1
|
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
|
2
|
Huang J, Xu Y, Wang F, Wang H, Li L, Deng Y, Cai L. Long Noncoding RNA SPRY4-IT1 Modulates Ketamine-Induced Neurotoxicity in Human Embryonic Stem Cell-Derived Neurons through EZH2. Dev Neurosci 2021; 43:9-17. [PMID: 33827085 DOI: 10.1159/000513535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/02/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to investigate whether long noncoding RNA sprouty receptor tyrosine kinase signaling antagonist 4-intronic transcript 1 (SPRY4-IT1) is involved in the regulation of ketamine-induced neurotoxicity. METHODS Human embryonic stem cells (hESCs) were induced into neurons in vitro and treated with ketamine. Apoptosis and neurite degeneration assays were used to determine ketamine-induced neurotoxicity and qRT-PCR to determine SPRY4-IT1 expression. SPRY4-IT1 was downregulated in hESC-induced neurons to examine its regulation on ketamine-induced neurotoxicity. The correlation between enhancer of zeste homolog 2 (EZH2) and SPRY4-IT1 was also examined. EZH2 was upregulated in SPRY4-IT1-downregualted hESC-induced neurons to further examine its participation in SPRY4-IT1-mediated ketamine neurotoxicity. RESULTS Ketamine-induced dose-dependent apoptosis, neurite degeneration, and SPRY4-IT1 upregulation in hESC-induced neurons. Lentivirus-mediated SPRY4-IT1 downregulation protected ketamine neurotoxicity. EZH2 expression was positively correlated with SPRY4-IT1 in hESC-induced neurons. EZH2 overexpression markedly reversed the protective effects of SPRY4-IT1 knockdown on ketamine neurotoxicity. CONCLUSIONS SPRY4-IT1 is involved in anesthesia-induced neurotoxicity, possibly through the regulation on EZH2 gene.
Collapse
Affiliation(s)
- Jingyuan Huang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Xu
- Department of Anesthesiology, Xi'an Central Hospital, Xi'an, China
| | - Fang Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haili Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lu Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yanan Deng
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Cai
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
3
|
Chen Q, Chu W, Sheng R, Song S, Yang J, Ji F, Jin X. Maternal anesthesia with sevoflurane during the mid-gestation induces social interaction deficits in offspring C57BL/6 mice. Biochem Biophys Res Commun 2021; 553:65-71. [PMID: 33756347 DOI: 10.1016/j.bbrc.2021.03.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
Sevoflurane anesthesia in pregnant mice could induce neurotoxicity in the developing brain and disturb learning and memory in the offspring mice. Whether it could impair social behaviors in the offspring mice is uncertain. Therefore, we assessed the neurobehavioral effect of in-utero exposure to sevoflurane on social interaction behaviors in C57BL/6 mice. The pregnant mice were anesthetized with 2.5% sevoflurane in 100% oxygen for 2 h, and their offspring mice were tested in three-chambered social paradigm, which includes three 10-min sessions of habituation, sociability, and preference for social novelty. At the juvenile age, the offspring mice showed abnormal sociability, as proved by not taking more time sniffing at the stranger 1 mouse compared with the empty enclosure (108.5 ± 25.4 vs. 108.2 ± 44.0 s, P = 0.9876). Meanwhile, these mice showed impaired preference for social novelty, as evidenced by not taking more time sniffing at the stranger 2 compared with the stranger 1 mouse (92.1 ± 52.2 vs. 126.7 ± 50.8 s, P = 0.1502). At the early adulthood, the offspring mice retrieved the normal sociability (145.6 ± 33.2 vs. 76.0 ± 31.8 s, P = 0.0001), but remained the impaired preference for social novelty (100.6 ± 29.3 vs. 118.0 ± 47.9 s, P = 0.3269). Collectively, these results suggested maternal anesthesia with sevoflurane could induce social interaction deficits in their offspring mice. Although the disturbance of sociability could be recoverable, the impairment of preference for social novelty could be long-lasting.
