1
|
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
|
2
|
Guo Y, Feng Y, Jiang F, Hu L, Shan T, Li H, Liao H, Bao H, Shi H, Si Y. Down-regulating nuclear factor of activated T cells 1 alleviates cognitive deficits in a mouse model of sepsis-associated encephalopathy, possibly by stimulating hippocampal neurogenesis. Brain Res 2024; 1826:148731. [PMID: 38154504 DOI: 10.1016/j.brainres.2023.148731] [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: 08/14/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a common complication of sepsis, and has been associated with increased morbidity and mortality. Nuclear factor of activated T cells (NFATs) 1, a transcriptional factor that regulates T cell development, activation and differentiation, has been implicated in neuronal plasticity. Here we examined the potential role of NFAT1 in sepsis-associated encephalopathy in mice. Adult male C57BL/6J mice received intracerebroventricular injections of short interfering RNA against NFAT1 or sex-determining region Y-box 2 (SOX2), or a scrambled control siRNA prior to cecal ligation and perforation (CLP). A group of mice receiving sham surgery were included as an additional control. CLP increased escape latency and decreased the number of crossings into, and total time spent within, the target quadrant in the Morris water maze test. CLP also decreased the freezing time in context-dependent, but not context-independent, fear conditioning test. Knockdown of either NFAT1 or SOX2 attenuated these behavioral deficits. NFAT1 knockdown also attenuated CLP-induced upregulation of SOX2, increased the numbers of nestin-positive cells and newborn astrocytes, reduced the number of immature newborn neurons, and promoted the G1 to S transition of neural stem cells in hippocampus. These findings suggest that NFAT1 may contribute to sepsis-induced behavioral deficits, possibly by promoting SOX2 signaling and neurogenesis.
Collapse
Affiliation(s)
- Yaoyi Guo
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Yue Feng
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Fan Jiang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Liang Hu
- Department of Pharmacology, Nanjing Medical University, No. 101 Longmiandadao Road, Jiangning District, Nanjing, Jiangsu Province 211166, People's Republic of China
| | - Tao Shan
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Haojia Li
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongsen Liao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongwei Shi
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China.
| |
Collapse
|
3
|
Zuo Y, Xie J, Zhang X, Thirupathi A, Liu X, Zhang D, Zhang J, Shi Z. Sevoflurane causes cognitive impairment by inducing iron deficiency and inhibiting the proliferation of neural precursor cells in infant mice. CNS Neurosci Ther 2024; 30:e14612. [PMID: 38334030 PMCID: PMC10853893 DOI: 10.1111/cns.14612] [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: 11/08/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
AIMS Numerous studies on animals have shown that exposure to general anesthetics in infant stage may cause neurocognitive impairment. However, the exact mechanism is not clear. The dysfunction of iron metabolism can cause neurodevelopmental disorders. Therefore, we investigated the effect of iron metabolism disorder induced by sevoflurane (Sev) on cognitive function and the proliferation of neural precursor cells (NPCs) and neural stem cells (NSCs) in infant mice. METHODS C57BL/6 mice of postnatal day 14 and neural stem cells NE4C were treated with 2% Sev for 6 h. We used the Morris water maze (MWM) to test the cognitive function of infant mice. The proliferation of NPCs was measured using bromodeoxyuridine (BrdU) label and their markers Ki67 and Pax6 in infant brain tissues 12 h after anesthesia. Meanwhile, we used immunohistochemical stain, immunofluorescence assay, western blot, and flow cytometer to evaluate the myelinogenesis, iron levels, and cell proliferation in cortex and hippocampus or in NE4C cells. RESULTS The results showed that Sev significantly caused cognitive deficiency in infant mice. Further, we found that Sev inhibited oligodendrocytes proliferation and myelinogenesis by decreasing MBP and CC-1 expression and iron levels. Meanwhile, Sev also induced the iron deficiency in neurons and NSCs by downregulating FtH and FtL expression and upregulating the TfR1 expression in the cortex and hippocampus, which dramatically suppressed the proliferation of NSCs and NPCs as indicated by decreasing the colocalization of Pax6+ and BrdU+ cells, and caused the decrease in the number of neurons. Interestingly, iron supplementation before anesthesia significantly improved iron deficiency in cortex and hippocampus and cognitive deficiency induced by Sev in infant mice. Iron therapy inhibited the decrease of MBP expression, iron levels in neurons and oligodendrocytes, and DNA synthesis of Pax6+ cells in hippocampus induced by Sev. Meanwhile, the number of neurons was partially recovered in hippocampus. CONCLUSION The results from the present study demonstrated that Sev-induced iron deficiency might be a new mechanism of cognitive impairment caused by inhaled anesthetics in infant mice. Iron supplementation before anesthesia is an effective strategy to prevent cognitive impairment caused by Sev in infants.
