman Yoo
- Department of Microbiology and CHA Organoid Research Center, CHA University School of Medicine, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea. .,R&D Institute, ORGANOIDSCIENCES, Ltd., Seongnam, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
7
|
Zhang J, Chang Q, Rizzello L, Wu Y. Research progress on the effects and mechanisms of anesthetics on neural stem cells. IBRAIN 2022; 8:453-464. [PMID: 37786590 PMCID: PMC10528967 DOI: 10.1002/ibra.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 10/04/2023]
Abstract
Exposure to anesthetic drugs has been proven to seriously affect developing animals in terms of neural stem cells' (NSCs') proliferation, differentiation, and apoptosis. This can severely hamper the development of physiological learning and memory skills. Studies on the effects of anesthetics on NSCs' proliferation and differentiation are thus reviewed here, with the aim to highlight which specific drug mechanisms are the least harmful to NSCs. PubMed has been used as the preferential searching database of relevant literature to identify studies on the effects and mechanisms of NSCs' proliferation and differentiation. It was concluded that propofol and sevoflurane may be the safest options for NSCs during pregnancy and in pediatric clinical procedures, while dexmedetomidine has been found to reduce opioid-related damage in NSCs. It was also found that the growth environment may impact neurodevelopment even more than narcotic drugs. Nonetheless, the current scientific literature available further highlights how more extensive clinical trials are absolutely required for corroborating the conclusion drawn here.
Collapse
Affiliation(s)
- Ji Zhang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Quan‐Yuan Chang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Loris Rizzello
- Department of Pharmaceutical SciencesUniversity of MilanMilanItaly
- National Institute of Molecular Genetics (INGM)MilanItaly
| | - You Wu
- Department of Family PlanningThe Affiliated Hospital of Zunyi Medical UniversityGuizhouZunyiChina
| |
Collapse
|
8
|
Li Q, Zhang X, Li S, Li W, Teng Y, Zhou Y, Xiong H. Carnosol alleviates sevoflurane-induced cognitive dysfunction by mediating NF-κB pathway in aged rats. Drug Dev Res 2022; 83:1342-1350. [PMID: 35781309 DOI: 10.1002/ddr.21963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/06/2022] [Accepted: 06/07/2022] [Indexed: 11/11/2022]
Abstract
Postoperative Cognitive Dysfunction (POCD) is a neurological disorder of unconsciousness due to cognitive regression after surgical anesthesia. However, the specific mechanism has not yet been clarified. Sevoflurane (SEV) is one of the most commonly used anesthetics in clinical practice, and how SEV mediates the generation of POCD is unclear. Carnosol, a natural ingredient, has been reported to have various beneficial effects such as anti-inflammatory, immune enhancement, and so forth, but how it ameliorates SEV-mediated neurotoxicity remains unclear. This study aimed to induce a POCD model in aged rats by SEV and to elucidate how Carnosol ameliorated SEV-mediated neurotoxicity. The effects of Carnosol on the expression of inflammatory factors in rat hippocampus mediated by SEV were determined by enzyme-linked immunoassay and polymerase chain reaction experiments; the effects of Carnosol on the expressions of Iba-1 and glial fibrillary acidic protein after SEV-mediated activation of rat microglia were clarified by immunofluorescence and Western blotting (WB); The effects of Carnosol on SEV-mediated neuronal apoptosis were studied by terminal deoxynucleotidyl transferase dUTP nick end labeling and WB; the specific signaling pathways regulated by Carnosol were elucidated by WB. The results showed that Carnosol can improve the cognitive dysfunction and reduce neuroinflammation in aged rats induced by SEV; Carnosol can reduce the activation of microglia and inhibit neuronal apoptosis in aged rats induced by SEV; Carnosol can phosphorylate p65 and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha regulates the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. Carnosol can attenuate SEV-induced neuroinflammation, prevent microglial activation and inhibit neuronal apoptosis by modulating the NF-κB pathway.