Collapse
Affiliation(s)
- Qingcai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Wei Chu
- Department of Pharmacology, Soochow University School of Pharmaceutical Science, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology, Soochow University School of Pharmaceutical Science, Suzhou, 215123, China
| | - Shaoyong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianping Yang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Xin Jin
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
4
|
Yao Y, Wang X, Gao J. LncRNA KCNQ1OT1 Sponges miR-206 to Ameliorate Neural Injury Induced by Anesthesia via Up-Regulating BDNF. Drug Des Devel Ther 2020; 14:4789-4800. [PMID: 33204065 PMCID: PMC7667201 DOI: 10.2147/dddt.s256319] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Widely used in anesthesia, ketamine is reported to induce neurotoxicity in patients. This study aimed to investigate the molecular regulatory mechanism of long non-coding RNA (lncRNA) KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) in ameliorating ketamine-induced neural injury. MATERIALS AND METHODS Sprague-Dawley rats were intraperitoneally injected with ketamine to induce neuronal injury. PC-12 cells treated with ketamine were used as the cell model. Ketamine-induced aberrant expression of KCNQ1OT1, miR-206 and brain-derived neurotrophic factor (BDNF) were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The effects of KCNQ1OT1 and miR-206 on ketamine-induced neural injury in PC-12 cells were then examined by MTT and LDH assay. The regulatory relationships between KCNQ1OT1 and miR-206, and miR-206 and BDNF were detected by dual-luciferase reporter assay. RESULTS Ketamine induced the apoptosis of neurons of the hippocampus in rats, and the apoptosis of PC-12 cells, accompanied by down-regulation of KCNQ1OT1 and BDNF expressions, and up-regulation of miR-206 expression. Overexpression of KCNQ1OT1 enhanced the resistance to apoptosis of PC-12 cells and significantly ameliorated ketamine-induced nerve injury, while transfection of miR-206 had opposite effects. Mechanistically, KCNQ1OT1 could target miR-206 and reduce its expression level, in turn indirectly increase the expression level of BDNF, and play a protective role in neural injury. CONCLUSION KCNQ1OT1/miR-206/BDNF axis is demonstrated to be an important regulatory mechanism in regulating ketamine-induced neural injury. Our study helps to clarify the mechanism by which ketamine exerts its toxicological effects and provides clues for the neuroprotection during anesthesia.
Collapse
Affiliation(s)
- Yao Yao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| | - Xuesong Wang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| | - Jin Gao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| |
Collapse
|
5
|
Niu D, Wang L, Cui J, Zhou B, Yao L. Inhibition of long noncoding RNA BLACAT1 protects anesthesia-induced neural cytotoxicity in human induced pluripotent stem cells derived neurons. Eur J Pharmacol 2019; 865:172737. [DOI: 10.1016/j.ejphar.2019.172737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
|
6
|
Traditional Chinese medicine, Kami-Shoyo-San protects ketamine-induced neurotoxicity in human embryonic stem cell-differentiated neurons through activation of brain-derived neurotrophic factor. Neuroreport 2019; 30:1102-1109. [PMID: 31568202 DOI: 10.1097/wnr.0000000000001328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Anesthesia-induced neurotoxicity may cause permanent dysfunctions in human brains. In this work, we used a cell-based in-vitro model to demonstrate that traditional Chinese medicine, Kami-Shoyo-San may protect ketamine-induced neuronal apoptosis in human embryonic stem cell-differentiated neurons. METHODS Human embryonic stem cell-differentiated neurons were cultured in vitro and treated with high-concentration ketamine to induce neuronal apoptosis. Pre-incubation of Kami-Shoyo-San was conducted to evaluate its neuroprotection on ketamine-injured neurons. Quantitative real-time PCR and western blot assays were used to assess brain-derived neurotrophic factor and its receptor, tropomyosin receptor kinase B, in response to Kami-Shoyo-San and ketamine treatment. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling pathway was then deactivated, by siRNA application, to further explore its functional role in Kami-Shoyo-San-mediated protection on ketamine-induced apoptosis among human embryonic stem cell-differentiated neurons. RESULTS High concentration of ketamine-induced significant apoptosis, whereas pre-incubation of Kami-Shoyo-San markedly rescued ketamine-induced apoptosis, in human embryonic stem cell-differentiated neurons. Kami-Shoyo-San activated brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling pathway by upregulating brain-derived neurotrophic factor and inducing tropomyosin receptor kinase B phosphorylation. Conversely, siRNA-mediated brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling pathway deactivation reversed the neuroprotection of Kami-Shoyo-San in ketamine-injured human embryonic stem cell-differentiated neurons. CONCLUSION Kami-Shoyo-San could protect ketamine-induced neurotoxicity, and the underlying mechanism may involve brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling pathway.