Collapse
Affiliation(s)
- Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jinhong Xie
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Xue Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | | | - Xiaopeng Liu
- The Second Affiliated Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Di Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Jianhua Zhang
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life ScienceHebei Normal UniversityShijiazhuangHebei ProvinceChina
| |
Collapse
|
4
|
Xu X, Pan Y, Zhan L, Sun Y, Chen S, Zhu J, Luo L, Zhang W, Li Y. The Wnt/β-catenin pathway is involved in 2,5-hexanedione-induced ovarian granulosa cell cycle arrest. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 268:115720. [PMID: 37995618 DOI: 10.1016/j.ecoenv.2023.115720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/15/2023] [Accepted: 11/19/2023] [Indexed: 11/25/2023]
Abstract
N-Hexane causes significant ovarian toxicity, and its main active metabolite 2,5-hexanedione (2,5-HD) can induce ovarian injury through mechanisms such as inducing apoptosis in ovarian granulosa cells (GCs); however, the specific mechanism has not been fully elucidated. In this study, we investigated the effects on the cell cycle of rat ovarian GCs exposed in vitro to different concentrations of 2,5-HD (0 mM, 20 mM, 40 mM, and 60 mM) and further explored the mechanism by mRNA and miRNA microarray analyses. The flow cytometry results sindicated that compared with control cells, in ovarian GCs, there was significant cell cycle arrest after 2,5-HD treatment. Cell cycle- and apoptosis- related gene (Cdk2, Ccnd1, Bax, Bcl-2, Caspase3, and Caspase9) expression was altered. The mRNA and miRNA microarray results suggested that 5678 mRNAs and 32 miRNAs were differentially expressed in the 2,5-HD-treated group. A total of 262 target mRNAs were obtained by miRNA and mRNA coexpression analysis, forming 368 miRNA-mRNA coexpression relationship pairs with 27 miRNAs. GO and KEGG analyses showed that differentially expressed genes were significantly enriched in the cell cycle and Wnt signaling pathways. Furthermore, significant changes in the expression of Wnt signaling pathway and cell cycle- related genes (Fzd1, Lrp6, Tcf3, Tcf4, Fzd6, Lrp5, β-catenin, Lef1, GSK3β, and Dvl3) after 2,5-HD treatment were confirmed by qRT-PCR and Western blotting. Ther results of dual-luciferase assays indicated decreased β-catenin/TCF transcriptional activity after 2,5-HD treatment. In addition, Wnt pathway-related miRNAs (rno-miR-145-5p, rno-miR-143-3p, rno-miR-214-3p, rno-miR-138-5p, and rno-miR-199a-3p) were changed significantly after 2,5-HD treatment. In summary, 2,5-HD induced cell cycle arrest in ovarian GCs, and the Wnt/β-catenin signaling pathway may play a very critical role in this process. Alterations in the expression of miRNAs such as rno-miR-145-5p may have significant implications.
Collapse
Affiliation(s)
- Xueming Xu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Yimei Pan
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Liqin Zhan
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Yi Sun
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Sichuan Chen
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Jianlin Zhu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Lingfeng Luo
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China
| | - Wenchang Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China.
| | - Yuchen Li
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, Fujian Province, China.
| |
Collapse
|
5
|
Cheng H, Li Q. Sevoflurane inhibits cholangiocarcinoma via Wnt/β-catenin signaling pathway. BMC Gastroenterol 2023; 23:279. [PMID: 37568083 PMCID: PMC10422733 DOI: 10.1186/s12876-023-02911-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is a refractory malignancy derived from bile duct epithelial cells. This study aimed to explore the role and molecular mechanisms of action of sevoflurane in CCA. METHODS CCK-8 assay was used to assess the proliferation of cholangiocarcinoma cells, and flow cytometry was used to detect cholangiocarcinoma cell apoptosis. The effects of sevoflurane on TFK1 and QBC939 cell migration and invasion were investigated using a Transwell assay. Western blotting and RT-qPCR were used to assess the expression of apoptosis-related proteins and genes, and gene expression of the Wnt/β-catenin signaling pathway. RESULTS Our study found that sevoflurane inhibited cholangiocarcinoma cell proliferation in a dose-dependent manner. In addition, sevoflurane induced cholangiocarcinoma cell apoptosis, inhibited cholangiocarcinoma cell migration and invasion, as well as the Wnt/β-catenin signaling pathway evidenced by decreased Wnt3a, β-catenin, c-Myc, and Cyclin D1 protein and mRNA expression, reduced p-GSK3β protein expression and p-GSK3β/GSK3β ratio. Further mechanistic studies revealed that Wnt/β-catenin pathway inducer SKL2001 reversed the inhibitory effect of sevoflurane on cholangiocarcinoma cells. CONCLUSIONS Sevoflurane induces apoptosis and inhibits the growth, migration, and invasion of cholangiocarcinoma cells by inhibiting the Wnt/β-catenin signaling pathway. This study not only revealed the role of sevoflurane in the development of CCA but also elucidated new therapeutic agents for CCA.
Collapse
Affiliation(s)
- Hui Cheng
- Department of Anesthesiology, People's Hospital of Dongxihu District, Wuhan, 430040, China
| | - Qinfang Li
- Department of Anesthesiology, People's Hospital of Dongxihu District, Wuhan, 430040, China.
- People's Hospital of Dongxihu District, No. 81 Huanshan Road, Wujiashan, Dongxihu District, Wuhan, 430040, China.
| |
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
|
Cai M, Sheng L. MFG-E8 Exerts Neuroprotection in Neural Stem Cells Induced by Anesthetic Sevoflurane via Regulating the PI3K/AKT Pathways. Stem Cells Int 2022; 2022:5609501. [PMID: 36277041 PMCID: PMC9581634 DOI: 10.1155/2022/5609501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
MFG-E8 has shown tissue protection effects in various models of organ injury. In this study, the function of MFG-E8 in SEV-induced neural stem cells (NSCs) was studied. The cell viability and apoptosis affected by rhMFG-E8 were tested by MTT and flow cytometry analysis, respectively. Then, the mRNA expression of MFG-E8 was detected by qRT-PCR. The expression of SOD, GSF-Px, and MDA was assessed using ELISA assay. Western blot analysis was applied for assessing the expression of MFG-E8, BCL2, BAX, cleaved caspase-3, GRP-78, XBP-1, ATF-6, ATF-4, CHOP, p-PI3K, PI3K, p-AKT, and AKT. The pharmacological experiments suggested that both mRNA and protein expression of MFG-E8 were significantly decreased after 24 h, 48 h, and 72 h treatment with SEV, and the Western blot results displayed that 50 and 100 μg/ml rhMFG-E8 could evidently promote the expression of MFG-E8 in NSCs induced by SEV. Next, rhMFG-E8 reduced the apoptosis of NSCs induced by SEV through upregulating Bcl-2 and cleaved caspase-3 and downregulating Bax. Moreover, rhMFG-E8 alleviated the endoplasmic reticulum pressure of NSCs induced by SEV through decreasing the expression of GRP-78, XBP-1, ATF-6, ATF-4, and CHOP. In addition, the rhMFG-E8 could promote the expression of SOD and GSH-Px and inhibit the expression of MDA and LDH detected by the ELISA assay and LDH kit. Moreover, rhMFG-E8 elevated the expression of p-PI3K/PI3K and p-AKT/AKT, which were inhibited by SEV in NSCs. The results of this project supported that rhMFG-E8 protects neural activity in neural stem cells induced by anesthetic sevoflurane via regulating the PI3K/AKT pathways.