Collapse
Affiliation(s)
- Qing Li
- Department of Anesthesiology, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Xianjie Zhang
- Department of Anesthesiology, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Siyuan Li
- Department of Anesthesia and Comfort Health Center, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Wen Li
- Department of Anesthesiology, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Yunpeng Teng
- Department of Anesthesia and Comfort Health Center, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Yukai Zhou
- Department of Anesthesiology, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Hongfei Xiong
- Department of Anesthesia and Comfort Health Center, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Yu Y, Zhang W, Zhu D, Wang H, Shao H, Zhang Y. LncRNA Rian ameliorates sevoflurane anesthesia-induced cognitive dysfunction through regulation of miR-143-3p/LIMK1 axis. Hum Cell 2021; 34:808-818. [PMID: 33616869 DOI: 10.1007/s13577-021-00502-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/02/2021] [Indexed: 02/05/2023]
Abstract
Sevoflurane could stimulate neurotoxicity and result in postoperative cognitive dysfunction (POCD). Long non-coding RNAs (lncRNAs) have been implicated in the regulation of nervous system disease. This study was performed to investigate role and mechanism of lncRNA Rian (RNA imprinted and accumulated in nucleus) in sevoflurane anesthesia-induced cognitive dysfunction. Mice post-sevoflurane anesthesia showed cognitive impairments and neuronal damage and apoptosis. However, intracerebroventricularly injection with Adenovirus (Ad) for the over-expression of Rian ameliorated sevoflurane-induced neuronal damage and apoptosis. Cognitive impairments induced by sevoflurane were attenuated by injection with Ad-Rian. Moreover, transfection with Ad-Rian also protected isolated primary hippocampal neurons against sevoflurane-induced decrease of cell viability and increase of lactic acid dehydrogenase (LDH) and apoptosis. Mechanistically, Rian bind to miR-143-3p, and decreased expression of LIMK1 (Lim kinase 1) through negative regulation of miR-143-3p. Knockdown of LIMK1 aggravated sevoflurane-induced decrease of cell viability and increase of LDH and apoptosis in neurons, while over-expression attenuated LIMK1 silence-induced neuronal damage post-sevoflurane anesthesia. In conclusion, Rian demonstrated neuroprotective effects against sevoflurane anesthesia-induced cognitive dysfunction through regulation of miR-143-3p/LIMK1 axis, providing promising target for sevoflurane anesthesia-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Yang Yu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Wei Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Dengyan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Haitao Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Hua Shao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yue Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| |
Collapse
|
10
|
Obal D, Wu JC. Induced pluripotent stem cells as a platform to understand patient-specific responses to opioids and anaesthetics. Br J Pharmacol 2020; 177:4581-4594. [PMID: 32767563 PMCID: PMC7520445 DOI: 10.1111/bph.15228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
Recent advances in human induced pluripotent stem cell (iPSC) technology may provide unprecedented opportunities to study patient-specific responses to anaesthetics and opioids. In this review, we will (1) examine the advantages and limitations of iPSC technology, (2) summarize studies using iPSCs that have contributed to our current understanding of anaesthetics and opioid action on the cardiovascular system and central nervous system (CNS), and (3) describe how iPSC technology can be used to further develop personalized analgesic and sedative pharmacotherapies with reduced or minimal detrimental cardiovascular effects.
Collapse
Affiliation(s)
- Detlef Obal
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Anesthesiology, Pain, and Perioperative MedicineStanford UniversityStanfordCaliforniaUSA
- Outcomes Research ConsortiumClevelandOhioUSA
| | - Joseph C. Wu
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford UniversityStanfordCaliforniaUSA
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
11
|
Sharma KD, Schaal D, Kore RA, Hamzah RN, Pandanaboina SC, Hayar A, Griffin RJ, Srivatsan M, Reyna NS, Xie JY. Glioma-derived exosomes drive the differentiation of neural stem cells to astrocytes. PLoS One 2020; 15:e0234614. [PMID: 32649728 PMCID: PMC7351166 DOI: 10.1371/journal.pone.0234614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/29/2020] [Indexed: 01/15/2023] Open
Abstract
Exosomes appear to be effective inter-cellular communicators delivering several types of molecules, such as proteins and RNAs, suggesting that they could influence neural stem cell (NSC) differentiation. Our RNA sequencing studies demonstrated that the RNAs related to cell proliferation and astrocyte differentiation were upregulated in human mesenchymal stem cells (hMSC) when co-cultured with exosomes obtained from the culture medium of human glioma cells (U87). Metallothionein 3 and elastin genes, which are related to cell proliferation, increased 10 and 7.2 fold, respectively. Expression of genes for astrocyte differentiation, such as tumor growth factor alpha, induced protein 3 of the NOTCH1 family, colony stimulating factor and interleukin 6 of the STAT3 family and Hes family bHLH transcription factor 1 also increased by 2.3, 10, 4.7 and 2.9 fold, respectively. We further examined the effects of these exosomes on rat fetal neural stem cell (rNSC) differentiation using the secreted exosomes from U87 glioma cells or exosomes from U87 cells that were stimulated with interleukin 1β (IL-1β). The rNSCs, extracted from rat brains at embryonic day 14 (E14), underwent a culture protocol that normally leads to predominant (~90%) differentiation to ODCs. However, in the presence of the exosomes from untreated or IL-1β-treated U87 cells, significantly more cells differentiated into astrocytes, especially in the presence of exosomes obtained from the IL-1β-challenged glioma cells. Moreover, glioma-derived exosomes appeared to inhibit rNSC differentiation into ODCs or astrocytes as indicated by a significantly increased population of unlabeled cells. A portion of the resulting astrocytes co-expressed both CD133 and glial fibrillary acidic protein (GFAP) suggesting that exosomes from U87 cells could promote astrocytic differentiation of NSCs with features expected from a transformed cell. Our data clearly demonstrated that exosomes secreted by human glioma cells provide a strong driving force for rat neural stem cells to differentiate into astrocytes, uncovering potential pathways and therapeutic targets that might control this aggressive tumor type.