Collapse
|
7
|
Logan S, Arzua T, Canfield SG, Seminary ER, Sison SL, Ebert AD, Bai X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr Physiol 2019; 9:565-611. [PMID: 30873582 PMCID: PMC6705133 DOI: 10.1002/cphy.c180025] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurological disorders have emerged as a predominant healthcare concern in recent years due to their severe consequences on quality of life and prevalence throughout the world. Understanding the underlying mechanisms of these diseases and the interactions between different brain cell types is essential for the development of new therapeutics. Induced pluripotent stem cells (iPSCs) are invaluable tools for neurological disease modeling, as they have unlimited self-renewal and differentiation capacity. Mounting evidence shows: (i) various brain cells can be generated from iPSCs in two-dimensional (2D) monolayer cultures; and (ii) further advances in 3D culture systems have led to the differentiation of iPSCs into organoids with multiple brain cell types and specific brain regions. These 3D organoids have gained widespread attention as in vitro tools to recapitulate complex features of the brain, and (iii) complex interactions between iPSC-derived brain cell types can recapitulate physiological and pathological conditions of blood-brain barrier (BBB). As iPSCs can be generated from diverse patient populations, researchers have effectively applied 2D, 3D, and BBB models to recapitulate genetically complex neurological disorders and reveal novel insights into molecular and genetic mechanisms of neurological disorders. In this review, we describe recent progress in the generation of 2D, 3D, and BBB models from iPSCs and further discuss their limitations, advantages, and future ventures. This review also covers the current status of applications of 2D, 3D, and BBB models in drug screening, precision medicine, and modeling a wide range of neurological diseases (e.g., neurodegenerative diseases, neurodevelopmental disorders, brain injury, and neuropsychiatric disorders). © 2019 American Physiological Society. Compr Physiol 9:565-611, 2019.
Collapse
Affiliation(s)
- Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott G. Canfield
- Department of Cellular & Integrative Physiology, IU School of Medicine-Terre Haute, Terre Haute, IN, USA
| | - Emily R. Seminary
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha L. Sison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
8
|
Zhao X, Shu F, Wang X, Wang F, Wu L, Li L, Lv H. Inhibition of microRNA-375 ameliorated ketamine-induced neurotoxicity in human embryonic stem cell derived neurons. Eur J Pharmacol 2019; 844:56-64. [DOI: 10.1016/j.ejphar.2018.11.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 01/15/2023]
|
9
|
Jiang JD, Zheng XC, Huang FY, Gao F, You MZ, Zheng T. MicroRNA-107 regulates anesthesia-induced neural injury in embryonic stem cell derived neurons. IUBMB Life 2018; 71:20-27. [PMID: 30308117 PMCID: PMC7379593 DOI: 10.1002/iub.1911] [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: 04/07/2018] [Accepted: 05/07/2018] [Indexed: 01/11/2023]
Abstract