Collapse
Affiliation(s)
- Minmin Cai
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315000, China
| | - Liufang Sheng
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315000, China
| |
Collapse
|
9
|
Sevoflurane induces microRNA-18a to delay rat neurodevelopment via suppression of the RUNX1/Wnt/β-catenin axis. Cell Death Dis 2022; 8:404. [PMID: 36182925 PMCID: PMC9526732 DOI: 10.1038/s41420-022-01179-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/09/2022]
Abstract
Sevoflurane anesthesia is reported to repress neurogenesis of neural stem cells (NSCs), thereby affecting the brain development, but the underlying mechanism of sevoflurane on the proliferation of NSCs remains unclear. Thus, this study aims to discern the relationship between sevoflurane and NSC proliferation. Bioinformatics tools were employed to predict the expression of microRNA-18a (miR-18a) in 9-day-old neonatal rat hippocampal tissues after sevoflurane treatment and the downstream genes of miR-18a, followed by a series of assays to explore the relationship among miR-18a, runt related transcription factor 1 (RUNX1), and β-catenin in the hippocampal tissues. NSCs were isolated from the hippocampal tissues and subjected to gain-/loss-of-function assays to investigate the interactions among miR-18a, RUNX1, and β-catenin in NSCs and their roles in NSC development. Bioinformatics analysis and experimental results confirmed high expression of miR-18a in rat hippocampal tissues and NSCs after sevoflurane treatment. Next, we found that miR-18a downregulated RUNX1 expression, while RUNX1 promoted NSC proliferation by activating the Wnt/β-catenin signaling pathway. The behavioral experiments also showed that sevoflurane caused nerve injury in rats, whilst RUNX1 overexpression protected rat neurodevelopment. Our findings uncovered that sevoflurane attenuated NSC proliferation via the miR-18a-meidated RUNX1/Wnt/β-catenin pathway, thereby impairing rat neurodevelopment.
Collapse
|
10
|
Shang J, Li B, Fan H, Liu P, Zhao W, Chen T, Chen P, Yang L. Sevoflurane promotes premature differentiation of dopaminergic neurons in hiPSC-derived midbrain organoids. Front Cell Dev Biol 2022; 10:941984. [PMID: 36176283 PMCID: PMC9513420 DOI: 10.3389/fcell.2022.941984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Conventional animal models used in corresponding basic studies are distinct from humans in terms of the brain’s development trajectory, tissue cytoarchitecture and cell types, making it difficult to accurately evaluate the potential adverse effects of anesthetic treatments on human fetal brain development. This study investigated the effects of sevoflurane on the midbrain’s development and cytopathology using human physiologically-relevant midbrain organoids. Methods: Monolayer human induced pluripotent stem cells (hiPSC)-derived human floor plate cells and three-dimensional hiPSC-derived midbrain organoids (hMBOs) were exposed to 2% (v/v) sevoflurane for 2 or 6 h, followed by expansion or differentiation culture. Then, immunofluorescence, real-time PCR, EdU assay, Tunnel assay, and transcriptome sequencing were performed to examine the effects of sevoflurane on the midbrain’s development. Results: We found that 2% sevoflurane exposure inhibited hFPCs’ proliferation (differentiation culture: 7.2% ± 0.3% VS. 13.3% ± 0.7%, p = 0.0043; expansion culture: 48% ± 2.2% VS. 35.2% ± 1.4%, p = 0.0002) and increased their apoptosis, but did not affect their differentiation into human dopaminergic neurons After 6 h, 2% sevoflurane exposure inhibited cell proliferation (62.8% ± 5.6% VS. 100% ± 5.5%, p = 0.0065) and enhanced the premature differentiation of hMBOs (246% ± 5.2% VS. 100% ± 28%, p = 0.0065). The RNA-seq results showed long-term exposure to sevoflurane up regulates some transcription factors in the differentiation of dopaminergic neurons, while short-term exposure to sevoflurane has a weak up-regulation effect on these transcription factors. Conclusion: This study revealed that long-term exposure to sevoflurane could promote the premature differentiation of hMBOs, while short-term exposure had negligible effects, suggesting that long-term exposure to sevoflurane in pregnant women may lead to fetals’ midbrain development disorder.