Collapse
Affiliation(s)
- Krishna D. Sharma
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Danielle Schaal
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Rajshekhar A. Kore
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Rabab N. Hamzah
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Sahitya Chetan Pandanaboina
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Abdallah Hayar
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Malathi Srivatsan
- Department of Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, Arkansas, United States of America
| | - Nathan S. Reyna
- Department of Biology, Ouachita Baptist University, Arkadelphia, Arkansas, United States of America
| | - Jennifer Yanhua Xie
- Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, Arkansas, United States of America
| |
Collapse
|
12
|
Li T, Huang Z, Wang X, Zou J, Tan S. Role of the GABAA receptors in the long-term cognitive impairments caused by neonatal sevoflurane exposure. Rev Neurosci 2020; 30:869-879. [PMID: 31145696 DOI: 10.1515/revneuro-2019-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023]
Abstract
Sevoflurane is a widely used inhalational anesthetic in pediatric surgeries, which is considered reasonably safe and reversible upon withdrawal. However, recent preclinical studies suggested that peri-neonatal sevoflurane exposure may cause developmental abnormalities in the brain. The present review aimed to present and discuss the accumulating experimental data regarding the undesirable effects of sevoflurane on brain development as revealed by the laboratory studies. First, we summarized the long-lasting side effects of neonatal sevoflurane exposure on cognitive functions. Subsequently, we presented the structural changes, namely, neuroapoptosis, neurogenesis and synaptogenesis, following sevoflurane exposure in the immature brain. Finally, we also discussed the potential mechanisms underlying subsequent cognitive impairments later in life, which are induced by neonatal sevoflurane exposure and pointed out potential strategies for mitigating sevoflurane-induced long-term cognitive impairments. The type A gamma-amino butyric acid (GABAA) receptor, the main targets of sevoflurane, is excitatory rather than inhibitory in the immature neurons. The excitatory effects of the GABAA receptors have been linked to increased neuroapoptosis, elevated serum corticosterone levels and epigenetic modifications following neonatal sevoflurane exposure in rodents, which might contribute to sevoflurane-induced long-term cognitive abnormalities. We proposed that the excitatory GABAA receptor-mediated HPA axis activity might be a novel mechanism underlying sevoflurane-induced long-term cognitive impairments. More studies are needed to investigate the effectiveness and mechanisms by targeting the excitatory GABAA receptor as a prevention strategy to alleviate cognitive deficits induced by neonatal sevoflurane exposure in future.