Ketamine, though widely used in pediatric anesthesia, may induce cortical neurotoxicity in young patients. This study focused on an in vitro model of rat brain embryonic stem cell (ESC)-derived neurons to investigate the effects of microRNA-107 (miR-107) on ketamine-induced neural injury. Rat brain ESCs were proliferated in vitro and differentiated toward neuronal fate. Ketamine induced neural injury in ESC-derived neurons was inspected by TUNEL and neurite growth assays. Ketamine-induce aberrant miR-107 expression was examined by qRT-PCR. MiR-107 was downregulated in ESCs through lentiviral transduction. Its effect on ketamine-induced neural injury in ESC-derived neurons was then examined. Potential downstream target of miR-107, brain derived neurotrophin factor (BDNF), was inspected by dual-luciferase reporter assay and qRT-PCR. BDNF was knocked down, through siRNA transfection, in NSCs to investigate its functional involvement in miR-107 mediated neural protection in ketamine-injured NSC-derived neurons. Ketamine induced apoptosis, neurite degeneration, and upregulated miR-107 in NSC-derived neurons. Lentivirus-mediated miR-107 downregulation attenuated ketamine-induced neural injury. BDNF was proven to be directly and inversely regulated by miR-107 in NSC-derived neurons. SiRNA-mediated BDNF inhibition reversed the protective effect of miR-107 downregulation on ketamine injury in NSC-derived neurons. MiR-107 / BDNF was demonstrated to be an important epigenetic signaling pathway in regulating ketamine-induced neural injury in cortical neurons. © 2018 The Authors. IUBMB Life published by Wiley Periodicals,Inc. on behalf of International Union of Biochemistry and Molecular Biology., 71(1):20-27, 2019.
Collapse
Affiliation(s)
- Jun Dan Jiang
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Xiao Chun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China.,Department of Anesthesiology, Fujian Provincial Emergency Center, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Feng Yi Huang
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Fei Gao
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Mei Zhen You
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Ting Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| |
Collapse
|
10
|
Long noncoding RNA SNHG16 reduced ketamine-induced neurotoxicity in human embryonic stem cell-derived neurons. J Chem Neuroanat 2018; 94:39-45. [PMID: 30171995 DOI: 10.1016/j.jchemneu.2018.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Clinical evidence demonstrates that prolonged exposure to ketamine may cause irreversible injury to immature human brains. In this study, we utilized an in vitro model to examine the function of long noncoding RNA (lncRNA) SNHG16 in ketamine-induced neurotoxicity in human embryonic stem cell (hESC)-derived neurons. METHODS HESCs were induced toward neuronsin vitro, and treated with ketamine, at various concentrations, for 48 h. Viability, apoptosis, caspase-3 activity and ROS activity were then examined among hESC-derived neurons. Ketamine-induced gene expression change of SNHG16 was assessed by qRT-PCR. SNHG16 was overexpressed in hESC-derived neurons, which were then treated with ketamine, followed by biochemical assays to assess the effects of SNHG16 upregulation on ketamine-induced neurotoxicity. Correlation between SNHG16 and NeuroD1 gene was assess by qRT-PCR. In SNHG16-upregulated hESC-derived neurons, they were double transfected with siRNA to knock down NeuroD1. The functions of NeuroD1 inhibition on SNHG16-associated neural protection on ketamine-induced neurotoxicity were further assessed. RESULTS 48-h in vitro treatment of ketamine induced significant neurotoxicity, and downregulated SNHG16 among hESC-derived neurons. Conversely, SNHG16 upregulation reduced ketamine-induced neurotoxicity. NeuroD1 expression was downregulated by ketamine in hESC-derived neurons, and concomitantly upregulated by SNHG16 overexpression. SiRNA-mediated NeuroD1 inhibition reversed the protection of SNHG16 upregulation on ketamine-induced neurotoxicity. CONCLUSIONS SNHG16 is an important epigenetic factor which may functionally modulate ketamine-induced neurotoxicity through NeuroD1.