Collapse
Affiliation(s)
- Jia Shang
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| | - Bin Li
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Han Fan
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Peidi Liu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Wen Zhao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Tao Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, Hubei, China
- *Correspondence: Longqiu Yang, ; Pu Chen,
| | - Longqiu Yang
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
- Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- *Correspondence: Longqiu Yang, ; Pu Chen,
| |
Collapse
|
11
|
He Z, Lang L, Hui J, Ma Y, Yang C, Weng W, Huang J, Zhao X, Zhang X, Liang Q, Jiang J, Feng J. Brain Extract of Subacute Traumatic Brain Injury Promotes the Neuronal Differentiation of Human Neural Stem Cells via Autophagy. J Clin Med 2022; 11:jcm11102709. [PMID: 35628836 PMCID: PMC9145659 DOI: 10.3390/jcm11102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Background: After a traumatic brain injury (TBI), the cell environment is dramatically changed, which has various influences on grafted neural stem cells (NSCs). At present, these influences on NSCs have not been fully elucidated, which hinders the finding of an optimal timepoint for NSC transplantation. Methods: Brain extracts of TBI mice were used in vitro to simulate the different phase TBI influences on the differentiation of human NSCs. Protein profiles of brain extracts were analyzed. Neuronal differentiation and the activation of autophagy and the WNT/CTNNB pathway were detected after brain extract treatment. Results: Under subacute TBI brain extract conditions, the neuronal differentiation of hNSCs was significantly higher than that under acute brain extract conditions. The autophagy flux and WNT/CTNNB pathway were activated more highly within the subacute brain extract than in the acute brain extract. Autophagy activation by rapamycin could rescue the neuronal differentiation of hNSCs within acute TBI brain extract. Conclusions: The subacute phase around 7 days after TBI in mice could be a candidate timepoint to encourage more neuronal differentiation after transplantation. The autophagy flux played a critical role in regulating neuronal differentiation of hNSCs and could serve as a potential target to improve the efficacy of transplantation in the early phase.
Collapse
Affiliation(s)
- Zhenghui He
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
| | - Lijian Lang
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
| | - Jiyuan Hui
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
| | - Yuxiao Ma
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
| | - Chun Yang
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
| | - Weiji Weng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
| | - Jialin Huang
- Shanghai Institute of Head Trauma, Shanghai 200127, China;
| | - Xiongfei Zhao
- Shanghai Angecon Biotechnology Co., Ltd., Shanghai 201318, China; (X.Z.); (X.Z.)
| | - Xiaoqi Zhang
- Shanghai Angecon Biotechnology Co., Ltd., Shanghai 201318, China; (X.Z.); (X.Z.)
| | - Qian Liang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Jiyao Jiang
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
- Shanghai Institute of Head Trauma, Shanghai 200127, China;
| | - Junfeng Feng
- Brain Injury Center, Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Z.H.); (L.L.); (J.H.); (Y.M.); (C.Y.); (J.J.)
- Shanghai Institute of Head Trauma, Shanghai 200127, China;
- Correspondence: ; Tel.: +86-136-1186-0825
| |
Collapse
|
12
|
Zhang J, Hu Q, Jiang X, Wang S, Zhou X, Lu Y, Huang X, Duan H, Zhang T, Ge H, Yu A. Actin Alpha 2 Downregulation Inhibits Neural Stem Cell Proliferation and Differentiation into Neurons through Canonical Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7486726. [PMID: 35186189 PMCID: PMC8850075 DOI: 10.1155/2022/7486726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022]
Abstract
Our previous study has shown that actin alpha 2 (ACTA2) is expressed in NSC and ACTA2 downregulation inhibits NSC migration by increasing RhoA expression and decreasing the expression of Rac1 to curb actin filament polymerization. Given that proliferation and differentiation are the two main characteristics of NSC, the role of ACTA2 downregulation in the proliferation and differentiation of NSC remains elusive. Here, the results demonstrated that ACTA2 downregulation using ACTA2 siRNA held the potential of inhibiting NSC proliferation using cell counting kit-8 (CCK8) and immunostaining. Then, our data illustrated that ACTA2 downregulation attenuated NSC differentiation into neurons, while directing NSC into astrocytes and oligodendrocytes using immunostaining and immunoblotting. Thereafter, the results revealed that the canonical Wnt/β-catenin pathway was involved in the effect of ACTA2 downregulation on the proliferation and differentiation of NSC through upregulating p-β-catenin and decreasing β-catenin due to inactivating GSK-3β, while this effect could be partially abolished with administration of CHIR99012, a GSK-3 inhibitor. Collectively, these results indicate that ACTA2 downregulation inhibits NSC proliferation and differentiation into neurons through inactivation of the canonical Wnt/β-catenin pathway. The aim of the present study is to elucidate the role of ACTA2 in proliferation and differentiation of NSC and to provide an intervention target for promoting NSC proliferation and properly directing NSC differentiation.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Shuhong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xin Zhou
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Yuanlan Lu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xiaofei Huang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Haizhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Tianxi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Hongfei Ge
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| |
Collapse
|
13
|
Chen L, Tang T, Zheng X, Xiong Y. Protective Effects of Dexmedetomidine on Hippocampal Neurons in Rats Anesthetized with Sevoflurane. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To explore effects of dexmedetomidine (Dex) on cognitive function and hippocampal neuronal apoptosis in rats anesthetized with sevoflurane (Sevo), and regulation of brain-derived neurotrophic factor (BDNF) and its downstream signaling. 30 Sprague-Dawley (SD) rats were randomly divided
into control group inhaled 29% concentration oxygen), Sevo group (2 L/min oxygen flow +1.5% Sevo), Dex+Sevo group (after injection of 20 μg/kg Dex, treated with 2L/min oxygen flow+1.5% Sevo). Haematoxylin and eosin (HE) staining and Nissl’s staining were adopted to detect morphological
and functional changes in hippocampus of rats. Apoptosis was detected by immunofluorescence, BDNF expression was detected by immunohistochemistry. Reverse transcription PCR (RT-PCR) was conducted to detect mRNA expression of key proteins in downstream signaling of BDNF. The results showed
that Sevo induced apoptosis of hippocampus neurons, while Dex improved Sevo induced apoptosis. In contrast to the control, the positive expression of BDNF in hippocampus of Sevo group was notably decreased (P < 0.05), and that of Dex+Sevo group was notably higher in contrast to Sevo
group (P < 0.05). Signaling pathways of MAPK, PI3K-Akt, and Ras were predicted by String software as the downstream pathways of BDNF. RT-PCR results showed that these 3 signaling pathways were involved in Dex improving Sevo-induced cognitive impairment and hippocampal neuron apoptosis.