Collapse
Affiliation(s)
- Tao Li
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Zeyi Huang
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Xianwen Wang
- Grade 2015 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Ju Zou
- Department of Parasitology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| | - Sijie Tan
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang 421001, Hunan Province, China
| |
Collapse
|
13
|
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
|
14
|
Hirai T, Konishi Y, Mizuno S, Rui Z, Sun Y, Nishiwaki K. Differential effects of sevoflurane on the growth and apoptosis of human cancer cell lines. J Anesth 2019; 34:47-57. [DOI: 10.1007/s00540-019-02701-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/13/2019] [Indexed: 12/15/2022]
|
15
|
Pandanaboina SC, Alghazali KM, Nima ZA, Alawajji RA, Sharma KD, Watanabe F, Saini V, Biris AS, Srivatsan M. Plasmonic nano surface for neuronal differentiation and manipulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 21:102048. [PMID: 31271878 DOI: 10.1016/j.nano.2019.102048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/15/2019] [Accepted: 06/15/2019] [Indexed: 12/28/2022]
Abstract
Neurodegenerative diseases and traumatic brain injuries can destroy neurons, resulting in sensory and motor function loss. Transplantation of differentiated neurons from stem cells could help restore such lost functions. Plasmonic gold nanorods (AuNR) were integrated in growth surfaces to stimulate and modulate neural cells in order to tune cell physiology. An AuNR nanocomposite system was fabricated, characterized, and then utilized to study the differentiation of embryonic rat neural stem cells (NSCs). Results demonstrated that this plasmonic surface 1) accelerated differentiation, yielding almost twice as many differentiated neural cells as a traditional NSC culture surface coated with poly-D-lysine and laminin for the same time period; and 2) promoted differentiation of NSCs into neurons and astrocytes in a 2:1 ratio, as evidenced by the expression of relevant marker proteins. These results indicate that the design and properties of this AuNR plasmonic surface would be advantageous for tissue engineering to address neural degeneration.
Collapse
Affiliation(s)
| | - Karrer M Alghazali
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204
| | - Zeid A Nima
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204
| | - Raad A Alawajji
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204
| | - Krishna Deo Sharma
- Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401
| | - Fumiya Watanabe
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204
| | - Viney Saini
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204.
| | - Malathi Srivatsan
- Biological Sciences and Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401.
| |
Collapse
|
16
|
Neuroglobin protects offspring rats from neuronal damage induced by sevoflurane exposure to pregnant rats by inhibiting endogenous apoptosis. Int J Dev Neurosci 2019; 76:17-24. [DOI: 10.1016/j.ijdevneu.2019.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/03/2019] [Accepted: 06/03/2019] [Indexed: 12/22/2022] Open
|
17
|
Bi C, Cai Q, Shan Y, Yang F, Sun S, Wu X, Liu H. Sevoflurane induces neurotoxicity in the developing rat hippocampus by upregulating connexin 43 via the JNK/c-Jun/AP-1 pathway. Biomed Pharmacother 2018; 108:1469-1476. [PMID: 30372849 DOI: 10.1016/j.biopha.2018.09.111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
As one of the most popular anesthetics, sevoflurane is widely used in pediatric anesthesia. Unfortunately, an increasing number of studies have demonstrated that sevoflurane has potential neurotoxic effects on the developing brain and cognition, even in adolescence. Connexin 43 (Cx43) has been documented to contribute to cognitive dysfunction. The present study hypothesized that Cx43 may participate in sevoflurane-induced neuroinjury and investigated the underlying mechanisms in young Sprague Dawley (SD) rats. Seven-day-old SD rats (P7) were exposed to 3% sevoflurane for 4 h. The levels of Cx43,mitogen-activated protein kinase (MAPK) signaling pathway components(including total and phosphorylated p38, extracellular signal-regulated kinase (ERK), and c-Jun n-terminal kinase (JNK) and activator protein 1(AP-1) transcription factors (including total and phosphorylated c-Fos, and c-Jun) were assessed by Western blot analysis. Neuronal apoptosis was detected using immunohistochemistry (IHC). The Morris water maze (MWM) was performed to evaluate cognitive function from P28 to P33. The results showed that anesthesia with 3% sevoflurane for 4 h increased Cx43 levels in the rat hippocampus from 6 h to 3 d, and compared with sevoflurane exposure in the control group rats, exposure in P7 SD rats also increased the ratios of phosphorylated JNK to JNK and, phosphorylated c-Jun to c-Jun in the hippocampus from 6 h to 3 d. All these effects could be alleviated by pretreatment with the JNK inhibitor SP600125 (10 mg/kg). Neuroapoptosis was similarly increased from 6 h to 1 d after inhaled sevoflurane exposure. Finally, the MWM indicated that sevoflurane could increase the escape latency and, decrease the number of platform crossings from P28 to P33. Overall, our findings suggested that sevoflurane increased Cx43 expression and induced to apoptosis by activating the JNK/c-Jun signaling pathway in the hippocampus of P7 rats. This finding may reveal a new strategy for preventing sevoflurane-induced neuronal dysfunction.