Collapse
|
11
|
Lin28b and Sox2 regulate anesthesia-induced neural degeneration in neural stem cell derived neurons. Eur J Pharmacol 2018; 820:167-172. [DOI: 10.1016/j.ejphar.2017.12.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/10/2017] [Accepted: 12/11/2017] [Indexed: 01/22/2023]
|
12
|
Zhang J, Cui C, Li Y, Xu H. Inhibition of GSK-3beta Signaling Pathway Rescues Ketamine-Induced Neurotoxicity in Neural Stem Cell-Derived Neurons. Neuromolecular Med 2017; 20:54-62. [PMID: 29218434 DOI: 10.1007/s12017-017-8472-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/30/2017] [Indexed: 01/13/2023]
Abstract
Clinical application of anesthetic reagent, ketamine (Keta), may induce irreversible neurotoxicity in central nervous system. In this work, we utilized an in vitro model of neural stem cells-derived neurons (nSCNs) to evaluate the role of GSK-3 signaling pathway in Keta-induced neurotoxicity. Embryonic mouse-brain neural stem cells were differentiated into neurons in vitro. Keta (50 μM)-induced neurotoxicity in cultured nSCNs was monitored by apoptosis, immunohistochemical and western blot assays, respectively. GSK-3 signaling pathways, including GSK-3α and GSK-3β, were inhibited by siRNA in the culture. The subsequent effects of GSK-3α or GSK-3β downregulation on Keta-induced neurotoxicity, including apoptosis and neurite loss, were then evaluated in nSCNs. Finally, caspase and Akt/ERK signal pathways were further examined by western blot to evaluate the regulatory effect of GSK-3 signaling pathways on Keta-induced neural injury. Keta (50 μM) caused markedly nSCN apoptosis and neurite degeneration in vitro. Keta decreased GSK-3β phosphorylation, but had no effect on GSK-3α phosphorylation. SiRNA-induced GSK-3β downregulation rescued Keta-induced neurotoxicity in nSCNs by reducing neuronal apoptosis and preventing neurite degeneration. On the other hand, GSK-3α downregulation had no effect on Keta-induced neurotoxicity. Western blot showed that, in Keta-injured nSCNs, GSK-3β downregulation reduced Caspase-1/3 proteins, but left phosphorylated Akt/ERK unchanged. GSK-3β, not GSK-3α, was specifically involved in the process of Keta-induced neurotoxicity in nSCNs. Inhibiting GSK-3β may be an effective approach to counter toxic effect of ketamine on central neurons in clinical and experimental applications.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Changlei Cui
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China
| | - Yanhui Li
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China
| | - Haiyang Xu
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China.
| |
Collapse
|
13
|
Álvarez Escudero J, Paredes Esteban RM, Cambra Lasaosa FJ, Vento M, López Gil M, de Agustín Asencio JC, Moral Pumarega MT. More than 3 hours and less than 3 years old. Safety of anesthetic procedures in children under 3 years of age, subject to surgeries of more than 3 hours. ACTA ACUST UNITED AC 2017. [PMID: 28641810 DOI: 10.1016/j.redar.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- J Álvarez Escudero
- Presidente de la Sociedad Española de Anestesiología, Reanimación y Terapéutica del Dolor (SEDAR), jefe del Servicio de Anestesiología y Reanimación, Complejo Hospitalario Universitario de Santiago de Compostela, catedrático de Anestesiología, director del Departamento de Cirugía y especialidades Médico Quirúrgicas, Facultad de Medicina y Odontología, Universidad de Santiago de Compostela.