In conclusion, Dex could improve cognitive dysfunction and hippocampal neuron apoptosis in rats induced by Sevo, and the mechanism was related to upregulation of BDNF expression and activation of pathways of MAPK, PI3K-Akt, and Ras.
Collapse
Affiliation(s)
- Li Chen
- Department of Anesthesiology, Dalian Youyi Hospital, Dalian, 116001, China
| | - Tao Tang
- Department of Anesthesiology, Dalian Youyi Hospital, Dalian, 116001, China
| | - Xin Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Ying Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| |
Collapse
|
14
|
Cai NN, Geng Q, Jiang Y, Zhu WQ, Yang R, Zhang BY, Xiao YF, Tang B, Zhang XM. Schisandrin A and B affect the proliferation and differentiation of neural stem cells. J Chem Neuroanat 2021; 119:102058. [PMID: 34896558 DOI: 10.1016/j.jchemneu.2021.102058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 10/31/2021] [Accepted: 12/06/2021] [Indexed: 01/20/2023]
Abstract
Schisandrin A and B (Sch A and B) are the important components of Asian dietary supplement and phytomedicine Schisandra chinensis (S. chinensis). They can enhance adult neurogenesis in vivo; however, these effects still need to be verified. Here NE-4 C neural stem cells (NSCs) were employed as the in vitro model and treated with Sch A and B at 0.1 μg/mL. EdU (5-Ethynyl-2'-deoxyuridine) labeling showed that both Sch A and B treatments enhanced NSC proliferation. Real-time PCR analysis showed the mRNA abundances of telomerase gene Tert and cell cycle gene Cyclin D1 were significantly up-regulated after the treatments. During the neurosphere induction, Sch B enhanced the neurosphere formation and neuronal differentiation, and increased the neurosphere semidiameters. Detection of the neuron differentiation marker Mapt indicates that both Sch A and B, especially Sch B, benefits the induced neuronal differentiation. Sch B treatment also enhanced mRNA expressions of the neurosphere-specific adhesion molecule Cdh2 and Wnt pathway-related genes including Mmp9, Cyclin D1 and β-catenin. Together, Sch A especially Sch B, promotes the proliferation, affects the survival, differentiation and neurogenesis of NSCs, which is consistent with their in vivo effects. This study provides further clue on the potential neuropharmacological effects of S. chinensis.
Collapse
Affiliation(s)
- Ning-Ning Cai
- College of Veterinary Medicine, Jilin University, Changchun 130062, China; Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, China.
| | - Qi Geng
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Jiang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wen-Qian Zhu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rui Yang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bo-Yang Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu-Feng Xiao
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xue-Ming Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
15
|
Zhao Z, Gao B, Zong X, Gao R. Sevoflurane impedes glioma progression via regulating circ_0000215/miR-1200/NCR3LG1 axis. Metab Brain Dis 2021; 36:2003-2014. [PMID: 34460046 DOI: 10.1007/s11011-021-00817-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022]
Abstract
Sevoflurane has been reported to have anti-tumorigenic effects in glioma. Circ_0000215 was found to play vital functions in the pathological progressions of glioma. However, whether circ_0000215 mediates the inhibitory effects of sevoflurane on glioma cells remains unclear. In vitro assays were performed using cell counting kit-8, flow cytometry, transwell and Western blot assays. The expression levels of circ_0000215, microRNA (miR)-1200 and NCR3LG1 (Natural Killer Cell Cytotoxicity Receptor 3 Ligand 1) were detected using quantitative real-time polymerase chain reaction (qRT-PCR) and/or Western blot. The dual-luciferase reporter assay and pull-down assay were used to investigate the relationship between miR-1200 and circ_0000215 or NCR3LG1. In vivo assay was conducted using xenograft nude mice model. In vitro assays suggested that sevoflurane repressed glioma cell proliferation, metastasis and induced apoptosis as well as hindered tumor growth in vivo, which were reversed by circ_0000215 overexpression. Mechanically, circ_0000215 was confirmed to directly target miR-1200, and NCR3LG1 was a target of miR-1200 in glioma cells. Importantly, circ_0000215 could regulate NCR3LG1 expression via miR-1200. Besides that, rescue assay suggested that circ_0000215 attenuated the inhibitory effects of sevoflurane on glioma cell growth and metastasis, which were reversed by miR-1200 overexpression or NCR3LG1 knockdown. Our study revealed that sevoflurane could suppress glioma tumorigenesis by regulating circ_0000215/miR-1200/NCR3LG1 axis, suggesting a new insight into the therapeutic potential of sevoflurane in glioma treatment.