Collapse
Affiliation(s)
- Congjie Bi
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China; Department of Anesthesiology, Dalian Central Hospital, Dalian, China
| | - Qiuping Cai
- Department of Anesthesiology, Dalian Central Hospital, Dalian, China
| | - Yangyang Shan
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shiwei Sun
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hongtao Liu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Shao CZ, Xia KP. Sevoflurane anesthesia represses neurogenesis of hippocampus neural stem cells via regulating microRNA-183-mediated NR4A2 in newborn rats. J Cell Physiol 2018; 234:3864-3873. [PMID: 30191980 DOI: 10.1002/jcp.27158] [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] [Received: 03/31/2018] [Accepted: 07/10/2018] [Indexed: 12/23/2022]
Abstract
Sevoflurane has been commonly utilized in nonobstetric surgeries in pregnant women, and its impacts on fetal brain are still not completely known. Ectopic NR4A2 expression has been reported to be related with familial Parkinson disease, and through dual luciferase we found that NR4A2 is a target gene of microRNA-183 (miR-183). We proposed a hypothesis that miR-183 may participate in the process by targeting NR4A2 in neurons after sevoflurane anesthesia. To verify the effect of sevoflurane on hippocampal neural stem cells (NSCs) proliferation and differentiation, we conducted EdU assay and immunofluorescence staining. Next, for better understanding of the impact of miR-183, we altered the miR-183 expression using mimic and inhibitor. Meanwhile, the targeting relationship between miR-183 and NR4A2 was validated by a bioinformatics website and dual-luciferase reporter gene assay. Finally, expressions of miR-184, NR4A2, SRY (sex-determining region Y)-box 2 (Sox2), and brain-derived neurotrophic factor (BDNF) were determined and evaluated by reverse transcription quantitative polymerase chain reaction and western blot analysis. First, sevoflurane was determined a crucial factor in biological behaviors of hippocampal NSCs. Moreover, upregulated miR-183 expression by mimic inhibited the proliferation and differentiation of NSCs. Sevoflurane negatively regulated NR4A2 and Sox2 expressions but positively regulated miR-183 and BDNF expressions. Our findings revealed the underlying novel mechanism by which sevoflurane inhibits hippocampal NSC proliferation and differentiation through interaction with miR-183 and NR4A2. The study provides reliable reference for safe application of sevoflurane anesthesia in neonates.
Collapse
Affiliation(s)
- Chang-Zhong Shao
- Department of Anesthesiology, Linyi People's Hospital Affiliated to Shandong University, Linyi, China
| | - Kun-Peng Xia
- Department of Anesthesiology, Linyi People's Hospital Affiliated to Shandong University, Linyi, China
| |
Collapse
|
19
|
Liu S, Fang F, Song R, Gao X, Jiang M, Cang J. Sevoflurane affects neurogenesis through cell cycle arrest via inhibiting wnt/β-catenin signaling pathway in mouse neural stem cells. Life Sci 2018; 209:34-42. [PMID: 30071197 DOI: 10.1016/j.lfs.2018.07.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/23/2018] [Accepted: 07/29/2018] [Indexed: 10/28/2022]
Abstract
AIMS The development of central nervous system requires proliferation of neural stem cells followed by differentiation. Cell cycle parameters are closely related with cell fate specification and differentiation. Recent researches indicated that wnt/β-catenin signaling pathway might cause proliferation inhibition and differentiation abnormality through interfering NSCs cell cycle. Our previous research also showed that multiple sevoflurane exposure to neural stem cells inhibited proliferation via repressing transcription factor Pax6 and cyclin D1 through inhibiting wnt/β-catenin pathway. All above encouraged us to figure out the effect of sevoflurane on cell cycle and neurogenesis. MAIN METHODS Primary mouse cultured neural stem cells were used and exposed to 4.1% sevoflurane for 6 h in this study. The expression of β-catenin, GSK-3β, c-myc and cyclin D1 were determined by western blot and qRT-PCR. FACS was used to measure the cell cycle. The proliferation of NSCs was evaluated by EdU staining while the differentiation was evaluated by Tuj1 and GFAP staining on immunocytochemistry. KEY FINDINGS We found that exposure to sevoflurane at a concentration of 4.1% for 6 h induced inhibition of wnt/β-catenin pathway, cell cycle arrest at G0/G1 phase and an earlier switch from proliferation to differentiation. GSK-3β specific inhibitor, CHIR99021, attenuated sevoflurane-induced cell cycle arrest and abnormality of neurogenesis in neural stem cells. SIGNIFICANCE Our research suggested that sevoflurane arrested cell cycle at G0/G1 phase through inhibition of wnt/β-catenin signaling pathway thus resulting in a premature differentiation in NSCs. This study presents a deeper understanding of the mechanism on cognitive impairment by sevoflurane exposure.