| | - R M Paredes Esteban
- Presidente de la Sociedad Española de Cirugía Pediátrica (SECP), presidente de la Sociedad Andaluza de Cirugía Pediátrica (ACPA), directora de la Unidad de Gestión Clínica de Cirugía Pediátrica, jefa del Servicio de Cirugía Pediátrica, Hospital Universitario Reina Sofía, Córdoba, España
| | - F J Cambra Lasaosa
- Presidente de la Sociedad Española de Cuidados Intensivos Pediátricos (SECIP), jefe del Servicio Unidad de Cuidados Intensivos Pediátricos Hospital Universitario Sant Joan de Déu, Barcelona, profesor asociado de Pediatría, Facultad de Medicina, Universidad de Barcelona
| | - M Vento
- Presidente de la Sociedad Española de Neonatología (SENeo), coordinador nacional de la Retic, Red de Salud Materno Infantil y del Desarrollo SAMID RD16/0022, Instituto Carlos III, Ministerio de Economía, Industria y Competitividad, Servicio de Neonatología, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - M López Gil
- Vicepresidenta de la Sección Anestesia Pediátrica, Sociedad Española de Anestesiología Reanimación y Terapéutica del Dolor (SEDAR), jefa del Servicio de Anestesia y Reanimación, Hospital General Universitario Gregorio Marañón, Madrid, profesora asociada del Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid
| | - J C de Agustín Asencio
- Vicepresidente de la Sociedad Española de Cirugía Pediátrica (SECP), jefe del Servicio de Cirugía Pediátrica, coordinador de Especialidades Quirúrgicas en Pediatría, Hospital Materno Infantil, Hospital General Universitario Gregorio Marañón, Madrid, profesor asociado de Pediatría, Facultad de Medicina, Universidad Complutense de Madrid
| | - M T Moral Pumarega
- Sociedad Española de Neonatología (SENeo), jefa de Sección, Servicio de Neonatología, Hospital Universitario 12 de Octubre, Madrid, colaboradora de la REDSAMID, profesora asociada de Pediatría y Ciencias de la Salud, Facultad de Medicina, Universidad Complutense de Madrid
| |
Collapse
|
14
|
Bal-Price A, Meek MEB. Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity. Pharmacol Ther 2017; 179:84-95. [PMID: 28529068 PMCID: PMC5869951 DOI: 10.1016/j.pharmthera.2017.05.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent developments have prompted the transition of empirically based testing of late stage toxicity in animals for a range of different endpoints including neurotoxicity to more efficient and predictive mechanistically based approaches with greater emphasis on measurable key events early in the progression of disease. The adverse outcome pathway (AOP) has been proposed as a simplified organizational construct to contribute to this transition by linking molecular initiating events and earlier (more predictive) key events at lower levels of biological organization to disease outcomes. As such, AOPs are anticipated to facilitate the compilation of information to increase mechanistic understanding of pathophysiological pathways that are responsible for human disease. In this review, the sequence of key events resulting in adverse outcome (AO) defined as parkinsonian motor impairment and learning and memory deficit in children, triggered by exposure to environmental chemicals has been briefly described using the AOP framework. These AOPs follow convention adopted in an Organization for Economic Cooperation and Development (OECD) AOP development program, publically available, to permit tailored application of AOPs for a range of different purposes. Due to the complexity of disease pathways, including neurodegenerative disorders, a specific symptom of the disease (e.g. parkinsonian motor deficit) is considered as the AO in a developed AOP. Though the description is necessarily limited by the extent of current knowledge, additional characterization of involved pathways through description of related AOPs interlinked into networks for the same disease has potential to contribute to more holistic and mechanistic understanding of the pathophysiological pathways involved, possibly leading to the mechanism-based reclassification of diseases, thus facilitating more personalized treatment.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Ispra, Italy.