Collapse
Affiliation(s)
- Zhitao Zhao
- Department of Anesthesiology, Shandong Provincial Third Hospital, Jinan, Shandong, 250031, People's Republic of China
| | - Baofeng Gao
- Department of Anesthesiology, Shandong Provincial Third Hospital, Jinan, Shandong, 250031, People's Republic of China
| | - Xiaoling Zong
- Department of Anesthesiology, Zibo Central Hospital, No.96, South Shanghai Road, Zhangdian District, Zibo City, 255000, Shandong Province, People's Republic of China
| | - Ruiming Gao
- Department of Anesthesiology, Zibo Central Hospital, No.96, South Shanghai Road, Zhangdian District, Zibo City, 255000, Shandong Province, People's Republic of China.
| |
Collapse
|
16
|
The Osteogenic Differentiation of Human Dental Pulp Stem Cells through G0/G1 Arrest and the p-ERK/Runx-2 Pathway by Sonic Vibration. Int J Mol Sci 2021; 22:ijms221810167. [PMID: 34576330 PMCID: PMC8471578 DOI: 10.3390/ijms221810167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mechanical/physical stimulations modulate tissue metabolism, and this process involves multiple cellular mechanisms, including the secretion of growth factors and the activation of mechano-physically sensitive kinases. Cells and tissue can be modulated through specific vibration-induced changes in cell activity, which depend on the vibration frequency and occur via differential gene expression. However, there are few reports about the effects of medium-magnitude (1.12 g) sonic vibration on the osteogenic differentiation of human dental pulp stem cells (HDPSCs). In this study, we investigated whether medium-magnitude (1.12 g) sonic vibration with a frequency of 30, 45, or 100 Hz could affect the osteogenic differentiation of HDPSCs. Their cell morphology changed to a cuboidal shape at 45 Hz and 100 Hz, but the cells in the other groups were elongated. FACS analysis showed decreased CD 73, CD 90, and CD 105 expression at 45 Hz and 100 Hz. Additionally, the proportions of cells in the G0/G1 phase in the control, 30 Hz, 45 Hz, and 100 Hz groups after vibration were 60.7%, 65.9%, 68.3%, and 66.7%, respectively. The mRNA levels of osteogenic-specific markers, including osteonectin, osteocalcin, BMP-2, ALP, and Runx-2, increased at 45 and 100 Hz, and the ALP and calcium content was elevated in the vibration groups compared with those in the control. Additionally, the western blotting results showed that p-ERK, BSP, osteoprotegerin, and osteonectin proteins were upregulated at 45 Hz compared with the other groups. The vibration groups showed higher ALP and calcium content than the control. Vibration, especially at 100 Hz, increased the number of calcified nodes relative to the control group, as evidenced by von Kossa staining. Immunohistochemical staining demonstrated that type I and III collagen, osteonectin, and osteopontin were upregulated at 45 Hz and 100 Hz. These results suggest that medium magnitude vibration at 45 Hz induces the G0/G1 arrest of HDPSCs through the p-ERK/Runx-2 pathway and can serve as a potent stimulator of differentiation and extracellular matrix production.
Collapse
|
17
|
Gao W, Bai Y, Ma X, Bian X, Xu J, Xue N, Yu T, Liu X, Bai Y, Chu D. Long-term sevoflurane exposure reduces the differentiation potential and hypoxia tolerance potential of neural stem cells. Int J Dev Neurosci 2021; 81:731-740. [PMID: 34532883 DOI: 10.1002/jdn.10150] [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: 06/05/2021] [Revised: 07/21/2021] [Accepted: 09/04/2021] [Indexed: 11/06/2022] Open
Abstract
PURPOSE To investigate the effect of prolonged sevoflurane (SEV) exposure on differentiation potential and hypoxia tolerance of neural stem cells (NSCs). MATERIALS AND METHODS NSCs were extracted from 15-day fetal mice. After sub-culture, SEV exposure treatment was performed. Cell cycle were detected by flow cytometry. Western blot and immunofluorescence assay were used to detect the expression and spatial distribution of Nestin, NSE, GFAP, Oct4, and SOX2; CCK-8 detected cell viability. Cell growth morphology was observed under a microscope. TUNEL detected cell apoptosis; the concentration of extracel-lular lactate dehydrogenase (LDH) was determined by ELISA. RESULTS Compared with the control group, the proportion of NSCs in the G2/M phase increased in the SEV exposure group; our results also suggested the sphere-formation rate decreased significantly, increased apoptosis and decreased cell viability. Besides, the level of LDH release increased. CONCLUSION Long-term exposure to SEV (>8 h) promoted the premature differentiation of NSCs and reduced their pluripotency, reserves, and hypoxia tolerance. This study reveals the reasons underlying damage to the nervous system of young children induced by long-term exposure to SEV from the perspective of CNS reserve cells.
Collapse
Affiliation(s)
- Wenbo Gao
- North China University of Science and Technology, Tangshan, China.,Department of Anesthesiology, Tangshan Maternity and Child Health Care Hospital, Tangshan, China
| | - Yunxiao Bai
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Xiaofang Ma
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China.,Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China.,Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Jingman Xu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Na Xue
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China.,Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Tian Yu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China.,Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China.,Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Yaowu Bai
- Department of Anesthesiology, Tangshan Maternity and Child Health Care Hospital, Tangshan, China
| | - Dongmei Chu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, The Fifth Central Hospital of Tianjin, Tianjin, China
| |
Collapse
|
18
|
Jiang M, Tang T, Liang X, Li J, Qiu Y, Liu S, Bian S, Xie Y, Fang F, Cang J. Maternal sevoflurane exposure induces temporary defects in interkinetic nuclear migration of radial glial progenitors in the fetal cerebral cortex through the Notch signalling pathway. Cell Prolif 2021; 54:e13042. [PMID: 33955094 PMCID: PMC8168415 DOI: 10.1111/cpr.13042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/02/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The effects of general anaesthetics on fetal brain development remain elusive. Radial glial progenitors (RGPs) generate the majority of neurons in developing brains. Here, we evaluated the acute alterations in RGPs after maternal sevoflurane exposure. METHODS Pregnant mice were exposed to 2.5% sevoflurane for 6 hours on gestational day 14.5. Interkinetic nuclear migration (INM) of RGPs in the ventricular zone (VZ) of the fetal brain was evaluated by thymidine analogues labelling. Cell fate of RGP progeny was determined by immunostaining using various neural markers. The Morris water maze (MWM) was used to assess the neurocognitive behaviours of the offspring. RNA sequencing (RNA-Seq) was performed for the potential mechanism, and the potential mechanism validated by quantitative real-time PCR (qPCR), Western blot and rescue experiments. Furthermore, INM was examined in human embryonic stem cell (hESC)-derived 3D cerebral organoids. RESULTS Maternal sevoflurane exposure induced temporary abnormities in INM, and disturbed the cell cycle progression of RGPs in both rodents and cerebral organoids without cell fate alternation. RNA-Seq analysis, qPCR and Western blot showed that the Notch signalling pathway was a potential downstream target. Reactivation of Notch by Jag1 and NICD overexpression rescued the defects in INM. Young adult offspring showed no obvious cognitive impairments in MWM. CONCLUSIONS Maternal sevoflurane exposure during neurogenic period temporarily induced abnormal INM of RGPs by targeting the Notch signalling pathway without inducing long-term effects on RGP progeny cell fate or offspring cognitive behaviours. More importantly, the defects of INM in hESC-derived cerebral organoids provide a novel insight into the effects of general anaesthesia on human brain development.