Collapse
Affiliation(s)
- Shiwen Liu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China
| | - Fang Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China
| | - Ruixue Song
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China
| | - Xuan Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China
| | - Ming Jiang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China.
| |
Collapse
|
20
|
Neonatal Exposure to Low-Dose (1.2%) Sevoflurane Increases Rats' Hippocampal Neurogenesis and Synaptic Plasticity in Later Life. Neurotox Res 2018; 34:188-197. [PMID: 29427282 DOI: 10.1007/s12640-018-9877-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/07/2018] [Accepted: 01/26/2018] [Indexed: 12/14/2022]
Abstract
The increasing usage of general anesthetics on young children and infants has drawn extensive attention to the effects of these drugs on cognitive function later in life. Recent animal studies have revealed improvement in hippocampus-dependent performance after lower concentrations of sevoflurane exposure. However, the long-term effects of low-dose sevoflurane on the developing brain remain elusive. On postnatal day (P) 7, rats were treated with 1.2% sevoflurane (1.2% sevo group), 2.4% sevoflurane (2.4% sevo group), and air control (C group) for 6 h. On P35-40, rats' hippocampus-dependent learning and memory was tested using the Morris water maze. Cognition-related and synapse-related proteins in the hippocampus were measured using Western blotting on P35. On the same day, neurogenesis and synapse ultrastructure were evaluated using immunofluorescence and transmission electron microscopy (TEM). On P35, the rats neonatally exposed to 1.2% sevoflurane showed better behavioral results than control rats, but not in the 2.4% sevo group. Exposure to 1.2% sevoflurane increased the number of 5'-bromo-2-deoxyuridine (BrdU)-positive cells in the dentate gyrus and improved both synaptic number and ultrastructure in the hippocampus. The expression levels of BDNF, TrkB, postsynaptic density (PSD)-95, and synaptophysin in the hippocampus were also increased in the 1.2% sevo group. In contrast, no significant changes in neurogenesis or synaptic plasticity were observed between the C group and the 2.4% sevo group on P35. These results showed that exposure of the developing brain to a low concentration of sevoflurane for 6 h could promote spatial learning and memory function, along with increased hippocampal neurogenesis and synaptic plasticity, in later life.
Collapse
|
21
|
Wang L, Zheng M, Wu S, Niu Z. MicroRNA-188-3p is involved in sevoflurane anesthesia-induced neuroapoptosis by targeting MDM2. Mol Med Rep 2018; 17:4229-4236. [PMID: 29344658 PMCID: PMC5802194 DOI: 10.3892/mmr.2018.8437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022] Open
Abstract
Sevoflurane is a commonly used inhalation anesthetic. Sevoflurane-induced neuroapoptosis and cognitive impairments in animals are widely reported, however, the underlying molecular mechanisms remain largely unknown. The results of the present study demonstrated that sevoflurane anesthesia induced spatial memory impairments in rats, as determined by the Morris water maze test. Mechanistically, the current study demonstrated that sevoflurane administration significantly enhanced the expression of microRNA (miR)-188-3p. Furthermore, inhibition of miR-188-3p using lentiviral miR-188-3p inhibitors attenuated sevoflurane-induced cognitive impairments in rats. The present study also demonstrated that miR-188-3p targeted MDM2 proto-oncogene (MDM2) and negatively regulated the expression of MDM2, as determined by luciferase assays, reverse transcription-quantitative polymerase chain reaction and western blot analysis. Furthermore, decreased abundance of MDM2 following transfection with miR-188-3p mimics was associated with increased stability of p53 protein. Suppression of p53 activity using the specific p53 inhibitor pifithrin-α alleviated sevoflurane-induced neuroapoptosis. These results indicate that the miR-188-3p-MDM2-p53 axis may have a critical role in sevoflurane-induced cognitive dysfunction. Therefore, miR-188-3p may be a potential target for the treatment of sevoflurane-induced cognitive impairment.
Collapse
Affiliation(s)
- Lei Wang
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Mengliang Zheng
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Shuishui Wu
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Zhiqiang Niu
- Department of Anesthesia, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|