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
15
|
Song C, Song C, Chen K, Zhang X. Inhibition of long non-coding RNA IGF2AS protects apoptosis and neuronal loss in anesthetic-damaged mouse neural stem cell derived neurons. Biomed Pharmacother 2017; 85:218-224. [DOI: 10.1016/j.biopha.2016.10.094] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/30/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023] Open
|
16
|
Zheng X, Lin C, Li Y, Ye J, Zhou J, Guo P. Long noncoding RNA BDNF-AS regulates ketamine-induced neurotoxicity in neural stem cell derived neurons. Biomed Pharmacother 2016; 82:722-8. [DOI: 10.1016/j.biopha.2016.05.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/31/2022] Open
|
17
|
Bal-Price A, Lein PJ, Keil KP, Sethi S, Shafer T, Barenys M, Fritsche E, Sachana M, Meek MEB. Developing and applying the adverse outcome pathway concept for understanding and predicting neurotoxicity. Neurotoxicology 2016; 59:240-255. [PMID: 27212452 DOI: 10.1016/j.neuro.2016.05.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 12/12/2022]
Abstract
The Adverse Outcome Pathway (AOP) concept has recently been proposed to support a paradigm shift in regulatory toxicology testing and risk assessment. This concept is similar to the Mode of Action (MOA), in that it describes a sequence of measurable key events triggered by a molecular initiating event in which a stressor interacts with a biological target. The resulting cascade of key events includes molecular, cellular, structural and functional changes in biological systems, resulting in a measurable adverse outcome. Thereby, an AOP ideally provides information relevant to chemical structure-activity relationships as a basis for predicting effects of structurally similar compounds. AOPs could potentially also form the basis for qualitative and quantitative predictive modeling of the human adverse outcome resulting from molecular initiating or other key events for which higher-throughput testing methods are available or can be developed. A variety of cellular and molecular processes are known to be critical for normal function of the central (CNS) and peripheral nervous systems (PNS). Because of the biological and functional complexity of the CNS and PNS, it has been challenging to establish causative links and quantitative relationships between key events that comprise the pathways leading from chemical exposure to an adverse outcome in the nervous system. Following introduction of the principles of MOA and AOPs, examples of potential or putative adverse outcome pathways specific for developmental or adult neurotoxicity are summarized and aspects of their assessment considered. Their possible application in developing mechanistically informed Integrated Approaches to Testing and Assessment (IATA) is also discussed.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy.
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Kimberly P Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Timothy Shafer
- Integrated Systems Toxicology Division, Office of Research and Development, U.S. Environmental Protection Agency, RTP, USA
| | - Marta Barenys
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Magdalini Sachana
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
18
|
Hunsberger JG, Efthymiou AG, Malik N, Behl M, Mead IL, Zeng X, Simeonov A, Rao M. Induced Pluripotent Stem Cell Models to Enable In Vitro Models for Screening in the Central Nervous System. Stem Cells Dev 2015; 24:1852-64. [PMID: 25794298 PMCID: PMC4533087 DOI: 10.1089/scd.2014.0531] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/20/2015] [Indexed: 12/23/2022] Open
Abstract
There is great need to develop more predictive drug discovery tools to identify new therapies to treat diseases of the central nervous system (CNS). Current nonpluripotent stem cell-based models often utilize non-CNS immortalized cell lines and do not enable the development of personalized models of disease. In this review, we discuss why in vitro models are necessary for translational research and outline the unique advantages of induced pluripotent stem cell (iPSC)-based models over those of current systems. We suggest that iPSC-based models can be patient specific and isogenic lines can be differentiated into many neural cell types for detailed comparisons. iPSC-derived cells can be combined to form small organoids, or large panels of lines can be developed that enable new forms of analysis. iPSC and embryonic stem cell-derived cells can be readily engineered to develop reporters for lineage studies or mechanism of action experiments further extending the utility of iPSC-based systems. We conclude by describing novel technologies that include strategies for the development of diversity panels, novel genomic engineering tools, new three-dimensional organoid systems, and modified high-content screens that may bring toxicology into the 21st century. The strategic integration of these technologies with the advantages of iPSC-derived cell technology, we believe, will be a paradigm shift for toxicology and drug discovery efforts.