Collapse
Affiliation(s)
- Ming Jiang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Tianxiang Tang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceFudan UniversityShanghaiChina
| | - Xinyue Liang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Juchen Li
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Yue Qiu
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiwen Liu
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Shan Bian
- Institute for Regenerative MedicineSchool of Life Sciences and TechnologyFrontier Science Center for Stem Cell ResearchShanghai East HospitalTongji UniversityShanghaiChina
| | - Yunli Xie
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain ScienceFudan UniversityShanghaiChina
| | - Fang Fang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| | - Jing Cang
- Department of AnesthesiaZhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
19
|
Zuo Y, Chang Y, Thirupathi A, Zhou C, Shi Z. Prenatal sevoflurane exposure: Effects of iron metabolic dysfunction on offspring cognition and potential mechanism. Int J Dev Neurosci 2020; 81:1-9. [PMID: 33259670 DOI: 10.1002/jdn.10080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
For decades, the neurotoxicity caused by anesthetics in mammalian brain development has gained increasing attention. Exposure to anesthetics leads to neurotoxicity and apoptosis of nerve cells, which in turn induces cognitive dysfunction. Although most of the data came from animal studies, general anesthetics have been shown to have adverse effects on cognitive function in infants and young children in recent years. This concern has led to a number of retrospective studies that observed an association between general anesthesia in pregnant women and neurobehavioral problems in fetuses or offspring. Every year, many pregnant women undergo non-obstetric anesthesia due to various reasons such as traffic accidents, fetal interventions, acute appendicitis, symptomatic cholelithiasis, and trauma. A matter of concern for these pregnant women is whether anesthesia has a detrimental effect on fetal brain development in the womb and whether the fetus has cognitive impairment after birth. In humans, the association of anesthetic exposure in infants with the long-term impairment of neurologic functions has been reported in several retrospective clinical studies. Recently, we have found that sevoflurane anesthesia during pregnancy in mice-induced cognitive impairment in the offspring by causing iron deficiency and inhibiting myelinogenesis. Sevoflurane is a commonly used general anesthetic in the hospitals, which can induce neurotoxicity and cause cognitive impairment in fetuses, infants, children, and adults. However, the exact mechanism of sevoflurane-induced damage to the central nervous system (CNS) is not fully understood. Based on our recent results, this paper reviewed the effects of sevoflurane on cognitive impairment and pathological changes such as neurogenesis, neuronal apoptosis, and iron metabolism dysfunction in the offspring.
Collapse
Affiliation(s)
- Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Yanzhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | | | - Changhao Zhou
- The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, China
| |
Collapse
|
20
|
Zhang Y, Wu Z, Li X, Wan Y, Zhang Y, Zhao P. Maternal sevoflurane exposure affects differentiation of hippocampal neural stem cells by regulating miR-410-3p and ATN1. Stem Cell Res Ther 2020; 11:423. [PMID: 32993796 PMCID: PMC7523391 DOI: 10.1186/s13287-020-01936-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/28/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Currently, numerous animal studies have shown that exposure to commonly used general anesthetics during pregnancy may cause neurocognitive impairment in the offspring. Reportedly, exposure to sevoflurane during mid-trimester of pregnancy can inhibit proliferation of neural stem cells (NSCs) and lead to early apoptosis. Whether exposure to sevoflurane during pregnancy affects the differentiation of NSCs remains unclear. METHODS In the present study, pregnant rats were exposed to 3% sevoflurane once for 2 h on gestational day 14 (G14) or 3 times for 2 h on G13, G14, and G15. Next, the differentiation of NSCs was measured using neuron marker β-tubulin III and astrocyte marker glial fibrillary acidic protein (GFAP) in fetal brain tissues 24 h and 72 h after anesthesia and in hippocampus on postnatal day 28. Primary cultured rat NSCs were exposed to 4.1% sevoflurane to explore the mechanism. RESULTS The results showed that during mid-trimester, multiple exposures to sevoflurane can cause premature differentiation of NSCs in developing brains of offspring and lead to long-term neuron reduction and astrocyte proliferation in hippocampus. The data from the present study indicated that repeated exposure to sevoflurane downregulated atrophin-1 (ATN1) expression and caused early differentiation of NSCs. Overexpression of ATN1 via lentivirus transfection attenuated the influence of sevoflurane. Using dual luciferase assay, ATN1 was found to be a target gene of microRNA-410-3p (miR-410-3p). MiR-410-3p suppression via lentivirus transfection recovered the ATN1 expression and differentiation of NSCs. CONCLUSIONS The results from the present study demonstrated that repeated exposure to sevoflurane leads to early differentiation of NSCs and long-term effects via the miR-410-3p/ATN1 pathway.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xingyue Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuxiao Wan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yinong Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Gao J, Liao Y, Qiu M, Shen W. Wnt/β-Catenin Signaling in Neural Stem Cell Homeostasis and Neurological Diseases. Neuroscientist 2020; 27:58-72. [PMID: 32242761 DOI: 10.1177/1073858420914509] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neural stem/progenitor cells (NSCs) maintain the ability of self-renewal and differentiation and compose the complex nervous system. Wnt signaling is thought to control the balance of NSC proliferation and differentiation via the transcriptional coactivator β-catenin during brain development and adult tissue homeostasis. Disruption of Wnt signaling may result in developmental defects and neurological diseases. Here, we summarize recent findings of the roles of Wnt/β-catenin signaling components in NSC homeostasis for the regulation of functional brain circuits. We also suggest that the potential role of Wnt/β-catenin signaling might lead to new therapeutic strategies for neurological diseases, including, but not limited to, spinal cord injury, Alzheimer's disease, Parkinson's disease, and depression.