Collapse
Affiliation(s)
| | | | - Nasir Malik
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland
| | - Mamta Behl
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Ivy L. Mead
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| | - Xianmin Zeng
- Buck Institute for Age Research, Novato, California
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Mahendra Rao
- New York Stem Cell Foundation, New York, New York
| |
Collapse
|
19
|
Bradford AB, McNutt PM. Importance of being Nernst: Synaptic activity and functional relevance in stem cell-derived neurons. World J Stem Cells 2015; 7:899-921. [PMID: 26240679 PMCID: PMC4515435 DOI: 10.4252/wjsc.v7.i6.899] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/28/2015] [Accepted: 05/11/2015] [Indexed: 02/06/2023] Open
Abstract
Functional synaptogenesis and network emergence are signature endpoints of neurogenesis. These behaviors provide higher-order confirmation that biochemical and cellular processes necessary for neurotransmitter release, post-synaptic detection and network propagation of neuronal activity have been properly expressed and coordinated among cells. The development of synaptic neurotransmission can therefore be considered a defining property of neurons. Although dissociated primary neuron cultures readily form functioning synapses and network behaviors in vitro, continuously cultured neurogenic cell lines have historically failed to meet these criteria. Therefore, in vitro-derived neuron models that develop synaptic transmission are critically needed for a wide array of studies, including molecular neuroscience, developmental neurogenesis, disease research and neurotoxicology. Over the last decade, neurons derived from various stem cell lines have shown varying ability to develop into functionally mature neurons. In this review, we will discuss the neurogenic potential of various stem cells populations, addressing strengths and weaknesses of each, with particular attention to the emergence of functional behaviors. We will propose methods to functionally characterize new stem cell-derived neuron (SCN) platforms to improve their reliability as physiological relevant models. Finally, we will review how synaptically active SCNs can be applied to accelerate research in a variety of areas. Ultimately, emphasizing the critical importance of synaptic activity and network responses as a marker of neuronal maturation is anticipated to result in in vitro findings that better translate to efficacious clinical treatments.
Collapse
|
20
|
Brown BP, Kang SC, Gawelek K, Zacharias RA, Anderson SR, Turner CP, Morris JK. In vivo and in vitro ketamine exposure exhibits a dose-dependent induction of activity-dependent neuroprotective protein in rat neurons. Neuroscience 2015; 290:31-40. [PMID: 25595994 DOI: 10.1016/j.neuroscience.2014.12.076] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 11/27/2022]
Abstract
Anesthetic doses of ketamine induce apoptosis, as well as gene expression of activity-dependent neuroprotective protein (ADNP), a putative homeodomain transcription factor in rat pups (P7). This study investigated if ketamine induced ADNP protein in a dose-dependent manner in vitro and in vivo using primary cultures of cortical neurons and neonatal pups (P7). In vivo immunohistochemistry demonstrated a sub-anesthetic dose of ketamine increased ADNP in the somatosensory cortex (SCC) which was previously identified to be damaged by repeated exposure to anesthetic doses of ketamine. Administration of low-dose ketamine prior to full sedation prevented caspase-3 activation in the hippocampus and SCC. Primary cultures of cortical neurons treated with ketamine (10 μM-10mM) at 3 days-in vitro (3 DIV) displayed a concentration-dependent decrease in expanded growth cones. Furthermore, neuronal production and localization of ADNP varied as a function of both ketamine concentration and length of exposure. Taken together, these data support the model that ADNP induction may be partially responsible for the efficacy of a low-dose ketamine pre-treatment in preventing ketamine-induced neuronal cell death.
Collapse
Affiliation(s)
- B P Brown
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA; Department of Chemistry, Baldwin Wallace University, Berea, OH 44017, USA
| | - S C Kang
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA
| | - K Gawelek
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA; Department of Biology, Baldwin Wallace University, Berea, OH 44017, USA; Department of Chemistry, Baldwin Wallace University, Berea, OH 44017, USA
| | - R A Zacharias
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA; Department of Psychology, Baldwin Wallace University, Berea, OH 44017, USA
| | - S R Anderson
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA; Department of Psychology, Baldwin Wallace University, Berea, OH 44017, USA; Department of Mathematics, Baldwin Wallace University, Berea, OH 44017, USA
| | - C P Turner
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA
| | - J K Morris
- Neuroscience Program, Baldwin Wallace University, Berea, OH 44017, USA; Department of Biology, Baldwin Wallace University, Berea, OH 44017, USA.
| |
Collapse
|
21
|
Wang C, Liu F, Patterson TA, Paule MG, Slikker W. Anesthetic Drug-Induced Neurotoxicity and Compromised Neural Stem Cell Proliferation. ACTA ACUST UNITED AC 2015. [DOI: 10.4303/jdar/235905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|