Collapse
Affiliation(s)
- Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Liao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
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]
|
23
|
Yu D, Zhu Y, Cui C, Long R, Ma J. Midazolam prevents sevoflurane-induced death in hippocampal neurons. Tissue Cell 2019; 58:1-7. [DOI: 10.1016/j.tice.2019.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/14/2019] [Accepted: 03/16/2019] [Indexed: 02/08/2023]
|
24
|
Zhang L, Yan J, Liu Q, Xie Z, Jiang H. LncRNA Rik-203 contributes to anesthesia neurotoxicity via microRNA-101a-3p and GSK-3β-mediated neural differentiation. Sci Rep 2019; 9:6822. [PMID: 31048708 PMCID: PMC6497879 DOI: 10.1038/s41598-019-42991-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/09/2019] [Indexed: 12/30/2022] Open
Abstract
The mechanism of anesthesia neurotoxicity remains largely to be determined. The effects of long noncoding RNAs (LncRNAs) on neural differentiation and the underlying mechanisms are unknown. We thus identified LncRNA Rik-203 (C130071C03Rik) and studied its role on neural differentiation and its interactions with anesthetic sevoflurane, miRNA and GSK-3β. We found that levels of Rik-203 were higher in hippocampus than other tissues and increased during neural differentiation. Sevoflurane decreased the levels of Rik-203. Rik-203 knockdown reduced mRNA levels of Sox1 and Nestin, the markers of neural progenitor cells, and decreased the count of Sox1 positive cells. RNA-RNA pull-down showed that miR-101a-3p was highly bound to Rik-203. Finally, sevoflurane, knockdown of Rik-203, and miR-101a-3p overexpression all decreased GSK-3β levels. These data suggest that Rik-203 facilitates neural differentiation by inhibiting miR-101a-3p's ability to reduce GSK-3β levels and that LncRNAs would serve as the mechanism of the anesthesia neurotoxicity.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, P.R. China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, P.R. China
| | - Qidong Liu
- Shanghai Tenth People's Hospital, Anesthesia and Brain Research Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Room, 4310, Charlestown, MA, USA.
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, P.R. China.
| |
Collapse
|
25
|
Autophagy promotes hepatic differentiation of hepatic progenitor cells by regulating the Wnt/β-catenin signaling pathway. J Mol Histol 2019; 50:75-90. [PMID: 30604254 PMCID: PMC6323068 DOI: 10.1007/s10735-018-9808-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
Hepatic progenitor cells (HPCs) can be activated when the liver suffers persistent and severe damage and can differentiate into hepatocytes to maintain liver regeneration and homeostasis. However, the molecular mechanism underlying the hepatic differentiation of HPCs is unclear. Therefore, in this study, we aimed to investigate the roles of autophagy and the Wnt/β-catenin signaling pathway during hepatic differentiation of HPCs in vivo and in vitro. First, immunohistochemistry, immunofluorescence and electron microscopy showed that Atg5 and β-catenin were highly expressed in human fibrotic liver and mouse liver injury induced by feeding a 50% choline-deficient diet plus 0.15% ethionine solution in drinking water (CDE diet) for 21 days; in addition, these factors were expressed in CK19-positive HPCs. Second, Western blotting and immunofluorescence confirmed that CK19-positive HPCs incubated in differentiation medium for 7 days can differentiate into hepatocytes and that differentiated HPCs were able to take up ICG and secrete albumin and urea. Further investigation via Western blotting, immunofluorescence and electron microscopy revealed autophagy and the Wnt/β-catenin pathway to be activated during hepatic differentiation of HPCs. Next, we found that inhibiting autophagy by downregulating Atg5 gene expression impaired hepatic differentiation of HPCs and inhibited activation of the Wnt/β-catenin pathway, which was rescued by overexpression of the β-catenin gene. Moreover, downregulating β-catenin gene expression without inhibiting autophagy still impeded the differentiation of HPCs. Finally, coimmunoprecipitation demonstrated that P62 forms a complex with phosphorylated glycogen synthase kinase 3 beta (pGSK3β). Third, in mouse CDE-induced liver injury, immunohistochemistry and immunofluorescence confirmed that downregulating Atg5 gene expression inhibited autophagy, thus impeding hepatic differentiation of HPCs and inhibiting activation of the Wnt/β-catenin pathway. As observed in vitro, overexpression of β-catenin rescued this phenomenon caused by autophagy inhibition, though decreasing β-catenin levels without autophagy inhibition still impeded HPC differentiation. We also found that HPCs differentiated into hepatocytes in human fibrotic liver tissue. Collectively, these results demonstrate that autophagy promotes HPC differentiation by regulating Wnt/β-catenin signaling. Our results are the first to identify a role for autophagy in promoting the hepatic differentiation of HPCs.
Collapse
|